Abstract
Importance of the field
The ubiquitously expressed 14-3-3ζ protein is involved in numerous important cellular pathways involved in cancer. Recent research suggests 14-3-3ζ may play a central role regulating multiple pathways responsible for cancer initiation and progression. This review will provide an overview of 14-3-3 proteins and address the role of 14-3-3ζ overexpression in cancer.
Areas covered in this review
The review covers the basic role of 14-3-3 in regulation of multiple pathways with a focus on 14-3-3ζ as a clinically relevant biomarker for cancer recurrence.
What the reader will gain
14-3-3ζ overexpression has been found in multiple cancers; however, the clinical implications were unclear. Recently, 14-3-3ζ has been identified as a biomarker for poor prognosis and chemoresistance in multiple tumor types, indicating a potential clinical application for using 14-3-3ζ in selecting treatment options and predicting cancer patients’ outcome.
Take home message
14-3-3ζ is a potential prognostic marker of cancer recurrence and predictive marker for therapeutic resistance. The overexpression of 14-3-3ζ in multiple cancers suggests that it may be a common target to intervene tumor progression; therefore, more efforts are needed for the development of 14-3-3 inhibitors.
Keywords: 14-3-3 zeta, apoptosis, cancer, chemoresistance, prognostic marker
1. Introduction
Cancer is a group of diseases caused by a multi-step process involving the malfunction of genes that regulate cell proliferation, cell division, cell death, and tissue microenvironment. The malfunction of these genes leads to a series of acquired capabilities that allow uncontrolled growth and invasion of abnormal cells to other organs. Many types of cancer exist; yet, all malignant growths manifest a set of essential alterations that are shared among different tumor types. These “hallmarks of cancer” are defined as: self-sufficiency in growth signals, insensitivity to antigrowth signals, evasion of apoptosis, limitless replicative potential, sustained angiogenesis and tissue invasion and metastasis 1. Identifying alterations associated with multiple cancers and central to cancer progression is imperative to improve diagnosis and treatment.
The 14-3-3 family of proteins is known to interact with a multitude of targets that participate in essential cellular processes such as signal transduction, apoptosis, cell cycle and cell migration 2, 3. They have also been found to be overexpressed in multiple types of cancers 2. These findings indicate that 14-3-3 proteins may play a central role in regulating various biological pathways that contribute to cancer progression. Of the seven 14-3-3 isoforms, several recent studies have demonstrated that 14-3-3ζ overexpression has a direct role in cellular transformation and is associated with poor prognosis in cancer patients. Therefore, in this review, we focus on the role of 14-3-3ζ in cancer and expand the discussion on the clinical significance of 14-3-3ζ. We bring a new paradigm in 14-3-3 research: the potential of 14-3-3ζ as a therapeutic target in human cancers.
2. 14-3-3 Overview
14-3-3 proteins are a family of evolutionally highly conserved acidic proteins expressed in all eukaryotic organisms 4. Moore and Perez first discovered 14-3-3 in 1967 when they fractionated soluble proteins from brain tissue. They named this abundant brain protein, 14-3-3, based on its particular migration pattern on two dimensional DEAE-cellulose chromatography and starch gel electrophoresis 5. At least two or more 14-3-3 isoforms have been observed in all eukaryotic organisms. In mammals, there are seven distinct isoforms: β, γ, ε, ζ, η, σ, and τ that are encoded by seven different genes. 14-3-3 proteins lack endogenous enzymatic activity; however, they function through binding to phosphorylated-serine/threonine motifs on their target proteins. Following binding, target proteins can be regulated by 14-3-3 through a number of mechanisms including: changing the conformation of the protein, affecting protein activity or stability, facilitating protein complex formation, or altering protein subcellular localization.
Crystal structures have revealed that all 14-3-3 isoforms are strikingly similar in structure forming a flattened horseshoe structure 6. 14-3-3 proteins exist as dimers, and each monomer in the dimer is composed of nine anti-parallel alpha helices with the dimer interface at the N-terminus. The concave inner surface of the horseshoe contains highly conserved residues while the outer surface is composed of variable residues. The inner domain of each monomer forms a groove, which is highly conserved amongst different species and mediates binding of 14-3-3 to its target proteins 6. The primary amino acid sequences of the N terminus are quite variable while the crystal structure indicates the residues that create the dimer interface are quite conserved. It has been previously shown that 14-3-3 isoforms can form homodimers as well as heterodimers 7. Therefore, the variability of the N terminus may be a mechanism for regulating the combinations of possible homo- and heterodimers between different 14-3-3 isoforms. Dimer formation is a functional characteristic of 14-3-3 proteins 8, 9. For example, it has been demonstrated that both monomeric and dimeric 14-3-3 molecules can bind to Raf-1; however, only the dimeric form supports Raf-1 kinase activity 10. Thus, 14-3-3 dimerization is required for ligand regulation and may represent a regulatory mechanism controlling 14-3-3 binding.
The primary mechanism controlling 14-3-3 interaction with target proteins is serine/threonine phosphorylation of the interacting target protein 4. Two prototype 14-3-3 binding motifs, RSXpSXP and RXY/FXXpSXP, where pS represents phosphoserine and X any amino acid, were derived from target binding proteins and extensive phospho-peptide library screening 11, 12. Many 14-3-3 interacting proteins contain one or more of these motifs; however, a broad range of substitutions, such as phospho-threonine, can be found in other proteins. Further, several non-phosphorylated proteins have been identified that interact with 14-3-3 13, 14. In addition to phosphorylation of 14-3-3 binding proteins, other mechanisms that directly regulate 14-3-3 have been shown to modulate 14-3-3 interaction with target proteins. Phosphorylation of the threonine 232 residue in 14-3-3 ζ and τ can disrupt 14-3-3 binding by altering the conformation of the C-terminal tail 15. 14-3-3 dimers are normally stable and do not readily exchange; however, phosphorylation of the serine 58 residue on 14-3-3ζ can disrupt binding by altering 14-3-3 dimerization 7, 16. Similar residues in other 14-3-3 isoforms can be phosphorylated that inhibit 14-3-3 binding and dimerization 17, 18. Sub-cellular localization of 14-3-3 may be another mechanism that directs or limits interacting proteins, especially in development and differentiation 19. Furthermore, 14-3-3ζ can be post-transcriptionally regulated by microRNA miR-375, leading to down regulation of both 14-3-3ζ mRNA and protein, effectively altering the available pool of 14-3-3 in cells 20. Since 14-3-3 proteins can form homodimers or heterodimers, deregulation of mechanisms controlling 14-3-3 dimerization or expression may alter the balance of 14-3-3 dimers. This unbalance may disrupt cellular homeostasis leading to activation of 14-3-3 targeted pathways that contribute to the initiation of cancer.
3. 14-3-3 targets in cancer
14-3-3 was initially characterized as an activator of tyrosine and tryptophan hydroxylases and regulator of protein kinase C (PKC) activity 21, 22. With the advent of proteomics, a plethora of publications have reported that 14-3-3 proteins can interact with several hundreds of binding partners 23, 24. Thus, 14-3-3 proteins have been found to interact with target proteins involved in regulating multiple cellular processes, such as cell cycle control, protein trafficking, anti-apoptosis, metabolism, signal transduction, inflammation and cell adhesion/motility 25, 26 (Figure 1). For examples, 14-3-3 proteins play a pivotal role during cell cycle progression 27 by binding and sequestering CDC25C in the cytoplasm thereby separating it from its nuclear substrate Cdc2 and allowing for entry into mitosis 25. 14-3-3 binding to the cardiac isoform of 6-phosphofructo-2-kinase/ fructose-2,6-bisphosphatase (PFK-2) implicates 14-3-3 in regulating glycolysis and metabolism 28. 14-3-3 proteins participate in the coordination of cell adhesion and motility by regulating the migration of keratinocytes through interactions with α6β4 and α3β1 integrins 29. 14-3-3 is also involved in the establishment and maintenance of mammalian epithelial polarity, as recently demonstrated in MDCK cells where 14-3-3 binding was shown to promote the dissociation of Par-1b from tight junctions after its phosphorylation by aPKC 30.
Figure 1.
14-3-3 regulates multiple pathways involved in cancer.
14-3-3 isoforms are homo- and hetero-dimeric, phospho-serine binding proteins. The 14-3-3 family proteins have been found to interact with and regulate several hundreds of proteins involved in multiple cellular pathways summarized in the figure. These pathways not only function to maintain normal cellular homeostasis, but, when deregulated, contribute to cancer initiation and progression. In this context, 14-3-3 may represent an ideal target for cancer therapy since it acts as a central hub regulating many pathways involved in cellular transformation common to various cancers.
