Siah1 in cancer and nervous system diseases (Review)

  • Authors:
    • Hui Zhang
    • Jie Wang
    • Yidong Ge
    • Meng Ye
    • Xiaofeng Jin
  • View Affiliations

  • Published online on: December 23, 2021     https://doi.org/10.3892/or.2021.8246
  • Article Number: 35
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The dysregulation of the ubiquitin‑proteasome system will result in the abnormal accumulation and dysfunction of proteins, thus leading to severe diseases. Seven in absentia homolog 1 (Siah1), an E3 ubiquitin ligase, has attracted wide attention due to its varied functions in physiological and pathological conditions, and the numerous newly discovered Siah1 substrates. In cancer and nervous system diseases, the functions of Siah1 as a promoter or a suppressor of diseases are related to the change in cellular microenvironment and subcellular localization. At the same time, complex upstream regulations make Siah1 different from other E3 ubiquitin ligases. Understanding the molecular mechanism of Siah1 will help the study of various signaling pathways and benefit the therapeutic strategy of human diseases (e.g., cancer and nervous system diseases). In the present review, the functions and regulations of Siah1 are described. Moreover, novel substrates of Siah1 discovered in recent studies will be highlighted in cancer and nervous system diseases, providing ideas for future research and clinical targeted therapies using Siah1.

Introduction

Ubiquitination is a key process of the post-translational modification of proteins, playing an important role in the stability of the intracellular environment, the proliferation and differentiation of cells, and a number of cellular functions (1), whereas the imbalance of ubiquitination-mediated protein degradation is a molecular basis for human diseases. Ubiquitin is activated in an ATP-dependent reaction catalyzed by the ubiquitin-activating enzyme (E1). Under the action of the ubiquitin-conjugating enzyme (E2), the activated ubiquitin is transferred into a specific substrate along with E3 ubiquitin ligase, which is responsible for the substrate specificity for ubiquitin ligation (2,3).

Increasing attention has been focused on E3 ubiquitin ligases due to their unique functions compared with E1 and E2. E3 ubiquitin ligases regulate a range of cellular physiological processes, such as cell proliferation and differentiation, participate in DNA damage and repair, and control the cell cycle (1,46). In pathological processes (i.e., cancer and nervous system diseases), E3 ubiquitin ligases promote or suppress various diseases (7). For example, the speckle-type POZ protein (SPOP), a typical E3 ubiquitin ligase, is known as a tumor suppressor protein in prostate cancer (PC) and an oncoprotein in kidney cancer (8). In addition, E3 ubiquitin ligases with high-frequency mutations (mutations in SPOP occur in up to 11.13% of PC cases; The Cancer Genome Atlas, http://portal.gdc.cancer.gov/) in diseases prove their important role in the development of disease (4,7).

Currently, E3 ubiquitin ligases can be classified in three main types [i.e., RING E3s (~600 in humans), homologous to the E6AP carboxyl terminus (HECT) E3s (~30 in humans) and RING-between-RING (RBR) E3s (~12 in humans)] depending on the characteristic domains and the mechanism of ubiquitin transfer to the substrates (9). Notably, some E3s, including some RING E3s, all HECT E3s and all RBR E3s, interact with E2 alone to transfer the ubiquitin to the substrates, whereas other RING E3s transfer ubiquitin by forming the ubiquitin-ligase complex (9). The mechanism of ubiquitin transfer to the substrates gets little research attention.

The seven in absentia homolog (Siah) family of proteins, which belong to the RING E3s, are the mammalian homologs of the Drosophila sina proteins, which are responsible for the ubiquitination of substrate proteins to promote functional changes or degradation of substrate proteins through the proteasomal pathway (10,11). In the human proteome, two proteins belonging to the Siah family of proteins have been identified and are known as Siah1 and Siah2. Mice have three Siah family proteins, termed Siah1a, Siah1b (collectively Siah1 due to their 98% similarity) and Siah2 (11).

Siah1 and Siah2 share high sequence similarity (86%) and presumably high structural homology. The difference between Siah1 and Siah2 is the additional amino acid sequence in the N terminal of Siah2 (1114). However, the functions of Siah1 and Siah2 are almost completely different due to their unique substrates and different affinity and types of ubiquitination for the shared substrates (11). Previous studies focused on the oncoprotein functions of Siah2 (11,15). However, in recent years, a number of novel substrates of Siah1 have been discovered, and the functions of Siah1 as an important E3 ubiquitin ligase have been gradually recognized (10,11,16,17). A systematic review summarizing the functions of Siah1 in human diseases, such as cancer and nervous system diseases, is not available. Siah1 knockout mice exhibit severe growth retardation, poor bone formation, early lethality and a block in meiotic cell division during the meiosis I of spermatogenesis (18,19). However, the Siah2 mutant or knockout mice are fertile and largely phenotypically normal. Notably, the loss of a single copy of Siah2 enhances the phenotype of early lethality caused by Siah1 homozygous mutation. This phenotype is further enhanced by the removal of both copies of Siah2, with Siah1−/−Siah2−/− mice subsequently dying within hours of birth, showing that Siah1 and Siah2 appear to perform partially overlapping functions in vivo (18,19). The functional compensation by Siah1 may maintain the normal regulation of Siah2 substrate proteins in Siah2−/− mice, suggesting the critical biological functions of Siah1 (18,19). In addition, in contrast to that of other E3 ubiquitin ligases (the regulations of SPOP have rarely been discovered), the regulations of Siah1 vary greatly (Table I; Fig. 1A), indicating that Siah1 plays a role as a bridge factor in various signaling pathways and is a promising therapeutic target (11,18). The present review will summarize the structure of Siah1 and the characteristics of its substrates, the regulations of Siah1 in physiological and pathological conditions, and its function and clinical significance in cancer and nervous system diseases to provide help for future researchers to understand Siah1.

Figure 1.

(A) The regulations of Siah1 vary greatly. At the transcriptional regulation level, p53, p21, E2F1 and HIF-1α trans-activate the transcription of Siah1. At the translational regulation level, microRNAs and lncRNAs inhibit the translation of Siah1 mRNA. At the post-translational regulation level, ASK1 induces the phosphorylation of Siah1. Overexpression of CacyBP/SIP promotes the interaction between Siah1 and cytoplasmic p27, which in turn increases the ubiquitination and degradation of cytoplasmic p27. Ubiquitin conjugase UbcH8 interacts with Siah1 to form a complex to ensure the functions of Siah1. HCF1 and HCF2 antagonize the E3 ligase activity of Siah1 through binding and blocking the substrate-binding domain. The AFF4-ELL2 interaction sequesters ELL2 away from Siah1, thereby inhibiting Siah1 ubiquitination of ELL2. (B) Siah1 consists of a N-terminal catalytic RING domain, two zinc finger domains and a C-terminal substrate binding domain that includes the first two zinc finger domains. A consensus Pro-X-Ala-X–Val-X-Pro (VxP, core sequence; where X is not conserved) motif is common to a family of SIPs (for example, CacyBP/SIP, Bim, PHD3, AXIN, HIPK2). Compared with Siah1, Siah2 has an additional amino acid sequence (~40 amino acids) at the N terminal. (C) Siah1 can interact with E2 ubiquitin-conjugating enzyme alone or become an essential part of the ubiquitin-ligase complex, which includes CacyBP/SIP, SKP1, TBL or EBI and Siah1. (D) β-catenin, AXIN1, APC and GSK-3β form a degradation complex without Wnt signaling, inducing the phosphorylation of β-catenin and finally leading to the degradation of β-catenin through the ubiquitin-proteasome pathway. The degradation complex is destroyed in response to Wnt signaling, releasing β-catenin, and thus activating transcription of downstream genes to promote the proliferation and survival of cells. Siah1 also induces the ubiquitination and proteasomal degradation of AXIN1 to promote the Wnt/β-catenin signaling pathways with Wnt signaling. TRAF4 protects β-catenin from Siah1-mediated degradation by competing with β-catenin for binding to Siah1 and replacing it for degradation. (E) The low protein level of Siah1 induces the degradation of dissociative ELL2 to prevent the formation of new SECs. The high protein level of Siah1 degrades all SECs. Siah1, seven in absentia homolog family proteins 1; Jab1, c-Jun activation domain-binding protein 1; ASK1, apoptosis signal-regulating kinase 1; UBCH8, ubiquitin conjugating enzyme human 8; Ub, ubiquitin; HCF1/2, host cell factor 1/2; P-TEFb, positive transcription elongation factor b; TRAF4, TNF receptor-associated factor 4; APC, adenomatous polyposis coli protein; GSK-3β, glycogen synthase kinase-3β; ELL2, elongation factor for RNA polymerase II 2; AFF4, AF4/FMR2 family member 4; miR, microRNA; lncRNA, long non-coding RNA; CacyBP, calcyclin-binding protein; Bim, Bcl-2-interacting mediator of cell death; PHD3, prolyl-hydroxylase protein 3; HIPK2, homeodomain-interacting protein kinase 2; SIP, Siah1-interacting protein; TBL/EBI, F-box-like/WD repeat-containing protein TBL1 or EBI.

