Abstract
Members of the small heat shock protein (sHSP) family are molecular chaperones with a critical role in the maintenance of cellular homeostasis under unfavorable conditions. The chaperone properties of sHSPs prevent protein aggregation, and sHSP deregulation underlies the pathology of several diseases, including neurodegenerative disorders. Recent evidence suggests that the clientele of sHSPs is broad, and the mechanisms of sHSP-mediated neuroprotection diverse. Nonetheless, the crosstalk of sHSPs with the neurodegeneration-promoting signaling pathways remains poorly understood. Here, we survey recent findings on the role and regulation of sHSPs in neurodegenerative diseases.
Introduction
Organisms and cells are constantly exposed to various types of stress, such as environmental, metabolic or pathophysiological stress. These disturbances result in loss of integrity of the proteome, cellular dysfunction and cell death. In the course of evolution, elaborate molecular mechanisms were invented to counteract the detrimental effect of extrinsic and intrinsic stress factors on protein function, and preserve protein homeostasis (proteostasis). An evolutionary conserved cytoprotective mechanism is the heat shock response pathway [1, 2]. Prominent effectors of the heat shock response pathway are molecular chaperones, which comprise of several families categorized according to their molecular weight: HSP10, HSP40, HSP60, HSP70, HSP90, HSP100 and sHSP. Chaperones are required for the maintenance of the native structure of cellular proteins, protein translocation, assembly of functional protein complexes and protein degradation. sHSPs are ATP-independent molecular chaperones characterized by a small molecular mass ranging from 12 to 42 kDa [3] and a highly conserved domain, called the alpha-crystallin domain [4]. sHSPs display a dynamic behavior, and can exist in the form of monomers, dimers and large multimeric complexes, exhibiting variability in subunit numbers (12 to >48) [5,6,7].
sHSPs have a wide clientele and sHSP dysfunction results in a broad range of pathologies, including ischemia, myopathies, motor neuron disease, diabetes and cataracts [8,9,10,11]. The main function of sHSPs is to bind to hydrophobic regions of aggregation-prone misfolded proteins and prevent the formation of insoluble aggregates [12]. However, association with sHSPs does not lead to substrate refolding to the native state [13,14,15]. Therefore, the sHSP/substrate complexes act as intermediates that are further processed by the HSP70 and HSP90 chaperones [16,17,18]. Here, we highlight recent findings regarding the interplay between sHSP function and neurodegeneration.
The role of sHSPs in neurodegenerative pathologies
Many devastating neurological disorders, such as Alzheimer’s, Parkinson’s, Creutzfeldt Jacobs and amyotrophic lateral sclerosis (ALS), are characterized by aggregation and precipitation of misfolded proteins. sHSPs are key players of the proteostasis network, and their ability to interact with aggregation-prone proteins highlights the therapeutic potential of these heat shock proteins [19]. The deposition of fibrillar α-Synuclein (α-syn) into inclusion bodies in the neuronal cell body or processes is a pathological hallmark of neurodegenerative disorders, collectively known as synucleinopathies [20]. The expression of sHSPs is significantly increased in the presence of cellular stress [21], and sHSPs have been colocalized with α-syn in inclusion bodies [22, 23]. The capacity of sHSPs to prevent α-syn aggregation depends on the kinetics of the aggregation process. Factors that increase the rate of aggregation, such as disease-related mutations, gene amplification, or macromolecular crowding, may alter the kinetics of α-syn aggregation in cells, thus overwhelming the protective capacity of sHSPs due to reduced availability of aggregation-combating chaperone subunits [24, 25]. HSPB1 (known also as HSP27) binds along the surface of α-syn fibrils and inhibits their growth by blocking elongation [26]. In addition to HSPB1, HSPB5 (αB-crystallin) also reduces the aggregation of α-syn in in vitro assays [27]. Another sHSP, HSPB8 (also known as HSP22) participates in the clearance of several misfolded proteins implicated in neurodegenerative diseases. HSPB8 acts in cooperation with the co-chaperone BAG3 and promotes the clearance of protein aggregates through upregulation of autophagy [28]. These findings show that sHSPs link the different nodes of the proteostasis machinery to counteract the accumulation of misfolded proteins.
A protective role of HSPB5 has been described in Alzheimer’s disease, characterized by Aβ amyloid fibrils in extracellular plaques [29]. HSPB5 binds and inhibits the elongation of amyloid fibrils [30]. Interestingly, HSPB8 is upregulated specifically in astrocytes of cerebral areas undergoing neurodegeneration [31], suggesting that astrocytic proteostasis is critical for the clearance of aggregates in the neuronal microenvironment. HSPB4 (αA-crystallin) and HSPB5 knockout mouse models are viable and have tissue specific developmental roles [32, 33]. Another sHSP, HSPB3, found in the central and peripheral nervous system, protects against motor neuron cell death [34]. Recent findings suggest that different members of the proteostasis machinery of the cell may prevent aggregation of proteins through distinct mechanisms. HSPB1 delayed tau (a microtubule-associated protein forming aggregates in various dementias) fibril elongation, by weakly interacting with early species during the aggregation process [35]. Interestingly, the phosphorylation dynamics of HSPB1 appears to be critical for the ability of this sHSP to reduce neuronal tau levels [36].
Alexander disease (AxD) is a rare disorder of astrocytes caused by mutations in the gene encoding for glial fibrillary acidic protein (GFAP). In AxD mouse models, overexpression of HSPB5 has a protective effect [37] and restores the defective proteasomal degradation of mutant GFAP [38], suggesting that this sHSP may serve as a therapeutic target in astropathology. In spinal and bulbar muscular atrophy, a disease characterized by abnormally long polyglutamine tract (polyQ) in mutant androgen receptor (ARpolyQ), HSPB8 promotes motorneuron survival of patients by restoring autophagic flux and removing misfolded aggregates of ARpolyQ [39].
Charcot-Marie-Tooth (CMT) is the most common inherited peripheral neuropathy. Missense mutations in HSPB8 and HSPB1 have been implicated in CMT pathogenesis [40,41,42]. HSPB8 knockout mice are viable and display normal locomotor performances [43]. The list of mutations affecting the function of these sHSPs continues to grow [44, 45]. Transgenic mice expressing mutant HSPB1 developed features of CMT [46, 47] and displayed decreased α-tubulin acetylation and defective axonal transport of mitochondria [46]. Intriguingly, pharmacological inhibition of histone deacetylase 6 (HDAC6) ameliorated the axonal transport defects caused by mutant HSPB1, underlying the importance of identifying downstream targets of sHSPs. In addition, recent knock-in mouse models expressing mutant HSPB8, recapitulated the CMT patient pathology and displayed reduced autophagy and HSPB8 aggregates [43].
In an animal model of experimental autoimmune encephalomyelitis (EAE), genetic ablation of HSPB5 dramatically increased animal clinical paralysis and induced inflammatory responses [48]. The inflammation was accompanied by an elevated temperature, and therefore heat shock proteins serve as ideal effectors in inflamed tissue. Interestingly, exogenous administration of HSPB5 suppressed inflammation in animals. HSPB5 is a potent modulator of inflammation and sequesters, in a temperature-dependent manner, a variety of pro-inflammatory proteins, therefore affecting both innate and adaptive immune responses [48].