Although 14-3-3 proteins are highly homologous and have some overlapping binding partners, there is evidence that 14-3-3 isoforms may have distinct binding targets and isoform specific functions 31–33. Additionally, the tendency for isoforms to heterodimerize may also regulate the selectivity of potential binding partners and specificity of 14-3-3 interacting targets 7, 33. To date, 14-3-3s has been divided into two subgroups, with 14-3-3σ defined as a tumor suppressor, while other 14-3-3 isoforms may have oncogenic roles. The down-regulation of 14-3-3σ has been associated with a multitude of human epithelial cancers, including hepatocellular carcinoma 34, lung cancer 35 and breast cancer 36. 14-3-3σ is considered a tumor suppressor mainly due to its unique role in positively regulating p53 and sustaining the G2/M checkpoint in epithelial cells following DNA damage 31, 37. However, recent studies suggest 14-3-3σ may also contribute to cancer development 38. Other 14-3-3 isoforms appear to be involved in pathways that contribute to transformation related properties. For example, some 14-3-3 isoforms have been identified to be involved in chemoresistance either in cell lines or patients tumors 39, 40. Single nucleotide polymorphisms in the 14-3-3τ gene from soft tissue sarcoma (STS) patients were associated with an early onset of disease and an increased risk of tumor-related death in STS patients 40. Additionally, knockdown of 14-3-3β with antisense induced apoptosis in cancer cell lines; whereas, 14-3-3β overexpression contributed to transformation 41, 42. Likewise, a number of 14-3-3ζ target proteins are proto-oncogene and oncogene products that have a causative role in the development of human cancers, indicating that 14-3-3ζ may directly contribute to cancer initiation and progression (Table 1) 43–47.
Table 1.
Representative transformation-associated 14-3-3ζ binding targets
Overexpression of 14-3-3ζ in human cancers may contribute to transformation by inhibiting apoptosis, activating signaling pathways that promote growth, and/or sequestering tumor suppressor proteins. 14-3-3ζ was found to play a major role in anti-apoptosis by directly binding with Akt-phosphorylated Bad, a pro-apoptotic protein, at one or more serine sites, thereby blocking the inhibition of Bcl-2 by Bad 48. 14-3-3ζ can also bind to the Forkhead transcription factor after its phosphorylation by Akt. 14-3-3ζ negatively regulates Forkhead’s function by sequestering it in the cytoplasm, preventing the transcription of the pro-apoptotic proteins Fas and Bax 49. Similarly, 14-3-3ζ enhances Akt-phosphorylated β-catenin-dependent transcription by stabilizing the β-catenin protein in the cytoplasm 50. β-catenin co-localizes with activated Akt in intestinal stem cells (ISCs) suggesting 14-3-3 may have a role in stem cell regulation 51. 14-3-3 proteins can also bind to the insulin-like growth factor receptor (IGF1R) that plays an important role in regulating normal cell survival as well as promoting tumorigenesis. The major site for 14-3-3ζ and β interaction with IGF1R occurs within a serine rich region that is critical for IGF1R mediated cell transformation 52. Mutation of the 14-3-3 binding site in IGF1R blocked colony formation in a soft agar assay and reduced the tumorigenicity in mice, suggesting 14-3-3ζ and/or β may contribute to IGF1R mediated transformation 53. The tuberous sclerosis (TSC) protein 1 and 2 are involved in tumor suppression by inhibiting ribosomal subunit S6 kinase (S6K) signaling pathways. TSC2 is phosphorylated by Akt and 14-3-3ζ and β were found to interact with TSC2 in an Akt dependent manner 54. 14-3-3 binding to TSC negatively regulates its function and overexpression of 14-3-3ζ and β compromised TSC inhibition of S6K leading to S6K activation 55. These studies indicate that 14-3-3ζ may play a central role in the development and progression of human cancers.
4. 14-3-3ζ and Cancer
14-3-3ζ has been implicated in the initiation and progression of cancer and has been shown to be overexpressed in multiple cancer types (Table 2). Several studies have suggested that 14-3-3ζ expression contributes to the transformation phenotype. Overexpression of 14-3-3ζ in breast cancer cell lines enhanced anchorage-independent growth and inhibited stress-induced apoptosis, whereas down-regulation of 14-3-3ζ reduced anchorage-independent growth, sensitized cells to stress-induced apoptosis, and reduced the onset and growth of tumor xenografts in vivo 56. Also in breast cancer, 14-3-3ζ was found to be elevated in metastatic cells compared to normal ductal epithelium 57. In lung cancer, knockdown of 14-3-3ζ sensitized cells to stress-induced apoptosis and enhanced cell adhesion and cell-cell contacts; two activities that are frequently being suppressed in cancer development 58. Similarly, knockdown of 14-3-3ζ was sufficient to restore the sensitivity of lung cancer cells to anoikis and impair their anchorage-independent growth 59. In the context of cancer, overexpression of 14-3-3ζ may lead to preferential activation of 14-3-3ζ-targeted pathways and may be a central node regulating multiple pathways involved in cancer development (Figure 1). Additionally, some studies have suggested the possibility that overexpression of other canonical 14-3-3 isoforms may also contribute to tumorigenicity. It is known that 14-3-3ζ proteins can form homodimers or form heterodimers with other isoforms. In the context of cancer, overexpression of 14-3-3ζ may interfere with the balance of various dimer compositions within the cell. This would lead to substrate bias for different 14-3-3 home- and heterodimers, which may explain why increasing 14-3-3ζ, even in the presence of other 14-3-3 proteins, leads to enhanced transformation.
Table 2.
Cancer types associated with 14-3-3 overexpression
| Tumor Type | 14-3-3 Isoform | Poor Prognosis | Reference |
|---|---|---|---|
| Breast | ζ | Yes | 56†, 57, 63, 64, 66†, 73†, 74† |
| Lung | ζ, multiple | Yes | 68†, 95 |
| Pancreas | ζ | 96 | |
| Colon | ζ | 97 | |
| Esophageal | ζ | 61, 98 | |
| Stomach | ζ | 99 | |
| Oral | ζ | 100 | |
| Head and Neck | ζ | Yes | 62, 67†, 72, 101 |
| Urothelial | ζ | 71 | |
| Renal | ε | 102 | |
| Brain, astrocytoma | β, η, multiple | 103, 104 | |
| Brain, meningioma | multiple | 105 | |
| Chronic myeloid leukemia | ζ | 106 | |
| Diffuse large B cell lymphoma | ζ | 75 | |
| Papillomavirus-induced carcinomas | ζ | 107 |
Studies associated with 14-3-3ζ overexpression and poor prognosis in cancer patients
A critical component for improved detection and treatment strategies for cancer is the identification of molecular and biochemical pathways that contribute to the transition from normal epithelium to the first definable stage of cancer. Overexpression of 14-3-3ζ occurs in pre-malignant hyperplastic stages of oral and esophageal cancers 60-62. However the significance of 14-3-3ζ overexpression in these cancers remains unknown. It was hypothesized that 14-3-3ζ binding to NFκB may be a link between chronic inflammation and cancer 60. Similarly in breast cancer, 14-3-3ζ has been found to have elevated expression in atypical ductal hyperplasia and ductal carcinoma in situ (DCIS) 63, 64. For breast cancer, DCIS represents an early stage cancerous lesion in which malignant cells accumulate within the lumen of mammary ducts with no evidence of invasion through the basement membrane into the surrounding stroma 65. Our recent studies have identified 14-3-3ζ as a possible key component in DCIS initiation and invasion into the surrounding tissue. 14-3-3ζ overexpression in mammary epithelial cells (MEC) was shown to severely disrupt the acini architecture of MEC in 3-dimentional (3D) culture resulting in apoptosis resistance and luminal filling 63. 14-3-3ζ overexpression in MEC conferred resistance to anoikis and induced hyperactivation of the phosphoinositide 3-kinase/Akt pathway which led to phosphorylation and translocation of the MDM2 E3 ligase and subsequently increased p53 degradation. Ectopic expression of p53 restored luminal apoptosis in 14-3-3ζ overexpressing MECs. Furthermore, 14-3-3ζ overexpression was found to be a “second hit” in a subset of ErbB2-overexpressing DCIS lesions facilitating the transition from non-invasive DCIS into life-threatening invasive breast cancer 66. Co-overexpression of 14-3-3ζ and ErbB2 in breast cancers from patients was significantly correlated with distant metastasis. At the cellular level, overexpression of ErbB2 and 14-3-3ζ in MECs increased cell migration and decreased cell adhesion, respectively. Increased expression of 14-3-3ζ reduced cell adhesion by binding to and stabilizing expression of the TGF- receptor I, which activated the TGF-β/Smads pathway. Consequently, activation of the TGF-β/Smads pathway up regulated SIP1, a master transcriptional regulator of epithelial-mesenchymal transition (EMT), leading to E-cadherin loss and epithelial to mesenchymal transition (EMT). In patient DCIS samples, increased expression of 14-3-3ζ was clearly associated with increased expression of TGF receptor and EMT markers. Importantly, overexpression of both HER2 and 14-3-3ζ in breast tumors was associated with poor prognosis and higher rates of metastatic recurrence in cancer patients. These studies indicate that 14-3-3ζ may contribute to the development of early stage cancers and promote the transition to invasive cancers.
5. 14-3-3ζ as a marker for cancer recurrence and chemoresistance
5.1. 14-3-3ζ as a cancer prognostic marker
The discovery of new markers of disease recurrence and distant metastasis could help to identify patients for more aggressive treatment earlier in the course of cancer development. Additionally, identification of markers of therapeutic resistance would allow clinicians to choose treatment regimes and spare patients side effects from treatments that may not be effective. 14-3-3ζ overexpression in multiple cancers and their interactions with multiple cellular pathways has implicated the important role of 14-3-3ζ in cancer progression; however, the clinical significance of 14-3-3ζ in human cancers has just begun to emerge.
The evolving 14-3-3 research indicates that 14-3-3ζ may be a prognosis marker to predict cancer recurrence and treatment resistance. In head-and-neck/oral squamous cell carcinomas (HNOSCCs), patients with 14-3-3ζ positive tumors had a shorter disease-free survival than those with 14-3-3ζ-negative tumors; however, the median time was not significantly different 67. Interestingly, HNOSCC patients having overexpression of both 14-3-3σ and 14-3-3ζ had a significantly decreased median disease-free survival compared to patients showing no overexpression of these two proteins. Similarly, 14-3-3ζ overexpression in non–small cell lung carcinoma (NSCLC) was significantly associated with reduced survival and disease recurrence in patients 68. This study also found high 14-3-3ζ expression was significantly correlated with histological grade and clinical stage in NSCLC. Most importantly, among the clinical and histological parameters tested, overexpression of 14-3-3ζ was the only independent predictor for disease recurrence and reduced overall survival in the cohort of NSCLC patients. We recently reported that 14-3-3ζ overexpression in advanced stage breast cancer was significantly associated with disease recurrence and poor survival in breast cancer patients 56. We found that among the clinical and histological parameters tested, 14-3-3ζ was associated with ErbB2 expression and late stage tumors. In addition to the well-known prognostic markers in breast cancer, i.e., lymph node status and ErbB2 expression, 14-3-3ζ was found to be an independent marker of disease recurrence. Furthermore, 14-3-3ζ overexpression, ErbB2 overexpression, and positive lymph node status identified a sub-group of patients with a significantly increased risk of distant metastasis. In another study investigating the contribution of 14-3-3ζ to ErbB2-mediated transformation, breast tumors co-overexpressing both ErbB2 and 14-3-3ζ had significantly increased disease recurrence associated with increased risk of metastasis 66. Together, these findings indicate that detecting 14-3-3ζ protein expression levels in cancers may serve as a clinical marker for cancer recurrence.
5.2. 14-3-3ζ gene amplification in cancer
A common feature of oncogenic transformation is DNA amplification, which can occur when gene copies of specific regions of the genome are increased due to redundant replication. Amplified genomic DNA can contain many copies of a gene and increased gene expression from these amplified regions may confer a selective advantage for cell growth. Amplification and overexpression of cellular oncogenes can be good predictors of clinical outcome and potential therapeutic targets. The 14-3-3ζ gene (YWHAZ) is located on chromosome 8q22.3, an area frequently amplified in breast and other cancers 69, 70. Several studies in different cancer types suggest that increased gene copy numbers of YWHAZ may be associated with increased protein expression of 14-3-3ζ. Comparing array CGH data with expression data obtained by high density expression profiling in urothelial carcinomas 71, it was found that the region spanning chromosome 8q22, which included YWHAZ, was amplified in late stage urothelial tumors. Of the ten genes in this region, two genes, YWHAZ and POLR2K, were found to have a strong correlation between high copy numbers and high gene expression. In another study using a tissue microarray of head and neck squamous cell carcinomas (HNSCC), increased gene copy numbers of YWHAZ, associated with chromosome 8 polysomy, occurred in 30–40% of the cases 72. Increased 14-3-3ζ expression was detected in 77% of the HNSCC samples from the tissue microarray with a subset of cases having both increased gene copy numbers of YWHAZ and increased 14-3-3ζ protein expression. Similarly, amplification and chromosome 8 polysomy contributes to increased YWHAZ gene copy numbers in breast cancer 56. Increased YWHAZ copy number determined by FISH was associated with strong 14-3-3ζ protein expression detected by IHC staining in breast tumors. Another recent study in breast cancer found that amplification of the chromosomal 8q22 region contained 12 genes, including YWHAZ, associated with metastatic recurrence 73. Amplification of 8q22 was correlated with increased expression of the 12 genes and was found to be a strong independent prognostic factor for breast cancer recurrence. These studies suggest amplification may be a common mechanism leading to 14-3-3ζ overexpression in multiple cancers, although mechanisms independent of increased gene copy number, such as modulation of 14-3-3ζ gene transcription, protein translation, or RNA and protein stability may also contribute to increased 14-3-3ζ expression. Therefore, detection of 14-3-3ζ gene amplification may also be developed as a clinical marker for cancer recurrence.
5.3. 14-3-3ζ as a marker for cancer therapeutic resistance
Therapies using cytotoxic drugs or radiation form the basis for effective treatments of multiple cancers. However, the responses to chemotherapeutic agents can vary and drug resistance can often lead to treatment failures. One obstacle for the successful treatment of cancers by chemotherapeutic agents is the intrinsic or acquired resistance to drug treatments. Increased expression of 14-3-3ζ in cancer and its role as an anti-apoptotic protein have been linked to resistance to anticancer therapies (Table 3).
Table 3.
Cancer treatments with reduced efficacy in patients with 14-3-3ζ-overexpressing tumors
In breast cancer, the expression of 14-3-3ζ may be increased by tamoxifen treatment leading to tamoxifen resistance 74. Tamoxifen treatment increased expression of 14-3-3ζ via the estrogen receptor (ER) in breast cancer cell lines; however, increased expression of 14-3-3ζ was not regulated by estrogen. High expression of two tamoxifen stimulated genes, YWHAZ/14-3-3ζ and LOC441453, were found to correlate significantly with disease recurrence following tamoxifen treatment in women with ER-positive breast cancers, suggesting 14-3-3ζ overexpression may contribute to tamoxifen resistance. Additionally, amplification of 8q22 was associated with the increased expression of 12 genes in this region and chemo resistance in breast cancer 73. 8q22 amplification led to overexpression of 8q22 genes in tumor tissue, which was associated with poor prognosis in untreated cases and increased disease recurrence despite adjuvant chemotherapy. An siRNA screen of the 12 genes revealed that down regulation of YWHAZ and LAPTM4B increased sensitivity to chemotherapeutic drugs in breast cancer cell lines. The increased sensitivity was specifically to anthracyclines but not cisplatin or paclitaxel. Increased expression of YWHAZ or LAPTM4B in breast tumors was associated with poor outcome after adjuvant chemotherapy. Furthermore, increased expression of YWHAZ and/or LAPTM4B in tumors from breast cancer patients could predict a lower clinical response to epirubicin but was not associated with response to cisplatin or docetaxel. In diffuse large B cell lymphoma (DLBCL), 14-3-3ζ was identified to be overexpressed in cell lines which were resistant to the standard anthracycline-based chemotherapeutic combination consisting of cyclophosphamide, doxorubicin, vincristine, and prednisone (CHOP) 75. Knockdown of 14-3-3ζ expression in DLBCL resistant cell lines restored the sensitivity of resistant cell lines to apoptosis induced by CHOP. Increased expression of 14-3-3ζ was also detected in patients with DLBCL, and 14-3-3ζ may lead to CHOP resistance in these patients. Similarly, elevated expression of 14-3-3ζ was observed in a prostate cancer cell line resistant to 9NC6, a camptothecin derivative 76. Other 14-3-3 isoforms have also been identified to be involved in chemoresistance either in cell lines or in patients 39, 40.
These data support the use of FISH analysis of YWHAZ in combination with 14-3-3ζ protein expression as clinically relevant markers to predict prognosis and response to anti-cancer treatment. 14-3-3ζ status in patients’ tumors may guide their treatment options. The significant association of 14-3-3ζ overexpression with disease recurrence and chemoresistance also makes 14-3-3ζ an attractive candidate for targeted therapy in multiple cancers.
6. 14-3-3ζ as a target for anti-cancer therapy
Since 14-3-3ζ overexpression can lead to apoptosis resistance, cancer recurrence and chemoresistance, it is imperative to design methods to effectively block the function of 14-3-3ζ in human cancers. 14-3-3ζ is not limited to cancer and may also represent a potential target for other diseases 77–79. However, identification of 14-3-3 targeting molecules has been an underdeveloped area of research. Presently, only small interfering RNAs, antisense, and peptide inhibitors are used for targeting 14-3-3ζ under experimental conditions.
6.1. Small Interfering RNA (siRNA)
Double stranded small interfering RNAs (siRNA) of 21 nucleotides can be used to inhibit specific genes without inhibiting global protein synthesis 80. The mechanism of siRNA involves the degradation of the targeted mRNA and therefore reduced protein abundance. This makes siRNA a powerful tool to study specific gene functions; however, the off-target effects of siRNA are a limiting factor for this technology 81.
In lung cancer, 14-3-3ζ knockdown by siRNA increased the sensitivity to cisplatin both in vitro and in vivo 68. We have shown that 14-3-3ζ siRNA treatment of cancer cell lines sensitized cells to stress-induced apoptosis and effectively reduced the onset and growth of tumor xenografts 56. Several other studies in multiple cancer types have shown that knock down of 14-3-3ζ via antisense or siRNA is effective at reducing tumor growth and sensitizing cancer cells to apoptosis inducing agents 75, 82, 83. These studies suggest using siRNA or antisense RNA to knockdown 14-3-3ζ expression in tumors may be a potential therapeutic strategy; however, the clinical feasibility of using siRNA or anti-sense as therapies is still a challenge that needs to be met 84.
6.2. Peptide inhibitors: R18/Difopein
R18 is a small non-phosphorylated peptide identified from a phage library that non-selectively binds all 14-3-3 isoforms with high affinity and is a potent inhibitor of 14-3-3 ligand interaction 85. To facilitate delivery of R18 into cells and enhance its efficacy, two R18 coding sequences separated by a sequence coding for a short peptide linker were cloned into a mammalian expression vector. The translated product consisting of two R18 peptides linked together, called difopein (dimeric fourteen-three-three peptide inhibitor), was found to induce cell death alone, and enhance cell death by cisplatin 86, 87. In BCR-ABL transformed cells the R18 dimer effectively induced apoptosis, in part through liberation and reactivation of FOXO3a but not by disrupting 14-3-3/BCR-ABL association or BCR-ABL tyrosine kinase activity 88. Furthermore, cells expressing imatinib-resistant BCR-ABL mutants were more susceptible to apoptosis by R18 alone or in combination with signaling pathway inhibitors. Similarly, glioma cell lines showed increased apoptosis in vitro following knockdown of 14-3-3 via siRNA or inhibition via difopein 89. Interestingly, difopein effectively hindered proliferation and triggered apoptosis of tumor cells implanted into nude mice. While R18/difopein is an effective inhibitor of 14-3-3 target binding, it is a global inhibitor of 14-3-3 and is not specific to any isoform, such as 14-3-3 ζ. Isoform-specific inhibitors are not currently available, and research on isoform-specific inhibitors is an underdeveloped area in the 14-3-3 field.
These studies indicate that 14-3-3ζ is a potential target in multiple cancers. Although current 14-3-3ζ-targeting approaches are limited to knockdown experiments and peptide inhibitors, efforts are being focused to develop assays to discover 14-3-3 interacting molecules that may interfere with the key 14-3-3 interactions involved in disease 90, 91. It is foreseeable that other creative approaches (chemical inhibitors, shRNAs, etc.) will be developed in the future to specifically inhibit 14-3-3ζ-mediated transformation. The discoveries of 14-3-3ζ antagonists will not only lead to therapeutic interventions against diseases involving 14-3-3ζ, it will expand our understanding of the biological functions of 14-3-3ζ.
7. Conclusions
Recent studies have demonstrated that 14-3-3ζ overexpression is significantly associated with disease recurrence and resistance to chemotherapeutic agents in cancer patients. Overexpression of 14-3-3ζ has been associated with anchorage independent growth and a survival advantage under stress conditions. In contrast, knockdown of 14-3-3ζ reduced tumor growth and sensitized cells to chemotherapeutic agents. One of the major challenges in cancer treatment is to identify patients at high risk who would benefit from potentially unnecessary and toxic systemic therapy. 14-3-3ζ has been identified as a clinically relevant prognostic marker for multiple cancers and may allow for identification of patients whose tumors are resistant to standard chemotherapies to receive more aggressive treatments. In addition, 14-3-3ζ regulates known oncogenes and cooperates with HER2 to increase cancer invasion. These findings indicate that overexpression of 14-3-3ζ may cooperate with known oncogenes leading to more aggressive cancers. Furthermore, multiple studies have found that 14-3-3ζ gene amplification was associated with poor prognosis and resistance to common treatment regimens of cancer. Gene amplification may be a common mechanism leading to 14-3-3ζ overexpression in multiple cancers. Although studies of larger cohorts are needed to validate these findings before moving to a clinical setting, these novel findings indicate 14-3-3ζ as an important molecular marker for disease recurrence in cancer patients.
Even though therapeutic targeting of 14-3-3 is an underdeveloped area of research, several studies have validated 14-3-3ζ as a target in cancer therapy. These data suggest that 14-3-3ζ has the potential to serve as a therapeutic target for chemo-sensitization and effective tumor inhibition. Additionally, 14-3-3ζ is known to modulate multiple downstream molecules of several survival signaling pathways. Therefore, targeting 14-3-3ζ or the downstream pathways regulated by 14-3-3ζ may sensitize cells to apoptosis and serve as effective anti-cancer strategies in patients whose tumors overexpress 14-3-3ζ.
8. Expert Opinion
8.1. Definitive Role in Cancer
Many studies have shown that 14-3-3ζ regulates central pathways critical for maintenance of the transformed phenotype and 14-3-3ζ overexpression may activate or enhance numerous pathways that contribute to cancer development and progression. A missing component in the confirmation of the role of 14-3-3ζ in cancer initiation and progression is the development of genetically engineered 14-3-3ζ mouse models. Currently, no transgenic mouse models for 14-3-3ζ exist in the literature. However, an initial unpublished study mentioned in a review by Tzivion et al indicated that a transgenic mouse model expressing the 14-3-3ζ isoform developed various types of tumors in up to 15% of mice by 7–15 months 2. In addition, the tumor profile of mice infected with a Polyomavirus middle T antigen (MT) mutant defective in 14-3-3 binding showed a striking deficiency in the induction of salivary gland tumors but not other tumor types 92. Our recent study also suggests that 14-3-3ζ can cooperate with HER2 to induce early stage breast cancer to become invasive 66. These data suggest that 14-3-3ζ plays a key role in the cooperative induction of tumors mediated by oncogenes. The development and characterization of 14-3-3ζ transgenic and knockout mouse models will not only provide important information on the role of 14-3-3ζ in normal development and cancer progression, but also facilitate the discovery of 14-3-3ζ targeted therapies.
8.2. Powerful Cancer Prognosis Marker
Oncoprotein overexpression and oncogene amplification are often used as makers for predicting clinical outcome in cancer patients. Recent studies have demonstrated that increased expression of 14-3-3ζ is associated with cancer recurrence and increased incidence of distant metastasis. In addition, 14-3-3ζ overexpression and gene amplification have been associated with the development of resistance to multiple therapies. These studies have provided the basis for developing 14-3-3ζ into a useful tumor marker. The utility of 14-3-3ζ overexpression is not limited to a particular cancer type; 14-3-3ζ can potentially be utilized as a global tumor marker and therapeutic target for multiple cancer types. However, in order for 14-3-3ζ to gain widespread clinical application, further clinical validation and standardization will need to be accomplished. Current studies have utilized small patient cohorts to establish the importance of 14-3-3ζ in association with clinical outcomes. To further validate 14-3-3ζ as a tumor marker, larger cohorts need to be investigated in both retrospective and prospective studies. A most important issue for 14-3-3ζ marker development is the standardization of detection and staining methods. Recent studies implicating 14-3-3ζ overexpression in cancers have used multiple methods of detection, including proteomic detection methods. A readily available method of detection and a standardized antibody for 14-3-3ζ staining would strengthen results across studies. Our laboratory has successfully used standard immunohistochemisty (IHC) methods for paraffin embedded tissues with the 14-3-3ζ clone C-16 from Santa Cruz. Validation of this antibody in our studies has shown this antibody to be specific for 14-3-3ζ in both western blotting and IHC staining 56. In addition, a standardized method for scoring 14-3-3ζ expression in tissues is needed to further compare results across studies.
14-3-3ζ has been demonstrated to be a promising biomarker to identify high-risk patients for more aggressive and alternative therapy at earlier stages of cancer progression. Since 14-3-3ζ is overexpressed in diverse tumor types, many patients will benefit from the validation of 14-3-3ζ as a prognosis and chemoresistance marker. The development of clinical assays to detect 14-3-3ζ expression in multiple tumor types will provide invaluable information for disease progression and treatment course in cancer patients.
8.3. Promising Therapeutic target
The overexpression of 14-3-3ζ in multiple types of cancers and its significant association with poor prognosis indicate an immediate need to target 14-3-3ζ. 14-3-3ζ impacts multiple pathways and may act as a central hub for controlling numerous targets implemented in cancer progression (Fig. 1). In this “Spoke and Hub” hypothesis, a disruption at the hub would create interference in many cellular pathways. Previous in vitro studies utilizing siRNA and the 14-3-3 peptide inhibitor, difopein, have indicated 14-3-3ζ as a potential target for cancer therapy. However, difopein targets all isoforms and has only been used in experimental studies. Targeting 14-3-3ζ with siRNA has been an effective and isoform specific method for sensitizing cancer cells to apoptosis in experimental studies; however, the use of siRNA in the clinic is still under investigation and is hampered by inefficient delivery methods 84, 93. The invention of new 14-3-3ζ–targeting agents/methods is an underdeveloped area of study.
Targeting 14-3-3 isoforms represents a major challenge since 14-3-3 does not have catalytic activities. Since 14-3-3 has been shown to regulate many targets, it is likely to have an important role in many normal cellular functions. Therefore, global inhibition of 14-3-3 functions might have unknown consequences in normal cells. Ideally, molecules that target 14-3-3 would be isoform specific and disrupt their dimerization or binding to specific targets. The protein structures of all 14-3-3 isoforms have been resolved and should facilitate the development of 14-3-3-targeting agents; however, this is a field in the early stages of development 90.
Alternatively, instead of targeting 14-3-3ζ, therapies could be directed towards targets known to be regulated by 14-3-3ζ. For example, many Akt downstream targets associated with cell survival are 14-3-3 biding partners. We have shown that 14-3-3ζ overexpression in mammary epithelial cells can activate Akt 63. Furthermore, we have found that 14-3-3ζ can activate PI3K, the upstream regulator of Akt (unpublished observation). This suggests that targeting the PI3K/Akt pathway in 14-3-3ζ overexpressing tumors may be an effective therapy. In addition, we have recently found that 14-3-3ζ can stabilize and activate the TGF receptor I, leading to activation of downstream signaling and EMT. The TGF pathway also represents a potential target for treating 14-3-3ζ overexpressing tumors 94. Targeting pathways activated by 14-3-3ζ would accelerate the discovery of therapies that may be effective in patients with 14-3-3ζ overexpressing tumors. This is an area of research which needs more focus and may bring the greatest benefit to patients with 14-3-3ζ overexpressing tumors.
Article Highlights.
14-3-3s, a highly conserved family of phospho-serine binding proteins, bind and regulate numerous targets involved in regulating multiple cellular pathways.
Overexpression of 14-3-3ζ is associated with diverse cancer types and regulates pathways that promote cancer initiation and progression.
14-3-3ζ overexpression and gene amplification are correlated with poor prognosis and chemoresistance in cancer patients.
14-3-3ζ is a potential therapeutic target in multiple cancers.
Development of 14-3-3ζ targeting agents is an under developed area of study.
Acknowledgments
The authors would like to thank Dr. Brian Pickering and Yu lab members for valuable comments on this manuscript.
This work was supported by National Institutes of Health grants RO1-CA112567 (DY), PO1-CA099031 project 4 (DY), DOD Synergistic Award W81XWH-08-1-0712 (DY), and Susan G. Komen Breast Cancer Foundation Promise Grant KG091020 (DY). Dr. D. Yu is the Hubert L. & Olive Stringer Distinguished Chair in Basic Science at M. D. Anderson Cancer Center.
Footnotes
The authors declare no conflict of interest
We apologize for not being able to cite all the relevant original research and review articles due to space limitation.
References
- 1.Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000 Jan 7;100(1):57–70. doi: 10.1016/s0092-8674(00)81683-9. [DOI] [PubMed] [Google Scholar]
- 2.Tzivion G, Gupta VS, Kaplun L, Balan V. 14-3-3 proteins as potential oncogenes. Semin Cancer Biol. 2006 Jun;16(3):203–13. doi: 10.1016/j.semcancer.2006.03.004. [DOI] [PubMed] [Google Scholar]
- 3.Kjarland E, Keen TJ, Kleppe R. Does isoform diversity explain functional differences in the 14-3-3 protein family? Curr Pharm Biotechnol. 2006 Jun;7(3):217–23. doi: 10.2174/138920106777549777. [DOI] [PubMed] [Google Scholar]
- 4.Aitken A. 14-3-3 proteins: a historic overview. Semin Cancer Biol. 2006 Jun;16(3):162–72. doi: 10.1016/j.semcancer.2006.03.005. [DOI] [PubMed] [Google Scholar]
- 5.Moore BW, Perez VJ. In: Physiological and biochemical aspects of nervous integration. Carlson F, editor. Englewood Cliffs, New Jersey: Prentice-Hall; 1967. pp. 343–59. [Google Scholar]
- 6.Gardino AK, Smerdon SJ, Yaffe MB. 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 Jun;16(3):173–82. doi: 10.1016/j.semcancer.2006.03.007. [DOI] [PubMed] [Google Scholar]
- 7.Chaudhri M, Scarabel M, Aitken A. Mammalian and yeast 14-3-3 isoforms form distinct patterns of dimers in vivo. Biochem Biophys Res Commun. 2003 Jan 17;300(3):679–85. doi: 10.1016/s0006-291x(02)02902-9. [DOI] [PubMed] [Google Scholar]
- 8.Aitken A. Functional specificity in 14-3-3 isoform interactions through dimer formation and phosphorylation. Chromosome location of mammalian isoforms and variants. Plant Mol Biol. 2002 Dec;50(6):993–1010. doi: 10.1023/a:1021261931561. [DOI] [PubMed] [Google Scholar]
- 9.Shen YH, Godlewski J, Bronisz A, Zhu J, Comb MJ, Avruch J, et al. Significance of 14-3-3 self-dimerization for phosphorylation-dependent target binding. Mol Biol Cell. 2003 Nov;14(11):4721–33. doi: 10.1091/mbc.E02-12-0821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Tzivion G, Luo Z, Avruch J. A dimeric 14-3-3 protein is an essential cofactor for Raf kinase activity. Nature. 1998 Jul 2;394(6688):88–92. doi: 10.1038/27938. [DOI] [PubMed] [Google Scholar]
- 11.Yaffe MB, Rittinger K, Volinia S, Caron PR, Aitken A, Leffers H, et al. The structural basis for 14-3-3:phosphopeptide binding specificity. Cell. 1997 Dec 26;91(7):961–71. doi: 10.1016/s0092-8674(00)80487-0. [DOI] [PubMed] [Google Scholar]
- 12.Rittinger K, Budman J, Xu J, Volinia S, Cantley LC, Smerdon SJ, et al. Structural analysis of 14-3-3 phosphopeptide complexes identifies a dual role for the nuclear export signal of 14-3-3 in ligand binding. Mol Cell. 1999 Aug;4(2):153–66. doi: 10.1016/s1097-2765(00)80363-9. [DOI] [PubMed] [Google Scholar]
- 13.Masters SC, Pederson KJ, Zhang L, Barbieri JT, Fu H. Interaction of 14-3-3 with a nonphosphorylated protein ligand, exoenzyme S of Pseudomonas aeruginosa. Biochemistry. 1999 Apr 20;38(16):5216–21. doi: 10.1021/bi982492m. [DOI] [PubMed] [Google Scholar]
- 14.Han DC, Rodriguez LG, Guan JL. Identification of a novel interaction between integrin beta1 and 14-3-3beta. Oncogene. 2001 Jan 18;20(3):346–57. doi: 10.1038/sj.onc.1204068. [DOI] [PubMed] [Google Scholar]
- 15.Obsilova V, Herman P, Vecer J, Sulc M, Teisinger J, Obsil T. 14-3-3zeta C-terminal stretch changes its conformation upon ligand binding and phosphorylation at Thr232. J Biol Chem. 2004 Feb 6;279(6):4531–40. doi: 10.1074/jbc.M306939200. [DOI] [PubMed] [Google Scholar]
- 16.Messaritou G, Grammenoudi S, Skoulakis EM. Dimerization is essential for 14-3-3zeta stability and function in vivo. J Biol Chem. 2010 Jan 15;285(3):1692–700. doi: 10.1074/jbc.M109.045989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Ellis JJ, Valencia TG, Zeng H, Roberts LD, Deaton RA, Grant SR. CaM kinase IIdeltaC phosphorylation of 14-3-3beta in vascular smooth muscle cells: activation of class II HDAC repression. Mol Cell Biochem. 2003 Jan;242(1–2):153–61. [PubMed] [Google Scholar]
- 18.Tsuruta F, Sunayama J, Mori Y, Hattori S, Shimizu S, Tsujimoto Y, et al. JNK promotes Bax translocation to mitochondria through phosphorylation of 14-3-3 proteins. Embo J. 2004 Apr 8; doi: 10.1038/sj.emboj.7600194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Van Hemert MJ, Niemantsverdriet M, Schmidt T, Backendorf C, Spaink HP. Isoform-specific differences in rapid nucleocytoplasmic shuttling cause distinct subcellular distributions of 14-3-3{sigma} and 14-3-3{zeta} J Cell Sci. 2004 Mar 2; doi: 10.1242/jcs.00990. [DOI] [PubMed] [Google Scholar]
- 20.Tsukamoto Y, Nakada C, Noguchi T, Tanigawa M, Nguyen LT, Uchida T, et al. MicroRNA-375 is downregulated in gastric carcinomas and regulates cell survival by targeting PDK1 and 14-3-3zeta. Cancer Res. 2010 Mar 15;70(6):2339–49. doi: 10.1158/0008-5472.CAN-09-2777. [DOI] [PubMed] [Google Scholar]
- 21.Obsilova V, Silhan J, Boura E, Teisinger J, Obsil T. 14-3-3 proteins: a family of versatile molecular regulators. Physiol Res. 2008;57( Suppl 3):S11–21. doi: 10.33549/physiolres.931598. [DOI] [PubMed] [Google Scholar]
- 22.Aitken A, Ellis CA, Harris A, Sellers LA, Toker A. Kinase and neurotransmitters. Nature. 1990 Apr 12;344(6267):594. doi: 10.1038/344594a0. [DOI] [PubMed] [Google Scholar]
- 23.Jin J, Smith FD, Stark C, Wells CD, Fawcett JP, Kulkarni S, et al. Proteomic, functional, and domain-based analysis of in vivo 14-3-3 binding proteins involved in cytoskeletal regulation and cellular organization. Curr Biol. 2004 Aug 24;14(16):1436–50. doi: 10.1016/j.cub.2004.07.051. [DOI] [PubMed] [Google Scholar]
- 24.Pozuelo Rubio M, Geraghty KM, Wong BH, Wood NT, Campbell DG, Morrice N, et al. 14-3-3-affinity purification of over 200 human phosphoproteins reveals new links to regulation of cellular metabolism, proliferation and trafficking. Biochem J. 2004 Apr 15;379(Pt 2):395–408. doi: 10.1042/BJ20031797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Wilker E, Yaffe MB. 14-3-3 Proteins--a focus on cancer and human disease. J Mol Cell Cardiol. 2004 Sep;37(3):633–42. doi: 10.1016/j.yjmcc.2004.04.015. [DOI] [PubMed] [Google Scholar]
- 26.Morrison DK. The 14-3-3 proteins: integrators of diverse signaling cues that impact cell fate and cancer development. Trends Cell Biol. 2009 Jan;19(1):16–23. doi: 10.1016/j.tcb.2008.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Hermeking H, Benzinger A. 14-3-3 proteins in cell cycle regulation. Semin Cancer Biol. 2006 Jun;16(3):183–92. doi: 10.1016/j.semcancer.2006.03.002. [DOI] [PubMed] [Google Scholar]
- 28.Pozuelo Rubio M, Peggie M, Wong BH, 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 Jul 15;22(14):3514–23. doi: 10.1093/emboj/cdg363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Santoro MM, Gaudino G, Marchisio PC. The MSP receptor regulates alpha6beta4 and alpha3beta1 integrins via 14-3-3 proteins in keratinocyte migration. Dev Cell. 2003 Aug;5(2):257–71. doi: 10.1016/s1534-5807(03)00201-6. [DOI] [PubMed] [Google Scholar]
- 30.Suzuki A, Hirata M, Kamimura K, Maniwa R, Yamanaka T, Mizuno K, et al. aPKC acts upstream of PAR-1b in both the establishment and maintenance of mammalian epithelial polarity. Curr Biol. 2004 Aug 24;14(16):1425–35. doi: 10.1016/j.cub.2004.08.021. [DOI] [PubMed] [Google Scholar]
- 31.Hermeking H. The 14-3-3 cancer connection. Nat Rev Cancer. 2003 Dec;3(12):931–43. doi: 10.1038/nrc1230. [DOI] [PubMed] [Google Scholar]
- 32.Subramanian RR, Masters SC, Zhang H, Fu H. Functional conservation of 14-3-3 isoforms in inhibiting bad-induced apoptosis. Exp Cell Res. 2001 Nov 15;271(1):142–51. doi: 10.1006/excr.2001.5376. [DOI] [PubMed] [Google Scholar]
- 33.Aitken A, Baxter H, Dubois T, Clokie S, Mackie S, Mitchell K, et al. Specificity of 14-3-3 isoform dimer interactions and phosphorylation. Biochem Soc Trans. 2002 Aug;30(4):351–60. doi: 10.1042/bst0300351. [DOI] [PubMed] [Google Scholar]
- 34.Iwata N, Yamamoto H, Sasaki S, Itoh F, Suzuki H, Kikuchi T, et al. Frequent hypermethylation of CpG islands and loss of expression of the 14-3-3 sigma gene in human hepatocellular carcinoma. Oncogene. 2000 Nov 2;19(46):5298–302. doi: 10.1038/sj.onc.1203898. [DOI] [PubMed] [Google Scholar]
- 35.Osada H, Tatematsu Y, Yatabe Y, Nakagawa T, Konishi H, Harano T, et al. Frequent and histological type-specific inactivation of 14-3-3sigma in human lung cancers. Oncogene. 2002 Apr 4;21(15):2418–24. doi: 10.1038/sj.onc.1205303. [DOI] [PubMed] [Google Scholar]
- 36.Ferguson AT, Evron E, Umbricht CB, Pandita TK, Chan TA, Hermeking H, et al. High frequency of hypermethylation at the 14-3-3 sigma locus leads to gene silencing in breast cancer. Proc Natl Acad Sci U S A. 2000 May 23;97(11):6049–54. doi: 10.1073/pnas.100566997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Yang HY, Wen YY, Chen CH, Lozano G, Lee MH. 14-3-3 sigma positively regulates p53 and suppresses tumor growth. Mol Cell Biol. 2003 Oct;23(20):7096–107. doi: 10.1128/MCB.23.20.7096-7107.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Li Z, Liu JY, Zhang JT. 14-3-3sigma, the double-edged sword of human cancers. Am J Transl Res. 2009;1(4):326–40. [PMC free article] [PubMed] [Google Scholar]
- 39.Sinha P, Kohl S, Fischer J, Hutter G, Kern M, Kottgen E, et al. Identification of novel proteins associated with the development of chemoresistance in malignant melanoma using two-dimensional electrophoresis. Electrophoresis. 2000 Aug;21(14):3048–57. doi: 10.1002/1522-2683(20000801)21:14<3048::AID-ELPS3048>3.0.CO;2-W. [DOI] [PubMed] [Google Scholar]
- 40.Vazquez A, Grochola LF, Bond EE, Levine AJ, Taubert H, Muller TH, et al. Chemosensitivity profiles identify polymorphisms in the p53 network genes 14-3-3tau and CD44 that affect sarcoma incidence and survival. Cancer Res. 2010 Jan 1;70(1):172–80. doi: 10.1158/0008-5472.CAN-09-2218. [DOI] [PubMed] [Google Scholar]
- 41.Sugiyama A, Miyagi Y, Komiya Y, Kurabe N, Kitanaka C, Kato N, et al. Forced expression of antisense 14-3-3beta RNA suppresses tumor cell growth in vitro and in vivo. Carcinogenesis. 2003 Sep;24(9):1549–59. doi: 10.1093/carcin/bgg113. [DOI] [PubMed] [Google Scholar]
- 42.Takihara Y, Matsuda Y, Hara J. Role of the beta isoform of 14-3-3 proteins in cellular proliferation and oncogenic transformation. Carcinogenesis. 2000 Nov;21(11):2073–7. doi: 10.1093/carcin/21.11.2073. [DOI] [PubMed] [Google Scholar]
- 43.Freed E, McCormick F, Ruggieri R. Proteins of the 14-3-3 family associate with Raf and contribute to its activation. Cold Spring Harb Symp Quant Biol. 1994;59:187–93. doi: 10.1101/sqb.1994.059.01.023. [DOI] [PubMed] [Google Scholar]
- 44.Pallas DC, Fu H, Haehnel LC, Weller W, Collier RJ, Roberts TM. Association of polyomavirus middle tumor antigen with 14-3-3 proteins. Science. 1994 Jul 22;265(5171):535–7. doi: 10.1126/science.8036498. [DOI] [PubMed] [Google Scholar]
- 45.Reuther GW, Fu H, Cripe LD, Collier RJ, Pendergast AM. Association of the protein kinases c-Bcr and Bcr-Abl with proteins of the 14-3-3 family. Science. 1994 Oct 7;266(5182):129–33. doi: 10.1126/science.7939633. [DOI] [PubMed] [Google Scholar]
- 46.Yoshida K, Yamaguchi T, Natsume T, Kufe D, Miki Y. JNK phosphorylation of 14-3-3 proteins regulates nuclear targeting of c-Abl in the apoptotic response to DNA damage. Nat Cell Biol. 2005 Mar;7(3):278–85. doi: 10.1038/ncb1228. [DOI] [PubMed] [Google Scholar]
- 47.Seimiya H, Sawada H, Muramatsu Y, Shimizu M, Ohko K, Yamane K, et al. Involvement of 14-3-3 proteins in nuclear localization of telomerase. Embo J. 2000 Jun 1;19(11):2652–61. doi: 10.1093/emboj/19.11.2652. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Zha J, Harada H, Yang E, Jockel J, Korsmeyer SJ. Serine phosphorylation of death agonist BAD in response to survival factor results in binding to 14-3-3 not BCL-X(L) Cell. 1996 Nov 15;87(4):619–28. doi: 10.1016/s0092-8674(00)81382-3. [DOI] [PubMed] [Google Scholar]
- 49.Brunet A, Bonni A, Zigmond MJ, Lin MZ, Juo P, Hu LS, et al. Akt promotes cell survival by phosphorylating and inhibiting a Forkhead transcription factor. Cell. 1999 Mar 19;96(6):857–68. doi: 10.1016/s0092-8674(00)80595-4. [DOI] [PubMed] [Google Scholar]
- 50.Fang D, Hawke D, Zheng Y, Xia Y, Meisenhelder J, Nika H, et al. Phosphorylation of beta-catenin by AKT promotes beta-catenin transcriptional activity. J Biol Chem. 2007 Apr 13;282(15):11221–9. doi: 10.1074/jbc.M611871200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Tian Q, Feetham MC, Tao WA, He XC, Li L, Aebersold R, et al. Proteomic analysis identifies that 14-3-3zeta interacts with beta-catenin and facilitates its activation by Akt. Proc Natl Acad Sci U S A. 2004 Oct 26;101(43):15370–5. doi: 10.1073/pnas.0406499101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Li S, Resnicoff M, Baserga R. Effect of mutations at serines 1280-1283 on the mitogenic and transforming activities of the insulin-like growth factor I receptor. J Biol Chem. 1996 May 24;271(21):12254–60. doi: 10.1074/jbc.271.21.12254. [DOI] [PubMed] [Google Scholar]
- 53.Spence SL, Dey BR, Terry C, Albert P, Nissley P, Furlanetto RW. Interaction of 14-3-3 proteins with the insulin-like growth factor I receptor (IGFIR): evidence for a role of 14-3-3 proteins in IGFIR signaling. Biochem Biophys Res Commun. 2003 Dec 26;312(4):1060–6. doi: 10.1016/j.bbrc.2003.11.043. [DOI] [PubMed] [Google Scholar]
- 54.Nellist M, Goedbloed MA, de Winter C, Verhaaf B, Jankie A, Reuser AJ, et al. Identification and characterization of the interaction between tuberin and 14-3-3zeta. J Biol Chem. 2002 Oct 18;277(42):39417–24. doi: 10.1074/jbc.M204802200. [DOI] [PubMed] [Google Scholar]
- 55.Shumway SD, Li Y, Xiong Y. 14-3-3beta binds to and negatively regulates the tuberous sclerosis complex 2 (TSC2) tumor suppressor gene product, tuberin. J Biol Chem. 2003 Jan 24;278(4):2089–92. doi: 10.1074/jbc.C200499200. [DOI] [PubMed] [Google Scholar]
- 56.Neal CL, Yao J, Yang W, Zhou X, Nguyen NT, Lu J, et al. 14-3-3zeta overexpression defines high risk for breast cancer recurrence and promotes cancer cell survival. Cancer Res. 2009 Apr 15;69(8):3425–32. doi: 10.1158/0008-5472.CAN-08-2765. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Zang L, Palmer Toy D, Hancock WS, Sgroi DC, Karger BL. Proteomic analysis of ductal carcinoma of the breast using laser capture microdissection, LC-MS, and 16O/18O isotopic labeling. J Proteome Res. 2004 May-Jun;3(3):604–12. doi: 10.1021/pr034131l. [DOI] [PubMed] [Google Scholar]
- 58.Niemantsverdriet M, Wagner K, Visser M, Backendorf C. Cellular functions of 14-3-3 zeta in apoptosis and cell adhesion emphasize its oncogenic character. Oncogene. 2008 Feb 21;27(9):1315–9. doi: 10.1038/sj.onc.1210742. [DOI] [PubMed] [Google Scholar]
- 59.Li Z, Zhao J, Du Y, Park HR, Sun SY, Bernal-Mizrachi L, et al. Down-regulation of 14-3-3zeta suppresses anchorage-independent growth of lung cancer cells through anoikis activation. Proc Natl Acad Sci U S A. 2008 Jan 8;105(1):162–7. doi: 10.1073/pnas.0710905105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Matta A, Bahadur S, Duggal R, Gupta SD, Ralhan R. Over-expression of 14-3-3zeta is an early event in oral cancer. BMC Cancer. 2007;7:169. doi: 10.1186/1471-2407-7-169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Bajpai U, Sharma R, Kausar T, Dattagupta S, Chattopadhayay TK, Ralhan R. Clinical significance of 14-3-3 zeta in human esophageal cancer. Int J Biol Markers. 2008 Oct-Dec;23(4):231–7. doi: 10.1177/172460080802300406. [DOI] [PubMed] [Google Scholar]
- 62.Ralhan R, Desouza LV, Matta A, Chandra Tripathi S, Ghanny S, Dattagupta S, et al. iTRAQ-multidimensional liquid chromatography and tandem mass spectrometry-based identification of potential biomarkers of oral epithelial dysplasia and novel networks between inflammation and premalignancy. J Proteome Res. 2009 Jan;8(1):300–9. doi: 10.1021/pr800501j. [DOI] [PubMed] [Google Scholar]
- 63.Danes CG, Wyszomierski SL, Lu J, Neal CL, Yang W, Yu D. 14-3-3 zeta down-regulates p53 in mammary epithelial cells and confers luminal filling. Cancer Res. 2008 Mar 15;68(6):1760–7. doi: 10.1158/0008-5472.CAN-07-3177. [DOI] [PubMed] [Google Scholar]
- 64.Wulfkuhle JD, Sgroi DC, Krutzsch H, McLean K, McGarvey K, Knowlton M, et al. Proteomics of human breast ductal carcinoma in situ. Cancer Res. 2002 Nov 15;62(22):6740–9. [PubMed] [Google Scholar]
- 65.Burstein HJ, Polyak K, Wong JS, Lester SC, Kaelin CM. Ductal carcinoma in situ of the breast. N Engl J Med. 2004 Apr 1;350(14):1430–41. doi: 10.1056/NEJMra031301. [DOI] [PubMed] [Google Scholar]
- 66.Lu J, Guo H, Treekitkarnmongkol W, Li P, Zhang J, Shi B, et al. 14-3-3zeta Cooperates with ErbB2 to promote ductal carcinoma in situ progression to invasive breast cancer by inducing epithelial-mesenchymal transition. Cancer Cell. 2009 Sep 8;16(3):195–207. doi: 10.1016/j.ccr.2009.08.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Matta A, DeSouza LV, Shukla NK, Gupta SD, Ralhan R, Siu KW. Prognostic significance of head-and-neck cancer biomarkers previously discovered and identified using iTRAQ-labeling and multidimensional liquid chromatography-tandem mass spectrometry. J Proteome Res. 2008 May;7(5):2078–87. doi: 10.1021/pr7007797. [DOI] [PubMed] [Google Scholar]
- 68.Fan T, Li R, Todd NW, Qiu Q, Fang HB, Wang H, et al. Up-regulation of 14-3-3zeta in lung cancer and its implication as prognostic and therapeutic target. Cancer Res. 2007 Aug 15;67(16):7901–6. doi: 10.1158/0008-5472.CAN-07-0090. [DOI] [PubMed] [Google Scholar]
- 69.Pollack JR, Sorlie T, Perou CM, Rees CA, Jeffrey SS, Lonning PE, et al. Microarray analysis reveals a major direct role of DNA copy number alteration in the transcriptional program of human breast tumors. Proc Natl Acad Sci U S A. 2002 Oct 1;99(20):12963–8. doi: 10.1073/pnas.162471999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Garnis C, Coe BP, Ishkanian A, Zhang L, Rosin MP, Lam WL. Novel regions of amplification on 8q distinct from the MYC locus and frequently altered in oral dysplasia and cancer. Genes Chromosomes Cancer. 2004 Jan;39(1):93–8. doi: 10.1002/gcc.10294. [DOI] [PubMed] [Google Scholar]
- 71.Heidenblad M, Lindgren D, Jonson T, Liedberg F, Veerla S, Chebil G, et al. Tiling resolution array CGH and high density expression profiling of urothelial carcinomas delineate genomic amplicons and candidate target genes specific for advanced tumors. BMC Med Genomics. 2008;1:3. doi: 10.1186/1755-8794-1-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Lin M, Morrison CD, Jones S, Mohamed N, Bacher J, Plass C. Copy number gain and oncogenic activity of YWHAZ/14-3-3zeta in head and neck squamous cell carcinoma. Int J Cancer. 2009 Aug 1;125(3):603–11. doi: 10.1002/ijc.24346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Li Y, Zou L, Li Q, Haibe-Kains B, Tian R, Desmedt C, et al. Amplification of LAPTM4B and YWHAZ contributes to chemotherapy resistance and recurrence of breast cancer. Nat Med. 2010 Feb;16(2):214–8. doi: 10.1038/nm.2090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Frasor J, Chang EC, Komm B, Lin CY, Vega VB, Liu ET, et al. Gene expression preferentially regulated by tamoxifen in breast cancer cells and correlations with clinical outcome. Cancer Res. 2006 Jul 15;66(14):7334–40. doi: 10.1158/0008-5472.CAN-05-4269. [DOI] [PubMed] [Google Scholar]
- 75.Maxwell SA, Li Z, Jaya D, Ballard S, Ferrell J, Fu H. 14-3-3zeta mediates resistance of diffuse large B cell lymphoma to an anthracycline-based chemotherapeutic regimen. J Biol Chem. 2009 Aug 14;284(33):22379–89. doi: 10.1074/jbc.M109.022418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Chatterjee D, Goldman M, Braastad CD, Darnowski J, Wyche JH, Pantazis P, et al. Reduction of 9-nitrocamptothecin-triggered apoptosis in DU-145 human prostate cancer cells by ectopic expression of 14-3-3zeta. Int J Oncol. 2004 Aug;25(2):503–9. [PubMed] [Google Scholar]
- 77.Andrews RK, Du X, Berndt MC. The 14-3-3zeta-GPIb-IX-V complex as an antiplatelet target. Drug News Perspect. 2007 Jun;20(5):285–92. doi: 10.1358/dnp.2007.20.5.1120215. [DOI] [PubMed] [Google Scholar]
- 78.Altar CA, Vawter MP, Ginsberg SD. Target identification for CNS diseases by transcriptional profiling. Neuropsychopharmacology. 2009 Jan;34(1):18–54. doi: 10.1038/npp.2008.172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Watanabe K, Thandavarayan RA, Gurusamy N, Zhang S, Muslin AJ, Suzuki K, et al. Role of 14-3-3 protein and oxidative stress in diabetic cardiomyopathy. Acta Physiol Hung. 2009 Sep;96(3):277–87. doi: 10.1556/APhysiol.96.2009.3.3. [DOI] [PubMed] [Google Scholar]
- 80.Elbashir SM, Harborth J, Lendeckel W, Yalcin A, Weber K, Tuschl T. Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells. Nature. 2001 May 24;411(6836):494–8. doi: 10.1038/35078107. [DOI] [PubMed] [Google Scholar]
- 81.Scacheri PC, Rozenblatt-Rosen O, Caplen NJ, Wolfsberg TG, Umayam L, Lee JC, et al. Short interfering RNAs can induce unexpected and divergent changes in the levels of untargeted proteins in mammalian cells. Proc Natl Acad Sci U S A. 2004 Feb 17;101(7):1892–7. doi: 10.1073/pnas.0308698100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Qi W, Martinez JD. Reduction of 14-3-3 proteins correlates with increased sensitivity to killing of human lung cancer cells by ionizing radiation. Radiat Res. 2003 Aug;160(2):217–23. doi: 10.1667/rr3038. [DOI] [PubMed] [Google Scholar]
- 83.Ge F, Lu XP, Zeng HL, He QY, Xiong S, Jin L. Proteomic and functional analyses reveal a dual molecular mechanism underlying arsenic-induced apoptosis in human multiple myeloma cells. J Proteome Res. 2009 Jun;8(6):3006–19. doi: 10.1021/pr9001004. [DOI] [PubMed] [Google Scholar]
- 84.Guo J, Fisher KA, Darcy R, Cryan JF, O’Driscoll C. Therapeutic targeting in the silent era: advances in non-viral siRNA delivery. Mol Biosyst. 2010 Apr 29; doi: 10.1039/c001050m. [DOI] [PubMed] [Google Scholar]
- 85.Wang B, Yang H, Liu YC, Jelinek T, Zhang L, Ruoslahti E, et al. Isolation of high-affinity peptide antagonists of 14-3-3 proteins by phage display. Biochemistry. 1999 Sep 21;38(38):12499–504. doi: 10.1021/bi991353h. [DOI] [PubMed] [Google Scholar]
- 86.Masters SC, Fu H. 14-3-3 proteins mediate an essential anti-apoptotic signal. J Biol Chem. 2001 Nov 30;276(48):45193–200. doi: 10.1074/jbc.M105971200. [DOI] [PubMed] [Google Scholar]
- 87.Masters SC, Subramanian RR, Truong A, Yang H, Fujii K, Zhang H, et al. Survival-promoting functions of 14-3-3 proteins. Biochem Soc Trans. 2002 Aug;30(4):360–5. doi: 10.1042/bst0300360. [DOI] [PubMed] [Google Scholar]
- 88.Dong S, Kang S, Lonial S, Khoury HJ, Viallet J, Chen J. Targeting 14-3-3 sensitizes native and mutant BCR-ABL to inhibition with U0126, rapamycin and Bcl-2 inhibitor GX15-070. Leukemia. 2008 Mar;22(3):572–7. doi: 10.1038/sj.leu.2405064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Cao W, Yang X, Zhou J, Teng Z, Cao L, Zhang X, et al. Targeting 14-3-3 protein, difopein induces apoptosis of human glioma cells and suppresses tumor growth in mice. Apoptosis. 2010 Feb;15(2):230–41. doi: 10.1007/s10495-009-0437-4. [DOI] [PubMed] [Google Scholar]
- 90.Du Y, Masters SC, Khuri FR, Fu H. Monitoring 14-3-3 protein interactions with a homogeneous fluorescence polarization assay. J Biomol Screen. 2006 Apr;11(3):269–76. doi: 10.1177/1087057105284862. [DOI] [PubMed] [Google Scholar]
- 91.Wu H, Ge J, Yao SQ. Microarray-assisted high-throughput identification of a cell-permeable small-molecule binder of 14-3-3 proteins. Angew Chem Int Ed Engl. 2010 Sep 3;49(37):6528–32. doi: 10.1002/anie.201003257. [DOI] [PubMed] [Google Scholar]
- 92.Cullere X, Rose P, Thathamangalam U, Chatterjee A, Mullane KP, Pallas DC, et al. Serine 257 phosphorylation regulates association of polyomavirus middle T antigen with 14-3-3 proteins. J Virol. 1998 Jan;72(1):558–63. doi: 10.1128/jvi.72.1.558-563.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Ashihara E, Kawata E, Maekawa T. Future prospect of RNA interference for cancer therapies. Curr Drug Targets. 2010 Mar;11(3):345–60. doi: 10.2174/138945010790711897. [DOI] [PubMed] [Google Scholar]
- 94.Yingling JM, Blanchard KL, Sawyer JS. Development of TGF-beta signalling inhibitors for cancer therapy. Nat Rev Drug Discov. 2004 Dec;3(12):1011–22. doi: 10.1038/nrd1580. [DOI] [PubMed] [Google Scholar]
- 95.Qi W, Liu X, Qiao D, Martinez JD. Isoform-specific expression of 14-3-3 proteins in human lung cancer tissues. Int J Cancer. 2005 Jan 20;113(3):359–63. doi: 10.1002/ijc.20492. [DOI] [PubMed] [Google Scholar]
- 96.Shen J, Person MD, Zhu J, Abbruzzese JL, Li D. Protein expression profiles in pancreatic adenocarcinoma compared with normal pancreatic tissue and tissue affected by pancreatitis as detected by two-dimensional gel electrophoresis and mass spectrometry. Cancer Res. 2004 Dec 15;64(24):9018–26. doi: 10.1158/0008-5472.CAN-04-3262. [DOI] [PubMed] [Google Scholar]
- 97.Lu B, Xu J, Zhu Y, Zhang H, Lai M. Systemic analysis of the differential gene expression profile in a colonic adenoma-normal SSH library. Clin Chim Acta. 2007 Mar;378(1–2):42–7. doi: 10.1016/j.cca.2006.10.024. [DOI] [PubMed] [Google Scholar]
- 98.Sharma R, Samantaray S, Shukla NK, Ralhan R. Transcriptional gene expression profile of human esophageal squamous cell carcinoma. Genomics. 2003 May;81(5):481–8. doi: 10.1016/s0888-7543(03)00023-5. [DOI] [PubMed] [Google Scholar]
- 99.Jang JS, Cho HY, Lee YJ, Ha WS, Kim HW. The differential proteome profile of stomach cancer: identification of the biomarker candidates. Oncol Res. 2004;14(10):491–9. doi: 10.3727/0965040042380441. [DOI] [PubMed] [Google Scholar]
- 100.Arora S, Matta A, Shukla NK, Deo SV, Ralhan R. Identification of differentially expressed genes in oral squamous cell carcinoma. Mol Carcinog. 2005 Feb;42(2):97–108. doi: 10.1002/mc.20048. [DOI] [PubMed] [Google Scholar]
- 101.Ralhan R, Desouza LV, Matta A, Chandra Tripathi S, Ghanny S, Datta Gupta S, et al. Discovery and verification of head-and-neck cancer biomarkers by differential protein expression analysis using iTRAQ labeling, multidimensional liquid chromatography, and tandem mass spectrometry. Mol Cell Proteomics. 2008 Jun;7(6):1162–73. doi: 10.1074/mcp.M700500-MCP200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Liang S, Xu Y, Shen G, Liu Q, Zhao X, Xu Z, et al. 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 Dec;30(23):4152–62. doi: 10.1002/elps.200900249. [DOI] [PubMed] [Google Scholar]
- 103.Yang X, Cao W, Lin H, Zhang W, Lin W, Cao L, et al. Isoform-specific expression of 14-3-3 proteins in human astrocytoma. J Neurol Sci. 2009 Jan 15;276(1–2):54–9. doi: 10.1016/j.jns.2008.08.040. [DOI] [PubMed] [Google Scholar]
- 104.Cao L, Cao W, Zhang W, Lin H, Yang X, Zhen H, et al. Identification of 14-3-3 protein isoforms in human astrocytoma by immunohistochemistry. Neurosci Lett. 2008 Feb 20;432(2):94–9. doi: 10.1016/j.neulet.2007.11.071. [DOI] [PubMed] [Google Scholar]
- 105.Liu Y, Tian RF, Li YM, Liu WP, Cao L, Yang XL, et al. The expression of seven 14-3-3 isoforms in human meningioma. Brain Res. 2010 Jun 8;1336:98–102. doi: 10.1016/j.brainres.2010.04.009. [DOI] [PubMed] [Google Scholar]
- 106.Janssen JJ, Klaver SM, Waisfisz Q, Pasterkamp G, de Kleijn DP, Schuurhuis GJ, et al. Identification of genes potentially involved in disease transformation of CML. Leukemia. 2005 Jun;19(6):998–1004. doi: 10.1038/sj.leu.2403735. [DOI] [PubMed] [Google Scholar]
- 107.Huber E, Vlasny D, Jeckel S, Stubenrauch F, Iftner T. Gene profiling of cottontail rabbit papillomavirus-induced carcinomas identifies upregulated genes directly Involved in stroma invasion as shown by small interfering RNA-mediated gene silencing. J Virol. 2004 Jul;78(14):7478–89. doi: 10.1128/JVI.78.14.7478-7489.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]

