Next Article in Journal
In Situ PD-L1 Expression in Oral Squamous Cell Carcinoma Is Induced by Heterogeneous Mechanisms among Patients
Next Article in Special Issue
Oxidative Stress in Amyotrophic Lateral Sclerosis: Synergy of Genetic and Environmental Factors
Previous Article in Journal
Resveratrol Prevents Cytoarchitectural and Interneuronal Alterations in the Valproic Acid Rat Model of Autism
Previous Article in Special Issue
The Expression of Active CD11b Monocytes in Blood and Disease Progression in Amyotrophic Lateral Sclerosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

DnaJC7 in Amyotrophic Lateral Sclerosis

by
Allison A. Dilliott
1,
Catherine M. Andary
2,
Meaghan Stoltz
2,
Andrey A. Petropavlovskiy
2,
Sali M. K. Farhan
1,3 and
Martin L. Duennwald
2,*
1
Department of Neurology and Neurosurgery, McGill University, Montréal, QC H4A 3J1, Canada
2
Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A5C1, Canada
3
Department of Human Genetics, McGill University, Montréal, QC H4A 3J1, Canada
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(8), 4076; https://doi.org/10.3390/ijms23084076
Submission received: 21 February 2022 / Revised: 24 March 2022 / Accepted: 24 March 2022 / Published: 7 April 2022
(This article belongs to the Special Issue Molecular Research on Amyotrophic Lateral Sclerosis)

Abstract

:
Protein misfolding is a common basis of many neurodegenerative diseases including amyotrophic lateral sclerosis (ALS). Misfolded proteins, such as TDP-43, FUS, Matrin3, and SOD1, mislocalize and form the hallmark cytoplasmic and nuclear inclusions in neurons of ALS patients. Cellular protein quality control prevents protein misfolding under normal conditions and, particularly, when cells experience protein folding stress due to the fact of increased levels of reactive oxygen species, genetic mutations, or aging. Molecular chaperones can prevent protein misfolding, refold misfolded proteins, or triage misfolded proteins for degradation by the ubiquitin–proteasome system or autophagy. DnaJC7 is an evolutionarily conserved molecular chaperone that contains both a J-domain for the interaction with Hsp70s and tetratricopeptide domains for interaction with Hsp90, thus joining these two major chaperones’ machines. Genetic analyses reveal that pathogenic variants in the gene encoding DnaJC7 cause familial and sporadic ALS. Yet, the underlying ALS-associated molecular pathophysiology and many basic features of DnaJC7 function remain largely unexplored. Here, we review aspects of DnaJC7 expression, interaction, and function to propose a loss-of-function mechanism by which pathogenic variants in DNAJC7 contribute to defects in DnaJC7-mediated chaperoning that might ultimately contribute to neurodegeneration in ALS.

1. Introduction

ALS is an incurable neurodegenerative disease associated with protein misfolding and impaired cellular protein quality control, which leads to progressive loss of motor neurons and, thus, motor function. Classic cases display a median survival period of only 2–4 years [1]. On the cellular level, ALS is characterized by misfolding, mislocalization, and inclusion formation of RNA-binding proteins such as TAR DNA-binding protein 43 (TDP-43), fused in sarcoma (FUS), Matrin3 (MATR3), and superoxide dismutase (SOD1) [2]. Most ALS cases (~90%) occur sporadically (sALS) with contributing genetic and environmental factors remaining mostly unknown. By contrast, ~10% of ALS cases are familial (fALS), presumed to be caused by pathogenic genetic variants, generally occurring in one of more than 30 genes.
We and then others found that pathogenic variants within the gene DNAJC7 (TPR2; tetratricopeptide repeat 2) are associated with ALS in a study comprising nearly 4000 ALS cases and 8000 ethnically matched controls [3]. The gene encodes a protein belonging to the J protein/Hsp40 family, a subclassification of molecular chaperones imperative for protein quality control by maintaining protein function and preventing toxicity often associated with protein misfolding. Therefore, the association between DNAJC7 genetic variants and ALS has motivated our postulation that in addition to the impaired RNA processing, vesicle trafficking, and transposon suppression known to contribute to cellular toxicity and, ultimately, neurodegeneration in ALS, protein misfolding and impaired protein quality control are early and fundamental features of ALS pathogenesis.
In this review article, we focus on the function and disease association of DNAJC7. We review the evolutionary lineage, expression pattern, and transcriptional regulation of DNAJC7 as well as the interaction of DnaJC7 with Hsp70 and Hsp90 chaperones and its proposed functions in cellular protein quality control. We discuss the most recent evidence for the involvement of DnaJC7 in ALS and propose plausible mechanisms by which pathogenic variants in DNAJC7 may contribute to ALS, focusing on its chaperone function.

2. J Proteins

As previously described, J proteins, also known as Hsp40s, constitute a special class of molecular chaperones involved in protein quality control. They are the most numerous and the most diverse molecular chaperone class in eukaryotic cells with 22 different J proteins expressed in yeast (S. cerevisiae), 49 in humans, and a staggering 89 in the plant A. thaliana [4,5]. This genetic diversity likely arose through gene duplications, which subsequently allowed J proteins to functionally diversify. J proteins were initially regarded as interchangeable isoforms that merely functioned to drive Hsp70 ATP hydrolysis; however, this view has evolved due to the fact of several discoveries demonstrating the immense functional diversity of J proteins and their role as highly specialized mediators of cellular protein quality control [6,7].
All J proteins contain a ~70 amino acid long J-domain that consists of three alpha-helices and a histidine, proline, and aspartic acid (HPD) motif between helices II and III [8]. Although the J-domain sequence varies among J proteins, the HPD motif is highly conserved and is key to the interaction with Hsp70. The general function of J proteins is mediated by their J-domain and was first deciphered in vitro using purified DnaJ from E. coli. Liberek et al. first documented that DnaJ increases the otherwise weak ATPase activity of DnaK (Hsp70) [9]. Ensuing studies showed that binding of DnaJ to Hsp70 is essential for Hsp70-mediated substrate folding, that the J-domain is essential for this function, and that DnaJ recruits client proteins to Hsp70 [10,11,12]. Together, these basic biochemical studies unraveled the J protein/Hsp70 ATPase cycle, a common example of a chaperone cycle [13]. In this cycle, the J protein binds to the client protein, transfers it to the Hsp70 substrate binding domain, and activates the Hsp70 ATPase. Then the helical lid of Hsp70 closes and the client protein is folded. Finally, after a conformational change induced by nucleotide exchange, the J protein dissociates from Hsp70, and the client protein is released from Hsp70.
J proteins are subdivided into three structural classes, all of which contain the compact helical J-domain [14]. Type I J proteins (DNAJAs) are unique due to the fact of their cysteine-rich zinc finger domain, which is linked to the J-domain by a glycine/phenylalanine-rich region. The subtype also contains a C-terminal domain hypothesized to be involved in client protein binding and primarily involved in mitigating proteotoxic stress by facilitating protein folding, refolding, and degradation [15,16]. In contrast, Type II J proteins (DNAJBs) lack the cysteine-rich region, rather containing a J-domain linked by glycine/phenylalanine-rich region to the C-terminal domain. DNAJBs are uniquely suited to aid in protein disaggregation [17]. Finally, Type III J proteins (DNAJCs) contain only the signature J-domain without the remaining features of DNAJAs and DNAJBs but are otherwise highly diverse displaying a wide range of cellular functions including those unrelated to their Hsp70 co-chaperone functions [18].
Given their central role in cellular protein quality control, including protein folding and clearance of misfolded proteins, the involvement of J proteins in neurodegenerative diseases resulting from protein aggregation and accumulation is unsurprising. Additionally, the overexpression of J proteins has displayed neuroprotective effects [3], including suppressed protein aggregation and accumulation of reactive oxidative species [17], decreased α-synuclein aggregation [19], improved motor neuron survival [20], and prevention of tau aggregation [21], which will be discussed further below. Table 1 outlines all J proteins that have been previously reported to be associated with a neurological phenotype. Although this review focuses on the discussion of DnaJC7 and its association with ALS, based on the abundance of associations between J proteins and various neurological conditions, it will remain important to continue exploring the involvement of other J proteins in the neuropathological pathways of diseases such as neurodegeneration.

3. DnaJC7

DnaJC7, otherwise referred to as Tpr2 or CCRP, is a conserved, albeit unusual, type of J protein that is highly expressed within the brain [3]. Although it does contain the expected J-domain, DnaJC7 is unique, as it contains two tetratricopeptide (TPR) domains, TPR1 and TPR2, which are suggested to allow DnaJC7 to bridge the two key molecular chaperone systems Hsp90 and Hsp70 (Figure 1) [22,23]. Specifically, DnaJC7 has been found to modulate the flow of substrates from Hsp70 to Hsp90, providing a control mechanism in proteins such as glucocorticoids and progesterone receptors [23,24]. The J protein has also demonstrated retention and co-activating capabilities with the constitutive active/androstane receptor (CAR), suggesting an important role in CAR-mediated gene activation [25].
Overall, Hsp70s and J proteins are among the most conserved chaperone systems throughout evolution, particularly among metazoans, likely due to the fact of their essential role in assisting the folding or disassembly of protein structures and protein homeostasis generally. These essential functions ensure conservation of protein-interacting domains of the molecular chaperones and their co-chaperones, which is especially obvious for DNAJ proteins [13]. DNAJC7 is no exception to this level of conservation, with orthologs containing both the classical J-domain for its interactions with the Hsp70 system as well as its two highly conserved TPR domains [21]. DnaJC7 orthologues can be found in all higher chordates and even most vertebrates including birds, alligators, turtles, lizards, mammals, amphibians, coelacanths, bony fishes, lampreys, and the cartilaginous fishes (Figure 2) [30].
Interestingly, the arrangement of the domains of DnaJC7, specifically the TPR domains, are also shared with other proteins. One such protein is the Hsp70–Hsp90-organizing protein (Hop), a co-chaperone that also binds Hsp70 and Hsp90 to facilitate substrate transfer [31]. The similarities between the TPR domain organization between DnaJC7 and Hop has led to previous propositions that the two proteins possess shared functionalities in chaperone regulation [23], although with differing mechanisms [32]. Yet, more recent evidence has shown little similarity in the sequences of the TPR domains of DnaJC7 and Hop, and when combined with the high conservation of the DnaJC7 TPR domain sequence, this may suggest that DnaJC7 has additional or differing functionality from Hop and other TPR domain-containing proteins [21].

4. Identification of a Gene–Disease Association between DNAJC7 and ALS

DNAJC7 was first identified as a gene associated with ALS in the largest ALS case–control study to date, which utilized whole exome sequencing (WES) data to perform exome-wide variant enrichment and gene burden analyses [3]. The ALS exome sequencing data were gathered as a part of the ALS Genetics (ALSGENS) and Familial ALS (FALS) consortia projects, and the analysis included 3864 ALS patients of European descent. The study replicated rare variant enrichment signals from known ALS genes including SOD1, NEK1, and FUS. Additionally, four carriers of rare, protein truncating variants in DNAJC7 were observed in the initial ALS cases, whereas zero were observed across the control cohort (n = 28,910). Following this observation, an additional 1231 ALS patients were assessed and four other PTV carriers were identified, resulting in a genome-wide significant signal of variant enrichment across the pooled ALS cohort (n = 5095). In total, six distinct protein-truncating variants were observed across the eight individuals including p.E33X, p.Q120X, p.R156X, p.F163fs, p.R216X, and c.1011-2A > G. Additionally, 15 rare missense variants were identified across the ALS cohort in DNAJC7, four of which were predicted to be pathogenic in ALS cases, namely, p.D8H, p.D211N, p. R412W, and p.E425K. All variants observed were exceedingly rare in the general population. Detailed annotations of all previously reported likely damaging DNAJC7 variants identified across the various ALS cohorts are reported in Table 2. Since the discovery of DNAJC7, multiple cohorts of Asian descent have also been screened for variants of interest in the gene. In total, six additional missense variants have been reported in these cohorts, of which four were predicted to be pathogenic (p.R156Q, p.K137R, p.R238G, and p.Y338N), and two were predicted to be benign (p.S94T and p.N369T) based on variant frequencies and in silico prediction methods [27,28,29]. Additionally, a novel splicing variant (c.754-3T > C) was identified in a sporadic ALS patient of Chinese descent [28], and a novel protein-truncating frameshift variant (p.Q134Rfs*6) was identified in a sporadic ALS patient of Taiwanese descent [26]. Figure 1 represents a schematic of the DnaJC7 protein to display the location of all previously reported likely damaging variants.

5. Transcriptional Regulation of DNAJC7

The heat shock response (HSR), driven by the entire family of heat shock proteins (Hsps), is the cell response to stress and ensures that proteins are properly folded otherwise assisting with re-folding or protein degradation. It is unsurprising that a disease largely driven by misfolded proteins and protein aggregates, such as ALS, has been associated with defects in the HSR [33,34,35]. As described, DnaJC7 is a member of the Hsp protein family and displays many interactions with other proteins involved in the HSR; therefore, gaining a greater understanding of its involvement in the HSR may provide important context regarding its involvement in ALS.
Hsf1 is the major heat shock transcription factor that activates the HSR due to the fact of protein misfolding [36]. Upon accumulation of misfolded proteins in the cytosol and the nucleus, Hsf1 binds to heat shock elements (HSEs) within the promoter region of target genes as a trimer and activates their expression [37,38]. To identify putative Hsf1 binding motifs, we performed promoter region analysis of human DNAJC7 using the Eukaryotic Promoter Database (EPD) Search Motif Tool and JASPAR CORE 2018 motif library [39,40]. The promoter region was chosen based on the transcription start site common for most DNAJC7 isoforms (DNAJC7_1). This analysis suggests the presence of a single Hsf1 recognizable HSE, 323 base pairs from the transcription start site (p = 0.001). There is also experimental evidence indicating that DNAJC7 is regulated by Hsf1. First, the ChIP-Seq studies performed as part of the ENCODE project identified a putative Hsf1 binding peak in the DNAJC7 promoter in a human lymphoblastoid cell line (ENCSR009MBP) and, to a lesser extent, in an MCF-7 human breast cancer cell line (ENCSR062HDL) [41,42]. Additionally, a study by Mendillo et al. using ChIP-Seq found that upon 42 °C heat shock, Hsf1 was abundantly bound in the promoter of DNAJC7 in primary immortalized mammary epithelial (HME/BPE) and non-malignant breast epithelial (MCF10A) cells but not in tumorigenic BT20, NCIH838, and SKBR3 cell lines [43]. Similarly, DNAJC7 was not induced by heat shock in HEP-G2 cells, based on Western blotting experiment, mouse embryonic fibroblasts, or PRO-Seq experiment [44,45], and the DNAJC7 promoter was not bound by Hsf1 in healthy or apoptotic rat cerebral granule neurons [46].
Another hallmark of neurodegenerative disease, and ALS specifically, is oxidative stress caused by the accumulation of reactive oxygen species (ROS) due to the imbalance between the levels of endogenously produced ROS or environmental conditions and the mechanisms that remove them [47,48,49]. Nrf2 is the master transcription factor involved in the oxidative stress response [50]. Upon oxidative stress, Nrf2 dissociates from its negative regulator Keap1 and binds to cis-regulatory sequences known as antioxidant response elements (AREs) [51]. To determine whether DNAJC7 may be regulated by Nsf1, we again performed promotor analysis using the EPD Search Motif Tool and JASPAR CORE 2018 motif library and identified putative Nrf2 motifs in DNAJC7 at −508 and −648 base pairs from the transcription start site (p = 0.001), although the experimental evidence contradicts the notion that DNAJC7 is regulated by Nrf2 including a lack of interaction between the protein and DNAJC7 promotor and a lack of DNAJC7 induction following oxidative stress [52,53,54].
Finally, we investigated the potential regulation of DNAJC7 by Nrf1, a transcription factor closely related to Nrf2 that has displayed neuroprotective effects [55,56,57]. While Nrf1 also binds to AREs, it has a slightly differing binding motif to Nrf2 and is known to regulate a distinct subset of genes including those related to the proteosome and cell proliferation [58,59]. Nrf1 possesses several distinct isoforms that regulate different subsets of genes and interact with different co-factors, adding an additional level of complexity to Nrf1 regulation [59,60]. Promoter analysis of DNAJC7 identified a strong putative binding motif for Nrf1 at –165 base pairs from the transcription start site (p = 0.0001). However, similar to Nrf2, the available experimental evidence does not support regulation of DNAJC7 by Nrf1 including evidence of a lack of interaction with the promotor and lack of induction of DNAJC7 [59,61,62]. Given the complexity of Nrf1 regulation, it is possible that these studies just did not utilize a unique set of conditions under which Nrf1 regulates DNAJC7.
Combined, our analysis of available experimental data suggests that DNAJC7 may be regulated by Hsf1, possibly in a cell-type-dependent and stress-dependent manner. Previously, inactivation of Hsf1 was shown to result in the accumulation of insoluble, hyperphosphorylated TDP-43 aggregates. In contrast, upregulation of Hsf1 resulted in the increased solubility of SOD1 and improved clearance of insoluble TDP-43 aggregates in a manner mediated by the DnaJC7 interactors, Hsp70 and DnaJB2 [33,63]. While it seems unlikely that DNAJC7 is regulated by the master transcription factor of oxidative stress response, Nrf2, it is possible that under some conditions, it may be regulated by Nrf1, and this mode of regulation may warrant further investigation.

6. Expression of DNAJC7 in the Human Central Nervous System

Of the 49 J proteins encoded within the human genome, the majority are expressed in the brain at varying levels, although DNAJC7 is considered highly expressed [64]. Using the expression of the genes encoding Hsp70 and Hsp90 proteins as a baseline, we compared the expression of multiple J protein-encoding genes—including those associated with neurological conditions according to ClinVar (Table 1), among others—which demonstrated a wide range of expression levels throughout the human central nervous system (CNS) (Figure 3). All expression data were obtained from the Genotype-Tissue Expression (GTEx) database (https://gtexportal.org/home/ accessed on 12 March 2022) [65].
The Hsp70 protein family are encoded by HSPA genes and are the most abundant chaperones, as they carry out a variety of functions including protein folding, translocation across organelle membranes, and prevention of aggregate formation. Here, we analyzed one constitutive and one stress-induced HSPA as a reference for molecular chaperone expression. The constitutive HSPA2 is expressed at moderate to high levels throughout the CNS with notably high expression in the spinal cord, whereas the stress-induced HSPA12A featured moderate to low expression in the human brain and spinal cord. Similarly, Hsp90s are ubiquitously expressed at very high levels. Overall, throughout the CNS, DNAJC7 displays higher expression levels than the Hsp70s but lower overall expression than the Hsp90s. The J protein’s expression levels were most like that of HSPA2 and most distinct to those of HSP90AB1 and HSP90AA1, although the differences were marginal.
Of the J protein-encoding genes, the mostly highly expressed throughout the CNS included DNAJA1 and DNAJB2, although DNAJC5, DNAJC6, DNAJB1, and DNAJC7 were all also considered to be highly expressed across the brain regions. Contrarily, many other DNAJCs were among those most lowly expressed throughout the CNS such as DNAJC28, SACS, and DNAJC16. Yet, many of these DNAJs have been previously associated with neurological phenotypes within the ClinVar database (Table 1). Overall, this expression analysis suggests a prominent role of DnaJC7 in the human CNS and indicates that loss of DnaJC7 function in the CNS could be particularly detrimental and cause ALS-associated neurodegeneration.

7. Interactions of DnaJC7 with HSP70s, HSP90s, and Other Proteins

DnaJC7 has demonstrated interactions with Hsp70s and Hsp90s as well as with important co-chaperones of these molecular chaperone systems [66]. Here, we analyzed known physical interactions between a selection of J proteins, Hsp70s, and Hsp90s and describe notable interactions with DnaJC7 that are potentially relevant to the encoding gene’s association with ALS. Figure 4 displays an interaction map created using the GeneMANIA prediction server to visualize the physical interactions between these proteins (https://genemania.org/ accessed on 12 March 2022) [67].
Expectedly, DnaJC7 displays physical interactions with Hsp70s, including HSPA2, HSPA4, and HSPA8, and Hsp90s such as HSP90AA1 and HSP90AB1 [23,32]. Interactions with the Hsp70s are mediated by DnaJC7’s conserved J-domain, consistent with the J-domain functionality of other J proteins [23]. Although this interaction is independent of any other protein features, evidence does suggest that the TPR domains may have stabilizing effects [32]. The interaction stimulates the ATPase function of Hsp70s, allowing the protein class to perform stable polypeptide binding and protein folding [23]. Notably, HSPA8 was shown to be reduced in the primary motor neurons and neuromuscular junctions in TDP-43-mediated mouse models of ALS as well as in C9orf72-mediated fly models and human-induced pluripotent stem cells [68]. DnaJC7 binds Hsp90s through its TPR domains and has a role in the disruption of interactions between Hsp90s and its substrates, allowing for proteins to re-enter the protein folding pathway if not properly folded after a single pass through the Hsp70–Hsp90 folding system [23,32]. This suggests that while DnaJC7 may increase the efficiency of protein folding, if overexpressed, it may prevent the completion of protein folding by Hsp90s [23]. Of particular interest, HSP90 has previously demonstrated an ability to bind to TDP-43 and contribute to its clearance; however, the clearance is inhibited when aberrant tau accumulates in the cytosol [69,70,71]. Recently, DnaJC7 was found to bind and stabilize natively folded conformations of tau [21]. While it remains unknown whether this function is compromised by ALS-associated DNAJC7 variants, we hypothesize that this may be a potential functional mechanism of the variants, such that mutant DnaJC7 results in tau accumulation and downstream inhibition of TDP-43 clearance.
DnaJC7 also demonstrates physical interactions with non-Hsp70/Hsp90 chaperones, including with the other J proteins DnaJB1 and DnaJB2, although the exact mechanism and function of the interactions remain unclear. Of note, overexpression of both DnaJBs have shown protective effects in models of ALS. Specifically, overexpression of DnaJB1, and its yeast homolog Sis1, reduced TDP-43-mediated toxicity including reduced effects on cell growth, cell shape, and ubiquitin–proteasome system inhibition [72]. Similarly, overexpression of DnaJB2 improved outcomes of mutant SOD1 in in vivo ALS models including improvements in muscle performance and motor neuron survival [20]. DnaJC7 also interacts with a selection of other proteins including RAD1, RAD9A, and STUB1. Together, RAD1 and RAD9A form a complex responsible for cell cycle checkpoint signaling [73] and were both found to bind to DnaJC7 via its TPR domain; however, new studies suggest that the J-domain has a role in the regulation of the interaction with RAD9 including influencing complex formation and localization [74]. Finally, similarly to DnaJC7, STUB1 is an Hsp90 co-chaperone protein involved in protein quality control, and it contains multiple TPR domains. Although there is not a thorough understanding of this interaction, in complex with Hsp70, STUB1 has displayed neuroprotective effects by modulating the degradation of mutant SOD1 [75]. The interaction and similarities between DnaJC7 and STUB1 suggest further investigation regarding DnaJC7’s involvement in the SOD1 pathway is warranted in the context of fALS caused by mutations in SOD1.
The rather striking number of associations between DnaJC7 interactors and ALS provides promise that interrogating these interactions further may allow for greater understanding of its general role in cellular protein quality control and the pathological mechanism underlying the association of DNAJC7 with ALS.