In a systematic approach for the identification of sHSPs with a potential role in cerebral ischemia in vivo, investigators observed upregulation of HSPB1 and HSPB5 in neurons in the infarcted cortex [49]. Phosphorylation of those sHSPs is critical for their neuroprotective role during cerebral ischemia [49, 50]. In addition, HSPB1 maintains the integrity of the blood-brain barrier during ischemia/reperfusion injury in experimental stroke, suggesting a potential therapeutic role of HSPB1 in injuries characterized by breach in brain vasculature [51].
sHSPs in the presence of external stress
In addition to neurodegeneration caused by detrimental protein aggregation, our research uncovered a protective role of sHSPs against necrotic cell death and neurodegeneration triggered by hyperthermia [52, 53]. We found that during the preconditioning of animals at mildly elevated temperatures, heat shock transcription factor 1 (HSF-1) upregulated the expression of HSP-16.1, which protects neurons against subsequent heat-induced damage. HSP-16.1 exerts its protective role through the Ca2+/Mn2+ ATPase PMR-1 in the Golgi. Importantly, this protective mechanism is evolutionary conserved and protects murine neurons against heat stroke-inflicted neurodegeneration [52]. These data suggest that organelle-specific Ca2+ channels and pumps might be important targets of sHSPs, and the protection of the function of these proteins under stress conditions preserves organelle, and consequently cellular ionstasis [53]. In a Drosophila model, external heat stress disrupted proteostasis and led to degeneration of muscle, motor neurons and associated glia. Intriguingly, muscle-specific overexpression of the sHSP HSP23 also had a protective effect on neurons and glia, suggesting the presence of a sophisticated intercellular proteostasis network [54]. Extracellular HSPB4 and HSPB5 protected astrocytes from various harmful insults, such as staurosporine and serum-starvation, through activation of pro-survival signaling pathways [55]. Finally, in the presence of external stress, overexpression of HSPB5 preserved the dendritic architecture in rat hippocampal neurons [56].
The multifaceted action of sHSPs
Inhibition of the apoptotic machinery is a key mechanism accounting for the cytoprotective effect of sHSPs. sHSPs participate in both intrinsic and extrinsic apoptotic signaling pathways. HSPB1 abolishes the release of cytochrome c from mitochondria [57], and protects against apoptosis through AKT activation and subsequent Bax inhibition [58]. HSPB1 knockout mice are viable and fertile [59, 60]. Interestingly, HSPB1 binds to cytochrome c released from mitochondria and prevents apoptosome formation [61]. In addition, HSPB6 and HSPB8 suppress apoptosis by inhibiting cytochrome c release from mitochondria [62]. In the extrinsic pathway, HSPB2 suppresses the activation of caspases-8 and 10 therefore blocking downstream apoptotic events, such as caspase-3 activation [63]. Also, HSPB4/5 have anti-apoptotic effects against Bax-induced apoptosis by sequestering Bax in the cytoplasm and preventing its translocation to the mitochondria [64]. sHSPs are critical for the survival of adult mammalian neurons. The fate determinant transcription factor Pax6 maintains homeostasis by directly regulating the expression of HSPB4, which inhibits the activation of procaspase-3 and apoptosis [65]. Therefore, sHSPs exert their anti-apoptotic role through blocking critical nodes in the cell death pathway, such as the release of cytochrome c from mitochondria and the activation of caspases.
Several sHSPs show organelle specificity [66,67,68,69]. Phosphorylation of HSPB5 results in localization at SC35 speckles, a nuclear compartment involved in the storage and recycling of splicing factors [66]. Further investigation will show whether this sHSP has a role in speckle stability and regulation of gene expression. HSPB8 was found to localize to stress granules, membrane-less compartments that form upon proteotoxic stress and sequester ribonucleoprotein complexes. Importantly, the disassembly of stress granules depends on the concerted action of the HSPB8-BAG3-HSP70 complex, suggesting that sHSPs are critical mediators of protein quality surveillance mechanisms of the cell [70, 71]. These findings may have clinical significance given the implication of stress granules in ALS pathology. Several sHSPs exert their function in mitochondria, possibly through preservation of membrane integrity. HSP25 (the murine homologue of HSPB1) localizes to the mitochondria and protects PC12 cells (rat pheochromocytoma) against 6-Hydroxydopamine (6-OHDA), a neurotoxin that targets dopaminergic neurons [72]. Better understanding of the organelle-specific roles of sHSPs will uncover the molecular mechanisms deregulated in pathological conditions caused by sHSPs malfunction.
In addition to binding to proteins and preventing their denaturation [73], sHSPs are also active in the extracellular matrix and alter gene expression through membrane receptor signaling [74]. sHSPs extracellular concentration increases in several neurodegenerative conditions [75]. Serum levels of HSPB1 have been shown to correlate with acute attack phases in patients with multiple sclerosis [76]. In contrast to secreted proteins that typically exit cells via the endoplasmic reticulum-Golgi network, HSPs lack an N-terminal signal peptide that marks the protein for secretion. In addition to sHSP accumulation into circulation in response to either cell damage or cellular stress, another mechanism for the release of HSPB1 to the extracellular space is through exosome secretion [77]. Specifically, HSPB1 was detected in membrane fractions of exosomes released from rat astrocytes upon treatment with amyloid-b. The precise localization of sHSPs in exosomes is still under investigation and depends on the type of sHSP, the cells from which the exosome is released and the stimuli that induce the release. Extracellular application of HSPB4/5 activated the PI3K/Akt/mTOR signaling pathway and promoted survival of olfactory ensheathing cells [78]. HSPB5 was also secreted via exosomes by highly polarized human retinal pigment epithelial cells (RPE). HSPB5 uptake from apical photoreceptors facing neural retina conferred neuroprotection in the presence of oxidative stress [79]. In addition to the release of HSPB5 via exosomes, a recent study demonstrated that HSPB5 is also critical for exosome biogenesis [80]. Administration of HSPB4 peptides inhibited protein insolubilization and opacification in selenite-induced cataract in mice [81]. Similarly, HSPB1 and HSPB6 peptides inhibited protein aggregation and oxidative stress, and prevented selenite-induced cataract formation in rats [82]. Extracellular HSPB5 neutralized misfolded oligomers and protected cells by shielding the reactive surfaces [83]. The mechanisms by which extracellular sHSPs exert beneficial functions need further investigation and may be stress-specific.
Inflammation underlies the pathology of many neurodegenerative diseases [8]. Prolonged inflammation results in cytotoxicity in the nervous system. sHSPs have the ability to control neuroinflammation through activation of macrophages [84]. Moreover, HSPB4/B5 prevent the accumulation of proinflammatory mediators and suppress ROS and NO production [48, 73, 85]. Also, in a mouse model of acute spinal cord injury, administration of human recombinant HSPB5 improved locomotor skills and ameliorated secondary tissue damage. Mechanistically, HSPB5 modulated an inflammatory response in the injured spinal cord, promoted granulocytes infiltration and reduced accumulation of inflammatory macrophages [86]. The therapeutic potential of HSPB5 is also evident by the increased survival of retinal ganglion cells and the inhibition of retinal microglial cells in the rat model of optic nerve crush [87]. Importantly, HSPB5 administration inhibits the expression of TNFa and iNOS in this model. Treatment of an EAE mouse model with HSPB1 reduced the paralytic symptoms through modulation of inflammatory cytokines [88]. Pro-inflammatory cytokines induce the formation of ROS and NO, and subsequently downregulate the levels of HSPB1 and promote apoptosis of retinal capillary endothelial cells [89]. HSPB8, HSP20 (HSPB6) and HSPB2 are associated with cerebral amyloid angiopathy (CAA) in hereditary cerebral hemorrhage with amyloidosis of the Dutch type (HCHWA-D). Importantly, these sHSPs induce interleukin-6 production in cultured astrocytes and pericytes, suggesting that sHSPs may be critical mediators of local inflammatory response in HCHWA-D [90]. In Alzheimer’s disease, the majority of patients are characterized by some degree of CAA, and HSP20, HSPB8 and HSPB2B3 colocalize with CAA and induce production of interleukin-8, intercellular adhesion molecule 1 (ICAM-1) and monocyte chemoattractant protein by human brain astrocytes, reinforcing the role of sHSPs in neuroinflammation in Alzheimer’s disease [91]. These findings, in their totality, suggest that exogenous administration of sHSPs has a protective effect in many diseases involving cell death, inflammation and protein aggregation.