Table I.

Regulations of Siah1 ubiquitin ligase.

Table I.

Regulations of Siah1 ubiquitin ligase.

Level of regulationRegulatorMode of regulation
Transcriptional regulationp53p53 acts directly on Siah1, to promote the transcription of Siah1.
Jab1Jab1 inhibit the expression of p53 to suppress the transcription of Siah1.
HIF-1αHIF-1α trans-activates the transcription of Siah1 by coordinating key histone modifications on the Siah1 promoter.
p21The transcription of Siah1 is directly activated by p21.
E2F1E2F1 can activate transcription from the Siah1 promoter.
Translational regulationmiR-135a, miR-424, miR-944, miR-299-5p, miR-15b-5p, miR-107MicroRNAs inhibit the translation of Siah1 mRNA by targeting the 3′UTR.
lncRNA RP11, hnRNPA2B1RP11 directly binds to the CDS of Siah1 and significantly downregulates the mRNA stability of Siah1 by forming the RP11-hnRNPA2B1-mRNA. complex
Post-translational regulationASK1Phosphorylation of Siah1 by ASK1 triggers GAPDH-Siah1 stress signaling.
CacyBP/SIPOverexpression of CacyBP/SIP promotes the interaction between Siah1 and cytoplasmic p27, which in turn increases the ubiquitination and degradation of cytoplasmic p27.
HCF1/2HCF1 and HCF2 antagonize the E3 ligase activity of Siah1 by binding and blocking the substrate-binding domain.
AFF4The AFF4-ELL2 interaction sequesters ELL2 away from Siah1 thereby inhibiting Siah1 ubiquitination of ELL2.
UBCH8Ubiquitin conjugase UbcH8 interacts with Siah1 to form a complex to ensure the function of Siah1.

[i] p53, tumor suppressor p53; Jab1, c-Jun activation domain-binding protein 1; HIF-1α, hypoxia-inducible factor 1α; p21, cyclin-dependent kinase inhibitor p21; E2F1, E2F transcription factor 1; CDS, coding sequence; ASK1, apoptosis signal-regulating kinase 1; CacyBP/SIP, calcyclin-binding protein/Siah-1-interacting protein; HCF1/2, host cell factor 1/2; AFF4, AF4/FMR2 family member 4; UBCH8, ubiquitin conjugating enzyme human 8; miR, microRNA; lncRNA, long non-coding RNA.

Structure of Siah1 and characteristics of Siah1-interacting proteins (SIPs)

Siah family proteins usually consist of an N-terminal catalytic RING domain, two zinc finger domains and a C-terminal substrate-binding domain (SBD) that includes the first two zinc finger domains (Fig. 1B) (13). The RING domain is essential for ubiquitin ligase activity, and the SBD mediates homodimerization and the interaction with substrates (14). In contrast with other E3 ubiquitin ligases, Siah1 can interact with E2 alone and become an essential part of the ubiquitin-ligase complex, which includes calcyclin-binding protein (CacyBP)/SIP, and adapters SKP1, F-box-like/WD repeat-containing protein TBL1 or EBI and Siah1 (Fig. 1C) (20). CacyBP/SIP is suggested to position the substrates and improve the affinity between Siah1 and the substrate. Thus, Siah1 is most powerful when complexed (21,22).

The characteristics of the SIPs have also been studied (13). A consensus Pro-X-Ala-X–Val-X-Pro (VxP, core sequence; where X is not conserved) motif is common to a family of SIPs (Table I) (11,14,21,2326). Some SIPs are also substrates of Siah1 and are targeted by Siah1 for degradation or functional modification. Dysregulation between Siah1 and substrates will lead to serious human diseases (e.g., cancer and nervous system diseases) (Table II) (11,15,16,18,27).

Table II.

SIPs of Siah1 ubiquitin ligase in human diseases.

Table II.

SIPs of Siah1 ubiquitin ligase in human diseases.

Disease typeSubstrateFunction of the SIPsDegradation of the substratePhysiological evidence (cell or animal model)
Cancer
  Breast cancerJNKPromotion of cell apoptosis and inhibition of MAPK signaling pathways.NoThe inhibition of Siah1 expression promotes human breast cancer cell proliferation, colony formation, migration and invasion, and inhibits apoptosis (15,23,57).
BimDNA-binding transcription factor activity.No
TRAF4Protection of β-catenin from Siah1-mediated degradation and leading chemotherapy resistance.Yes
  GliomaHIPK2Response to DNA damage and promotion of cells apoptosis.YesThe knockdown of Siah1 by shRNA severely suppresses the migration and invasion of human glioma U251 cells under hypoxia, while overexpression of Siah1 promotes it. However, when Siah1 interacts with CacyBP/SIP, the overexpression of Siah1 suppresses the migration and invasion of human glioma U251 and U87 cells (22,26).
PHD3Degradation of the HIF-1α.Yes
CacyBP/SIPThe part of Siah1 ubiquitin-ligase complexes.No
p27/kip1Negative regulation of the cell cycle.Yes
  Hepatocellular carcinomaAxin, β-cateninCell migration and cell differentiation.YesThe overexpression of Siah1 induces growth arrest and apoptosis in HepG2, SNU475 and Huh7 cells (30).
ZEB1 Epithelial-mesenchymal transition.Yes
  LeukemogenesisPML-RARaThe fusion protein of leukemia.YesIn the murine myeloblastic cell line M1 (generated from a spontaneous leukemia), expression of a stably introduced temperature-sensitive mutant of the tumor suppressor p53 activates the Siah1 gene (27,74).
ELL2An elongation factor that modulates gene expression.Yes
AML1-ETOThe fusion protein of leukemia.Yes
AF4-MLL
  Colorectal cancerAKT, YAPInhibition of cells apoptosisYesThe knockdown of Siah1 by shRNA promotes HCT116/SW480 colorectal cancer cell proliferation and migration, and results in faster tumor growth and a markedly larger tumor volume in nude mice (108).
ZEB1 Epithelial-mesenchymal transition.Yes
  OsteosarcomaZEB1 Epithelial-mesenchymal transition.YesNone.
Nervous system diseases
  Development delayAxinNeuronal development and cell differentiation.YesThe development of skin and hair follicle development in the angora rabbit is affected by the level of Siah1 protein (132).
Akt3Neuronal development.Yes
  Neuronal damageGAPDHGlycolysis and promotion of cell apoptosis.NoSiah1 is upregulated after spinal cord injury in adult rats (138,139,143).
  Parkinson's diseaseα-synucleinThe development of Parkinson's disease and the formation of LBs.NoInhibition of Siah1 by siRNA increases cell proliferation and inhibits apoptosis in SH-SY5Y neuroblastoma cells (113,115).
Synphilin-1The development of Parkinson's disease and the formation of LBs.Yes
Alzheimer's diseaseCacyBP/SIPPart of the Siah1 ubiquitin-ligase complexes and de-phosphorylation of tau protein.NoIn tau transgenic mice, localization of CacyBP/SIP and Siah1 is similar to that observed for patients with Alzheimer's disease (150).

[i] JNK, c-Jun N-terminal kinase; Bim, Bcl-2-interacting mediator of cell death; TRAF4, TNF receptor-associated factor 4; HIPK2, homeodomain-interacting protein kinase 2; PHD3, prolyl-hydroxylase proteins 3; CacyBP/SIP, calcyclin-binding protein/Siah1 interacting protein; p27/kip1, p27 kinase inhibitory protein 1; ZEB1, zinc finger E-box-binding homeobox 1; ELL2, elongation factor for RNA polymerase II 2; PML-RARα, t(15;17)(q24;q21), generating the promyelocytic leukemia-retinoic acid receptor α fusion protein; AML1-ETO, t(8;21)(q22;q22), RUNX family transcription factor 1 fusion protein; AF4-MLL, t(4;11)(q21;q23), ectopic activator of transcript initiation fusion protein; AKT, targeting protein kinase; YAP, transcriptional coactivator YAP1; AKT3, targeting protein kinase-b3; GAPDH, glyceraldehyde 3-phosphate dehydrogenase; LBs, Lewy bodies; siRNA, small interfering RNA; shRNA, small hairpin RNA.

Siah1 in cancer

Siah1 in hepatocellular carcinoma (HCC)

HCC is one of the most common malignancies worldwide (in 2020, there were 910,000 new cases of HCC worldwide, ranking sixth among all cancer types; The Cancer Genome Atlas, http://portal.gdc.cancer.gov/), with extremely high recurrence and metastasis rates (28). Siah1 was identified as one of the tumor-suppressing genes of HCC in 2003 (29). Siah1 is significantly downregulated in advanced HCC, including poorly differentiated tumors, large tumors and tumors in advanced stages (29,30).

Wnt/β-catenin signaling pathways are one of the key cascades regulating cell growth, cell development and differentiation of normal stem cells, and have also been tightly associated with cancers made up of several key proteins, including Wnt, β-catenin, AXIN1, adenomatous polyposis coli protein (APC) and glycogen synthase kinase-3β (GSK-3β) (3133). β-catenin, AXIN1, APC and GSK-3β form a degradation complex without Wnt signaling, inducing the phosphorylation of β-catenin and leading to the degradation of β-catenin through the ubiquitin proteasome pathway (3136). The degradation complex is destroyed in response to Wnt signaling, releasing β-catenin and activating the transcription of downstream genes to promote the proliferation and survival of cells (Fig. 1D) (3136). β-catenin is widely considered to be a major oncoprotein in HCC based on the frequency of mutations [15–33% of patients with HCC carry activating mutations in ctnnb1 (coding for β-catenin)] associated with aberrant Wnt/β-catenin signaling pathways in patients with HCC (37). Siah1 functions in the ubiquitination-dependent degradation of β-catenin, thus inhibiting the abnormal activation of Wnt/β-catenin signaling pathways [whether wild-type or mutant Wnt/β-catenin signaling pathways; HepG2 (β-catenin with activating mutations), SNU475 (AXIN1 with dysfunctional mutation) and Huh7 (wild-type β-catenin and AXIN1)] and promoting cell apoptosis and growth arrest of HCC cells (30).

However, some studies have reported that Siah1 also promotes Wnt/β-catenin signaling pathways by inducing the ubiquitination and proteasomal degradation of AXIN1, suggesting the positive regulation of Siah1 in Wnt/β-catenin signaling pathways (38). The positive and negative regulations of Siah1 in Wnt/β-catenin signaling pathways indicate that Siah1 plays a key role in the dynamic balance of these pathways, suggesting that targeted therapy of Siah1 for HCC is a promising but prudent choice (Fig. 1D).

Acquired chemoresistance during long-term chemotherapy is one of the most important factors to limit the application of some chemotherapy drugs, such as doxorubicin (Dox), for the clinical treatment of patients with HCC (39,40). In addition, chemotherapy-resistant HCC shows a malignant phenotype, suggesting that the changes in cancer-related factors occur in the HCC cells (41). Recent studies have shown that zinc finger E-box-binding homeobox 1 (ZEB1), a powerful epithelial mesenchymal transition-related transcription factor, is upregulated in Dox-resistant HCC cells and accompanied by a decrease in the protein expression of Siah1 (4143). ZEB1 is degraded by the proteasomal pathway as a substrate for Siah1, but the low protein level of Siah1 induces the accumulation of ZEB1 (44,45). The same phenomenon also occurs in colorectal cancer (CRC) cells and Dox-resistant osteosarcoma cells (44,46). Some studies have suggested that long non-coding RNA (lncRNA) RP11 accelerates the mRNA degradation of Siah1 in CRC, thus leading to the accumulation of ZEB1 (46). Whether the cause of ZEB1 accumulation in HCC is related to RP11 remains unclear, but the lncRNA-Siah1-ZEB1 axis may be an important target against Dox resistance and ZEB1-related cancer.

Notably, Siah1 in HCC functions as a tumor suppressor protein and as an oncoprotein (11,15). Some studies have reported that the nuclear expression of Siah1 induces proliferation and migration and prevents the apoptosis of HCC cells (47). In addition, in HCC tissues, the decrease in cytoplasmic Siah1 and the nuclear accumulation of Siah1 are positively correlated with HCC progression, suggesting that the functions of Siah1 may be closely related to subcellular localization (47). The nuclear Siah1 accumulation is significantly correlated with the expression of the transcription factor far-upstream element-binding protein 3 (FBP-3). FBP-3 predominantly supports proliferation but cannot explain the reason for HCC cell migration being affected by Siah1 (47). Thus, the mechanisms for the high expression of Siah1 in the nucleus of HCC cells and the promotion of HCC by nuclear Siah1 accumulation are still unclear and should be studied in the future.

Siah1 in breast cancer (BC)

BC is the most common cancer in women and is considered the second leading cause of cancer-related death in women (in 2020, there were 2,260,000 new cases of BC worldwide, ranking first among all cancer types of women; The Cancer Genome Atlas, http://portal.gdc.cancer.gov/) (4851). BC is a type of cancer with complex phenotypes and heterogeneity. Thus, traditional pathology has been unable to meet the needs of BC diagnosis (52,53). The accurate diagnosis of BC requires entirely new tumor markers, such as Siah1.

Siah1 was originally identified as a tumor suppressor gene in BC (5456). The expression of Siah1 is downregulated in BC tissues and is correlated with well to moderately differentiated and early stage BC (23). In addition, the inhibition of Siah1 expression promotes human BC cell proliferation, colony formation, migration and invasion, and inhibits apoptosis (15,23,57), suggesting the key role of Siah1 in the occurrence and development of BC. Some studies have further reported that Siah1 induces apoptosis, that inhibited invasion in BC cells may by upregulation of the level of Bcl-2-interacting mediator of cell death through the activation of the c-Jun NH2-terminal kinase signaling pathway, and that the suppression of Siah1 expression increases migration via the activation of the extracellular-regulated protein kinases signaling pathway (23,57).

The classification of BC is complex. Under the general trend of the development of precision medicine, oncologists prefer to classify breast cancer by molecular classification (5153). Therefore, based on the expression of the three key factors for BC [estrogen receptor (ER), progesterone receptor (PR) and the human epidermal growth factor receptor 2 (HER2) subtype], oncologists classify BC into six subtypes: Luminal A (low-grade and ER-positive), luminal B (high-grade and ER-positive), HER2-overexpressing, triple-negative BC (TNBC; lacking ER, PR and HER2), normal breast-like tumors and claudin-low (TNBC with a low expression level of cell adhesion molecules) (15,49,51,52). TNBC is the most lethal subtype of BC due to its high heterogeneity, aggressive nature and lack of treatment options (58). Notably, the expression of Siah1 is significantly decreased in TNBC cells (MDA-MB-231), and the inhibition of Siah1 expression has recently been shown to be mediated by microRNA (miRNA/miR)-107 (an overexpression miRNA in BC, especially in TNBC) (59). miR-107 is considered to be a good predictive parameter of TNBC recurrence and promotes cell proliferation, colony formation, migration, invasion and cell cycle progression in human BC cells (i.e., MCF-7 and MDA-MB-231) through the downregulation of Siah1 expression (5961). In a previous study, the inhibition of Siah1 was relieved by the silencing of miR-107, which inhibited tumor growth in a nude mouse model of TNBC. This phenomenon suggests that the miR-107-Siah1 axis will be a promising therapeutic target in TNBC (59). In addition, miR-944 exhibits a similar function to miR-107, strongly supporting the importance of the regulation of Siah1 expression by miRNA (62).

Chemotherapy is the basic treatment of BC, and has made marked progress over the last few decades, with the emergence of new beneficial treatment methods, such as neoadjuvant chemotherapy (6365). However, chemoresistance in BC is still common, leading to a poor prognosis and high mortality rate (66). One study has shown that Siah1 is associated with the chemoresistance of BC, which may be due to the interaction of Siah1 with tumor necrosis factor receptor associated-factor 4 (TRAF4) (63). Siah1 mediates the ubiquitination and degradation of β-catenin, thus inhibiting the activation of the Wnt/β-catenin signaling pathways, promoting cell apoptosis and preventing tumor progression (38). However, TRAF4 protects β-catenin from Siah1-mediated degradation by competing with β-catenin for binding to Siah1 and replacing it for degradation (Fig. 1D) (63). TRAF4 is highly expressed in chemotherapy-resistant breast cancer cells, and patients with BC and low TRAF4 expression levels benefit from chemotherapy (6769). Notably, the chemoresistance mediated by TRAF4 appears to be strongest to etoposide (a chemotherapeutic agent that induces Siah1-mediated degradation of β-catenin), suggesting that the key role of the Siah1-TRAF4/β-catenin axis in the chemoresistance of BC and further study of this axis may lead to new treatments (63).

Siah1 in leukemia

The dysregulation of the ubiquitin-proteasome system (UPS) is observed in solid tumors and leukemia (27). Increased substrates of Siah in leukemia have been found, suggesting the critical roles of Siah in leukemogenesis (27,70).

Acute promyelocytic leukemia (APL) is one of the most characterized forms of acute myeloid leukemia (AML) (71). t(15;17)(q24;q21), generating the promyelocytic leukemia-retinoic acid receptor α (PML-RARα) fusion gene, is the hallmark of APL (72,73). Notably, Pietschmann et al reported that the Siah1/2 cooperates with the E2 ubiquitin conjugase, i.e., ubiquitin-conjugating enzyme human 8 (UBCH8), leading to the proteasomal degradation of PML-RARα (74). In addition, this degradation of PML-RARα by the UBCH8-Siah1 complex can be significantly enhanced by all-trans-retinoic acid and sodium valproate (drug combination against APL), promoting the differentiation and maturation of APL cells (74,75). Moreover, other leukemia fusion proteins, including t(8;21)(q22;q22), RUNX family transcription factor 1 (AML1-ETO) fusion protein, have been identified as substrates of Siah1, but not Siah2, suggesting the powerful tumor suppressor effects of Siah1 in leukemia (27,76,77).

Super elongation complexes (SECs) promote the transcription of normal and leukemia-associated gene expression (78). SECs contain two different transcription elongation factors, namely positive transcription elongation factor b and elongation factor for RNA polymerase II 1/2 (ELL1/2), linked by the scaffolding protein AF4/FMR2 family member 1/4 (AFF1/4) (79). ELL2, a stoichiometrically limiting protein of SECs and an oncoprotein in leukemia, is specifically targeted for ubiquitin-mediated degradation by Siah1, but not by Siah2 (70,80,81). Notably, when AFF4 interacts with ELL2 to form SECs, the half-life of ELL2 against the Siah1-mediated ubiquitination is significantly prolonged (70). Through the proteasomal degradation induced by Siah1, AFF4 appears to have a lower affinity for Siah1 than ELL2. Thus, AFF4 is not adequately degraded by the low protein level of Siah1 (70). Notably, at relatively low protein levels of Siah1 in cells under physiological conditions, ELL2, especially the parts of ELL2 outside SECs, is highly sensitive to Siah1-induced ubiquitin-mediated degradation, suggesting that the primary effect of Siah1 is to prevent the formation of new SECs (Fig. 1E) (27,70). However, when the protein level of Siah1 becomes high, all remaining SECs are destroyed through Siah1-induced ubiquitin-mediated degradation (27,70). The abundance of Siah1 changes rapidly in response to various stresses, which may be to regulate SECs for the maintenance of a suitable transcription level of cells to adapt to stresses (11,71,73,74,77,82). Therefore, regulating the abundance of Siah1 in leukemia with disordered SECs may be a feasible method (Fig. 1E).

Siah1 in glioblastoma (GBM)

GBM is the most common brain cancer (48%), with high tumor heterogeneity and poor survival time (median overall survival time, 12–14 months) in adults (8385). Siah1 is widely regarded as a tumor suppressor in the majority of cancer types, with the exception of GBM (26,86). The knockdown of Siah1 by short-hairpin RNA (shRNA) severely suppresses the migration and invasion of human GBM cells (U251), whereas the overexpression of Siah1 has the opposite effect (26). Furthermore, recent studies have suggested that the tumor promotion of Siah1 in GBM is associated with hypoxic stress (11,16,26,87,88). Hypoxia-inducible factor 1α (HIF-1α) is the key transcription factor that regulates hypoxia-induced genes and enables cells to adapt to hypoxia (89,90). HIF-1α is extremely unstable under normal oxygen conditions (21% O2), as the prolyl-hydroxylation of HIF-1α promotes the binding of von Hippel-Lindau disease tumor suppressor to HIF-1α, resulting in the degradation of HIF-1α through the ubiquitin-proteasome pathway (89,91,92). The prolyl-hydroxylation of HIF-1α is mediated by the prolyl-hydroxylase protein (PHD) family, and the activity of PHD is inhibited in hypoxia (16,89,92). Notably, PHD3 has been identified as a substrate of Siah1, and Siah1 mediates the ubiquitination of PHD3 and induces the degradation of PHD3 through the UPS (11,26,93). The degradation of PHD3 increases the abundance of HIF-1α, thus promoting cells to adapt to hypoxia (16,26,87,88,93). Moreover, HIF-1α trans-activates the transcription of Siah1 by coordinating key histone modifications on the Siah1 promoter to increase its expression continuously and form a positive feedback loop, thus leading to the progression of GBM via the tolerance of tumor cells to hypoxia (16,26,87,88,93). Siah1 may be a potential molecular target for the treatment of GBM through the interference of the Siah1-PHD3-HIF-1α axis (Fig. 2A).

Figure 2.

(A) Siah1 mediates the ubiquitination of PHD3 and induces the degradation of PHD3, increasing the abundance of HIF-1α and promoting cells to adapt to hypoxia. HIF-1α trans-activates the transcription of Siah1 by coordinating key histone modifications on the Siah1 promoter to continuously increase HIF-1α expression and form a positive feedback loop. (B) Siah1 targets HIPK2 for poly-ubiquitination and proteasomal degradation, thereby inhibiting the phosphorylation of p53 at Ser46 and preventing cell apoptosis. p53 continues to activate the transcription of Siah1 and forms a positive feedback loop. The initiation of this positive feedback mechanism may be mediated by HIF-1α under hypoxic stress. (C) Under cell stress, GAPDH translocates to the nucleus in a Siah1-dependent manner upon glutamate stimulation and stabilizes Siah1 to facilitate degradation of nuclear proteins by Siah1, resulting in cell apoptosis and neuronal damage. (D) Siah1 functions as a tumor suppressor in the vast majority of tumors (breast cancer, hepatocellular cancer, leukemia, colorectal cancer and osteosarcoma). In some cancer types, such as glioblastoma and a part of hepatocellular carcinoma (where Siah1 is localized to the nucleus), Siah1 functions as an oncoprotein. Siah1, seven in absentia homolog family proteins 1; PHD3, prolyl-hydroxylase proteins 3; HIF-1α, hypoxia-inducible factor 1α; pVHL, von Hippel-Lindau disease tumor suppressor protein; HIPK2, homeodomain-interacting protein kinase 2; p53, tumor suppressor p53; P, phosphate group; GAPDH, glyceraldehyde 3-phosphate dehydrogenase.

Additionally, some studies have suggested that the Siah1-homeodomain-interacting protein kinase 2 (HIPK2)-p53Ser46 axis plays a key role in the promotion of glioma progression (86,94). HIPK2 functions as the key factor that is activated in DNA damage or cell response to stress and that triggers the phosphorylation of p53 at Ser46 (95,96). The best-known tumor suppressor gene, TP53, is mutated in >50% of malignancies and encodes a protein known as p53, a transcription factor that controls the initiation of the cell cycle (97100). However, when p53 is phosphorylated at Ser46, its functional activity as a transcription factor is inactivated, whereas the function of apoptosis promotion is activated, thus leading cells to apoptosis under stress response (101,102). However, Siah1 has been considered as the negative regulatory E3 ubiquitin ligase of HIPK2 that targets HIPK2 for poly-ubiquitination and proteasomal degradation, thereby inhibiting cell apoptosis by HIPK2 and resulting in the survival and proliferation of GBM cells (86,94,103,104). Notably, p53 acts directly on Siah1 to promote its transcription (11). As a tumor suppressor, p53 is supposed to promote the apoptosis of tumor cells, but the regulation of p53 on Siah1 in GBM seems to be a contradiction. p53 continuously activates the transcription of Siah1, whereas the increased abundance of Siah1 targets the degradation of HIPK2 and blocks the phosphorylation of p53 at Ser46, which keeps the activity of p53 as a transcription factor, continues to activate the transcription of Siah1 and promotes the progression of GBM. This phenomenon may also be explained by the hypoxic microenvironment of GBM, as HIF-1α can promote p53 transcription and stabilize the function of p53 in hypoxic stress (105,106). Siah1, p53, HIPK2, PHD3 and HIF-1α constitute an extremely complex network in the process of promoting the development of GBM (Fig. 2B), in which Siah1, as a key bridge factor, has the potential to be a future therapeutic target.

Siah1 may also partially act as a tumor suppressor in GBM when it interacts with CacyBP/SIP. CacyBP/SIP inhibits the migration and invasion behaviors of GBM cells by activating Siah1-mediated ubiquitination and degradation of cytoplasmic p27/kip1 (a key transcription factor and an oncoprotein highly expressed in GBM tissues) (22).

Siah1 as an oncoprotein

Studies have shown that Siah1 promotes cancer progression only in GBM and HCC, and only when Siah1 is localized in the nucleus (11,47,86,94). The varying biological functions in these studies are most probably associated with differences in cell types and the differential subcellular localization of Siah1.

The functions of Siah1 to induce the proliferation of cancer cells may be due to increased protein levels of FBP-3 (47). In addition, the Siah1-PHD3-HIF-1 and HIPK2-p53Ser46 axes explain how Siah1 inhibits apoptosis (26,86,87,94,107). However, the functions of Siah1 as an oncoprotein have not been systematically studied and summarized. Thus, future studies are needed to provide evidence for the targeted therapy of Siah1 as an oncoprotein.

Siah1 in other cancer types

Siah1 also acts as a tumor suppressor in CRC and pancreatic carcinogenesis. The knock down of Siah1 by shRNA promotes HCT116/SW480 CRC cell proliferation and migration, and results in fast tumor growth and a markedly large tumor volume in nude mice. Mechanically, Siah1 represses the occurrence and development of CRC by promoting the ubiquitylation of AKT and inhibiting the activity of the MAPK, PI3K-AKT and Hippo pathways (108). Additionally, the mutations of p53 in pancreatic cancer act in the opposite manner to the wild-type p53, inhibiting the transcription of Siah1 and leading to the accumulation of the oncoprotein. This phenomenon suggests the novel regulation of Siah1 in cancer (109).

Siah1 in nervous system diseases

Siah1 in Parkinson's Disease (PD)

PD, one of the most common neurodegenerative diseases, is manifested by a series of movement disorders, such as static tremor, bradykinesia, myotonia, and postural and gait disorders (110,111). Dopamine neurons and formation of Lewy bodies (LBs) in the substantia nigra striatum of the midbrain are regarded as typical pathological features of PD (112). LBs contain misfolded and abnormally stacked α-synuclein (α-syn), synphilin-1, ubiquitin and UPS-related enzymes, e.g., Siah1 (113115), F-box only protein 7 (116), and Parkin (117), suggesting that the disorder of UPS plays a key role in the pathogenesis of PD (118120). Notably, Siah1 is reported to monoubiquitinate or diubiquitinate α-syn, but without degradation, and is capable of ubiquitination and proteasomal degradation of synphilin-1, thus limiting the availability of α-syn for binding to synphilin-1 and the formation of LBs (114,115), suggesting the key pathological role of Siah1 in the development of PD. In addition, one study reported the cases of 7 patients with PD and Siah1 mutations (113), but the function of these mutations in PD is not fully known yet. Increasing evidence shows that the PTEN-induced putative kinase 1, synphilin-1 and Siah1 complex constitutes a novel mitophagy pathway (mitochondrial dysfunction is also considered to be one of the main pathological features of PD] (7,12,13), and that the complex has the function of clearing damaged mitochondria in PD (121), suggesting that drugs that activate Siah1 provide a novel strategy to promote the clearance of damaged mitochondria in PD (116,122127).

Siah1 in developmental delay

Developmental delay is defined as the skills of a child in one or a number aspects, including physical, motor, socioemotional, speech and language, and cognitive development, being significantly slower than those of other children of the same age (128131). Developmental delay occurs in up to 5% of children <5 years of age, and patients can benefit from the early detection of developmental delay and appropriate therapeutic measures (129,130). Genetic factors are the main causes of development delay, and a recent case report showed de novo monoallelic variants (Cys41Gly, Pro50Leu, Cys128Phe, Thr168Ala and Gly174Arg) in Siah1 in 5 unrelated patients within a phenotypic spectrum of developmental delay, infantile hypotonia, dysmorphism, strabismus and laryngomalacia (128). All patients with Siah1 mutations, except Pro50Leu, presented with moderate or severe developmental delay (128).

The overinhibition of the Wnt/β-catenin signaling pathways is one of the important pathogenesis factors of developmental delay (34,35). Previous studies have reported that wild-type Siah1 enhances the Wnt/β-catenin signaling pathways by mediating the Wnt-induced degradation of Axin (38,132). However, in 293T cells, exogenous Siah1 mutations (i.e., Cys41Gly, Pro50Leu, Cys128Phe, Thr168Ala and Gly174Arg) lose the ability to degrade Axin compared with the wild-type Siah1, resulting in the overinhibition of the Wnt/β-catenin signaling pathways (128). The best treatment for developmental delay is early detection. Thus, the prenatal examination for Siah1 is a highly effective option.

In addition, Siah1 has recently been identified as an upstream regulator of Akt3 (Akt signaling is an important regulator of neural development) (133135), and it is responsible for the ubiquitination and degradation of Akt3, suggesting that Siah1 may play a key role in neural development (135).

Siah1 in neuronal damage

Neuronal damage includes a series of diseases, including spinal cord injury and cerebral ischemia reperfusion, and the common feature of these diseases is the excessive apoptosis of nerve cells (136,137). Previous studies have reported that glyceraldehyde-3-phosphate dehydrogenase (GAPDH) is conventionally considered a critical factor in the process of nerve cell apoptosis (138143). GAPDH is important in glutamate-induced neuronal excitotoxicity, and evidence also demonstrates that GAPDH nuclear translocation plays a critical role in cell death (143,144). Notably, recent studies have shown that GAPDH is translocated to the nucleus in a Siah1-dependent manner upon glutamate stimulation and that it stabilizes Siah1 to facilitate the degradation of nuclear proteins by Siah1, resulting in cell apoptosis and neuronal damage (Fig. 2C) (138,144,145). Notably, the GAPDH/Siah1 cascade can be inhibited by the administration of the interfering peptide, the cannabinoid agonist WIN55212-2 and Sivelestat sodium, thus preventing neuronal damage (138,143,144) and suggesting that the GAPDH/Siah1 cascade can serve as a potential therapeutic target for neuronal damage treatment.

Siah1 was previously considered to be only a neuroprotective factor (113,128,128), but in neuronal damage, it appears to play a role in promoting the progression of neuronal damage (143,144). This abnormal phenomenon may be related to the subcellular localization of Siah1, and the function of nuclear Siah1 seems to be opposite to Siah1 under normal physiological conditions (114). Notably, Siah1, a tumor suppressor, functions as an oncoprotein in some types of liver cancer, and Siah1 in liver cancer is localized in the nucleus (11,146). Thus, the subcellular localization of Siah1 is an important focus and may become a novel treatment strategy for some diseases.

Siah1 in Alzheimer's Disease (AD)

AD, the most common chronic and irreversible neurodegenerative disease in the world, is characterized by impaired cognitive function and loss of self-care ability (147). The hyperphosphorylation of Tau is considered to be one of the main pathological features of AD (148,149). A recent study showed the CacyBP/SIP could mediate the dephosphorylation of phosphorylated-Tau (150), suggesting the neuroprotective effects of CacyBP/SIP. CacyBP/SIP has long been identified as a SIP (21,22). However, the function of Siah1 in AD has been rarely studied and should be clarified in future studies.

Clinical significance of Siah1 in human diseases

Siah1 functions as a tumor suppressor in the vast majority of tumors [e.g., BC (23), HCC (30), leukemia (27), CRC (108) and osteosarcoma (44)]. However, in some cancer types, such as GBM (86) and a part of HCC (where Siah1 is localized to the nucleus) (47), Siah1 functions as an oncoprotein (Table II). The activator lncRNA SNHG1(151) and inhibitors miR-135a (152), miR-424 (153), miR-944 (62), miR-299-5p (146), miR-15b-5p (151) and miR-107 (59), are all good choices to regulate the functions of Siah1 depending on the different cancer contexts (Table I). Notably, the oncoprotein functions of Siah1 may be due to the nuclear localization of Siah1, thus suggesting that the inhibition of the Siah1 nuclear localization signal is a way to inhibit the oncoprotein functions of Siah1 but one that does not destroy the normal physiological function of Siah1 in the cytoplasm (47). This idea is also suitable for the treatment of nervous system diseases, as the nuclear localization of Siah1 often promotes nerve apoptosis and leads to the occurrence of nervous system diseases (138140,142,143).

Discussion

The functions of a protein depend on a number of elements, including post-translational modification, cell types, cellular microenvironment and binding to other proteins. Siah1 was originally identified as a tumor-suppressing protein for BC (5456). However, new functions of Siah1 are continuously being discovered, suggesting that Siah1 is not only a tumor-suppressing protein.

As an E3 ubiquitin ligase, the most important function of Siah1 is the ubiquitination of substrates to promote their degradation or change their function (11,13). However, the types of ubiquitin modifications of Siah1 in cancer and nervous system diseases seem to differ markedly. In cancer, Siah1 is responsible for the ubiquitination-mediated degradation of substrates, whether the substrates are oncoproteins or tumor-suppressing proteins (16,74,76,94,154,155). In nervous system diseases, Siah1 does more to change the function of substrates than to degrade them via non-degradative ubiquitination (140,142,156). This difference may be related to the formation of the Siah1 ubiquitin-ligase complex. Siah1 can interact with E2 alone or become an essential part of the ubiquitin-ligase complex (13,20). Notably, CacyBP/SIP, one of the parts of the complex, has the highest protein levels in the brain and maintains lower protein levels in other organs, suggesting that the complex may affect the Siah1-mediated ubiquitination of substrates (21). Siah1 has been reported to monoubiquitinate or diubiquitinate α-syn. Upon interaction with CacyBP/SIP, Siah1 can inhibit the development of GBM by inducing the degradation of p27/kip1, suggesting that the status of Siah1 (interacting with E2 alone or forming the ubiquitin-ligase complex) significantly affects the functions of Siah1 (22,114,115). Numerous studies are limited to substrate degradation by Siah1, but they ignore the special status of Siah1 in this process, thus requiring future supplemental studies (23,26,57,86). The studies on the Siah1 ubiquitin-ligase complex may be the key to study the function of Siah1 thoroughly and for targeting of Siah1 in the future.

The subcellular localization of Siah1 also determines the function of Siah1. The nuclear localization of Siah1 promotes the occurrence of cancer and nerve apoptosis, suggesting that the nuclear localization of Siah1 is a pathological phenomenon (47,138). Although the mechanism that causes Siah1 to transfer to the nucleus is not clear, we believe that the inhibition of the Siah1 nuclear localization signal can be identified as a favorable choice in the treatment of diseases.

The oncoprotein functions of Siah1 in GBM seem to be closely related to the tumor hypoxic microenvironment (26,93). However, the function of Siah1 as an oncoprotein in the hypoxic microenvironment has not been reported in other tumors, especially in HCC, which is most closely associated with the hypoxic microenvironment (89). GBM belongs to the diseases of the nervous system. Thus, studying the functional differences of Siah1 in cancer and neurological diseases is a notable and promising topic.

The dysregulation of UPS is usually caused by the mutations of E3 ubiquitin ligase, such as SPOP (8,157) and leucine zipper-like transcription regulator 1 (LZTR1) (158). The mutations of SPOP and LZTR1 occur mostly in domains bound to the substrate, severely affecting their ability to bind to substrates, inhibiting the ubiquitination and degradation of substrates, and leading to the accumulation of substrates (4,6,159). Notably, although one study showed the lack of somatic mutation in the coding sequence of Siah1 (55), the mutations of Siah1 are rarely reported. The regulations of SPOP and LZTR1 are rarely recorded, whereas the regulations of Siah1 are various (Table I), suggesting that Siah1 plays a core role as a bridge factor in various signaling pathways (11). Slight changes in Siah1 protein levels and protein localization lead to significant changes in its functions, which may be as the mutations affect the function of Siah1 significantly and cause death in the earliest embryos (knockout of both Siah1 genes is embryonically lethal in mice). This phenomenon results in little spread of Siah1 mutation heritage lines (11,27).

Although numerous studies have focused on the upstream regulations of Siah1 (Table I), studies on agonists and inhibitors specifically targeting Siah1 remain lacking. The only drug targeting Siah that has been described so far is vitamin K3 (menadione), which has been identified as an inhibitor of Siah2 ubiquitin ligase activity in a screen of U.S. Food and Drug Administration-approved therapeutic drugs (158). Therefore, the future drug research of Siah1 is still needed.

The present review summarized the novel substrates and complex upstream regulations of Siah1, describes the functions of Siah1 as a tumor suppressor protein and an oncoprotein, and discusses the potential mechanisms of the different roles of Siah1 in the nervous system and cancer. In addition, the review focused on the effects of Siah1 nuclear localization and the special status of Siah1 (interacting with E2 alone or forming the ubiquitin-ligase complex). Moreover, it highlighted the clinical significance of Siah1 in human diseases. This review may provide inspiration for future Siah1 research.

Acknowledgements

The authors would like to thank Dr Yuqi Wang (West Lake University, Hangzhou, Zhejiang, China) for the kind help and good advice provided.

Funding

This study was supported by the Natural Science Foundation of Zhejiang Province (grant no. LY20C070001), the National Natural Science Foundation of China (grant no. 31801165), the Graduate Research Innovation Fund project in Ningbo University (grant no. IF2021097) and the K.C.Wong Magna Fund in Ningbo University.

Availability of data and materials

Not applicable.

Authors' contributions

XJ conceived the present study. HZ drafted the manuscript. YG, JW and MY made substantial contributions to the interpretation and drafting of the study, and revised the manuscript critically for important intellectual content. All authors read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Hershko A and Ciechanover A: The ubiquitin system. Ann Rev Biochem. 67:425–479. 1998. View Article : Google Scholar : PubMed/NCBI

2 

Mani RS: The emerging role of speckle-type POZ protein (SPOP) in cancer development. Drug Discov Today. 19:1498–1502. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Zou T and Zhang J: Diverse and pivotal roles of neddylation in metabolism and immunity. FEBS J. 288:3884–3912. 2020. View Article : Google Scholar : PubMed/NCBI

4 

Chen RH: Cullin 3 and its role in tumorigenesis. Adv Exp Med Biol. 1217:187–210. 2020. View Article : Google Scholar : PubMed/NCBI

5 

Nandi D, Tahiliani P, Kumar A and Chandu D: The ubiquitin-proteasome system. J Biosci. 31:137–155. 2006. View Article : Google Scholar : PubMed/NCBI

6 

Cuneo MJ and Mittag T: The ubiquitin ligase adaptor SPOP in cancer. FEBS J. 286:3946–3958. 2019. View Article : Google Scholar : PubMed/NCBI

7 

Wang D, Ma L, Wang B, Liu J and Wei W: E3 ubiquitin ligases in cancer and implications for therapies. Cancer Metastasis Rev. 36:683–702. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Wang Z, Song Y, Ye M, Dai X, Zhu X and Wei W: The diverse roles of SPOP in prostate cancer and kidney cancer. Nat Rev Urol. 17:339–350. 2020. View Article : Google Scholar : PubMed/NCBI

9 

Morreale F and Walden H: Types of ubiquitin ligases. Cell. 165:248. 2016. View Article : Google Scholar : PubMed/NCBI

10 

Yun S, Möller A, Chae SK, Hong WP, Bae YJ, Bowtell DD, Ryu SH and Suh PG: Siah proteins induce the epidermal growth factor-dependent degradation of phospholipase Cepsilon. J Biol Chem. 283:1034–1042. 2008. View Article : Google Scholar : PubMed/NCBI

11 

Qi J, Kim H, Scortegagna M and Ronai ZA: Regulators and effectors of Siah ubiquitin ligases. Cell Biochem Biophys. 67:15–24. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Garrison JB, Correa RG, Gerlic M, Yip KW, Krieg A, Tamble CM, Shi R, Welsh K, Duggineni S, Huang Z, et al: ARTS and Siah collaborate in a pathway for XIAP degradation. Mol Cell. 41:107–116. 2011. View Article : Google Scholar : PubMed/NCBI

13 

Santelli E, Leone M, Li C, Fukushima T, Preece NE, Olson AJ, Ely KR, Reed JC, Pellecchia M, Liddington RC and Matsuzawa SI: Structural analysis of Siah1-Siah-interacting protein interactions and insights into the assembly of an E3 ligase multiprotein complex. J Biol Chem. 280:34278–34287. 2005. View Article : Google Scholar : PubMed/NCBI

14 

Zhang Q, Wang Z, Hou F, Harding R, Huang X, Dong A, Walker JR and Tong Y: The substrate binding domains of human SIAH E3 ubiquitin ligases are now crystal clear. Biochim Biophys Acta Gen Subj. 1861:3095–3105. 2017. View Article : Google Scholar : PubMed/NCBI

15 

Knauer SK, Mahendrarajah N, Roos WP and Krämer OH: The inducible E3 ubiquitin ligases SIAH1 and SIAH2 perform critical roles in breast and prostate cancers. Cytokine Growth Factor Rev. 26:405–413. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Nakayama K and Ronai Z: Siah: New players in the cellular response to hypoxia. Cell Cycle. 3:1345–1347. 2004. View Article : Google Scholar : PubMed/NCBI

17 

Famulski JK, Trivedi N, Howell D, Yang Y, Tong Y, Gilbertson R and Solecki DJ: Siah regulation of Pard3A controls neuronal cell adhesion during germinal zone exit. Science. 330:1834–1838. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Wong CS and Möller A: Siah: A promising anticancer target. Cancer Res. 73:2400–2406. 2013. View Article : Google Scholar : PubMed/NCBI

19 

House CM, Möller A and Bowtell DD: Siah proteins: Novel drug targets in the Ras and hypoxia pathways. Cancer Res. 69:8835–8838. 2009. View Article : Google Scholar : PubMed/NCBI

20 

Matsuzawa S, Li C, Ni CZ, Takayama S, Reed JC and Ely KR: Structural analysis of Siah1 and its interactions with Siah-interacting protein (SIP). J Biol Chem. 278:1837–1840. 2003. View Article : Google Scholar : PubMed/NCBI

21 

Topolska-Woś AM, Chazin WJ and Filipek A: CacyBP/SIP-structure and variety of functions. Biochim Biophys Acta. 1860:79–85. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Yan S, Li A and Liu Y: CacyBP/SIP inhibits the migration and invasion behaviors of glioblastoma cells through activating Siah1 mediated ubiquitination and degradation of cytoplasmic p27. Cell Biol Int. 42:216–226. 2018. View Article : Google Scholar : PubMed/NCBI

23 

Wen YY, Yang ZQ, Song M, Li BL, Yao XH, Chen XL, Zhao J, Lu YY, Zhu JJ and Wang EH: The expression of SIAH1 is downregulated and associated with Bim and apoptosis in human breast cancer tissues and cells. Mol Carcinog. 49:440–449. 2010. View Article : Google Scholar : PubMed/NCBI

24 

Briant DJ, Ceccarelli DF and Sicheri F: I Siah substrate! Structure. 14:627–628. 2006. View Article : Google Scholar : PubMed/NCBI

25 

Czechowicz JS, Nagel CH, Voges M, Spohn M, Eibl MM and Hauber J: Interaction between the cellular E3 ubiquitin ligase SIAH-1 and the viral immediate-early protein ICP0 enables efficient replication of herpes simplex virus type 2 in vivo. PLoS One. 13:e02018802018. View Article : Google Scholar : PubMed/NCBI

26 

Shi H, Zheng B, Wu Y, Tang Y, Wang L, Gao Y, Gong H, Du J and Yu R: Ubiquitin ligase Siah1 promotes the migration and invasion of human glioma cells by regulating HIF-1α signaling under hypoxia. Oncol Rep. 33:1185–1190. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Krämer OH, Stauber RH, Bug G, Hartkamp J and Knauer SK: SIAH proteins: Critical roles in leukemogenesis. Leukemia. 27:792–802. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Sim HW and Knox J: Hepatocellular carcinoma in the era of immunotherapy. Curr Probl Cancer. 42:40–48. 2018. View Article : Google Scholar : PubMed/NCBI

29 

Matsuo K, Satoh S, Okabe H, Nomura A, Maeda T, Yamaoka Y and Ikai I: SIAH1 inactivation correlates with tumor progression in hepatocellular carcinomas. Genes Chromosomes Cancer. 36:283–291. 2003. View Article : Google Scholar : PubMed/NCBI

30 

Yoshibayashi H, Okabe H, Satoh S, Hida K, Kawashima K, Hamasu S, Nomura A, Hasegawa S, Ikai I and Sakai Y: SIAH1 causes growth arrest and apoptosis in hepatoma cells through beta-catenin degradation-dependent and -independent mechanisms. Oncol Rep. 17:549–556. 2007.PubMed/NCBI

31 

Yao H, Ashihara E and Maekawa T: Targeting the Wnt/β-catenin signaling pathway in human cancers. Expert Opin Ther Targets. 15:873–887. 2011. View Article : Google Scholar : PubMed/NCBI

32 

Clevers H and Nusse R: Wnt/β-catenin signaling and disease. Cell. 149:1192–1205. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Arend RC, Londoño-Joshi AI, Straughn JM Jr and Buchsbaum DJ: The Wnt/β-catenin pathway in ovarian cancer: A review. Gynecol Oncol. 131:772–779. 2013. View Article : Google Scholar : PubMed/NCBI

34 

Taciak B, Pruszynska I, Kiraga L, Bialasek M and Krol M: Wnt signaling pathway in development and cancer. J Physiol Pharmacol. 69:doi: 10.26402. 2018.PubMed/NCBI

35 

Steinhart Z and Angers S: Wnt signaling in development and tissue homeostasis. Development. 145:dev1465892018. View Article : Google Scholar : PubMed/NCBI

36 

Huang P, Yan R, Zhang X, Wang L, Ke X and Qu Y: Activating Wnt/β-catenin signaling pathway for disease therapy: Challenges and opportunities. Pharmacol Ther. 196:79–90. 2019. View Article : Google Scholar : PubMed/NCBI

37 

Kim E, Lisby A, Ma C, Lo N, Ehmer U, Hayer KE, Furth EE and Viatour P: Promotion of growth factor signaling as a critical function of β-catenin during HCC progression. Nat Commun. 10:19092019. View Article : Google Scholar : PubMed/NCBI

38 

Ji L, Jiang B, Jiang X, Charlat O, Chen A, Mickanin C, Bauer A, Xu W, Yan X and Cong F: The SIAH E3 ubiquitin ligases promote Wnt/β-catenin signaling through mediating wnt-induced axin degradation. Genes Dev. 31:904–915. 2017. View Article : Google Scholar : PubMed/NCBI

39 

Yang Y, Zhang J, Wu T, Xu X, Cao G, Li H and Chen X: Histone deacetylase 2 regulates the doxorubicin (Dox) resistance of hepatocarcinoma cells and transcription of ABCB1. Life Sci. 216:200–206. 2019. View Article : Google Scholar : PubMed/NCBI

40 

Cheng C, Li C, Zhu X, Han W, Li J and Lv Y: Doxorubicin-loaded Fe(3)O(4)-ZIF-8 nano-composites for hepatocellular carcinoma therapy. J Biomater Appl. 33:1373–1381. 2019. View Article : Google Scholar : PubMed/NCBI

41 

Long L, Xiang H, Liu J, Zhang Z and Sun L: ZEB1 mediates doxorubicin (Dox) resistance and mesenchymal characteristics of hepatocarcinoma cells. Exp Mol Pathol. 106:116–122. 2019. View Article : Google Scholar : PubMed/NCBI

42 

Li LY, Yang CC, Yang JF, Li HD, Zhang BY, Zhou H, Hu S, Wang K, Huang C, Meng XM, et al: ZEB1 regulates the activation of hepatic stellate cells through Wnt/β-catenin signaling pathway. Eur J Pharmacol. 865:1727872019. View Article : Google Scholar : PubMed/NCBI

43 

Qin Y, Yu J, Zhang M, Qin F and Lan X: ZEB1 promotes tumorigenesis and metastasis in hepatocellular carcinoma by regulating the expression of vimentin. Mol Med Rep. 19:2297–2306. 2019.PubMed/NCBI

44 

Han X, Liu F, Zhang C, Ren Z, Li L and Wang G: SIAH1/ZEB1/IL-6 axis is involved in doxorubicin (Dox) resistance of osteosarcoma cells. Biol Chem. 400:545–553. 2019. View Article : Google Scholar : PubMed/NCBI

45 

Abshire CF, Carroll JL and Dragoi AM: FLASH protects ZEB1 from degradation and supports cancer cells' epithelial-to-mesenchymal transition. Oncogenesis. 5:e2542016. View Article : Google Scholar : PubMed/NCBI

46 

Wu Y, Yang X, Chen Z, Tian L, Jiang G, Chen F, Li J, An P, Lu L, Luo N, et al: m6 A-induced lncRNA RP11 triggers the dissemination of colorectal cancer cells via upregulation of Zeb1. Mol Cancer. 18:872019. View Article : Google Scholar : PubMed/NCBI

47 

Brauckhoff A, Malz M, Tschaharganeh D, Malek N, Weber A, Riener MO, Soll C, Samarin J, Bissinger M and Schmidt J: Nuclear expression of the ubiquitin ligase seven in absentia homolog (SIAH)-1 induces proliferation and migration of liver cancer cells. J Hepatol. 55:1049–1057. 2011. View Article : Google Scholar : PubMed/NCBI

48 

Veronesi U, Boyle P, Goldhirsch A, Orecchia R and Viale G: Breast cancer. Lancet. 365:1727–1741. 2005. View Article : Google Scholar : PubMed/NCBI

49 

Woolston C: Breast cancer. Nature. 527:S1012015. View Article : Google Scholar : PubMed/NCBI

50 

DeSantis C, Siegel R, Bandi P and Jemal A: Breast cancer statistics, 2011. CA Cancer J Clin. 61:409–418. 2011. View Article : Google Scholar : PubMed/NCBI

51 

Ullah MF: Breast cancer: Current perspectives on the disease status. Adv Exp Med Biol. 1152:51–64. 2019. View Article : Google Scholar : PubMed/NCBI

52 

Tsang JYS and Tse GM: Molecular classification of breast cancer. Adv Anat Pathol. 27:27–35. 2020. View Article : Google Scholar : PubMed/NCBI

53 

Barzaman K, Karami J, Zarei Z, Hosseinzadeh A, Kazemi MH, Moradi-Kalbolandi S, Safari E and Farahmand L: Breast cancer: Biology, biomarkers, and treatments. Int Immunopharmacol. 84:1065352020. View Article : Google Scholar : PubMed/NCBI

54 

Bruzzoni-Giovanelli H, Faille A, Linares-Cruz G, Nemani M, Deist FL, Germani A, Chassoux D, Millot G, Roperch JP, Amson R, et al: SIAH-1 inhibits cell growth by altering the mitotic process. Oncogene. 18:7101–7109. 1999. View Article : Google Scholar : PubMed/NCBI

55 

Medhioub M, Vaury C, Hamelin R and Thomas G: Lack of somatic mutation in the coding sequence of SIAH1 in tumors hemizygous for this candidate tumor suppressor gene. Int J Cancer. 87:794–797. 2000. View Article : Google Scholar : PubMed/NCBI

56 

Germani A, Bruzzoni-Giovanelli H, Fellous A, Gisselbrecht S, Varin-Blank N and Calvo F: SIAH-1 interacts with alpha-tubulin and degrades the kinesin Kid by the proteasome pathway during mitosis. Oncogene. 19:5997–6006. 2000. View Article : Google Scholar : PubMed/NCBI

57 

Wen YY, Yang ZQ, Song M, Li BL, Zhu JJ and Wang EH: SIAH1 induced apoptosis by activation of the JNK pathway and inhibited invasion by inactivation of the ERK pathway in breast cancer cells. Cancer Sci. 101:73–79. 2010. View Article : Google Scholar : PubMed/NCBI

58 

Nedeljković M and Damjanović A: Mechanisms of chemotherapy resistance in triple-negative breast cancer-how we can rise to the challenge. Cells. 8:9572019. View Article : Google Scholar : PubMed/NCBI

59 

Zhang L, Ma P, Sun LM, Han YC, Li BL, Mi XY, Wang EH and Song M: MiR-107 down-regulates SIAH1 expression in human breast cancer cells and silencing of miR-107 inhibits tumor growth in a nude mouse model of triple-negative breast cancer. Mol Carcinog. 55:768–777. 2016. View Article : Google Scholar : PubMed/NCBI

60 

Hong HC, Chuang CH, Huang WC, Weng SL, Chen CH, Chang KH, Liao KW and Huang HD: A panel of eight microRNAs is a good predictive parameter for triple-negative breast cancer relapse. Theranostics. 10:8771–8789. 2020. View Article : Google Scholar : PubMed/NCBI

61 

Sahlberg KK, Bottai G, Naume B, Burwinkel B, Calin GA, Børresen-Dale AL and Santarpia L: A serum microRNA signature predicts tumor relapse and survival in triple-negative breast cancer patients. Clin Cancer Res. 21:1207–1214. 2015. View Article : Google Scholar : PubMed/NCBI

62 

Flores-Pérez A, Marchat LA, Rodríguez-Cuevas S, Bautista VP, Fuentes-Mera L, Romero-Zamora D, Maciel-Dominguez A, de la Cruz OH, Fonseca-Sánchez M, Ruíz-García E, et al: Suppression of cell migration is promoted by miR-944 through targeting of SIAH1 and PTP4A1 in breast cancer cells. BMC Cancer. 16:3792016. View Article : Google Scholar : PubMed/NCBI

63 

Ren H, Mi X, Zhao P, Zhao X, Wei N, Huang H, Meng Z, Kou J, Sun M, Liu Y, et al: TRAF4, a new substrate of SIAH1, participates in chemotherapy resistance of breast cancer cell by counteracting SIAH1-mediated downregulation of β-catenin. Breast Cancer Res Treat. 183:275–289. 2020. View Article : Google Scholar : PubMed/NCBI

64 

Butti R, Gunasekaran VP, Kumar TVS, Banerjee P and Kundu GC: Breast cancer stem cells: Biology and therapeutic implications. Int J Biochem Cell Biol. 107:38–52. 2019. View Article : Google Scholar : PubMed/NCBI

65 

Fisusi FA and Akala EO: Drug combinations in breast cancer therapy. Pharm Nanotechnol. 7:3–23. 2019. View Article : Google Scholar : PubMed/NCBI

66 

Tang Y, Wang Y, Kiani MF and Wang B: Classification, treatment strategy, and associated drug resistance in breast cancer. Clin Breast Cancer. 16:335–343. 2016. View Article : Google Scholar : PubMed/NCBI

67 

Zhou J, Li W, Ming J, Yang W, Lu L, Zhang Q, Ruan S and Huang T: High expression of TRAF4 predicts poor prognosis in tamoxifen-treated breast cancer and promotes tamoxifen resistance. Anticancer Drugs. 31:558–566. 2020. View Article : Google Scholar : PubMed/NCBI

68 

Zhang X, Wen Z and Mi X: Expression and anti-apoptotic function of TRAF4 in human breast cancer MCF-7 cells. Oncol Lett. 7:411–414. 2014. View Article : Google Scholar : PubMed/NCBI

69 

Wang A, Wang J, Ren H, Yang F, Sun L, Diao K, Zhao Z, Song M, Cui Z, Wang E, et al: TRAF4 participates in wnt/β-catenin signaling in breast cancer by upregulating β-catenin and mediating its translocation to the nucleus. Mol Cell Biochem. 395:211–219. 2014. View Article : Google Scholar : PubMed/NCBI

70 

Liu M, Hsu J, Chan C, Li Z and Zhou Q: The ubiquitin ligase Siah1 controls ELL2 stability and formation of super elongation complexes to modulate gene transcription. Mol Cell. 46:325–334. 2012. View Article : Google Scholar : PubMed/NCBI

71 

de Thé H, Pandolfi PP and Chen Z: Acute promyelocytic leukemia: A paradigm for oncoprotein-targeted cure. Cancer Cell. 32:552–560. 2017. View Article : Google Scholar : PubMed/NCBI

72 

De Braekeleer E, Douet-Guilbert N and De Braekeleer M: RARA fusion genes in acute promyelocytic leukemia: A review. Expert Rev Hematol. 7:347–357. 2014. View Article : Google Scholar : PubMed/NCBI

73 

Liquori A, Ibañez M, Sargas C, Sanz MA, Barragán E and Cervera J: Acute promyelocytic leukemia: A constellation of molecular events around a single PML-RARA Fusion Gene. Cancers (Basel). 12:6242020. View Article : Google Scholar : PubMed/NCBI

74 

Pietschmann K, Buchwald M, Müller S, Knauer SK, Kögl M, Heinzel T and Krämer OH: Differential regulation of PML-RARα stability by the ubiquitin ligases SIAH1/SIAH2 and TRIAD1. Int J Biochem Cell Biol. 44:132–138. 2012. View Article : Google Scholar : PubMed/NCBI

75 

Bug G, Ritter M, Wassmann B, Schoch C, Heinzel T, Schwarz K, Romanski A, Kramer OH, Kampfmann M, Hoelzer D, et al: Clinical trial of valproic acid and all-trans retinoic acid in patients with poor-risk acute myeloid leukemia. Cancer. 104:2717–2725. 2005. View Article : Google Scholar : PubMed/NCBI

76 

Bursen A, Moritz S, Gaussmann A, Moritz S, Dingermann T and Marschalek R: Interaction of AF4 wild-type and AF4.MLL fusion protein with SIAH proteins: Indication for t(4;11) pathobiology? Oncogene. 23:6237–6249. 2004. View Article : Google Scholar : PubMed/NCBI

77 

Krämer OH, Müller S, Buchwald M, Reichardt S and Heinzel T: Mechanism for ubiquitylation of the leukemia fusion proteins AML1-ETO and PML-RARalpha. Faseb J. 22:1369–1379. 2008. View Article : Google Scholar : PubMed/NCBI

78 

Luo Z, Lin C and Shilatifard A: The super elongation complex (SEC) family in transcriptional control. Nat Rev Mol Cell Biol. 13:543–547. 2012. View Article : Google Scholar : PubMed/NCBI

79 

Dahl NA, Danis E, Balakrishnan I, Wang D, Pierce A, Walker FM, Gilani A, Serkova NJ, Madhavan K, