Next Article in Journal
Drought Resistance in Rice from Conventional to Molecular Breeding: A Review
Next Article in Special Issue
GRP94 Is Involved in the Lipid Phenotype of Brain Metastatic Cells
Previous Article in Journal
Flavonoids from Chionanthus retusus (Oleaceae) Flowers and Their Protective Effects against Glutamate-Induced Cell Toxicity in HT22 Cells
Previous Article in Special Issue
mRNA Engineering for the Efficient Chaperone-Mediated Co-Translational Folding of Recombinant Proteins in Escherichia coli
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

14-3-3 Proteins Are on the Crossroads of Cancer, Aging, and Age-Related Neurodegenerative Disease

1
Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu 611130, China
2
Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
*
Author to whom correspondence should be addressed.
These authors have contributed equally to this work.
Int. J. Mol. Sci. 2019, 20(14), 3518; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms20143518
Submission received: 18 June 2019 / Revised: 15 July 2019 / Accepted: 16 July 2019 / Published: 18 July 2019
(This article belongs to the Special Issue Molecular Chaperones 2.0)

Abstract

:
14-3-3 proteins are a family of conserved regulatory adaptor molecules which are expressed in all eukaryotic cells. These proteins participate in a variety of intracellular processes by recognizing specific phosphorylation motifs and interacting with hundreds of target proteins. Also, 14-3-3 proteins act as molecular chaperones, preventing the aggregation of unfolded proteins under conditions of cellular stress. Furthermore, 14-3-3 proteins have been shown to have similar expression patterns in tumors, aging, and neurodegenerative diseases. Therefore, we put forward the idea that the adaptor activity and chaperone-like activity of 14-3-3 proteins might play a substantial role in the above-mentioned conditions. Interestingly, 14-3-3 proteins are considered to be standing at the crossroads of cancer, aging, and age-related neurodegenerative diseases. There are great possibilities to improve the above-mentioned diseases and conditions through intervention in the activity of the 14-3-3 protein family.

1. Introduction

Aging is followed by a gradual decline in the functions of multiple organ systems and an increase in the incidence of chronic diseases such as cancer [1], Type 2 diabetes, and Alzheimer’s disease (AD) [2]. Aging is also associated with considerable alterations in internal homeostasis, especially in the immune and endocrine systems, which play a significant role in cancer control. Therefore, aging and carcinogenesis are coupled to each other at the molecular level [3]. By analyzing the transcriptomic data set covering 531 samples at five different time points of aging, Peer et al. found that the aging-associated changes in transcriptomic expression and the transformation characteristics of chronic degenerative diseases (cardiovascular, metabolic, and neurodegenerative diseases) are related to each other and are different from the gene expression characteristics associated with cancer [4]. It seems that aging, chronic degenerative diseases, and tumors affect the human body via different directions. Interestingly, aging and cancer also share similar expression characteristics, for example, the genomic stability during aging and cancer follows a similar pattern at the transcriptome level [5,6] and therapeutic interventions for one of these might allow dual benefits of anti-aging as well as cancer prevention. These strategies include, but are not limited to, caloric restriction [7], drug senolytics [8,9], and so on. Long-term caloric restriction has been shown to delay the development of aging-related diseases in rodents and primates, including cancer [10].
The 14-3-3 protein family is constituted by 28–33 kDa acidic proteins found in all eukaryotes [11]. The 14-3-3 proteins are phosphorylated serine/threonine binding proteins that bind to a variety of kinases, phosphatases, transmembrane receptors, and transcription factors. Hundreds of 14-3-3 ligands have been reported in the human proteome [12,13]. The 14-3-3 proteins are widely expressed, especially in the central nervous system (CNS), and plays a key role in development [14] and disease progression [15]. Playing a similar role as other domains in signaling networks, 14-3-3 proteins generally interact with proteins that are involved in one of the three major functions, i.e., regulation, localization, or catalysis. It is widely accepted that 14-3-3 proteins act in two ways: By acting as adaptors [16] and by displaying chaperone-like activity [17]. By interaction with its partners, 14-3-3 proteins regulate critical biological processes, such as cell proliferation, growth, and apoptosis [18,19]. In addition, 14-3-3 proteins are also involved in the regulation of various tumors [20], metabolic diseases [21], and neurodegenerative diseases [22,23]. The gist of this review article is that 14-3-3 proteins are consistently up- and down-regulated in tumors, aging, and neurodegenerative disease. This suggests that targeting 14-3-3 proteins with specific drug compounds may facilitate a common therapeutic approach against aging, neurodegenerative disease, and cancer.

2. The Structure of 14-3-3 Proteins

14-3-3 proteins are present in almost all eukaryotic cells [24]. There are seven human 14-3-3 members according to the amino acid sequences (Table 1), while two isoforms in yeast and up to 13 isoforms in plants have been observed [25].
It is well-accepted that most isoforms of the 14-3-3 proteins can form and function as both homodimers and heterodimers, with an exception of the 14-3-3σ isoform, which preferentially forms homodimers [56,57]. The crystal structures of all seven mammalian 14-3-3 isoforms are available, showing that homodimers or heterodimers of 14-3-3 proteins generally consist of 9 α-helices. Each monomer consists of a bundle of nine antiparallel helices (H1-H9) [58]. The 14-3-3 dimers form cup-shaped structures, with a large, negatively charged, central passage with a diameter of about 35 Å, a width of 35 Å, and a depth of 20 Å, containing two ligand-binding grooves [59,60]. These grooves include the side chains of Lys49, Arg56, Arg127, and Tyr128 (residue numbering corresponds to the isoform of 14-3-3). The monomeric subunits form a dimer through their N-terminal helices and the linkage of the salt bridge connects the dimer between the first two helices of one monomer and the fourth helix of the other monomer [61]. The dimers bind the target proteins by three consensus phosphopeptide sequences: Motif I (RSXpSXP), motif II (RX(Y/F)XpSXP), and motif III (pSX1-2–COOH), wherein pS represents a phosphorylated serine/threonine and X is any residue [62,63]. These phosphopeptide binding sites are present in both monomer units of 14-3-3, therefore this protein can bind to both phosphopeptides simultaneously; they can be from the same target protein, or two different target proteins [64]. Furthermore, 14-3-3 proteins serve as adaptors or linkers. Depending upon the phosphorylation state of their specific recognition partners, 14-3-3 proteins bind their targets in order to stabilize the structure, phosphorylate and control their targets at the degradation level [65,66,67], localize and distribute between the different cellular compartments [68], and ultimately modulate their own interactions with other proteins.
However, 14-3-3 proteins also have the chaperone-like activity, i.e. 14-3-3ζ has been reported to dissolve heat-aggregated citrate synthase in vitro and has also been shown to interact with the heat shock proteins (HSP), HSP70/HSP40 chaperone to promote its reactivation [67]. This chaperon-like activity of the 14-3-3 family proteins is very different from the well-characterized phosphorylation-dependent interaction of 14-3-3 with multiple target proteins. Neither the phospho-serine binding groove nor the flexible C-terminal extension have been proven to be necessary for 14-3-3 chaperone activity [69]. Regardless of the use of any model substrate, 14-3-3 monomeric forms generally have higher activity than the dimeric form [70]. Studies by Sluchanko et al. have shown that exposure of the dimer interface may play a role in 14-3-3 proteins’ molecular chaperone mechanism [71,72]. Joanna et al. reported that the N-terminal helices of 14-3-3zeta may also play a role in chaperone action, whereby a D21N mutation may provide the key to the chaperone activity [72]. To summarize, the mechanism of 14-3-3 anti-aggregation activity appears to be similar to the unrelated small heat shock proteins (sHsps) and is independent of ATP. The N-terminal portion of 14-3-3 contains a hydrophobic region and hides the intrinsic barrier that is critical for protein dimerization and appears to be important for the development of unfolded/misfolded proteins. Many different factors that promote dimerization enhance the chaperone-like activity of 14-3-3.

3. 14-3-3 Proteins Have Consistent Expression Patterns in Aging and Cancer

3.1. Cancer

Much work has been dedicated to understand the role of 14-3-3 proteins in cancer. Because of the lack of significant catalytic activity, the contribution of 14-3-3 proteins to cancer is primarily related to the regulation of oncoproteins and tumor suppressor proteins. The detailed information regarding the regulation of different types of cancers by 14-3-3 protein isoforms has been summarized in Table 1.
As shown in Table 1, the majority of 14-3-3 isoforms are elevated in almost all types of tumors, except the σ isoform, which is down-regulated in some cancer types. In breast, gastric, prostate, lung, and liver cancers, an association is seen with elevated levels of most 14-3-3 isoforms, whereas in leukemia, renal, and glioma cancers, only few specific isoforms have been reported to exhibit abnormal expression. There are a large number of reports on the regulatory mechanisms about the ζ and σ isoforms in tumors (see Table 1), therefore, in the following paragraphs, we will discuss their detailed regulatory mechanism in cancers.
The ζ isoform among the 14-3-3 protein family is the one with most abundant research reports in a multitude of cancers. The 14-3-3ζ isoform is highly expressed in a variety of cancers, including breast, ovarian, prostate, lung, and stomach cancers [26,73]. This high expression of 14-3-3ζ has been associated with (but not limited to) poor prognosis and resistance to these cancers [74]. 14-3-3 promotes survival of cancer cells through either binding to the p85 regulatory subunit of PI3K and activating Akt [75], or inactivating the tumor suppressor genes p53 and p21 [76].
The 14-3-3ζ isoform plays an important role in another important cancer signaling pathway, which is the Wnt5a/ROR1 signal transduction pathway, and promotes the migration and proliferation of chronic lymphocytic leukemia [77]. The 14-3-3ζ isoform likely functions via binding to the FOXO3a transcription factor and facilitating its transport to the cytoplasm, which in turn results in the enhanced proliferation of tongue cancer cells. In breast cancer cells, 14-3-3ζ brings forth contextual changes of Smad partners from p53 to Gli2 and therefore facilitates the switch from the tumor suppressive function of TGFβ to its metastasis-promoting activity [78].
The 14-3-3σ isoform attracts particular attention, which is considered to be a tumor suppressor protein whose down-regulation has been frequently detected in tumor specimens of many types of cancer. Also, 14-3-3σ was found to be a potent tumor suppressor involved in ErbB2-driven breast cancer initiation and metastasis [79]. There is evidence that correlates the low expression of 14-3-3σ to hypermethylation of the 14-3-3σ promoter, leading to gene silencing [80,81]. The promoter regions of 14-3-3σ gene displayed abnormal methylation in breast, lung, liver, ovarian, bladder, and prostate cancers [82,83,84,85,86]. Therefore, 14-3-3σ methylation can be used as a diagnostic indicator for these tumors [32]. 14-3-3σ has also been shown to be involved in the regulation of the energy metabolism of cancer cells. 14-3-3σ targets c-Myc for ubiquitination and proteasome-mediated degradation to suppress tumor metabolic reprogramming [87]. Recently, it has been shown that the 14-3-3ζ and 14-3-3σ isoforms play an opposite role in the regulation of tumor suppressor or metastasis-promoting functions of transforming growth factor beta (TGFβ) signaling during cancer [78,88]. In short, the 14-3-3 proteins in cancers mostly work as adaptors to bind their phosphorylated target proteins to regulate the occurrence, development, metastasis, and invasion of tumors.

3.2. Age-Related Neurodegenerative Disease

The 14-3-3 proteins exhibit chaperone-like activity, wherein they contain a nuclear localization sequence (NLS) through which they can transport target proteins to the nucleus [25]. The pathogenesis of certain neurological diseases, such as Alzheimer’s disease (AD), Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS), schizophrenia, and bipolar disorder involve misfolding and excessive aggregation of proteins. Because of their chaperone-like activity, 14-3-3 proteins may play a role in these disease states [89,90,91]. In fact, 14-3-3 proteins are highly expressed in the brain, accounting for about 1% of the total amount of soluble brain proteins [89]. Also, the 14-3-3 isoform-specific functional knock-out mice have shown some syndrome phenotypes. Multiple studies in 14-3-3 isoform-specific K/O mouse models, as summarized in Table 2, have been very helpful in understanding 14-3-3 isoform-specific functions in the brain. Looking at the chaperone-like activity of 14-3-3 proteins, it comes as no surprise to see their involvement in a number of neurological disorders.
The association of 14-3-3 proteins with neurodegenerative diseases is further strengthened by their presence in Lewy bodies (LBs) and neurofibrillary tangles (NFTs) of AD brain sections. Several 14-3-3 isoforms are able to interact with specific proteins involved either in PD, ASL, or AD (Figure 1).

3.2.1. Parkinson’s Disease

Most of the 14-3-3 proteins are capable of interacting with α-synuclein, which is a regulator of the mitogen-activated protein kinase (MAPK) pathway, and therefore play an important role in the synthesis of dopamine [101]. Connotations between 14-3-3 (β and ε isoforms) and α-synuclein occurs either in cytosolic or membrane fractions of rat brain homogenate [102]. In fact, 14-3-3 and α-synuclein can be obtained by co-immunoprecipitation in the mammalian brain [103]. The 14-3-3η isoform strongly affect the products and the kinetics of α-synuclein aggregation in vitro by binding to α-synuclein oligomers. Overexpression of the 14-3-3η isoform results in reduced α-synuclein toxicity in cellular models [104]. One possible mechanism for this could be that the 14-3-3 protein is sequestered by the interaction with α-synuclein, resulting in a loss of 14-3-3 function, which is involved in the pathogenesis of PD.
Besides the α-synuclein, the 14-3-3ζ isoform also binds and stimulates the activation of tyrosine hydroxylase (TH), the rate-limiting enzyme in the biosynthesis of catecholamine [105]. The 14-3-3η isoform interacts with parkin, which is an ubiquitin E3 ligase, leading to protein degradation. The 14-3-3η–parkin association leads to the suppression of ubiquitin-ligase activity of parkin, which is one of the causes of PD [106].
LRRK2 and phosphorylated FOXO3a are also the interacting partners of 14-3-3 proteins [107]. FOXO3a localizes in LBs and recently a hypothesis was proposed suggesting the formation of a complex, including FOXO3a, α-synuclein, and 14-3-3 proteins, which promotes cell survival [108].

3.2.2. Alzheimer’s Disease

Tau is a major microtubule-associated protein in neurons, which can bind and stabilize microtubules. Tau phosphorylation reduces its affinity for microtubules and it is reported that tau is hyperphosphorylated in AD [109]. 14-3-3 proteins have been detected in NFT of AD patients, with 14-3-3ζ being the most immuno-reactive [110,111]. Further study has demonstrated that 14-3-3ζ facilitates GSK3β-dependent phosphorylation of tau by enhancing the affinity of GSK3β for tau [112]. The 14-3-3ζ isoform also binds to δ-catenin [113], a brain protein first discovered in the interaction with presenilin 1 [114].
In addition to the evidence of specific interactions with proteins associated to neurodegenerative diseases, 14-3-3 proteins also exhibit protective effects on dopaminergic neurons [115]. Indeed, 14-3-3θ, γ, and ε isoforms reduce the cellular toxicity induced by neurotoxins, causing cell death in dopaminergic cells [23,116]. It has also been suggested that 14-3-3 proteins may be involved in the chelation and degradation of toxic oligomers and aggregates by promoting the formation of aggresomes [117]. Recently, 14-3-3 proteins recognized phosphorylated transcription factor EB (TFEB) and affected the autophagy, which is strongly correlated with neurodegenerative disease [118]. Thus, looking at the functions of 14-3-3 proteins in age-related neurodegenerative disease, the potential development of drugs to therapeutically target 14-3-3 protein-protein interactions (PPIs) could be a good approach for the treatment of these kinds of diseases.

4. Aging Process

The 14-3-3 proteins are shown to be involved in many metabolic and autophagy regulatory pathways, such as Insulin/insulin-like growth factor signalling (IIS), AMP-activated protein kinase (AMPK), mechanistic target of rapamycin (mTOR) and MAPK, and these pathways play direct roles in the aging process. Thus, the 14-3-3 protein family may play a role in regulation of aging. The first study to explore the involvement of the 14-3-3 protein family on the lifespan regulation was carried out on C. elegans. Wang et al. found that in lifespan regulation, 14-3-3 proteins were co-expressed with DAF-16 and SIR-2.1 and DAF-16/ Forkhead box O (FOXO) interacted physically with 14-3-3 proteins [119], suggesting that in C. elegans, the 14-3-3 protein regulate the lifespan by synergy with SIR-2.1 and DAF-16/FOXO. In another report, Berdichevsky et al. demonstrated that SIR-2.1 and 14-3-3 activated DAF16 and extended the life span in a stress-dependent pathway in C. elegans [120]. Also, 14-3-3 proteins promoted the life span by both FOXO/daf16-dependent and independent manners [121]. To sum up, the role of 14-3-3 proteins in lifespan regulation in C. elegans is mostly by interacting with FOXO/DAF16.
In Drosophila, there are two isoforms of proteins, ε and ζ. Nielsen and colleagues found that the mutations in 14-3-3ε resulted in increased stress-induced apoptosis, growth inhibition, and prolonged lifespan, which were associated with increased FOXO activity [122]. Both 14-3-3 protein isoforms regulated two interacting components of mTOR signaling in Drosophila and regulated the translation of tumor protein (Tctp) and Rheb GTPase during organ growth [123]. It is already well-known that FOXO and TOR are two proteins that participate extensively in the aging process [124,125]. Therefore, it can be stated that 14-3-3 proteins participate in the lifespan by regulating the activity of these longevity proteins in Drosophila.
The 14-3-3 proteins have also been reported to participate in the process of metabolic diseases, such as obesity and diabetes [126]. The 14-3-3 protein interaction partner, heart-isomerized phospho-fructose-2-kinase/fructose-2,6-bisphosphatase (PFK-2), is involved in gluconeogenesis and glycolysis [127]. 14-3-3β and 14-3-3γ have been reported to participate in human PPARγ2 transactivation and hepatic lipid metabolism [128]. 14-3-3ζ and 14-3-3γ have been reported to be elevated in visceral and subcutaneous adipose tissue of obese individuals [129]. 14-3-3ζ-overexpressing mice had significantly higher body weights and fat masses when fed a high fat diet [130]. Significant changes in RNA and protein levels of 14-3-3ζ, ε, θ, and η in a murine model of Type 1 diabetes mellitus (T1DM) were detected [131]. Thus, it can be put forth that 14-3-3 proteins contribute to the development of metabolic diseases.
In Saccharomyces cerevisiae, upon deleting the 14-3-3 protein, Bmh1 increased the stress response and prolonged the lifespan [132]. The isoform β negatively regulated the glioblastoma cells senescence via the ERK-SKP2-p27 pathway [133]. The 14-3-3η protein and the downstream MAPK were thought to be effective in age-related cardiac dysfunction [134]. Network analyses have shown that skin aging triggered significant downregulation of 14-3-3 sigma [135]. Therefore, it can be firmly stated that 14-3-3 proteins play a very important regulatory role during aging.

5. Conclusions and Challenges

The 14-3-3 protein family plays a major role in aging, cancer, and aging-related neurodegenerative disease. During these disease states, the majority of the 14-3-3 proteins are up-regulated. This means a careful reduction of 14-3-3 activity in these processes or diseases may be beneficial in alleviating the relevant phenotype. However, the activity of 14-3-3 in regulating tumors and neurological diseases is slightly different (Figure 2). In tumors, 14-3-3 proteins play the role of adaptors by regulating the phosphorylation of the target sites in order to regulate protein activity, proliferation, apoptosis, metastasis, and survival of tumor cells. All of this regulation is predominantly based on the presence of a special phosphopeptide-binding amphipathic groove and on the dimeric status of 14-3-3.
In neurological diseases, 14-3-3 proteins mostly exhibit chaperone-like activities to interact with the protein aggregates. This activity of 14-3-3 proteins prevent the aggregation of partially folded or misfolded proteins or pro-proteins, thereby protecting cells from the accumulation of potentially harmful oligomers of unfolded protein intermediates. Therefore, it represents an integral part of the overall cyto-protection system and this protection is mostly associated with the monomeric forms. Various factors (stress, drugs, aging, etc.; see Figure 2) can alter the balance between 14-3-3 protein dimers and their monomeric forms, thereby affecting their mode of action.
The members of the 14-3-3 protein family can bind hundreds of target proteins and perform essential roles in human development, health, and pathological processes. However, many challenges still exist around this protein family. As 14-3-3 proteins function as dimers and monomers, the 14-3-3 isoforms may have functional redundancy. Alterations in the specific isoform levels may thus have an indirect effect by changing the balance of the 14-3-3 proteins. Furthermore, due to the similarity of the 14-3-3 family protein structure, it is very challenging to specifically inhibit one isoform’s function. Strategies to effectively regulate 14-3-3 activity need to be developed in order to utilize them as therapeutic candidates.

Author Contributions

X.F. and M.Y. structured the text and content; X.F., L.C. and Y.Z. reviewed the literature and provided intellectual contributions; X.F. and W.S. generated the figures; X.F., U.G. and M.Y. wrote the manuscript. All of the authors approved the final version of the manuscript.

Funding

This work was supported by the National Natural Science Foundation of China (31771338, 81701392).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Jackaman, C.; Tomay, F.; Duong, L.; Abdol Razak, N.B.; Pixley, F.J.; Metharom, P.; Nelson, D.J. Aging and cancer: The role of macrophages and neutrophils. Ageing Res. Rev. 2017, 36, 105–116. [Google Scholar] [CrossRef] [PubMed]
  2. Niccoli, T.; Partridge, L. Ageing as a risk factor for disease. Curr. Biol. 2012, 22, R741–R752. [Google Scholar] [CrossRef] [PubMed]
  3. Bottazzi, B.; Riboli, E.; Mantovani, A. Aging, inflammation and cancer. Semin. Immunol. 2018, 40, 74–82. [Google Scholar] [CrossRef] [PubMed]
  4. Aramillo, P.; Schauble, S.; Esser, D.; Groth, M.; Frahm, C.; Priebe, S.; Baumgart, M.; Hartmann, N.; Marthandan, S.; Menzel, U.; et al. Transcriptomic alterations during ageing reflect the shift from cancer to degenerative diseases in the elderly. Nat. Commun. 2018, 9, 327. [Google Scholar] [CrossRef] [PubMed]
  5. Lee, J.S. Cellular senescence, aging, and age-related disease: Special issue of BMB Reports in 2019. BMB Rep. 2019, 52, 1–2. [Google Scholar] [CrossRef] [Green Version]
  6. Hartley, A.V.; Martin, M.; Lu, T. Aging: Cancer - an unlikely couple. Aging (Albany NY). 2017, 9, 1949–1950. [Google Scholar] [CrossRef]
  7. Meynet, O.; Ricci, J.E. Caloric restriction and cancer: Molecular mechanisms and clinical implications. Trends Mol. Med. 2014, 20, 419–427. [Google Scholar] [CrossRef]
  8. Zhu, Y.; Tchkonia, T.; Pirtskhalava, T.; Gower, A.C.; Ding, H.; Giorgadze, N.; Palmer, A.K.; Ikeno, Y.; Hubbard, G.B.; Lenburg, M.; et al. The Achilles’ heel of senescent cells: From transcriptome to senolytic drugs. Aging Cell. 2015, 14, 644–658. [Google Scholar] [CrossRef]
  9. Short, S.; Fielder, E.; Miwa, S.; von Zglinicki, T. Senolytics and senostatics as adjuvant tumour therapy. EBio Med. 2019, 41, 683–692. [Google Scholar] [Green Version]
  10. Aunan, J.R.; Cho, W.C.; Soreide, K. The Biology of Aging and Cancer: A Brief Overview of Shared and Divergent Molecular Hallmarks. Aging Dis. 2017, 8, 628–642. [Google Scholar] [CrossRef] [Green Version]
  11. van Heusden, G.P. 14-3-3 proteins: Regulators of numerous eukaryotic proteins. IUBMB Life. 2005, 57, 623–629. [Google Scholar] [CrossRef] [PubMed]
  12. Pozuelo-Rubio, M. Proteomic and biochemical analysis of 14-3-3-binding proteins during C2-ceramide-induced apoptosis. FEBS J. 2010, 277, 3321–3342. [Google Scholar] [CrossRef] [PubMed]
  13. Pozuelo Rubio, M.; Geraghty, K.M.; Wong, B.H.; Wood, N.T.; Campbell, D.G.; Morrice, N.; Mackintosh, C. 14-3-3-affinity purification of over 200 human phosphoproteins reveals new links to regulation of cellular metabolism, proliferation and trafficking. Biochem. J. 2004, 379, 395–408. [Google Scholar] [CrossRef] [PubMed]
  14. Cornell, B.; Toyo-Oka, K. 14-3-3 Proteins in Brain Development: Neurogenesis, Neuronal Migration and Neuromorphogenesis. Front Mol. Neurosci. 2017, 10, 318. [Google Scholar] [CrossRef] [PubMed]
  15. Morales, D.; Skoulakis, E.C.; Acevedo, S.F. 14-3-3s are potential biomarkers for HIV-related neurodegeneration. J. Neurovirol. 2012, 18, 341–353. [Google Scholar] [CrossRef] [Green Version]
  16. Kaplan, A.; Ottmann, C.; Fournier, A.E. 14-3-3 adaptor protein-protein interactions as therapeutic targets for CNS diseases. Pharmacol. Res. 2017, 125, 114–121. [Google Scholar] [CrossRef] [PubMed]
  17. Sluchanko, N.; Gusev, N.B. Moonlighting chaperone-like activity of the universal regulatory 14-3-3 proteins. FEBS J. 2017, 284, 1279–1295. [Google Scholar] [CrossRef] [PubMed]
  18. Aghazadeh, Y.; Papadopoulos, V. The role of the 14-3-3 protein family in health, disease, and drug development. Drug Discov. Today. 2016, 21, 278–287. [Google Scholar] [CrossRef]
  19. Morrison, D.K. The 14-3-3 proteins: Integrators of diverse signaling cues that impact cell fate and cancer development. Trends Cell Biol. 2009, 19, 16–23. [Google Scholar] [CrossRef]
  20. Tzivion, G.; Gupta, V.S.; Kaplun, L.; Balan, V. 14-3-3 proteins as potential oncogenes. Semin Cancer Biol. 2006, 16, 203–213. [Google Scholar] [CrossRef]
  21. Diallo, K.; Oppong, A.K.; Lim, G.E. Can 14-3-3 proteins serve as therapeutic targets for the treatment of metabolic diseases? Pharmacol Res. 2019, 139, 199–206. [Google Scholar] [CrossRef] [PubMed]
  22. Shimada, T.; Fournier, A.E.; Yamagata, K. Neuroprotective function of 14-3-3 proteins in neurodegeneration. BioMed Res. Int. 2013, 2013, 564534. [Google Scholar] [CrossRef]
  23. Yacoubian, T.A.; Slone, S.R.; Harrington, A.J.; Hamamichi, S.; Schieltz, J.M.; Caldwell, K.A.; Caldwell, G.A.; Standaert, D.G. Differential neuroprotective effects of 14-3-3 proteins in models of Parkinson’s disease. Cell Death Dis. 2010, 1, e2. [Google Scholar] [CrossRef] [PubMed]
  24. Hermeking, H. 14-3-3 proteins and cancer biology. Semin Cancer Biol. 2006, 16, 161. [Google Scholar] [CrossRef]
  25. Muslin, A.J.; Xing, H. 14-3-3 proteins: Regulation of subcellular localization by molecular interference. Cell Signal. 2000, 12, 703–709. [Google Scholar] [CrossRef]
  26. Matta, A.; Siu, K.W.; Ralhan, R. 14-3-3 zeta as novel molecular target for cancer therapy. Expert Opin. Ther. Targets 2012, 16, 515–523. [Google Scholar] [CrossRef] [PubMed]
  27. Macha, M.A.; Matta, A.; Chauhan, S.; Siu, K.M.; Ralhan, R. 14-3-3 zeta is a molecular target in guggulsterone induced apoptosis in head and neck cancer cells. BMC Cancer 2010, 10, 655. [Google Scholar] [CrossRef]
  28. Tang, Y.; Liu, S.; Li, N.; Guo, W.; Shi, J.; Yu, H.; Zhang, L.; Wang, K.; Liu, S.; Cheng, S. 14-3-3ζ promotes hepatocellular carcinoma venous metastasis by modulating hypoxia-inducible factor-1α. Oncotarget 2016, 7, 15854–15867. [Google Scholar] [PubMed]
  29. Nishimura, Y.; Komatsu, S.; Ichikawa, D.; Nagata, H.; Hirajima, S.; Takeshita, H.; Kawaguchi, T.; Arita, T.; Konishi, H.; Kashimoto, K.; et al. Overexpression of YWHAZ relates to tumor cell proliferation and malignant outcome of gastric carcinoma. Br. J. Cancer 2013, 108, 1324–1331. [Google Scholar] [CrossRef] [Green Version]
  30. He, Y.; Wu, X.; Liu, X.; Yan, G.; Xu, C. LC-MS/MS analysis of ovarian cancer metastasis-related proteins using a nude mouse model: 14-3-3 zeta as a candidate biomarker. J. Proteome Res. 2010, 9, 6180–6190. [Google Scholar] [CrossRef]
  31. Janssen, J.J.; Klaver, S.M.; Waisfisz, Q.; Pasterkamp, G.; de Kleijn, D.P.; Schuurhuis, G.J.; Ossenkoppele, G.J. Identification of genes potentially involved in disease transformation of CML. Leukemia 2005, 19, 998–1004. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Li, Z.; Liu, J.Y.; Zhang, J.T. 14-3-3sigma, the double-edged sword of human cancers. Am. J. Transl. Res. 2009, 1, 326–340. [Google Scholar] [PubMed]
  33. Zhou, R.; Shao, Z.; Liu, J.; Zhan, W.; Gao, Q.; Pan, Z.; Wu, L.; Xu, L.; Ding, Y.; Zhao, L. COPS5 and LASP1 synergistically interact to downregulate 14-3-3sigma expression and promote colorectal cancer progression via activating PI3K/AKT pathway. Int. J. Cancer 2018, 142, 1853–1864. [Google Scholar] [CrossRef]
  34. Peng, C.; Jia, X.; Xiong, Y.; Yin, J.; Li, N.; Deng, Y.; Luo, K.; Zhang, Q.; Wang, C.; Zhang, Z.; et al. The 14-3-3 sigma/GSK3beta/beta-catenin/ZEB1 regulatory loop modulates chemo-sensitivity in human tongue cancer. Oncotarget 2015, 6, 20177–20189. [Google Scholar] [CrossRef] [PubMed]
  35. Qi, Y.J.; Wang, M.; Liu, R.M.; Wei, H.; Chao, W.X.; Zhang, T.; Lou, Q.; Li, X.M.; Ma, J.; Zhu, H.; et al. Downregulation of 14-3-3sigma correlates with multistage carcinogenesis and poor prognosis of esophageal squamous cell carcinoma. PLoS ONE 2014, 9, e95386. [Google Scholar]
  36. Lodygin, D.; Yazdi, A.S.; Sander, C.A.; Herzinger, T.; Hermeking, H. Analysis of 14-3-3sigma expression in hyperproliferative skin diseases reveals selective loss associated with CpG-methylation in basal cell carcinoma. Oncogene 2003, 22, 5519–5524. [Google Scholar] [CrossRef] [PubMed]
  37. Neupane, D.; Korc, M. 14-3-3sigma Modulates pancreatic cancer cell survival and invasiveness. Clin. Cancer Res. 2008, 14, 7614–7623. [Google Scholar] [CrossRef]
  38. Liu, C.C.; Jan, Y.J.; Ko, B.S.; Wu, Y.M.; Liang, S.M.; Chen, S.C.; Lee, Y.M.; Liu, T.A.; Chang, T.C.; Wang, J.; et al. 14-3-3sigma induces heat shock protein 70 expression in hepatocellular carcinoma. BMC Cancer 2014, 14, 425. [Google Scholar] [CrossRef]
  39. Raungrut, P.; Wongkotsila, A.; Lirdprapamongkol, K.; Svasti, J.; Geater, S.L.; Phukaoloun, M.; Suwiwat, S.; Thongsuksai, P. Prognostic significance of 14-3-3gamma overexpression in advanced non-small cell lung cancer. Asian Pac. J. Cancer Prev. 2014, 15, 3513–3518. [Google Scholar] [CrossRef]
  40. Yang, X.; Cao, W.; Lin, H.; Zhang, W.; Lin, W.; Cao, L.; Zhen, H.; Huo, J.; Zhang, X. Isoform-specific expression of 14-3-3 proteins in human astrocytoma. J. Neurol. Sci. 2009, 276, 54–59. [Google Scholar] [CrossRef]
  41. Liang, S.; Shen, G.; Liu, Q.; Xu, Y.; Zhou, L.; Xiao, S.; Xu, Z.; Gong, F.; You, C.; Wei, Y. Isoform-specific expression and characterization of 14-3-3 proteins in human glioma tissues discovered by stable isotope labeling with amino acids in cell culture-based proteomic analysis. Proteomics Clin. Appl. 2009, 3, 743–753. [Google Scholar] [CrossRef] [PubMed]
  42. Wang, Z.; Nesland, J.M.; Suo, Z.; Trope, C.G.; Holm, R. The prognostic value of 14-3-3 isoforms in vulvar squamous cell carcinoma cases: 14-3-3beta and epsilon are independent prognostic factors for these tumors. PLoS ONE 2011, 6, e24843. [Google Scholar] [CrossRef] [PubMed]
  43. Liang, S.; Xu, Y.; Shen, G.; Liu, Q.; Zhao, X.; Xu, Z.; Xie, X.; Gong, F.; Li, R.; Wei, Y. Quantitative protein expression profiling of 14-3-3 isoforms in human renal carcinoma shows 14-3-3 epsilon is involved in limitedly increasing renal cell proliferation. Electrophoresis 2009, 30, 4152–4162. [Google Scholar] [CrossRef] [PubMed]
  44. Ko, B.S.; Chang, T.C.; Hsu, C.; Chen, Y.C.; Shen, T.L.; Chen, S.C.; Wang, J.; Wu, K.K.; Jan, Y.J.; Liou, J.Y. Overexpression of 14-3-3epsilon predicts tumour metastasis and poor survival in hepatocellular carcinoma. Histopathology 2011, 58, 705–711. [Google Scholar] [CrossRef] [PubMed]
  45. Qi, W.; Liu, X.; Qiao, D.; Martinez, J.D. Isoform-specific expression of 14-3-3 proteins in human lung cancer tissues. Int. J. Cancer 2005, 113, 359–363. [Google Scholar] [CrossRef] [PubMed]
  46. Gong, X.; Yan, L.; Gu, H.; Mu, Y.; Tong, G.; Zhang, G. 14-3-3ε functions as an oncogene in SGC7901 gastric cancer cells through involvement of cyclin E and p27kip1. Mol. Med. Rep. 2014, 10, 3145–3150. [Google Scholar] [CrossRef]
  47. Ko, B.S.; Lai, I.R.; Chang, T.C.; Liu, T.A.; Chen, S.C.; Wang, J.; Jan, Y.J.; Liou, J.Y. Involvement of 14-3-3γ overexpression in extrahepatic metastasis of hepatocellular carcinoma. Hum. Pathol. 2011, 42, 129–135. [Google Scholar] [CrossRef]
  48. Hiraoka, E.; Mimae, T.; Ito, M.; Kadoya, T.; Miyata, Y.; Ito, A.; Okada, M. Breast cancer cell motility is promoted by 14-3-3γ. Breast Cancer 2019. [Google Scholar] [CrossRef]
  49. Qi, W.; Liu, X.; Chen, W.; Li, Q.; Martinez, J.D. Overexpression of 14-3-3gamma causes polyploidization in H322 lung cancer cells. Mol. Carcinog. 2007, 46, 847–856. [Google Scholar] [CrossRef]
  50. Titus, M.A.; Tan, J.A.; Gregory, C.W.; Ford, O.H.; Subramanian, R.R.; Fu, H.; Wilson, E.M.; Mohler, J.L.; French, F.S. 14-3-3{eta} amplifies androgen receptor actions in prostate cancer. Clin. Cancer Res.: An Off. J. Am. Assoc. Cancer Res. 2009, 15, 7571–7581. [Google Scholar] [CrossRef]
  51. Li, X.H.; Noguchi, A.; Nishida, T.; Takahashi, H.; Zheng, Y.; Yang, X.H.; Masuda, S.; Kikuchi, K.; Takano, Y. Cytoplasmic expression of p33ING1b is correlated with tumorigenesis and progression of head and neck squamous cell carcinoma. Histol. Histopathol. 2011, 26, 597–607. [Google Scholar] [PubMed]
  52. Park, G.Y.; Han, J.Y.; Han, Y.K.; Kim, S.D.; Kim, J.S.; Jo, W.S.; Chun, S.H.; Jeong, D.H.; Lee, C.W.; Yang, K.; et al. 14-3-3 eta depletion sensitizes glioblastoma cells to irradiation due to enhanced mitotic cell death. Cancer Gene Ther. 2014, 21, 158–163. [Google Scholar] [CrossRef] [PubMed]
  53. Xiao, Y.; Lin, V.Y.; Ke, S.; Lin, G.E.; Lin, F.T.; Lin, W.C. 14-3-3tau promotes breast cancer invasion and metastasis by inhibiting RhoGDI alpha. Mol. Cell Biol. 2014, 34, 2635–2649. [Google Scholar] [CrossRef] [PubMed]
  54. Lee, T.G.; Jeong, E.H.; Kim, S.Y.; Kim, H.R.; Kim, H.; Kim, C.H. Fhit, a tumor suppressor protein, induces autophagy via 14-3-3tau in non-small cell lung cancer cells. Oncotarget 2017, 8, 31923–31937. [Google Scholar] [PubMed]
  55. Yan, Y.; Xu, Y.; Gao, Y.Y.; Zong, Z.H.; Zhang, Q.; Li, C.; Wang, H.Q. Implication of 14-3-3epsilon and 14-3-3theta/tau in proteasome inhibition-induced apoptosis of glioma cells. Cancer Sci. 2013, 104, 55–61. [Google Scholar] [CrossRef]
  56. Obsilova, V.; Silhan, J.; Boura, E.; Teisinger, J.; Obsil, T. 14-3-3 proteins: A family of versatile molecular regulators. Physiol Res. 2008, 57, S11–S21. [Google Scholar]
  57. Bridges, D.; Moorhead, G.B. 14-3-3 proteins: A number of functions for a numbered protein. Sci. STKE 2005, 2005, re10. [Google Scholar] [CrossRef]
  58. Wilker, E.W.; Grant, R.A.; Artim, S.C.; Yaffe, M.B. A structural basis for 14-3-3sigma functional specificity. J. Biol. Chem. 2005, 280, 18891–18898. [Google Scholar] [CrossRef]
  59. Stevers, L.M.; Lam, C.V.; Leysen, S.F.; Meijer, F.A.; Scheppingen, D.S.; Vries, R.M.; Carlile, G.W.; Milroy, L.G.; Thomas, D.Y.; Brunsveld, L.; et al. Characterization and small-molecule stabilization of the multisite tandem binding between 14-3-3 and the R domain of CFTR. Proc. Natl. Acad. Sci. USA 2016, 113, 1152–1161. [Google Scholar] [CrossRef]
  60. Yang, X.; Lee, W.H.; Sobott, F.; Papagrigoriou, E.; Robinson, C.V.; Grossmann, J.G.; Sundstrom, M.; Doyle, D.A.; Elkins, J.M. Structural basis for protein-protein interactions in the 14-3-3 protein family. Proc. Natl. Acad. Sci. USA 2006, 103, 17237–17242. [Google Scholar] [CrossRef]
  61. Gardino, A.K.; Smerdon, S.J.; Yaffe, M.B. Structural determinants of 14-3-3 binding specificities and regulation of subcellular localization of 14-3-3-ligand complexes: A comparison of the X-ray crystal structures of all human 14-3-3 isoforms. Semin. Cancer Biol. 2006, 16, 173–182. [Google Scholar] [CrossRef] [PubMed]
  62. Coblitz, B.; Wu, M.; Shikano, S.; Li, M. C-terminal binding: An expanded repertoire and function of 14-3-3 proteins. FEBS Lett. 2006, 580, 1531–1535. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Yaffe, M.B.; Rittinger, K.; Volinia, S.; Caron, P.R.; Aitken, A.; Leffers, H.; Gamblin, S.J.; Smerdon, S.J.; Cantley, L.C. The structural basis for 14-3-3: phosphopeptide binding specificity. Cell 1997, 91, 961–971. [Google Scholar] [CrossRef]
  64. Alblova, M.; Smidova, A.; Docekal, V.; Vesely, J.; Herman, P.; Obsilova, V.; Obsil, T. Molecular basis of the 14-3-3 protein-dependent activation of yeast neutral trehalase Nth1. Proc. Natl. Acad. Sci. USA 2017, 114, E9811–E9820. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Ganguly, S.; Weller, J.L.; Ho, A.; Chemineau, P.; Malpaux, B.; Klein, D.C. Melatonin synthesis: 14-3-3-dependent activation and inhibition of arylalkylamine N-acetyltransferase mediated by phosphoserine-205. Proc. Natl. Acad. Sci. USA 2005, 102, 1222–1227. [Google Scholar] [CrossRef] [PubMed]
  66. Demmel, L.; Beck, M.; Klose, C.; Schlaitz, A.L.; Gloor, Y.; Hsu, P.P.; Havlis, J.; Shevchenko, A.; Krause, E.; Kalaidzidis, Y.; et al. Nucleocytoplasmic shuttling of the Golgi phosphatidylinositol 4-kinase Pik1 is regulated by 14-3-3 proteins and coordinates Golgi function with cell growth. Mol. Biol. Cell. 2008, 19, 1046–1061. [Google Scholar] [CrossRef]
  67. Yano, M.; Nakamuta, S.; Wu, X.; Okumura, Y.; Kido, H. A novel function of 14-3-3 protein: 14-3-3zeta is a heat-shock-related molecular chaperone that dissolves thermal-aggregated proteins. Mol. Biol. Cell. 2006, 17, 4769–4779. [Google Scholar] [CrossRef] [PubMed]
  68. Williams, D.M.; Ecroyd, H.; Goodwin, K.L.; Dai, H.; Fu, H.; Woodcock, J.M.; Zhang, L.; Carver, J.A. NMR spectroscopy of 14-3-3zeta reveals a flexible C-terminal extension: Differentiation of the chaperone and phosphoserine-binding activities of 14-3-3zeta. Biochem. J. 2011, 437, 493–503. [Google Scholar] [CrossRef] [PubMed]
  69. Sluchanko, N.N.; Roman, S.G.; Chebotareva, N.A.; Gusev, N.B. Chaperone-like activity of monomeric human 14-3-3zeta on different protein substrates. Arch. Biochem. Biophys. 2014, 549, 32–39. [Google Scholar] [CrossRef]
  70. Sluchanko, N.N.; Artemova, N.V.; Sudnitsyna, M.V.; Safenkova, I.V.; Antson, A.A.; Levitsky, D.I.; Gusev, N.B. Monomeric 14-3-3zeta has a chaperone-like activity and is stabilized by phosphorylated HspB6. Biochemistry 2012, 51, 6127–6138. [Google Scholar] [CrossRef]
  71. Lin, J.P.; Fan, Y.K.; Liu, H.M. The 14-3-3eta chaperone protein promotes antiviral innate immunity via facilitating MDA5 oligomerization and intracellular redistribution. PLoS Pathog. 2019, 15, e1007582. [Google Scholar] [CrossRef] [PubMed]
  72. Woodcock, J.M.; Goodwin, K.L.; Sandow, J.J.; Coolen, C.; Perugini, M.A.; Webb, A.I.; Pitson, S.M.; Lopez, A.F.; Carver, J.A. Role of salt bridges in the dimer interface of 14-3-3zeta in dimer dynamics, N-terminal alpha-helical order, and molecular chaperone activity. J. Biol. Chem. 2018, 293, 89–99. [Google Scholar] [CrossRef] [PubMed]
  73. Neal, C.L.; Yu, D. 14-3-3zeta as a prognostic marker and therapeutic target for cancer. Expert Opin. Ther. Targets 2010, 14, 1343–1354. [Google Scholar] [CrossRef] [PubMed]
  74. Neal, C.L.; Yao, J.; Yang, W.; Zhou, X.; Nguyen, N.T.; Lu, J.; Danes, C.G.; Guo, H.; Lan, K.H.; Ensor, J.; et al. 14-3-3zeta overexpression defines high risk for breast cancer recurrence and promotes cancer cell survival. Cancer Res. 2009, 69, 3425–3432. [Google Scholar] [CrossRef] [PubMed]
  75. Neal, C.L.; Xu, J.; Li, P.; Mori, S.; Yang, J.; Neal, N.N.; Zhou, X.; Wyszomierski, S.L.; Yu, D. Overexpression of 14-3-3ζ in cancer cells activates PI3K via binding the p85 regulatory subunit. Oncogene 2012, 31, 897–906. [Google Scholar] [CrossRef]
  76. Lee, J.J.; Lee, J.S.; Cui, M.N.; Yun, H.H.; Kim, H.Y.; Lee, S.H.; Lee, J.H. BIS targeting induces cellular senescence through the regulation of 14-3-3 zeta/STAT3/SKP2/p27 in glioblastoma cells. Cell Death Dis. 2014, 5, e1537. [Google Scholar] [CrossRef]
  77. Yu, J.; Chen, L.; Chen, Y.; Hasan, M.K.; Ghia, E.M.; Zhang, L.; Wu, R.; Rassenti, L.Z.; Widhopf, G.F.; Shen, Z.; et al. Wnt5a induces ROR1 to associate with 14-3-3ζ for enhanced chemotaxis and proliferation of chronic lymphocytic leukemia cells. Leukemia 2017, 31, 2608–2614. [Google Scholar] [CrossRef]
  78. Xu, J.; Acharya, S.; Sahin, O.; Zhang, Q.; Saito, Y.; Yao, J.; Wang, H.; Li, P.; Zhang, L.; Lowery, F.J.; et al. 14-3-3ζ turns TGF-β’s function from tumor suppressor to metastasis promoter in breast cancer by contextual changes of Smad partners from p53 to Gli2. Cancer Cell 2015, 27, 177–192. [Google Scholar] [CrossRef]
  79. Ling, C.; Su, V.M.; Zuo, D.; Muller, W.J. Loss of the 14-3-3σ tumor suppressor is a critical event in ErbB2-mediated tumor progression. Cancer Discov. 2012, 2, 68–81. [Google Scholar] [CrossRef]
  80. Umbricht, C.B.; Evron, E.; Gabrielson, E.; Ferguson, A.; Marks, J.; Sukumar, S. Hypermethylation of 14-3-3 sigma (stratifin) is an early event in breast cancer. Oncogene 2001, 20, 3348–3353. [Google Scholar] [CrossRef]
  81. Vercoutter-Edouart, A.S.; Lemoine, J.; Le, X.; Bourhis; Louis, H.; Boilly, B.; Nurcombe, V.; Révillion, F.; Peyrat, J.P.; Hondermarck, H. Proteomic analysis reveals that 14-3-3sigma is down-regulated in human breast cancer cells. Cancer Res. 2001, 61, 76–80. [Google Scholar] [PubMed]
  82. Radhakrishnan, V.M.; Jensen, T.J.; Cui, H.; Futscher, B.W.; Martinez, J.D. Hypomethylation of the 14-3-3σ promoter leads to increased expression in non-small cell lung cancer. Genes Chromosomes Cancer 2011, 50, 830–836. [Google Scholar] [CrossRef]
  83. Luo, J.; Feng, J.; Lu, J.; Wang, Y.; Tang, X.; Xie, F.; Li, W. Aberrant methylation profile of 14-3-3 sigma and its reduced transcription/expression levels in Chinese sporadic female breast carcinogenesis. Med. Oncol. 2010, 27, 791–797. [Google Scholar] [CrossRef] [PubMed]
  84. Yi, B.; Tan, S.X.; Tang, C.E.; Huang, W.G.; Cheng, A.; Li, C.; Zhang, P.F.; Li, M.Y.; Li, J.L.; Yi, H.; et al. Inactivation of 14-3-3 sigma by promoter methylation correlates with metastasis in nasopharyngeal carcinoma. J. Cell. Biochem. 2009, 106, 858–866. [Google Scholar] [CrossRef] [PubMed]
  85. Raungrut, P.; Petjaroen, P.; Geater, S.L.; Keeratichananont, W.; Phukaoloun, M.; Suwiwat, S.; Thongsuksai, P. Methylation of 14-3-3σ gene and prognostic significance of 14-3-3σ expression in non-small cell lung cancer. Oncology Lett. 2017, 14, 5257–5264. [Google Scholar] [CrossRef]
  86. Lodygin, D.; Hermeking, H. Epigenetic silencing of 14-3-3sigma in cancer. Semin. Cancer Biol. 2006, 16, 214–224. [Google Scholar] [CrossRef]
  87. Phan, L.; Chou, P.C.; Velazquez-Torres, G.; Samudio, I.; Parreno, K.; Huang, Y.; Tseng, C.; Vu, T.; Gully, C.; Su, C.H.; et al. The cell cycle regulator 14-3-3sigma opposes and reverses cancer metabolic reprogramming. Nat. Commun. 2015, 6, 7530. [Google Scholar] [CrossRef]
  88. Hong, H.Y.; Jeon, W.K.; Bae, E.J.; Kim, S.T.; Lee, H.J.; Kim, S.J.; Kim, B.C. 14-3-3 sigma and 14-3-3 zeta plays an opposite role in cell growth inhibition mediated by transforming growth factor-beta 1. Mol. Cells 2010, 29, 305–309. [Google Scholar] [CrossRef]
  89. Berg, D.; Holzmann, C.; Riess, O. 14-3-3 proteins in the nervous system. Nat. Rev. Neurosci. 2003, 4, 752–762. [Google Scholar] [CrossRef]
  90. Steinacker, P.; Aitken, A.; Otto, M. 14-3-3 proteins in neurodegeneration. Semin. Cell Dev. Biol. 2011, 22, 696–704. [Google Scholar] [CrossRef]
  91. Ge, W.W.; Volkening, K.; Leystra-Lantz, C.; Jaffe, H.; Strong, M.J. 14-3-3 protein binds to the low molecular weight neurofilament (NFL) mRNA 3’ UTR. Mol. Cell Neurosci. 2007, 34, 80–87. [Google Scholar] [CrossRef] [PubMed]
  92. Cheah, P.S.; Ramshaw, H.S.; Thomas, P.Q.; Toyo-Oka, K.; Xu, X.; Martin, S.; Coyle, P.; Guthridge, M.A.; Stomski, F.; van den Buuse, M.; et al. Neurodevelopmental and neuropsychiatric behaviour defects arise from 14-3-3zeta deficiency. Mol. Psychiatry 2012, 17, 451–466. [Google Scholar] [CrossRef] [PubMed]
  93. Xu, X.; Jaehne, E.J.; Greenberg, Z.; McCarthy, P.; Saleh, E.; Parish, C.L.; Camera, D.; Heng, J.; Haas, M.; Baune, B.T.; et al. 14-3-3zeta deficient mice in the BALB/c background display behavioural and anatomical defects associated with neurodevelopmental disorders. Sci. Rep. 2015, 5, 12434. [Google Scholar] [CrossRef] [PubMed]
  94. Ikeda, M.; Hikita, T.; Taya, S.; Uraguchi-Asaki, J.; Toyo-oka, K.; Wynshaw-Boris, A.; Ujike, H.; Inada, T.; Takao, K.; Miyakawa, T.; et al. Identification of YWHAE, a gene encoding 14-3-3epsilon, as a possible susceptibility gene for schizophrenia. Hum. Mol. Genet. 2008, 17, 3212–3222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Wachi, T.; Cornell, B.; Toyo-Oka, K. Complete ablation of the 14-3-3epsilon protein results in multiple defects in neuropsychiatric behaviors. Behav. Brain Res. 2017, 319, 31–36. [Google Scholar] [CrossRef] [PubMed]
  96. Kim, D.E.; Cho, C.H.; Sim, K.M.; Kwon, O.; Hwang, E.M.; Kim, H.W.; Park, J.Y. 14-3-3gamma Haploinsufficient Mice Display Hyperactive and Stress-sensitive Behaviors. Exp. Neurobiol. 2019, 28, 43–53. [Google Scholar] [CrossRef] [PubMed]
  97. Toyo-oka, K.; Wachi, T.; Hunt, R.F.; Baraban, S.C.; Taya, S.; Ramshaw, H.; Kaibuchi, K.; Schwarz, Q.P.; Lopez, A.F.; Wynshaw-Boris, A. 14-3-3epsilon and zeta regulate neurogenesis and differentiation of neuronal progenitor cells in the developing brain. J. Neurosci. 2014, 34, 12168–12181. [Google Scholar] [CrossRef] [PubMed]
  98. Cornell, B.; Toyo-oka, K. Deficiency of 14-3-3epsilon and 14-3-3zeta by the Wnt1 promoter-driven Cre recombinase results in pigmentation defects. BMC Res. Notes. 2016, 9, 180. [Google Scholar] [CrossRef]
  99. Foote, M.; Qiao, H.; Graham, K.; Wu, Y.; Zhou, Y. Inhibition of 14-3-3 Proteins Leads to Schizophrenia-Related Behavioral Phenotypes and Synaptic Defects in Mice. Biol. Psychiatry 2015, 78, 386–395. [Google Scholar] [CrossRef] [Green Version]
  100. Graham, K.; Zhang, J.; Qiao, H.; Wu, Y.; Zhou, Y. Region-specific inhibition of 14-3-3 proteins induces psychomotor behaviors in mice. NPJ Schizophr. 2019, 5, 1. [Google Scholar] [CrossRef]
  101. Perez, R.G.; Waymire, J.C.; Lin, E.; Liu, J.J.; Guo, F.; Zigmond, M.J. A role for alpha-synuclein in the regulation of dopamine biosynthesis. J. Neurosci. 2002, 22, 3090–3099. [Google Scholar] [CrossRef] [PubMed]
  102. Ostrerova, N.; Petrucelli, L.; Farrer, M.; Mehta, N.; Choi, P.; Hardy, J.; Wolozin, B. alpha-Synuclein shares physical and functional homology with 14-3-3 proteins. J. Neurosci. 1999, 19, 5782–5791. [Google Scholar] [CrossRef] [PubMed]
  103. Xu, J.; Kao, S.Y.; Lee, F.J.; Song, W.; Jin, L.W.; Yankner, B.A. Dopamine-dependent neurotoxicity of alpha-synuclein: A mechanism for selective neurodegeneration in Parkinson disease. Nat. Med. 2002, 8, 600–606. [Google Scholar] [CrossRef] [PubMed]
  104. Plotegher, N.; Kumar, D.; Tessari, I.; Brucale, M.; Munari, F.; Tosatto, L.; Belluzzi, E.; Greggio, E.; Bisaglia, M.; Capaldi, S.; et al. The chaperone-like protein 14-3-3eta interacts with human alpha-synuclein aggregation intermediates rerouting the amyloidogenic pathway and reducing alpha-synuclein cellular toxicity. Hum. Mol. Genet. 2014, 23, 5615–5629. [Google Scholar] [CrossRef] [PubMed]
  105. Wang, J.; Lou, H.; Pedersen, C.J.; Smith, A.D.; Perez, R.G. 14-3-3zeta contributes to tyrosine hydroxylase activity in MN9D cells: Localization of dopamine regulatory proteins to mitochondria. J. Biol. Chem. 2009, 284, 14011–14019. [Google Scholar] [CrossRef] [PubMed]
  106. Sato, S.; Chiba, T.; Sakata, E.; Kato, K.; Mizuno, Y.; Hattori, N.; Tanaka, K. 14-3-3eta is a novel regulator of parkin ubiquitin ligase. EMBO J. 2006, 25, 211–221. [Google Scholar] [CrossRef] [PubMed]
  107. Paisan-Ruiz, C.; Jain, S.; Evans, E.W.; Gilks, W.P.; Simon, J.; van der Brug, M.; Lopez de Munain, A.; Aparicio, S.; Gil, A.M.; Khan, N.; et al. Cloning of the gene containing mutations that cause PARK8-linked Parkinson’s disease. Neuron 2004, 44, 595–600. [Google Scholar] [CrossRef]
  108. Su, B.; Liu, H.; Wang, X.; Chen, S.G.; Siedlak, S.L.; Kondo, E.; Choi, R.; Takeda, A.; Castellani, R.J.; Perry, G.; et al. Ectopic localization of FOXO3a protein in Lewy bodies in Lewy body dementia and Parkinson’s disease. Mol. Neurodegener 2009, 4, 32. [Google Scholar] [CrossRef]
  109. Ehrenberg, A.J.; Nguy, A.K.; Theofilas, P.; Dunlop, S.; Suemoto, C.K.; Di Lorenzo Alho, A.T.; Leite, R.P.; Diehl Rodriguez, R.; Mejia, M.B.; Rub, U.; et al. Quantifying the accretion of hyperphosphorylated tau in the locus coeruleus and dorsal raphe nucleus: The pathological building blocks of early Alzheimer’s disease. Neuropathol. Appl. Neurobiol. 2017, 43, 393–408. [Google Scholar] [CrossRef]
  110. Umahara, T.; Uchihara, T.; Tsuchiya, K.; Nakamura, A.; Iwamoto, T.; Ikeda, K.; Takasaki, M. 14-3-3 proteins and zeta isoform containing neurofibrillary tangles in patients with Alzheimer’s disease. Acta Neuropathol. 2004, 108, 279–286. [Google Scholar] [CrossRef]
  111. McFerrin, M.B.; Chi, X.; Cutter, G.; Yacoubian, T.A. Dysregulation of 14-3-3 proteins in neurodegenerative diseases with Lewy body or Alzheimer pathology. Ann. Clin. Transl. Neurol. 2017, 4, 466–477. [Google Scholar] [CrossRef] [PubMed]
  112. Yuan, Z.; Agarwal-Mawal, A.; Paudel, H.K. 14-3-3 binds to and mediates phosphorylation of microtubule-associated tau protein by Ser9-phosphorylated glycogen synthase kinase 3beta in the brain. J. Biol. Chem. 2004, 279, 26105–26114. [Google Scholar] [CrossRef] [PubMed]
  113. He, Y.; Han, J.R.; Chang, O.; Oh, M.; James, S.E.; Lu, Q.; Seo, Y.W.; Kim, H.; Kim, K. 14-3-3varepsilon/zeta Affects the stability of delta-catenin and regulates delta-catenin-induced dendrogenesis. FEBS Open Bio. 2013, 3, 16–21. [Google Scholar] [CrossRef]
  114. Zhou, J.; Liyanage, U.; Medina, M.; Ho, C.; Simmons, A.D.; Lovett, M.; Kosik, K.S. Presenilin 1 interaction in the brain with a novel member of the Armadillo family. Neuroreport 1997, 8, 2085–2090. [Google Scholar] [CrossRef] [PubMed]
  115. Ding, H.; Underwood, R.; Lavalley, N.; Yacoubian, T.A. 14-3-3 inhibition promotes dopaminergic neuron loss and 14-3-3θ overexpression promotes recovery in the MPTP mouse model of Parkinson’s disease. Neuroscience 2015, 307, 73–82. [Google Scholar] [CrossRef]
  116. Betarbet, R.; Sherer, T.B.; MacKenzie, G.; Garcia-Osuna, M.; Panov, A.V.; Greenamyre, J.T. Chronic systemic pesticide exposure reproduces features of Parkinson’s disease. Nat. Neurosci. 2000, 3, 1301–1306. [Google Scholar] [CrossRef] [PubMed]
  117. Jia, B.; Wu, Y.; Zhou, Y. 14-3-3 and aggresome formation: Implications in neurodegenerative diseases. Prion 2014, 8, 173–177. [Google Scholar] [CrossRef]
  118. Xu, Y.; Ren, J.; He, X.; Chen, H.; Wei, T.; Feng, W. YWHA/14-3-3 proteins recognize phosphorylated TFEB by a noncanonical mode for controlling TFEB cytoplasmic localization. Autophagy 2019, 15, 1017–1030. [Google Scholar] [CrossRef]
  119. Wang, Y.; Oh, S.W.; Deplancke, B.; Luo, J.; Walhout, A.J.; Tissenbaum, H.A. C. elegans 14-3-3 proteins regulate life span and interact with SIR-2.1 and DAF-16/FOXO. Mech. Ageing Dev. 2006, 127, 741–747. [Google Scholar] [CrossRef]
  120. Berdichevsky, A.; Viswanathan, M.; Horvitz, H.R.; Guarente, L.C. elegans SIR-2.1 interacts with 14-3-3 proteins to activate DAF-16 and extend life span. Cell 2006, 125, 1165–1177. [Google Scholar] [CrossRef]
  121. Araiz, C.; Chateau, M.T.; Galas, S. 14-3-3 regulates life span by both DAF-16-dependent and -independent mechanisms in Caenorhabditis elegans. Exp. Gerontol. 2008, 43, 505–519. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Nielsen, M.D.; Luo, X.; Biteau, B.; Syverson, K.; Jasper, H. 14-3-3 Epsilon antagonizes FoxO to control growth, apoptosis and longevity in Drosophila. Aging Cell. 2008, 7, 688–699. [Google Scholar] [CrossRef] [PubMed]
  123. Le, T.P.; Vuong, L.T.; Kim, A.R.; Hsu, Y.C.; Choi, K.W. 14-3-3 proteins regulate Tctp-Rheb interaction for organ growth in Drosophila. Nat. Commun. 2016, 7, 11501. [Google Scholar] [CrossRef] [PubMed]