The transcriptional regulation of sHSPs
In addition to a plethora of post-translational modifications, including phosphorylation, isomerization, deamidation and glycation [92, 93], sHSPs also undergo transcriptional regulation. Although the transcriptional regulation of sHSPs during development of various tissues is well described [92], understanding of the gene expression regulation in disease is lacking. Historically, the transcription of heat shock proteins, including sHSPs, has been attributed to the heat shock transcription factors (HSFs) [1]. However, the crosstalk of HSFs with disease-promoting signaling pathways and other transcription factors is poorly understood. Recent findings have implicated signaling pathways, with a critical role in disease, in the regulation of HSF1 [2, 94, 95]. In neurons, the HSF1 amino acids Ser303 and Ser307, two critical residues for the stability of HSF1 [94], are phosphorylated by the casein kinase 2 (CK2). These phosphorylation events link HSF1 to the E3 ligase FBXW7 which ubiquitylates HSF1, resulting in proteasomal-mediated degradation [2, 96]. In Huntington’s disease, the CK2α¢ catalytic subunit and FBXW7 are elevated. Subsequently, phosphorylated HSF1 interacts with FBXW7 and is degraded in the proteasome leading to protein aggregation, neuronal dysfunction and death [96]. Future experiments will reveal whether exogenous administration of sHSPs is sufficient to prevent aggregate formation in this model and in neurodegenerative disorders characterized by HSF1 reduced protein stability. Interestingly, decreased activity of HSF1 has been reported in several neurodegenerative disorders, including Parkinson’s, Alzheimer’s disease and AES.
In a model of Huntington’s disease, HSF1 and nuclear factor of activated T cells (NFAT) cooperatively regulate the expression of HSPB5 [97]. Also, HSF2 regulates proteostasis in a mouse modeling Huntington’s disease, partially through cooperative interaction with HSF1 upon the regulation of expression of HSPB5 [98]. Hsf2 genetic ablation results in reduced lifespan and increased protein aggregation in the striatum of this disease mouse model [98]. The specific interactions between stress transcription factors in different neuronal cell types will uncover the expression programs activated against aggregation in neurodegenerative disorders.
Activating transcription factor 3 (ATF-3) is induced in the presence of neuronal injury and, in cooperation with c-Jun, upregulates the expression of the anti-apoptotic chaperone HSPB1 [99]. HSF4, the predominant stress factor expressed in ocular lens tissue, protects lens epithelial cells against cytotoxic drug-induced apoptosis, partially through upregulation of HSPB5 [100]. Moreover, HSF4 controls the selective expression of heat shock proteins in the lens tissue by promoting HSF1 protein degradation and selectively binding the promoters of stress response genes [101]. Mechanistically, HSF4 interacts with the ATP-dependent DEXD/H-box RNA helicase UAP56, coupling the transcriptional and post-transcriptional machinery [102].
The importance of efficient transcriptional regulation of sHSPs is exemplified by the identification of a mutation in the promoter of HSPB1 in a cohort of sporadic ALS patients. This mutation effects a conserved nucleotide in the heat shock element (HSE), the sequence recognized by HSF1, resulting in impaired promoter activity [103]. HSPB1 is upregulated in a mouse model of ALS overexpressing human mutant SOD1, suggesting an involvement of this sHSP in the ALS pathogenesis [104]. However, HSPB1 overexpression alone is not sufficient to protect against chronic motor neuron injury in an ALS mouse model overexpressing human mutant SOD1 [105].
Future perspectives
Preservation of protein homeostasis is critical for many cellular processes and underlies the pathology of diseases as diverse as cancer, neurodegeneration and cardiovascular disease. Over the past few years, findings suggest that the regulation of stress transcription factors and their effectors is cell-type and disease specific. It will be important to gain a systematic understanding of the disease signaling that converge on altered expression of effectors of the proteostasis machinery, including sHSPs (Figure 1). Moreover, the recent advances in genome-wide methods and high-resolution chromatin analysis techniques [106] will allow the investigators to uncover the mechanisms of nucleosome remodeling, chromatin states and transcription factor synergy on the regulation of sHSPs in neurodegeneration. The beneficial effects of sHSPs in neurodegeneration animal models and human clinical trials is emerging [107,108,109]. To exploit sHSPs therapeutically, it is imperative that we decipher the regulation and targets of these multifaceted molecular chaperones.

Multiple levels of regulation of small heat shock proteins in neurodegeneration. In Huntington disease mutant huntingtin protein (mHTT) increases the levels of CK2α¢ and FBXW7 leading to altered HSF1 protein levels. Future studies will reveal whether disease-specific molecular pathways or unfavorable external conditions affect the stability and function of HSF1. Also, better understanding of the cooperation of HSF1 with critical for disease transcription factors on the regulation of sHSPs transcription will uncover the stress-induced transcriptional network. In addition to transcriptional regulation, future analysis will identify the sHSPs’ post-translational modifications and how they affect sHSP functionality. Systematic functional approaches will catalogue the sHSP clientele, organelle tropism and participation in chaperone networks in disease-specific context.
Acknowledgements
We apologize to those colleagues whose work we could not reference directly owing to space limitations. N.K. is supported by a European Molecular Biology Organization (EMBO) Long Term Fellowship and a Human Frontiers Science Program (HFSP) Long Term Fellowship. NT is funded by grants from the European Research Council (ERC), the European Commission Framework Programmes, and the Greek Ministry of Education.
Author disclosure: No conflict of financial interest exists.
References
1 Akerfelt M, Morimoto RI, Sistonen L. Heat shock factors: integrators of cell stress, development and lifespan. Nat Rev Mol Cell Biol. 2010;11(8):545-55.10.1038/nrm2938Search in Google Scholar PubMed PubMed Central
2 Gomez-Pastor R, Burchfiel ET, Thiele DJ. Regulation of heat shock transcription factors and their roles in physiology and disease. Nat Rev Mol Cell Biol. 2018;19(1):4-19.10.1038/nrm.2017.73Search in Google Scholar PubMed PubMed Central
3 Haslbeck M, Vierling E. A first line of stress defense: small heat shock proteins and their function in protein homeostasis. J Mol Biol. 2015;427(7):1537-48.10.1016/j.jmb.2015.02.002Search in Google Scholar PubMed PubMed Central
4 Franck E, Madsen O, van Rheede T, Ricard G, Huynen MA, de Jong WW. Evolutionary diversity of vertebrate small heat shock proteins. J Mol Evol. 2004;59(6):792-805.10.1007/s00239-004-0013-zSearch in Google Scholar PubMed
5 Candido EP. The small heat shock proteins of the nematode Caenorhabditis elegans: structure, regulation and biology. Prog Mol Subcell Biol. 2002;28:61-78.10.1007/978-3-642-56348-5_4Search in Google Scholar PubMed
6 McDonald ET, Bortolus M, Koteiche HA, McHaourab HS. Sequence, structure, and dynamic determinants of Hsp27 (HspB1) equilibrium dissociation are encoded by the N-terminal domain. Biochemistry. 2012;51(6):1257-68.10.1021/bi2017624Search in Google Scholar PubMed PubMed Central
7 van Montfort RL, Basha E, Friedrich KL, Slingsby C, Vierling E. Crystal structure and assembly of a eukaryotic small heat shock protein. Nat Struct Biol. 2001;8(12):1025-30.10.1038/nsb722Search in Google Scholar PubMed
8 Bakthisaran R, Tangirala R, Rao Ch M. Small heat shock proteins: Role in cellular functions and pathology. Biochim Biophys Acta. 2015;1854(4):291-319.10.1016/j.bbapap.2014.12.019Search in Google Scholar PubMed
9 Kampinga HH, Garrido C. HSPBs: small proteins with big implications in human disease. Int J Biochem Cell Biol. 2012;44(10):1706-10.10.1016/j.biocel.2012.06.005Search in Google Scholar PubMed
10 Kannan R, Sreekumar PG, Hinton DR. Novel roles for alpha-crystallins in retinal function and disease. Prog Retin Eye Res. 2012;31(6):576-604.10.1016/j.preteyeres.2012.06.001Search in Google Scholar PubMed PubMed Central
11 Sun Y, MacRae TH. The small heat shock proteins and their role in human disease. FEBS J. 2005;272(11):2613-27.10.1111/j.1742-4658.2005.04708.xSearch in Google Scholar
12 Jakob U, Gaestel M, Engel K, Buchner J. Small heat shock proteins are molecular chaperones. J Biol Chem. 1993;268(3):1517-20.10.1016/S0021-9258(18)53882-5Search in Google Scholar
13 Friedrich KL, Giese KC, Buan NR, Vierling E. Interactions between small heat shock protein subunits and substrate in small heat shock protein-substrate complexes. J Biol Chem. 2004;279(2):1080-9.10.1074/jbc.M311104200Search in Google Scholar PubMed
14 Haslbeck M, Miess A, Stromer T, Walter S, Buchner J. Disassembling protein aggregates in the yeast cytosol. The cooperation of Hsp26 with Ssa1 and Hsp104. J Biol Chem. 2005;280(25):23861-8.10.1074/jbc.M502697200Search in Google Scholar PubMed
15 Mogk A, Schlieker C, Friedrich KL, Schonfeld HJ, Vierling E, Bukau B. Refolding of substrates bound to small Hsps relies on a disaggregation reaction mediated most efficiently by ClpB/DnaK. J Biol Chem. 2003;278(33):31033-42.10.1074/jbc.M303587200Search in Google Scholar PubMed
16 Nillegoda NB, Kirstein J, Szlachcic A, Berynskyy M, Stank A, Stengel F, et al. Crucial HSP70 co-chaperone complex unlocks metazoan protein disaggregation. Nature. 2015;524(7564):247-51.10.1038/nature14884Search in Google Scholar PubMed PubMed Central
17 Nillegoda NB, Bukau B. Metazoan Hsp70-based protein disaggregases: emergence and mechanisms. Front Mol Biosci. 2015;2:57.10.3389/fmolb.2015.00057Search in Google Scholar PubMed PubMed Central
18 Lee GJ, Vierling E. A small heat shock protein cooperates with heat shock protein 70 systems to reactivate a heat-denatured protein. Plant Physiol. 2000;122(1):189-98.10.1104/pp.122.1.189Search in Google Scholar PubMed PubMed Central
19 Carra S, Rusmini P, Crippa V, Giorgetti E, Boncoraglio A, Cristofani R, et al. Different anti-aggregation and pro-degradative functions of the members of the mammalian sHSP family in neurological disorders. Philos Trans R Soc Lond B Biol Sci. 2013;368(1617):20110409.10.1098/rstb.2011.0409Search in Google Scholar PubMed PubMed Central
20 Marti MJ, Tolosa E, Campdelacreu J. Clinical overview of the synucleinopathies. Mov Disord. 2003;18 Suppl 6:S21-7.10.1002/mds.10559Search in Google Scholar PubMed
21 Bartelt-Kirbach B, Golenhofen N. Reaction of small heat-shock proteins to different kinds of cellular stress in cultured rat hippocampal neurons. Cell Stress Chaperones. 2014;19(1):145-53.10.1007/s12192-013-0452-9Search in Google Scholar PubMed PubMed Central
22 Spillantini MG, Schmidt ML, Lee VM, Trojanowski JQ, Jakes R, Goedert M. Alpha-synuclein in Lewy bodies. Nature. 1997;388(6645):839-40.10.1038/42166Search in Google Scholar PubMed
23 Outeiro TF, Klucken J, Strathearn KE, Liu F, Nguyen P, Rochet JC, et al. Small heat shock proteins protect against alpha-synuclein-induced toxicity and aggregation. Biochem Biophys Res Commun. 2006;351(3):631-8.10.1016/j.bbrc.2006.10.085Search in Google Scholar PubMed PubMed Central
24 Cox D, Selig E, Griffin MD, Carver JA, Ecroyd H. Small Heat-shock Proteins Prevent alpha-Synuclein Aggregation via Transient Interactions and Their Efficacy Is Affected by the Rate of Aggregation. J Biol Chem. 2016;291(43):22618-29.10.1074/jbc.M116.739250Search in Google Scholar PubMed PubMed Central
25 Rekas A, Adda CG, Andrew Aquilina J, Barnham KJ, Sunde M, Galatis D, et al. Interaction of the molecular chaperone alphaB-crystallin with alpha-synuclein: effects on amyloid fibril formation and chaperone activity. J Mol Biol. 2004;340(5):1167-83.10.1016/j.jmb.2004.05.054Search in Google Scholar PubMed
26 Cox D, Whiten DR, Brown J, Horrocks MH, San Gil R, Dobson CM, et al. The small heat shock protein Hsp27 binds alpha-synuclein fibrils, preventing elongation and cytotoxicity. J Biol Chem. 2018.10.1074/jbc.M117.813865Search in Google Scholar PubMed PubMed Central
27 Cox D, Ecroyd H. The small heat shock proteins alphaB-crystallin (HSPB5) and Hsp27 (HSPB1) inhibit the intracellular aggregation of alpha-synuclein. Cell Stress Chaperones. 2017;22(4):589-600.10.1007/s12192-017-0785-xSearch in Google Scholar PubMed PubMed Central
28 Crippa V, D’Agostino VG, Cristofani R, Rusmini P, Cicardi ME, Messi E, et al. Transcriptional induction of the heat shock protein B8 mediates the clearance of misfolded proteins responsible for motor neuron diseases. Sci Rep. 2016;6:22827.10.1038/srep22827Search in Google Scholar PubMed PubMed Central
29 Wilhelmus MM, Boelens WC, Otte-Holler I, Kamps B, de Waal RM, Verbeek MM. Small heat shock proteins inhibit amyloid-beta protein aggregation and cerebrovascular amyloid-beta protein toxicity. Brain Res. 2006;1089(1):67-78.10.1016/j.brainres.2006.03.058Search in Google Scholar PubMed
30 Shammas SL, Waudby CA, Wang S, Buell AK, Knowles TP, Ecroyd H, et al. Binding of the molecular chaperone alphaB-crystallin to Abeta amyloid fibrils inhibits fibril elongation. Biophys J. 2011;101(7):1681-9.10.1016/j.bpj.2011.07.056Search in Google Scholar PubMed PubMed Central
31 Seidel K, Vinet J, Dunnen WF, Brunt ER, Meister M, Boncoraglio A, et al. The HSPB8-BAG3 chaperone complex is upregulated in astrocytes in the human brain affected by protein aggregation diseases. Neuropathol Appl Neurobiol. 2012;38(1):39-53.10.1111/j.1365-2990.2011.01198.xSearch in Google Scholar PubMed
32 Brady JP, Garland D, Duglas-Tabor Y, Robison WG, Jr., Groome A, Wawrousek EF. Targeted disruption of the mouse alpha A-crystallin gene induces cataract and cytoplasmic inclusion bodies containing the small heat shock protein alpha B-crystallin. Proc Natl Acad Sci U S A. 1997;94(3):884-9.10.1073/pnas.94.3.884Search in Google Scholar PubMed PubMed Central
33 Brady JP, Garland DL, Green DE, Tamm ER, Giblin FJ, Wawrousek EF. AlphaB-crystallin in lens development and muscle integrity: a gene knockout approach. Invest Ophthalmol Vis Sci. 2001;42(12):2924-34.Search in Google Scholar
34 La Padula V, Staszewski O, Nestel S, Busch H, Boerries M, Roussa E, et al. HSPB3 protein is expressed in motoneurons and induces their survival after lesion-induced degeneration. Exp Neurol. 2016;286:40-9.10.1016/j.expneurol.2016.08.014Search in Google Scholar PubMed
35 Baughman HER, Clouser AF, Klevit RE, Nath A. HspBl and Hsc70 chaperones engage distinct tau species and have different inhibitory effects on amyloid formation. J Biol Chem. 2018;293(8):2687-700.10.1074/jbc.M117.803411Search in Google Scholar PubMed PubMed Central
36 Abisambra JF, Blair LJ, Hill SE, Jones JR, Kraft C, Rogers J, et al. Phosphorylation dynamics regulate Hsp27-mediated rescue of neuronal plasticity deficits in tau transgenic mice. J Neurosci. 2010;30(46):15374-82.10.1523/JNEUROSCI.3155-10.2010Search in Google Scholar PubMed PubMed Central
37 Hagemann TL, Boelens WC, Wawrousek EF, Messing A. Suppression of GFAP toxicity by alphaB-crystallin in mouse models of Alexander disease. Hum Mol Genet. 2009;18(7):1190-9.10.1093/hmg/ddp013Search in Google Scholar PubMed PubMed Central
38 Tang G, Perng MD, Wilk S, Quinlan R, Goldman JE. Oligomers of mutant glial fibrillary acidic protein (GFAP) Inhibit the proteasome system in alexander disease astrocytes, and the small heat shock protein alphaB-crystallin reverses the inhibition. J Biol Chem. 2010;285(14):10527-37.10.1074/jbc.M109.067975Search in Google Scholar PubMed PubMed Central
39 Rusmini P, Crippa V, Giorgetti E, Boncoraglio A, Cristofani R, Carra S, et al. Clearance of the mutant androgen receptor in motoneuronal models of spinal and bulbar muscular atrophy. Neurobiol Aging. 2013;34(11):2585-603.10.1016/j.neurobiolaging.2013.05.026Search in Google Scholar PubMed PubMed Central
40 Evgrafov OV, Mersiyanova I, Irobi J, Van Den Bosch L, Dierick I, Leung CL, et al. Mutant small heat-shock protein 27 causes axonal Charcot-Marie-Tooth disease and distal hereditary motor neuropathy. Nat Genet. 2004;36(6):602-6.10.1038/ng1354Search in Google Scholar PubMed
41 Irobi J, Van Impe K, Seeman P, Jordanova A, Dierick I, Verpoorten N, et al. Hot-spot residue in small heat-shock protein 22 causes distal motor neuropathy. Nat Genet. 2004;36(6):597-601.10.1038/ng1328Search in Google Scholar PubMed
42 Srivastava AK, Renusch SR, Naiman NE, Gu S, Sneh A, Arnold WD, et al. Mutant HSPB1 overexpression in neurons is sufficient to cause age-related motor neuronopathy in mice. Neurobiol Dis. 2012;47(2):163-73.10.1016/j.nbd.2012.03.035Search in Google Scholar PubMed PubMed Central
43 Bouhy D, Juneja M, Katona I, Holmgren A, Asselbergh B, De Winter V, et al. A knock-in/knock-out mouse model of HSPB8-associated distal hereditary motor neuropathy and myopathy reveals toxic gain-of-function of mutant Hspb8. Acta Neuropathol. 2018;135(1):131-48.10.1007/s00401-017-1756-0Search in Google Scholar PubMed PubMed Central
44 Nakhro K, Park JM, Kim YJ, Yoon BR, Yoo JH, Koo H, et al. A novel Lys141Thr mutation in small heat shock protein 22 (HSPB8) gene in Charcot-Marie-Tooth disease type 2L. Neuromuscul Disord. 2013;23(8):656-63.10.1016/j.nmd.2013.05.009Search in Google Scholar PubMed
45 Stancanelli C, Fabrizi GM, Ferrarini M, Cavallaro T, Taioli F, Di Leo R, et al. Charcot-Marie-Tooth 2F: phenotypic presentation of the Arg136Leu HSP27 mutation in a multigenerational family. Neurol Sci. 2015;36(6):1003-6.10.1007/s10072-014-2050-8Search in Google Scholar PubMed
46 d’Ydewalle C, Krishnan J, Chiheb DM, Van Damme P, Irobi J, Kozikowski AP, et al. HDAC6 inhibitors reverse axonal loss in a mouse model of mutant HSPB1-induced Charcot-Marie-Tooth disease. Nat Med. 2011;17(8):968-74.10.1038/nm.2396Search in Google Scholar PubMed
47 Lee J, Jung SC, Joo J, Choi YR, Moon HW, Kwak G, et al. Overexpression of mutant HSP27 causes axonal neuropathy in mice. J Biomed Sci. 2015;22:43.10.1186/s12929-015-0154-ySearch in Google Scholar PubMed PubMed Central
48 Rothbard JB, Kurnellas MP, Brownell S, Adams CM, Su L, Axtell RC, et al. Therapeutic effects of systemic administration of chaperone alphaB-crystallin associated with binding proinflammatory plasma proteins. J Biol Chem. 2012;287(13):9708-21.10.1074/jbc.M111.337691Search in Google Scholar PubMed PubMed Central
49 Bartelt-Kirbach B, Slowik A, Beyer C, Golenhofen N. Upregulation and phosphorylation of HspB1/Hsp25 and HspB5/alphaB-crystallin after transient middle cerebral artery occlusion in rats. Cell Stress Chaperones. 2017;22(4):653-63.10.1007/s12192-017-0794-9Search in Google Scholar PubMed PubMed Central
50 Zeng L, Tan J, Lu W, Hu Z. Blockade of Ser16-Hsp20 phosphorylation attenuates neuroprotection dependent upon Bcl-2 and Bax. Curr Neurovasc Res. 2013;10(3):208-15.10.2174/15672026113109990006Search in Google Scholar PubMed
51 Leak RK, Zhang L, Stetler RA, Weng Z, Li P, Atkins GB, et al. HSP27 protects the blood-brain barrier against ischemia-induced loss of integrity. CNS Neurol Disord Drug Targets. 2013;12(3):325-37.10.2174/1871527311312030006Search in Google Scholar PubMed PubMed Central
52 Kourtis N, Nikoletopoulou V, Tavernarakis N. Small heat-shock proteins protect from heat-stroke-associated neurodegeneration. Nature. 2012;490(7419):213-8.10.1038/nature11417Search in Google Scholar PubMed
53 Kourtis N, Nikoletopoulou V, Tavernarakis N. Heat shock response and ionstasis: axis against neurodegeneration. Aging (Albany NY). 2012;4(12):856-8.10.18632/aging.100517Search in Google Scholar PubMed PubMed Central
54 Kawasaki F, Koonce NL, Guo L, Fatima S, Qiu C, Moon MT, et al. Small heat shock proteins mediate cell-autonomous and -nonautonomous protection in a Drosophila model for environmental-stress-induced degeneration. Dis Model Mech. 2016;9(9):953-64.Search in Google Scholar
55 Zhu Z, Li R, Stricker R, Reiser G. Extracellular alpha-crystallin protects astrocytes from cell death through activation of MAPK, PI3K/Akt signaling pathway and blockade of ROS release from mitochondria. Brain Res. 2015;1620:17-28.10.1016/j.brainres.2015.05.011Search in Google Scholar PubMed
56 Bartelt-Kirbach B, Moron M, Glomb M, Beck CM, Weller MP, Golenhofen N. HspB5/alphaB-crystallin increases dendritic complexity and protects the dendritic arbor during heat shock in cultured rat hippocampal neurons. Cell Mol Life Sci. 2016;73(19):3761-75.10.1007/s00018-016-2219-9Search in Google Scholar PubMed
57 Paul C, Simon S, Gibert B, Virot S, Manero F, Arrigo AP. Dynamic processes that reflect anti-apoptotic strategies set up by HspB1 (Hsp27). Exp Cell Res. 2010;316(9):1535-52.10.1016/j.yexcr.2010.03.006Search in Google Scholar PubMed
58 Havasi A, Li Z, Wang Z, Martin JL, Botla V, Ruchalski K, et al. Hsp27 inhibits Bax activation and apoptosis via a phosphatidylinositol 3-kinase-dependent mechanism. J Biol Chem. 2008;283(18):12305-13.10.1074/jbc.M801291200Search in Google Scholar PubMed PubMed Central
59 Crowe J, Aubareda A, McNamee K, Przybycien PM, Lu X, Williams RO, et al. Heat shock protein B1-deficient mice display impaired wound healing. PLoS One. 2013;8(10):e77383.10.1371/journal.pone.0077383Search in Google Scholar PubMed PubMed Central
60 Kammoun M, Picard B, Astruc T, Gagaoua M, Aubert D, Bonnet M, et al. The Invalidation of HspB1 Gene in Mouse Alters the Ultrastructural Phenotype of Muscles. PLoS One. 2016;11(8):e0158644.10.1371/journal.pone.0158644Search in Google Scholar PubMed PubMed Central
61 Bruey JM, Ducasse C, Bonniaud P, Ravagnan L, Susin SA, Diaz-Latoud C, et al. Hsp27 negatively regulates cell death by interacting with cytochrome c. Nat Cell Biol. 2000;2(9):645-52.10.1038/35023595Search in Google Scholar PubMed
62 Yang B, Zhang H, Mo X, Xiao H, Hu Z. HspB8 is neuroprotective during oxygen glucose deprivation and reperfusion. Curr Neurovasc Res. 2015;12(1):63-72.10.2174/1567202612666150102152350Search in Google Scholar PubMed
63 Oshita SE, Chen F, Kwan T, Yehiely F, Cryns VL. The small heat shock protein HspB2 is a novel anti-apoptotic protein that inhibits apical caspase activation in the extrinsic apoptotic pathway. Breast Cancer Res Treat. 2010;124(2):307-15.10.1007/s10549-010-0735-0Search in Google Scholar PubMed PubMed Central
64 Arrigo AP, Gibert B. HspB1, HspB5 and HspB4 in Human Cancers: Potent Oncogenic Role of Some of Their Client Proteins. Cancers (Basel). 2014;6(1):333-65.10.3390/cancers6010333Search in Google Scholar PubMed PubMed Central
65 Ninkovic J, Pinto L, Petricca S, Lepier A, Sun J, Rieger MA, et al. The transcription factor Pax6 regulates survival of dopaminergic olfactory bulb neurons via crystallin alphaA. Neuron. 2010;68(4):682-94.10.1016/j.neuron.2010.09.030Search in Google Scholar PubMed PubMed Central
66 den Engelsman J, Bennink EJ, Doerwald L, Onnekink C, Wunderink L, Andley UP, et al. Mimicking phosphorylation of the small heat-shock protein alphaB-crystallin recruits the F-box protein FBX4 to nuclear SC35 speckles. Eur J Biochem. 2004;271(21):4195-203.10.1111/j.1432-1033.2004.04359.xSearch in Google Scholar PubMed
67 Laganowsky A, Benesch JL, Landau M, Ding L, Sawaya MR, Cascio D, et al. Crystal structures of truncated alphaA and alphaB crystallins reveal structural mechanisms of polydispersity important for eye lens function. Protein Sci. 2010;19(5):1031-43.10.1002/pro.380Search in Google Scholar PubMed PubMed Central
68 Xi JH, Bai F, McGaha R, Andley UP. Alpha-crystallin expression affects microtubule assembly and prevents their aggregation. FASEB J. 2006;20(7):846-57.10.1096/fj.05-5532comSearch in Google Scholar PubMed
69 Zeng L, Tan J, Lu W, Lu T, Hu Z. The potential role of small heat shock proteins in mitochondria. Cell Signal. 2013;25(11):2312-9.10.1016/j.cellsig.2013.07.027Search in Google Scholar PubMed
70 Ganassi M, Mateju D, Bigi I, Mediani L, Poser I, Lee HO, et al. A Surveillance Function of the HSPB8-BAG3-HSP70 Chaperone Complex Ensures Stress Granule Integrity and Dynamism. Mol Cell. 2016;63(5):796-810.10.1016/j.molcel.2016.07.021Search in Google Scholar PubMed
71 Mateju D, Franzmann TM, Patel A, Kopach A, Boczek EE, Maharana S, et al. An aberrant phase transition of stress granules triggered by misfolded protein and prevented by chaperone function. EMBO J. 2017;36(12):1669-87.10.15252/embj.201695957Search in Google Scholar PubMed PubMed Central
72 Gorman AM, Szegezdi E, Quigney DJ, Samali A. Hsp27 inhibits 6-hydroxydopamine-induced cytochrome c release and apoptosis in PC12 cells. Biochem Biophys Res Commun. 2005;327(3):801-10.10.1016/j.bbrc.2004.12.066Search in Google Scholar PubMed
73 Masilamoni JG, Jesudason EP, Baben B, Jebaraj CE, Dhandayuthapani S, Jayakumar R. Molecular chaperone alpha-crystallin prevents detrimental effects of neuroinflammation. Biochim Biophys Acta. 2006;1762(3):284-93.10.1016/j.bbadis.2005.11.007Search in Google Scholar PubMed
74 Batulan Z, Pulakazhi Venu VK, Li Y, Koumbadinga G, Alvarez-Olmedo DG, Shi C, et al. Extracellular Release and Signaling by Heat Shock Protein 27: Role in Modifying Vascular Inflammation. Front Immunol. 2016;7:285.10.3389/fimmu.2016.00285Search in Google Scholar PubMed PubMed Central
75 Reddy VS, Madala SK, Trinath J, Reddy GB. Extracellular small heat shock proteins: exosomal biogenesis and function. Cell Stress Chaperones. 2018;23(3):441-54.10.1007/s12192-017-0856-zSearch in Google Scholar PubMed PubMed Central
76 Ce P, Erkizan O, Gedizlioglu M. Elevated HSP27 levels during attacks in patients with multiple sclerosis. Acta Neurol Scand. 2011;124(5):317-20.10.1111/j.1600-0404.2010.01475.xSearch in Google Scholar PubMed
77 Nafar F, Williams JB, Mearow KM. Astrocytes release HspB1 in response to amyloid-beta exposure in vitro. J Alzheimers Dis. 2016;49(1):251-63.10.3233/JAD-150317Search in Google Scholar PubMed
78 Wang YH, Li YC, Huo SJ, Yin ZQ. Alpha-crystallin promotes rat olfactory ensheathing cells survival and proliferation through regulation of PI3K/Akt/mTOR signaling pathways. Neurosci Lett. 2012;531(2):170-5.10.1016/j.neulet.2012.10.057Search in Google Scholar PubMed