8. Possible Role of Pathogenic DNAJC7 Variants in ALS

It is plausible that some ALS-associated mutations in DNAJC7 disrupt or even completely abolish the chaperone function of DnaJC7, supporting a loss-of-function mechanism. In the analyses presented by Farhan et al., immunoblot assays of DnaJC7 from a fibroblast sample of an ALS patient carrying a protein-truncating variant (p.R156X) identified that the variant resulted in significantly reduced protein levels [3]. Yet, further mechanistic studies are warranted to provide conclusive evidence.
It remains unclear why the variants in DNAJC7 result in ALS pathogenesis, i.e., mostly affecting motor neurons and no other neurons or non-neuronal tissues. Ablation of DnaJC7 expression in mice produces viable animals that seem to develop normally, indicating non-essential functions of DnaJC7 at least under normal conditions [76]. Further, recent evidence suggests DnaJC7 binds and stabilizes natively folded tau, the dysmetabolism of which is observed in frontotemporal spectrum disorder of ALS (ALS-FTSD) [77]. We propose that DnaJC7 becomes essential under specific stress conditions, such as oxidative stress, protein misfolding stress, and/or the diminished function of protein quality control in aged cells, specifically in motor neurons. Additionally, misfolded proteins commonly associated with ALS, such as TDP-43, FUS, and SOD1, might require particularly diligent chaperoning by Hsp70 and Hsp90, which may be regulated by DnaJC7. Finally, there is previous evidence that the homolog of DNAJC7 in Drosophila may suppress polyglutamine-induced toxicity [78,79]. Although these studies were not directly in relation to ATXN2, the gene known to carry CAG-repeat risk factors for ALS, further investigation into this mechanism and the ATXN2 polyglutamine status of DNAJC7 variant carriers may prove beneficial.

9. Conclusions

In this review article, we summarized the evolutionary conservation, expression profile, and proposed function in cellular protein quality control of the J protein, DnaJC7, and discussed how potentially pathogenic variants in DNAJC7 can contribute to neurodegeneration in ALS, possibly via a loss-of-function mechanism. Based on the collected evidence, it seems that DNAJC7 regulation may be controlled in response to cell stress via the HSR under conditions that are yet to be defined. However, when mutated, we hypothesize that there may be dysregulation of downstream pathways. These may include the dysregulation of tau, potentially resulting in the accumulation of TDP-43 through the inhibition of proper HSP90AB1 function, destabilization of interactions with other J proteins involved in neuroprotection, or the inability to accurately chaperone the variety of Hsp70s or Hsp90s involved in ALS-associated protein aggregates; although, other potential mechanisms cannot be ruled out. Future studies are required to determine the functions of DnaJC7 under normal conditions and under cellular stress conditions, its specific clients, and how DnaJC7 interacts with and processes misfolded proteins, specifically those that characterize ALS.

Author Contributions

A.A.D., C.M.A., M.S., A.A.P. contributed different parts of the content. A.A.D. organized the manuscript, and A.A.D., S.M.K.F. and M.L.D. wrote the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

ALS Canada/Brain Canada Project grant to S.M.K.F. and M.L.D.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

All authors reviewed the final version of this review article and consented to its publication.

Data Availability Statement

Not applicable.

Conflicts of Interest

All authors declare that they have no conflict of interest.

References

  1. Grad, L.I.; Rouleau, G.A.; Ravits, J.; Cashman, N.R. Clinical Spectrum of Amyotrophic Lateral Sclerosis (ALS). Cold Spring Harb. Perspect. Med. 2017, 7, a024117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Mejzini, R.; Flynn, L.L.; Pitout, I.L.; Fletcher, S.; Wilton, S.D.; Akkari, P.A. ALS Genetics, Mechanisms, and Therapeutics: Where Are We Now? Front. Neurosci. 2019, 13, 1310. [Google Scholar] [CrossRef] [Green Version]
  3. Farhan, S.M.K.; Howrigan, D.P.; Abbott, L.E.; Klim, J.R.; Topp, S.D.; Byrnes, A.E.; Churchhouse, C.; Phatnani, H.; Smith, B.N.; Rampersaud, E.; et al. Exome sequencing in amyotrophic lateral sclerosis implicates a novel gene, DNAJC7, encoding a heat-shock protein. Nat. Neurosci. 2019, 22, 1966–1974. [Google Scholar] [CrossRef] [PubMed]
  4. Ajit Tamadaddi, C.; Sahi, C. J domain independent functions of J proteins. Cell Stress Chaperones 2016, 21, 563–570. [Google Scholar] [CrossRef] [Green Version]
  5. Koutras, C.; Braun, J.E. J protein mutations and resulting proteostasis collapse. Front. Cell Neurosci. 2014, 8, 191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Greene, M.K.; Maskos, K.; Landry, S.J. Role of the J-domain in the cooperation of Hsp40 with Hsp70. Proc. Natl. Acad. Sci. USA 1998, 95, 6108–6113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Sahi, C.; Kominek, J.; Ziegelhoffer, T.; Yu, H.Y.; Baranowski, M.; Marszalek, J.; Craig, E.A. Sequential duplications of an ancient member of the DnaJ-family expanded the functional chaperone network in the eukaryotic cytosol. Mol. Biol. Evol. 2013, 30, 985–998. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Pellecchia, M.; Szyperski, T.; Wall, D.; Georgopoulos, C.; Wuthrich, K. NMR structure of the J-domain and the Gly/Phe-rich region of the Escherichia coli DnaJ chaperone. J. Mol. Biol. 1996, 260, 236–250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Liberek, K.; Marszalek, J.; Ang, D.; Georgopoulos, C.; Zylicz, M. Escherichia coli DnaJ and GrpE heat shock proteins jointly stimulate ATPase activity of DnaK. Proc. Natl. Acad. Sci. USA 1991, 88, 2874–2878. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Acebron, S.P.; Fernandez-Saiz, V.; Taneva, S.G.; Moro, F.; Muga, A. DnaJ recruits DnaK to protein aggregates. J. Biol. Chem. 2008, 283, 1381–1390. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Kravats, A.N.; Doyle, S.M.; Hoskins, J.R.; Genest, O.; Doody, E.; Wickner, S. Interaction of E. coli Hsp90 with DnaK Involves the DnaJ Binding Region of DnaK. J. Mol. Biol. 2017, 429, 858–872. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Wall, D.; Zylicz, M.; Georgopoulos, C. The NH2-terminal 108 amino acids of the Escherichia coli DnaJ protein stimulate the ATPase activity of DnaK and are sufficient for lambda replication. J. Biol. Chem. 1994, 269, 5446–5451. [Google Scholar] [CrossRef]
  13. Kampinga, H.H.; Craig, E.A. The HSP70 chaperone machinery: J proteins as drivers of functional specificity. Nat. Rev. Mol. Cell Biol. 2010, 11, 579–592. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Cheetham, M.E.; Caplan, A.J. Structure, function and evolution of DnaJ: Conservation and adaptation of chaperone function. Cell Stress Chaperones 1998, 3, 28–36. [Google Scholar] [CrossRef]
  15. Fan, C.Y.; Lee, S.; Ren, H.Y.; Cyr, D.M. Exchangeable chaperone modules contribute to specification of type I and type II Hsp40 cellular function. Mol. Biol. Cell 2004, 15, 761–773. [Google Scholar] [CrossRef] [Green Version]
  16. Lu, Z.; Cyr, D.M. The conserved carboxyl terminus and zinc finger-like domain of the co-chaperone Ydj1 assist Hsp70 in protein folding. J. Biol. Chem. 1998, 273, 5970–5978. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Hageman, J.; Rujano, M.A.; van Waarde, M.A.; Kakkar, V.; Dirks, R.P.; Govorukhina, N.; Oosterveld-Hut, H.M.; Lubsen, N.H.; Kampinga, H.H. A DNAJB chaperone subfamily with HDAC-dependent activities suppresses toxic protein aggregation. Mol. Cell 2010, 37, 355–369. [Google Scholar] [CrossRef]
  18. Walsh, P.; Bursac, D.; Law, Y.C.; Cyr, D.; Lithgow, T. The J-protein family: Modulating protein assembly, disassembly and translocation. EMBO Rep. 2004, 5, 567–571. [Google Scholar] [CrossRef] [Green Version]
  19. Aprile, F.A.; Kallstig, E.; Limorenko, G.; Vendruscolo, M.; Ron, D.; Hansen, C. The molecular chaperones DNAJB6 and Hsp70 cooperate to suppress alpha-synuclein aggregation. Sci. Rep. 2017, 7, 9039. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Novoselov, S.S.; Mustill, W.J.; Gray, A.L.; Dick, J.R.; Kanuga, N.; Kalmar, B.; Greensmith, L.; Cheetham, M.E. Molecular chaperone mediated late-stage neuroprotection in the SOD1(G93A) mouse model of amyotrophic lateral sclerosis. PLoS ONE 2013, 8, e73944. [Google Scholar] [CrossRef] [Green Version]
  21. Hou, Z.; Wydorski, P.M.; Perez, V.A.; Mendoza-Oliva, A.; Ryder, B.D.; Mirbaha, H.; Kashmer, O.; Joachimiak, L.A. DnaJC7 binds natively folded structural elements in tau to inhibit amyloid formation. Nat. Commun. 2021, 12, 5338. [Google Scholar] [CrossRef]
  22. Alvira, S.; Cuellar, J.; Rohl, A.; Yamamoto, S.; Itoh, H.; Alfonso, C.; Rivas, G.; Buchner, J.; Valpuesta, J.M. Structural characterization of the substrate transfer mechanism in Hsp70/Hsp90 folding machinery mediated by Hop. Nat. Commun. 2014, 5, 5484. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Brychzy, A.; Rein, T.; Winklhofer, K.F.; Hartl, F.U.; Young, J.C.; Obermann, W.M. Cofactor Tpr2 combines two TPR domains and a J domain to regulate the Hsp70/Hsp90 chaperone system. EMBO J. 2003, 22, 3613–3623. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Schulke, J.P.; Wochnik, G.M.; Lang-Rollin, I.; Gassen, N.C.; Knapp, R.T.; Berning, B.; Yassouridis, A.; Rein, T. Differential impact of tetratricopeptide repeat proteins on the steroid hormone receptors. PLoS ONE 2010, 5, e11717. [Google Scholar] [CrossRef]
  25. Ohno, M.; Kanayama, T.; Moore, R.; Ray, M.; Negishi, M. The roles of co-chaperone CCRP/DNAJC7 in Cyp2b10 gene activation and steatosis development in mouse livers. PLoS ONE 2014, 9, e115663. [Google Scholar] [CrossRef] [PubMed]
  26. Jih, K.Y.; Tsai, P.C.; Tsai, Y.S.; Liao, Y.C.; Lee, Y.C. Rapid progressive ALS in a patient with a DNAJC7 loss-of-function mutation. Neurol. Genet. 2020, 6, e503. [Google Scholar] [CrossRef] [PubMed]
  27. Wang, M.; Liu, Z.; Yuan, Y.; Ni, J.; Li, W.; Hu, Y.; Liu, P.; Hou, X.; Huang, L.; Jiao, B.; et al. A Novel Potentially Pathogenic Rare Variant in the DNAJC7 Gene Identified in Amyotrophic Lateral Sclerosis Patients From Mainland China. Front. Genet. 2020, 11, 821. [Google Scholar] [CrossRef]
  28. He, J.; Ma, X.; Yu, W.; Tang, L.; Fu, J.; Liu, X.; Ye, S.; Wan, M.; Fan, D. Validation of the pathogenic role of rare DNAJC7 variants in Chinese patients with amyotrophic lateral sclerosis. Neurobiol. Aging 2021, 106, 314.e1–314.e6. [Google Scholar] [CrossRef]
  29. Sun, X.; Zhao, X.; Liu, Q.; Zhang, K.; Liu, S.; Wang, Z.; Yang, X.; Shang, L.; Cui, L.; Zhang, X. Mutations of DNAJC7 are rare in Chinese amyotrophic lateral sclerosis patients. Amyotroph. Lateral Scler. Front. Degener. 2021, 22, 312–315. [Google Scholar] [CrossRef] [PubMed]
  30. Howe, K.L.; Achuthan, P.; Allen, J.; Allen, J.; Alvarez-Jarreta, J.; Amode, M.R.; Armean, I.M.; Azov, A.G.; Bennett, R.; Bhai, J.; et al. Ensembl 2021. Nucleic Acids Res. 2021, 49, D884–D891. [Google Scholar] [CrossRef]
  31. Schmid, A.B.; Lagleder, S.; Grawert, M.A.; Rohl, A.; Hagn, F.; Wandinger, S.K.; Cox, M.B.; Demmer, O.; Richter, K.; Groll, M.; et al. The architecture of functional modules in the Hsp90 co-chaperone Sti1/Hop. EMBO J. 2012, 31, 1506–1517. [Google Scholar] [CrossRef] [Green Version]
  32. Moffatt, N.S.; Bruinsma, E.; Uhl, C.; Obermann, W.M.; Toft, D. Role of the cochaperone Tpr2 in Hsp90 chaperoning. Biochemistry 2008, 47, 8203–8213. [Google Scholar] [CrossRef]
  33. Chen, H.J.; Mitchell, J.C.; Novoselov, S.; Miller, J.; Nishimura, A.L.; Scotter, E.L.; Vance, C.A.; Cheetham, M.E.; Shaw, C.E. The heat shock response plays an important role in TDP-43 clearance: Evidence for dysfunction in amyotrophic lateral sclerosis. Brain 2016, 139 Pt 5, 1417–1432. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Kieran, D.; Kalmar, B.; Dick, J.R.; Riddoch-Contreras, J.; Burnstock, G.; Greensmith, L. Treatment with arimoclomol, a coinducer of heat shock proteins, delays disease progression in ALS mice. Nat. Med. 2004, 10, 402–405. [Google Scholar] [CrossRef]
  35. Seminary, E.R.; Sison, S.L.; Ebert, A.D. Modeling Protein Aggregation and the Heat Shock Response in ALS iPSC-Derived Motor Neurons. Front. Neurosci. 2018, 12, 86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Anckar, J.; Sistonen, L. Regulation of HSF1 function in the heat stress response: Implications in aging and disease. Annu. Rev. Biochem. 2011, 80, 1089–1115. [Google Scholar] [CrossRef] [PubMed]
  37. Akerfelt, M.; Morimoto, R.I.; Sistonen, L. Heat shock factors: Integrators of cell stress, development and lifespan. Nat. Rev. Mol. Cell Biol. 2010, 11, 545–555. [Google Scholar] [CrossRef]
  38. Pelham, H.R. A regulatory upstream promoter element in the Drosophila hsp 70 heat-shock gene. Cell 1982, 30, 517–528. [Google Scholar] [CrossRef]
  39. Dreos, R.; Ambrosini, G.; Groux, R.; Cavin Perier, R.; Bucher, P. The eukaryotic promoter database in its 30th year: Focus on non-vertebrate organisms. Nucleic Acids Res. 2017, 45, D51–D55. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Khan, A.; Fornes, O.; Stigliani, A.; Gheorghe, M.; Castro-Mondragon, J.A.; van der Lee, R.; Bessy, A.; Cheneby, J.; Kulkarni, S.R.; Tan, G.; et al. JASPAR 2018: Update of the open-access database of transcription factor binding profiles and its web framework. Nucleic Acids Res. 2018, 46, D1284. [Google Scholar] [CrossRef] [PubMed]
  41. Lou, S.; Li, T.; Kong, X.; Zhang, J.; Liu, J.; Lee, D.; Gerstein, M. TopicNet: A framework for measuring transcriptional regulatory network change. Bioinformatics 2020, 36 (Suppl. 1), i474–i481. [Google Scholar] [CrossRef]
  42. Moore, J.E.; Pratt, H.E.; Purcaro, M.J.; Weng, Z. A curated benchmark of enhancer-gene interactions for evaluating enhancer-target gene prediction methods. Genome Biol. 2020, 21, 17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Mendillo, M.L.; Santagata, S.; Koeva, M.; Bell, G.W.; Hu, R.; Tamimi, R.M.; Fraenkel, E.; Ince, T.A.; Whitesell, L.; Lindquist, S. HSF1 drives a transcriptional program distinct from heat shock to support highly malignant human cancers. Cell 2012, 150, 549–562. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Mahat, D.B.; Salamanca, H.H.; Duarte, F.M.; Danko, C.G.; Lis, J.T. Mammalian Heat Shock Response and Mechanisms Underlying Its Genome-wide Transcriptional Regulation. Mol. Cell 2016, 62, 63–78. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Zhang, J.; Fan, N.; Peng, Y. Heat shock protein 70 promotes lipogenesis in HepG2 cells. Lipids Health Dis. 2018, 17, 73. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Qu, Z.; Titus, A.; Xuan, Z.; D’Mello, S.R. Neuroprotection by Heat Shock Factor-1 (HSF1) and Trimerization-Deficient Mutant Identifies Novel Alterations in Gene Expression. Sci. Rep. 2018, 8, 17255. [Google Scholar] [CrossRef] [PubMed]
  47. Barber, S.C.; Mead, R.J.; Shaw, P.J. Oxidative stress in ALS: A mechanism of neurodegeneration and a therapeutic target. Biochim. Biophys. Acta 2006, 1762, 1051–1067. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Ferrante, R.J.; Shinobu, L.A.; Schulz, J.B.; Matthews, R.T.; Thomas, C.E.; Kowall, N.W.; Gurney, M.E.; Beal, M.F. Increased 3-nitrotyrosine and oxidative damage in mice with a human copper/zinc superoxide dismutase mutation. Ann. Neurol. 1997, 42, 326–334. [Google Scholar] [CrossRef] [PubMed]
  49. Shaw, P.J.; Ince, P.G.; Falkous, G.; Mantle, D. Oxidative damage to protein in sporadic motor neuron disease spinal cord. Ann. Neurol. 1995, 38, 691–695. [Google Scholar] [CrossRef]
  50. Ma, Q. Role of nrf2 in oxidative stress and toxicity. Annu. Rev. Pharmacol. Toxicol. 2013, 53, 401–426. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Vomhof-Dekrey, E.E.; Picklo Sr, M.J. The Nrf2-antioxidant response element pathway: A target for regulating energy metabolism. J. Nutr. Biochem. 2012, 23, 1201–1206. [Google Scholar] [CrossRef] [PubMed]
  52. Chorley, B.N.; Campbell, M.R.; Wang, X.; Karaca, M.; Sambandan, D.; Bangura, F.; Xue, P.; Pi, J.; Kleeberger, S.R.; Bell, D.A. Identification of novel NRF2-regulated genes by ChIP-Seq: Influence on retinoid X receptor alpha. Nucleic Acids Res. 2012, 40, 7416–7429. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Malhotra, D.; Portales-Casamar, E.; Singh, A.; Srivastava, S.; Arenillas, D.; Happel, C.; Shyr, C.; Wakabayashi, N.; Kensler, T.W.; Wasserman, W.W.; et al. Global mapping of binding sites for Nrf2 identifies novel targets in cell survival response through ChIP-Seq profiling and network analysis. Nucleic Acids Res. 2010, 38, 5718–5734. [Google Scholar] [CrossRef] [PubMed]
  54. Namani, A.; Liu, K.; Wang, S.; Zhou, X.; Liao, Y.; Wang, H.; Wang, X.J.; Tang, X. Genome-wide global identification of NRF2 binding sites in A549 non-small cell lung cancer cells by ChIP-Seq reveals NRF2 regulation of genes involved in focal adhesion pathways. Aging 2019, 11, 12600–12623. [Google Scholar] [CrossRef] [PubMed]
  55. Babcock, D.T.; Shen, W.; Ganetzky, B. A neuroprotective function of NSF1 sustains autophagy and lysosomal trafficking in Drosophila. Genetics 2015, 199, 511–522. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Hertel, M.; Braun, S.; Durka, S.; Alzheimer, C.; Werner, S. Upregulation and activation of the Nrf-1 transcription factor in the lesioned hippocampus. Eur. J. Neurosci. 2002, 15, 1707–1711. [Google Scholar] [CrossRef] [PubMed]
  57. Lee, C.S.; Lee, C.; Hu, T.; Nguyen, J.M.; Zhang, J.; Martin, M.V.; Vawter, M.P.; Huang, E.J.; Chan, J.Y. Loss of nuclear factor E2-related factor 1 in the brain leads to dysregulation of proteasome gene expression and neurodegeneration. Proc. Natl. Acad. Sci. USA 2011, 108, 8408–8413. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Koizumi, S.; Hamazaki, J.; Murata, S. Transcriptional regulation of the 26S proteasome by Nrf1. Proc. Jpn. Acad. Ser. B Phys. Biol. Sci. 2018, 94, 325–336. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Wang, M.; Qiu, L.; Ru, X.; Song, Y.; Zhang, Y. Distinct isoforms of Nrf1 diversely regulate different subsets of its cognate target genes. Sci. Rep. 2019, 9, 2960. [Google Scholar] [CrossRef] [PubMed]
  60. Zhang, Y.; Li, S.; Xiang, Y.; Qiu, L.; Zhao, H.; Hayes, J.D. The selective post-translational processing of transcription factor Nrf1 yields distinct isoforms that dictate its ability to differentially regulate gene expression. Sci. Rep. 2015, 5, 12983. [Google Scholar] [CrossRef] [Green Version]
  61. Liu, P.; Kerins, M.J.; Tian, W.; Neupane, D.; Zhang, D.D.; Ooi, A. Differential and overlapping targets of the transcriptional regulators NRF1, NRF2, and NRF3 in human cells. J. Biol. Chem. 2019, 294, 18131–18149. [Google Scholar] [CrossRef] [PubMed]
  62. Satoh, J.; Kawana, N.; Yamamoto, Y. Pathway Analysis of ChIP-Seq-Based NRF1 Target Genes Suggests a Logical Hypothesis of their Involvement in the Pathogenesis of Neurodegenerative Diseases. Gene Regul. Syst. Biol. 2013, 7, 139–152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Lin, P.Y.; Simon, S.M.; Koh, W.K.; Folorunso, O.; Umbaugh, C.S.; Pierce, A. Heat shock factor 1 over-expression protects against exposure of hydrophobic residues on mutant SOD1 and early mortality in a mouse model of amyotrophic lateral sclerosis. Mol. Neurodegener. 2013, 8, 43. [Google Scholar] [CrossRef] [Green Version]
  64. Blair, L.J.; Sabbagh, J.J.; Dickey, C.A. Targeting Hsp90 and its co-chaperones to treat Alzheimer’s disease. Expert. Opin. Ther. Targets 2014, 18, 1219–1232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Consortium, G.T. The Genotype-Tissue Expression (GTEx) project. Nat. Genet. 2013, 45, 580–585. [Google Scholar]
  66. Uhlen, M.; Fagerberg, L.; Hallstrom, B.M.; Lindskog, C.; Oksvold, P.; Mardinoglu, A.; Sivertsson, A.; Kampf, C.; Sjostedt, E.; Asplund, A.; et al. Proteomics. Tissue-based map of the human proteome. Science 2015, 347, 1260419. [Google Scholar] [CrossRef] [PubMed]
  67. Warde-Farley, D.; Donaldson, S.L.; Comes, O.; Zuberi, K.; Badrawi, R.; Chao, P.; Franz, M.; Grouios, C.; Kazi, F.; Lopes, C.T.; et al. The GeneMANIA prediction server: Biological network integration for gene prioritization and predicting gene function. Nucleic Acids Res. 2010, 38, W214–W220. [Google Scholar] [CrossRef] [PubMed]
  68. Coyne, A.N.; Lorenzini, I.; Chou, C.C.; Torvund, M.; Rogers, R.S.; Starr, A.; Zaepfel, B.L.; Levy, J.; Johannesmeyer, J.; Schwartz, J.C.; et al. Post-transcriptional Inhibition of Hsc70-4/HSPA8 Expression Leads to Synaptic Vesicle Cycling Defects in Multiple Models of ALS. Cell Rep. 2017, 21, 110–125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Jinwal, U.K.; Abisambra, J.F.; Zhang, J.; Dharia, S.; O’Leary, J.C.; Patel, T.; Braswell, K.; Jani, T.; Gestwicki, J.E.; Dickey, C.A. Cdc37/Hsp90 protein complex disruption triggers an autophagic clearance cascade for TDP-43 protein. J. Biol. Chem. 2012, 287, 24814–24820. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Lin, L.T.; Razzaq, A.; Di Gregorio, S.E.; Hong, S.; Charles, B.; Lopes, M.H.; Beraldo, F.; Prado, V.F.; Prado, M.A.M.; Duennwald, M.L. Hsp90 and its co-chaperone Sti1 control TDP-43 misfolding and toxicity. FASEB J. 2021, 35, e21594. [Google Scholar] [CrossRef] [PubMed]
  71. Zhang, Y.J.; Gendron, T.F.; Xu, Y.F.; Ko, L.W.; Yen, S.H.; Petrucelli, L. Phosphorylation regulates proteasomal-mediated degradation and solubility of TAR DNA binding protein-43 C-terminal fragments. Mol. Neurodegener. 2010, 5, 33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Park, S.K.; Hong, J.Y.; Arslan, F.; Kanneganti, V.; Patel, B.; Tietsort, A.; Tank, E.M.H.; Li, X.; Barmada, S.J.; Liebman, S.W. Overexpression of the essential Sis1 chaperone reduces TDP-43 effects on toxicity and proteolysis. PLoS Genet. 2017, 13, e1006805. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Parrilla-Castellar, E.R.; Arlander, S.J.; Karnitz, L. Dial 9-1-1 for DNA damage: The Rad9-Hus1-Rad1 (9-1-1) clamp complex. DNA Repair. 2004, 3, 1009–1014. [Google Scholar] [CrossRef] [PubMed]
  74. Xiang, S.L.; Kumano, T.; Iwasaki, S.I.; Sun, X.; Yoshioka, K.; Yamamoto, K.C. The J domain of Tpr2 regulates its interaction with the proapoptotic and cell-cycle checkpoint protein, Rad9. Biochem. Biophys. Res. Commun. 2001, 287, 932–940. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Urushitani, M.; Kurisu, J.; Tateno, M.; Hatakeyama, S.; Nakayama, K.; Kato, S.; Takahashi, R. CHIP promotes proteasomal degradation of familial ALS-linked mutant SOD1 by ubiquitinating Hsp/Hsc70. J. Neurochem. 2004, 90, 231–244. [Google Scholar] [CrossRef] [PubMed]
  76. Ohno, M.; Moore, R.; Myers, P.; Negishi, M. Co-Chaperone-Mediated Suppression of LPS-Induced Cardiac Toxicity Through NFkappaB Signaling. Shock 2018, 50, 248–254. [Google Scholar] [CrossRef] [PubMed]
  77. Strong, M.J.; Donison, N.S.; Volkening, K. Alterations in Tau Metabolism in ALS and ALS-FTSD. Front. Neurol. 2020, 11, 598907. [Google Scholar] [CrossRef] [PubMed]
  78. Kazemi-Esfarjani, P.; Benzer, S. Genetic suppression of polyglutamine toxicity in Drosophila. Science 2000, 287, 1837–1840. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Shieh, S.Y.; Bonini, N.M. Genes and pathways affected by CAG-repeat RNA-based toxicity in Drosophila. Hum. Mol. Genet. 2011, 20, 4810–4821. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Protein schematic of the previously reported likely damaging variants in DNAJC7 in various ALS cohorts. Variants were previously reported to be associated with ALS by Farhan et al.; Jih et al.; Wang et al.; He et al.; Sun et al. [3,26,27,28,29]. Exon boundaries are indicated by black vertical, dashed lines.
Figure 1. Protein schematic of the previously reported likely damaging variants in DNAJC7 in various ALS cohorts. Variants were previously reported to be associated with ALS by Farhan et al.; Jih et al.; Wang et al.; He et al.; Sun et al. [3,26,27,28,29]. Exon boundaries are indicated by black vertical, dashed lines.
Ijms 23 04076 g001
Figure 2. Phylogenetic tree of human DNAJC7 including protein sequence alignment. The single gene split event at P. reticulata is not shown. The phylogenetic tree was built using the Gene Tree application within the current release of Ensembl [30].
Figure 2. Phylogenetic tree of human DNAJC7 including protein sequence alignment. The single gene split event at P. reticulata is not shown. The phylogenetic tree was built using the Gene Tree application within the current release of Ensembl [30].
Ijms 23 04076 g002
Figure 3. Expression of various J protein-, Hsp70-, and Hsp90-encoding genes throughout the human central nervous system (CNS). Gene expression data were obtained from Genotype-Tissue Expression (GTEx) database (https://gtexportal.org/home/ accessed on 12 March 2022) [65]. (A) Expression levels of various J protein-, Hsp70-, and Hsp90-encoding genes throughout the human CNS. The tree along the left y-axis demonstrates the relative difference between the various genes’ overall CNS expression profiles; (B) violin plots comparing the expression levels of DNAJC7 to a sample of Hsp70- and Hsp90-encoding genes.
Figure 3. Expression of various J protein-, Hsp70-, and Hsp90-encoding genes throughout the human central nervous system (CNS). Gene expression data were obtained from Genotype-Tissue Expression (GTEx) database (https://gtexportal.org/home/ accessed on 12 March 2022) [65]. (A) Expression levels of various J protein-, Hsp70-, and Hsp90-encoding genes throughout the human CNS. The tree along the left y-axis demonstrates the relative difference between the various genes’ overall CNS expression profiles; (B) violin plots comparing the expression levels of DNAJC7 to a sample of Hsp70- and Hsp90-encoding genes.
Ijms 23 04076 g003
Figure 4. Map of known physical interactions between a selection of J proteins, Hsp70s, Hsp90s, and other relevant co-factors. The interaction map was created using the GeneMANIA prediction server (https://genemania.org/ accessed on 12 March 2022) [67] by inputting a selection of 20 J proteins, two Hsp70s, and three Hsp90s. Using this selection, the prediction server was then able to select other relevant interactors: (A) all physical interactions between the J proteins, Hsp70s, Hsp90s, and other relevant cofactors; (B) proteins known to physically interact with DnaJC7.
Figure 4. Map of known physical interactions between a selection of J proteins, Hsp70s, Hsp90s, and other relevant co-factors. The interaction map was created using the GeneMANIA prediction server (https://genemania.org/ accessed on 12 March 2022) [67] by inputting a selection of 20 J proteins, two Hsp70s, and three Hsp90s. Using this selection, the prediction server was then able to select other relevant interactors: (A) all physical interactions between the J proteins, Hsp70s, Hsp90s, and other relevant cofactors; (B) proteins known to physically interact with DnaJC7.
Ijms 23 04076 g004
Table 1. Number of variants in J proteins, otherwise referred to as Hsp40s, previously associated with neurological phenotypes and/or phenotypes presenting with neurological features.
Table 1. Number of variants in J proteins, otherwise referred to as Hsp40s, previously associated with neurological phenotypes and/or phenotypes presenting with neurological features.
GeneDiseaseClinVar PathogenicClinVar Likely Pathogenic
MissenseLoFMissenseLoF
DNAJA3Developmental delay and polyneuropathyNANANANA
DNAJB2Charcot–Marie–Tooth disease; distal spinal muscular atrophyNA724
DNAJB5Peripheral neuropathy; skeletal myopathy; peripheral neuropathyNANA1NA
DNAJB6Limb–girdle muscular dystrophy, type 1E; frontotemporal dementia814NA
DNAJC3Combined cerebellar and peripheral ataxia with hearing loss and diabetes mellitusNA3NA1
DNAJC5Neuronal ceroid lipofuscinosis12NANA
DNAJC6Juvenile-onset Parkinson’s disease 19a29NANA
DNAJC7Amyotrophic lateral sclerosisNANANANA
DNAJC12Mild hyperphenylalaninemia, non-bh4-deficient; early-onset parkinsonism210NA2
DNAJC13Late-onset Parkinson’s disease; essential tremor1NANANA
DNAJC16Hereditary spastic paraplegiaNANANANA
DNAJC193-Methylglutaconic aciduria type VNA412
DNAJC21Bone marrow failure syndrome 3; tongue abnormality, acute myeloid leukemia, cognitive impairment, pancytopenia, pectus excavatum, short stature, and webbed neck 28NANA
DNAJC28Delayed speech and language, generalized hypotonia, intellectual disability, seizures, and optic atrophyNANANANA
DNAJC30Leber hereditary optic neuropathy3NA1NA
GAKParkinson’s diseaseNANANANA
SACSSpastic ataxia of Charlevoix–Saguenay; Spastic paraplegia1815517188
Previous disease associations were based on reports of variant pathogenicity in the ClinVar database. NA: no variants were reported with the pathogenicity classification in ClinVar; LoF: putative loss-of-function variants, including essential splice site, frameshift, stop gain, and stop loss variants.
Table 2. Likely damaging variants previously identified in DNAJC7 in multiple ALS cohorts.
Table 2. Likely damaging variants previously identified in DNAJC7 in multiple ALS cohorts.
cDNA ChangeProtein ChangeVariant TypeALS Cases (N)GnomAD (Non-Neuro v2.1.1) MAFIn Silico Prediction (CADD)Reference
c.22G > Cp.D8HMissense1 (5095)0.000019825Farhan et al., 2019 [3]
c.97G > Tp.E33XStop gain1 (5095)039Farhan et al., 2019 [3]
c.358C > Tp.Q120XStop gain1 (5095)037Farhan et al., 2019 [3]
c.401_402delAAp.Q134Rfs*6Truncating frameshift1 (325)031Jih et al., 2020 [26]
c.410A > Gp.K137RMissense1 (326)023Sun et al., 2021 [29]
c.466C > Tp.R156XStop gain2 (5095)041Farhan et al., 2019 [3]
c.467G > Ap.R156QMissense1 (701)0.000014623He et al., 2021 [28]
c.488delTp.F163fs*17Frameshift1 (5095)0NAFarhan et al., 2019 [3]
c.631G > Ap.D211NMissense1 (5095)026Farhan et al., 2019 [3]
c.646C > Tp.R216XStop gain2 (5095)040Farhan et al., 2019 [3]
c. 712A > Gp.R238GMissense1 (578)024Wang et al., 2020 [27]
c.754-3T > CNAEssential splice site1 (701)0.000024415He et al., 2021 [28]
c.1011-2A > GNAEssential splice site1 (5095)026Farhan et al., 2019 [3]
c.1012T > Ap.Y338NMissense1 (701)028He et al., 2021 [28]
c.1234C > Tp.R412WMissense1 (5095)0.000004034Farhan et al., 2019 [3]
c.1273G > Ap.E425KMissense2 (5095)035Farhan et al., 2019 [3]
ALS, amyotrophic lateral sclerosis; CADD, combined annotation-dependent depletion; MAF, minor allele frequency; N, total cohort size; NA, not applicable.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Dilliott, A.A.; Andary, C.M.; Stoltz, M.; Petropavlovskiy, A.A.; Farhan, S.M.K.; Duennwald, M.L. DnaJC7 in Amyotrophic Lateral Sclerosis. Int. J. Mol. Sci. 2022, 23, 4076. https://doi.org/10.3390/ijms23084076

AMA Style

Dilliott AA, Andary CM, Stoltz M, Petropavlovskiy AA, Farhan SMK, Duennwald ML. DnaJC7 in Amyotrophic Lateral Sclerosis. International Journal of Molecular Sciences. 2022; 23(8):4076. https://doi.org/10.3390/ijms23084076

Chicago/Turabian Style

Dilliott, Allison A., Catherine M. Andary, Meaghan Stoltz, Andrey A. Petropavlovskiy, Sali M. K. Farhan, and Martin L. Duennwald. 2022. "DnaJC7 in Amyotrophic Lateral Sclerosis" International Journal of Molecular Sciences 23, no. 8: 4076. https://doi.org/10.3390/ijms23084076

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

Article Metrics

Back to TopTop