  124. Martins, R.; Lithgow, G.J.; Link, W. Long live FOXO: Unraveling the role of FOXO proteins in aging and longevity. Aging cell. 2016, 15, 196–207. [Google Scholar] [CrossRef] [PubMed]
  125. Evans, D.S.; Kapahi, P.; Hsueh, W.C.; Kockel, L. TOR signaling never gets old: Aging, longevity and TORC1 activity. Ageing Res. Rev. 2011, 10, 225–237. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Kleppe, R.; Martinez, A.; Doskeland, S.O.; Haavik, J. The 14-3-3 proteins in regulation of cellular metabolism. Semin. Cell Dev. Biol. 2011, 22, 713–719. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Pozuelo, M.; Peggie, M.; Wong, B.H.; Morrice, N.; MacKintosh, C. 14-3-3s regulate fructose-2, 6-bisphosphate levels by binding to PKB-phosphorylated cardiac fructose-2,6-bisphosphate kinase/phosphatase. EMBO J. 2003, 22, 3514–3523. [Google Scholar] [CrossRef] [PubMed]
  128. Park, S.; Yoo, S.; Kim, J.; An, H.T.; Kang, M.; Ko, J. 14-3-3beta and gamma differentially regulate peroxisome proliferator activated receptor gamma2 transactivation and hepatic lipid metabolism. Biochim. Biophys. Acta 2015, 1849, 1237–1247. [Google Scholar] [CrossRef]
  129. Insenser, M.; Montes-Nieto, R.; Vilarrasa, N.; Lecube, A.; Simo, R.; Vendrell, J.; Escobar-Morreale, H.F. A nontargeted proteomic approach to the study of visceral and subcutaneous adipose tissue in human obesity. Mol. Cell Endocrinol. 2012, 363, 10–19. [Google Scholar] [CrossRef]
  130. Lim, G.E.; Albrecht, T.; Piske, M.; Sarai, K.; Lee, T.C.; Ramshaw, H.S.; Sinha, S.; Guthridge, M.A.; Acker-Palmer, A.; Lopez, A.F.; et al. 14-3-3zeta coordinates adipogenesis of visceral fat. Nat. Commun. 2015, 6, 7671. [Google Scholar] [CrossRef]
  131. Taurino, F.; Stanca, E.; Vonghia, L.; Siculella, L.; Sardanelli, A.M.; Papa, S.; Zanotti, F.; Gnoni, A. Short-term type-1 diabetes differentially modulates 14-3-3 proteins in rat brain and liver. Eur. J. Clin. Invest. 2014, 44, 350–358. [Google Scholar] [CrossRef] [PubMed]
  132. Wang, C.; Skinner, C.; Easlon, E.; Lin, S.J. Deleting the 14-3-3 protein Bmh1 extends life span in Saccharomyces cerevisiae by increasing stress response. Genetics 2009, 183, 1373–1384. [Google Scholar] [CrossRef] [PubMed]
  133. Seo, S.B.; Lee, J.J.; Yun, H.H.; Im, C.N.; Kim, Y.S.; Ko, J.H.; Lee, J.H. 14-3-3beta Depletion Drives a Senescence Program in Glioblastoma Cells Through the ERK/SKP2/p27 Pathway. Mol. Neurobiol. 2018, 55, 1259–1270. [Google Scholar] [CrossRef] [PubMed]
  134. Sreedhar, R.; Giridharan, V.V.; Arumugam, S.; Karuppagounder, V.; Palaniyandi, S.S.; Krishnamurthy, P.; Quevedo, J.; Watanabe, K.; Konishi, T.; Thandavarayan, R.A. Role of MAPK-mediated endoplasmic reticulum stress signaling in the heart during aging in senescence-accelerated prone mice. BioFactors 2016, 42, 368–375. [Google Scholar] [CrossRef] [PubMed]
  135. Fang, J.Y.; Wang, P.W.; Huang, C.H.; Chen, M.H.; Wu, Y.R.; Pan, T.L. Skin aging caused by intrinsic or extrinsic processes characterized with functional proteomics. Proteomics 2016, 16, 2718–2731. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The 14-3-3 isoforms in neurodegeneration. ND: Neurodegenerative disease; TH: Tyrosine hydroxylase; LRRK2: Leucine-rich repeat kinase 2; LBHI: lewy body-like hyalineinclusions; CAMKK: Calcium/calmodulin-dependent protein kinase kinase; PKA: Protein kinase A.
Figure 1. The 14-3-3 isoforms in neurodegeneration. ND: Neurodegenerative disease; TH: Tyrosine hydroxylase; LRRK2: Leucine-rich repeat kinase 2; LBHI: lewy body-like hyalineinclusions; CAMKK: Calcium/calmodulin-dependent protein kinase kinase; PKA: Protein kinase A.
Ijms 20 03518 g001
Figure 2. Schematic showing the action mechanisms of 14-3-3 proteins. The right half of the scheme shows the phosphopeptide-binding adaptor function of 14-3-3 proteins and the left half describes the chaperone-like function. The function of the phosphopeptide-binding adapter is primarily contributed by the dimeric form of 14-3-3, while the chaperone-like activity is primarily attributed to their monomeric form. Various factors (phosphorylation, stress, drug, aging, etc.) can disturb the balance between the dimeric and monomeric forms of the 14-3-3 proteins. Further details can be seen in the text. IGF-I: Insulin and insulin related growth factor I; TERT: Telomerase reverse transcriptase; Ron: Recepteur d’Origine nantais; SSH1L: Cofilin-phosphatase slingshot-1L; ADAM22: A disintegrin and metalloprotease 22.
Figure 2. Schematic showing the action mechanisms of 14-3-3 proteins. The right half of the scheme shows the phosphopeptide-binding adaptor function of 14-3-3 proteins and the left half describes the chaperone-like function. The function of the phosphopeptide-binding adapter is primarily contributed by the dimeric form of 14-3-3, while the chaperone-like activity is primarily attributed to their monomeric form. Various factors (phosphorylation, stress, drug, aging, etc.) can disturb the balance between the dimeric and monomeric forms of the 14-3-3 proteins. Further details can be seen in the text. IGF-I: Insulin and insulin related growth factor I; TERT: Telomerase reverse transcriptase; Ron: Recepteur d’Origine nantais; SSH1L: Cofilin-phosphatase slingshot-1L; ADAM22: A disintegrin and metalloprotease 22.
Ijms 20 03518 g002
Table 1. 14-3-3 protein isoforms in cancers.
Table 1. 14-3-3 protein isoforms in cancers.
14-3-3 IsoformCancer TypeExpressionReference
14-3-3ζBreast, lung, pancreas, esophageal, head and neck, oral, colon, chronic myeloid leukemia, ovarian[26,27,28,29,30,31]
14-3-3σLung, breast, esophageal, chronic myeloid leukemia, uterine, ovarian, skin[32,33,34,35,36]
Liver, pancreatic ductal[37,38]
14-3-3βLung, astrocytoma, glioma, colorectal, gastric squamous, liver[39,40,41,42]
14-3-3εRenal, liver, squamous, breast, gastric[43,44,45,46]
14-3-3γLiver, breast, lung[47,48,49]
14-3-3ηLiver, prostate, squamous, glioma[49,50,51,52]
14-3-3τ/θBreast, lung, glioma, prostate[53,54,55]
Red = elevated expression; blue = decreased expression.
Table 2. 14-3-3 isoform knock-out phenotypes in mice.
Table 2. 14-3-3 isoform knock-out phenotypes in mice.
K/O IsoformPhenotypesReferences
14-3-3ζSchizophrenia, autism spectrum disorder, and bipolar disorder; reduced learning, memory, and prepulse inhibition and locomotor hyperactivity[92,93]
14-3-3εSchizophrenic behavior; increased locomotor activity and sociability and decreased working memory[94,95]
14-3-3γHyperactive and depressive-like behavior; sensitive responses to acute stress[96]
14-3-3ζ and 14-3-3ε Neuronal migration and pigmentation defects and neural progenitor cells [14,97,98]
14-3-3 functional knock-outSchizophrenic behavior; synaptic alterations[99,100]

Share and Cite

MDPI and ACS Style

Fan, X.; Cui, L.; Zeng, Y.; Song, W.; Gaur, U.; Yang, M. 14-3-3 Proteins Are on the Crossroads of Cancer, Aging, and Age-Related Neurodegenerative Disease. Int. J. Mol. Sci. 2019, 20, 3518. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms20143518

AMA Style

Fan X, Cui L, Zeng Y, Song W, Gaur U, Yang M. 14-3-3 Proteins Are on the Crossroads of Cancer, Aging, and Age-Related Neurodegenerative Disease. International Journal of Molecular Sciences. 2019; 20(14):3518. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms20143518

Chicago/Turabian Style

Fan, Xiaolan, Lang Cui, Yao Zeng, Wenhao Song, Uma Gaur, and Mingyao Yang. 2019. "14-3-3 Proteins Are on the Crossroads of Cancer, Aging, and Age-Related Neurodegenerative Disease" International Journal of Molecular Sciences 20, no. 14: 3518. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms20143518

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