79 Sreekumar PG, Kannan R, Kitamura M, Spee C, Barron E, Ryan SJ, et al. alphaB crystallin is apically secreted within exosomes by polarized human retinal pigment epithelium and provides neuroprotection to adjacent cells. PLoS One. 2010;5(10):e12578.10.1371/journal.pone.0012578Search in Google Scholar PubMed PubMed Central
80 Gangalum RK, Bhat AM, Kohan SA, Bhat SP. Inhibition of the Expression of the Small Heat Shock Protein alphaB-Crystallin Inhibits Exosome Secretion in Human Retinal Pigment Epithelial Cells in Culture. J Biol Chem. 2016;291(25):12930-42.10.1074/jbc.M115.698530Search in Google Scholar PubMed PubMed Central
81 Nahomi RB, Wang B, Raghavan CT, Voss O, Doseff AI, Santhoshkumar P, et al. Chaperone peptides of alpha-crystallin inhibit epithelial cell apoptosis, protein insolubilization, and opacification in experimental cataracts. J Biol Chem. 2013;288(18):13022-35.10.1074/jbc.M112.440214Search in Google Scholar PubMed PubMed Central
82 Nahomi RB, DiMauro MA, Wang B, Nagaraj RH. Identification of peptides in human Hsp20 and Hsp27 that possess molecular chaperone and anti-apoptotic activities. Biochem J. 2015;465(1):115-25.10.1042/BJ20140837Search in Google Scholar PubMed PubMed Central
83 Mannini B, Cascella R, Zampagni M, van Waarde-Verhagen M, Meehan S, Roodveldt C, et al. Molecular mechanisms used by chaperones to reduce the toxicity of aberrant protein oligomers. Proc Natl Acad Sci U S A. 2012;109(31):12479-84.10.1073/pnas.1117799109Search in Google Scholar PubMed PubMed Central
84 van Noort JM, Bsibsi M, Nacken P, Gerritsen WH, Amor S. The link between small heat shock proteins and the immune system. Int J Biochem Cell Biol. 2012;44(10):1670-9.10.1016/j.biocel.2011.12.010Search in Google Scholar PubMed
85 Masilamoni JG, Jesudason EP, Bharathi SN, Jayakumar R. The protective effect of alpha-crystallin against acute inflammation in mice. Biochim Biophys Acta. 2005;1740(3):411-20.10.1016/j.bbadis.2004.11.002Search in Google Scholar PubMed
86 Klopstein A, Santos-Nogueira E, Francos-Quijorna I, Redensek A, David S, Navarro X, et al. Beneficial effects of alphaB-crystallin in spinal cord contusion injury. J Neurosci. 2012;32(42):14478-88.10.1523/JNEUROSCI.0923-12.2012Search in Google Scholar PubMed PubMed Central
87 Wu N, Yu J, Chen S, Xu J, Ying X, Ye M, et al. alpha-Crystallin protects RGC survival and inhibits microglial activation after optic nerve crush. Life Sci. 2014;94(1):17-23.10.1016/j.lfs.2013.10.034Search in Google Scholar PubMed
88 Kurnellas MP, Brownell SE, Su L, Malkovskiy AV, Rajadas J, Dolganov G, et al. Chaperone activity of small heat shock proteins underlies therapeutic efficacy in experimental autoimmune encephalomyelitis. J Biol Chem. 2012;287(43):36423-34.10.1074/jbc.M112.371229Search in Google Scholar PubMed PubMed Central
89 Nahomi RB, Palmer A, Green KM, Fort PE, Nagaraj RH. Pro-inflammatory cytokines downregulate Hsp27 and cause apoptosis of human retinal capillary endothelial cells. Biochim Biophys Acta. 2014;1842(2):164-74.10.1016/j.bbadis.2013.11.011Search in Google Scholar PubMed PubMed Central
90 Wilhelmus MM, Boelens WC, Kox M, Maat-Schieman ML, Veerhuis R, de Waal RM, et al. Small heat shock proteins associated with cerebral amyloid angiopathy of hereditary cerebral hemorrhage with amyloidosis (Dutch type) induce interleukin-6 secretion. Neurobiol Aging. 2009;30(2):229-40.10.1016/j.neurobiolaging.2007.06.001Search in Google Scholar PubMed
91 Bruinsma IB, de Jager M, Carrano A, Versleijen AA, Veerhuis R, Boelens W, et al. Small heat shock proteins induce a cerebral inflammatory reaction. J Neurosci. 2011;31(33):11992-2000.10.1523/JNEUROSCI.0945-11.2011Search in Google Scholar PubMed PubMed Central
92 Thornell E, Aquilina A. Regulation of alphaA- and alphaB-crystallins via phosphorylation in cellular homeostasis. Cell Mol Life Sci. 2015;72(21):4127-37.10.1007/s00018-015-1996-xSearch in Google Scholar PubMed
93 Treweek TM, Meehan S, Ecroyd H, Carver JA. Small heat-shock proteins: important players in regulating cellular proteostasis. Cell Mol Life Sci. 2015;72(3):429-51.10.1007/s00018-014-1754-5Search in Google Scholar PubMed
94 Kourtis N, Moubarak RS, Aranda-Orgilles B, Lui K, Aydin IT, Trimarchi T, et al. FBXW7 modulates cellular stress response and metastatic potential through HSF1 post-translational modification. Nat Cell Biol. 2015;17(3):322-32.10.1038/ncb3121Search in Google Scholar PubMed PubMed Central
95 Tang Z, Dai S, He Y, Doty RA, Shultz LD, Sampson SB, et al. MEK guards proteome stability and inhibits tumor-suppressive amyloidogenesis via HSF1. Cell. 2015;160(4):729-44.10.1016/j.cell.2015.01.028Search in Google Scholar PubMed PubMed Central
96 Gomez-Pastor R, Burchfiel ET, Neef DW, Jaeger AM, Cabiscol E, McKinstry SU, et al. Abnormal degradation of the neuronal stress-protective transcription factor HSF1 in Huntington’s disease. Nat Commun. 2017;8:14405.10.1038/ncomms14405Search in Google Scholar PubMed PubMed Central
97 Hayashida N, Fujimoto M, Tan K, Prakasam R, Shinkawa T, Li L, et al. Heat shock factor 1 ameliorates proteotoxicity in cooperation with the transcription factor NFAT. EMBO J. 2010;29(20):3459-69.10.1038/emboj.2010.225Search in Google Scholar PubMed PubMed Central
98 Shinkawa T, Tan K, Fujimoto M, Hayashida N, Yamamoto K, Takaki E, et al. Heat shock factor 2 is required for maintaining proteostasis against febrile-range thermal stress and polyglutamine aggregation. Mol Biol Cell. 2011;22(19):3571-83.10.1091/mbc.e11-04-0330Search in Google Scholar
99 Nakagomi S, Suzuki Y, Namikawa K, Kiryu-Seo S, Kiyama H. Expression of the activating transcription factor 3 prevents c-Jun N-terminal kinase-induced neuronal death by promoting heat shock protein 27 expression and Akt activation. J Neurosci. 2003;23(12):5187-96.10.1523/JNEUROSCI.23-12-05187.2003Search in Google Scholar
100 Cui X, Liu H, Li J, Guo K, Han W, Dong Y, et al. Heat shock factor 4 regulates lens epithelial cell homeostasis by working with lysosome and anti-apoptosis pathways. Int J Biochem Cell Biol. 2016;79:118-27.10.1016/j.biocel.2016.08.022Search in Google Scholar PubMed
101 Cui X, Xie PP, Jia PP, Lou Q, Dun G, Li S, et al. Hsf4 counteracts Hsf1 transcription activities and increases lens epithelial cell survival in vitro. Biochim Biophys Acta. 2015;1853(3):746-55.10.1016/j.bbamcr.2015.01.004Search in Google Scholar PubMed
102 Cui X, Han W, Li J, Feng R, Zhou Z, Han J, et al. Heat shock factor 4 regulates the expression of HSP25 and alpha B-crystallin by associating with DEXD/H-box RNA helicase UAP56. Cell Stress Chaperones. 2017.10.1007/s12192-017-0865-ySearch in Google Scholar PubMed PubMed Central
103 Dierick I, Irobi J, Janssens S, Theuns J, Lemmens R, Jacobs A, et al. Genetic variant in the HSPB1 promoter region impairs the HSP27 stress response. Hum Mutat. 2007;28(8):830.10.1002/humu.9503Search in Google Scholar PubMed
104 Vleminckx V, Van Damme P, Goffin K, Delye H, Van Den Bosch L, Robberecht W. Upregulation of HSP27 in a transgenic model of ALS. J Neuropathol Exp Neurol. 2002;61(1l):968-74.10.1093/jnen/61.11.968Search in Google Scholar PubMed
105 Krishnan J, Vannuvel K, Andries M, Waelkens E, Robberecht W, Van Den Bosch L. Over-expression of Hsp27 does not influence disease in the mutant SOD1(G93A) mouse model of amyotrophic lateral sclerosis. J Neurochem. 2008;106(5):2170-83.Search in Google Scholar
106 Vihervaara A, Duarte FM, Lis JT. Molecular mechanisms driving transcriptional stress responses. Nat Rev Genet. 2018.10.1038/s41576-018-0001-6Search in Google Scholar PubMed PubMed Central
107 Arac A, Brownell SE, Rothbard JB, Chen C, Ko RM, Pereira MP, et al. Systemic augmentation of alphaB-crystallin provides therapeutic benefit twelve hours post-stroke onset via immune modulation. Proc Natl Acad Sci U S A. 2011;108(32):13287-92.10.1073/pnas.1107368108Search in Google Scholar PubMed PubMed Central
108 Badin RA, Lythgoe MF, van der Weerd L, Thomas DL, Gadian DG, Latchman DS. Neuroprotective effects of virally delivered HSPs in experimental stroke. J Cereb Blood Flow Metab. 2006;26(3):371-81.10.1038/sj.jcbfm.9600190Search in Google Scholar PubMed
109 van Noort JM, Bsibsi M, Nacken PJ, Verbeek R, Venneker EH. Therapeutic Intervention in Multiple Sclerosis with Alpha B-Crystallin: A Randomized Controlled Phase Ila Trial. PLoS One. 2015;10(11):e0143366.10.1371/journal.pone.0143366Search in Google Scholar PubMed PubMed Central
© 2018 Nikos Kourtis, Nektarios Tavernarakis, published by De Gruyter
This work is licensed under the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 License.
Articles in the same Issue
- Review
- Secretion of full-length Tau or Tau fragments in cell culture models. Propagation of Tau in vivo and in vitro
- Research Article
- DNA testing of edible crabs from seafood shops on the Odisha coast, India
- Communication
- Overexpression and pre-treatment of recombinant human Secretory Leukocyte Protease Inhibitor (rhSLPI) reduces an in vitro ischemia/reperfusion injury in rat cardiac myoblast (H9c2) cell
- Review
- Global Perspective of Novel Therapeutic Strategies for the Management of NeuroAIDS
- Research Article
- Current insights on use of growth factors as therapy for Intervertebral Disc Degeneration
- Research Article
- Treatment of SEC62 over-expressing tumors by Thapsigargin and Trifluoperazine
- Review
- Interferon-gamma (IFN-γ): Exploring its implications in infectious diseases
- Research Article
- Polymorphisms in DNA repair genes increase the risk for type 2 diabetes mellitus and hypertension
- Review
- Small heat shock proteins and neurodegeneration: recent developments
- Review Article
- The rise of genetically engineered mouse models of pancreatitis: A review of literature
- Research Article
- Early Life Trauma Predicts Affective Phenomenology and the Effects are Partly Mediated by Staging Coupled with Lowered Lipid-Associated Antioxidant Defences
- Review Article
- Significance of bacteriophages in fermented soybeans: A review
- Review
- Biased receptor functionality versus biased agonism in G-protein-coupled receptors
- Research Article
- Neural like cells and acetyl-salicylic acid alter rat brain structure and function following transient middle cerebral artery occlusion
- Research Article
- Enhancing the anticoagulant profile of meizothrombin
- Mini review
- Can uterine secretion of modified histones alter blastocyst implantation, embryo nutrition, and transgenerational phenotype?
- Research Article
- In vitro and in vivo evaluation of colon cancer targeted epichlorohydrin crosslinked Portulaca-alginate beads
- Mini review
- Prospects of Pharmacological Interventions to Organismal Aging
- When Humans Met Superbugs: Strategies to Tackle Bacterial Resistances to Antibiotics
- Special Issue: Recent Advances in Basic and Clinical Medicine
- Beneficial effects of Spirogyra Neglecta Extract on antioxidant and anti-inflammatory factors in streptozotocin-induced diabetic rats
Articles in the same Issue
- Review
- Secretion of full-length Tau or Tau fragments in cell culture models. Propagation of Tau in vivo and in vitro
- Research Article
- DNA testing of edible crabs from seafood shops on the Odisha coast, India
- Communication
- Overexpression and pre-treatment of recombinant human Secretory Leukocyte Protease Inhibitor (rhSLPI) reduces an in vitro ischemia/reperfusion injury in rat cardiac myoblast (H9c2) cell
- Review
- Global Perspective of Novel Therapeutic Strategies for the Management of NeuroAIDS
- Research Article
- Current insights on use of growth factors as therapy for Intervertebral Disc Degeneration
- Research Article
- Treatment of SEC62 over-expressing tumors by Thapsigargin and Trifluoperazine
- Review
- Interferon-gamma (IFN-γ): Exploring its implications in infectious diseases
- Research Article
- Polymorphisms in DNA repair genes increase the risk for type 2 diabetes mellitus and hypertension
- Review
- Small heat shock proteins and neurodegeneration: recent developments
- Review Article
- The rise of genetically engineered mouse models of pancreatitis: A review of literature
- Research Article
- Early Life Trauma Predicts Affective Phenomenology and the Effects are Partly Mediated by Staging Coupled with Lowered Lipid-Associated Antioxidant Defences
- Review Article
- Significance of bacteriophages in fermented soybeans: A review
- Review
- Biased receptor functionality versus biased agonism in G-protein-coupled receptors
- Research Article
- Neural like cells and acetyl-salicylic acid alter rat brain structure and function following transient middle cerebral artery occlusion
- Research Article
- Enhancing the anticoagulant profile of meizothrombin
- Mini review
- Can uterine secretion of modified histones alter blastocyst implantation, embryo nutrition, and transgenerational phenotype?
- Research Article
- In vitro and in vivo evaluation of colon cancer targeted epichlorohydrin crosslinked Portulaca-alginate beads
- Mini review
- Prospects of Pharmacological Interventions to Organismal Aging
- When Humans Met Superbugs: Strategies to Tackle Bacterial Resistances to Antibiotics
- Special Issue: Recent Advances in Basic and Clinical Medicine
- Beneficial effects of Spirogyra Neglecta Extract on antioxidant and anti-inflammatory factors in streptozotocin-induced diabetic rats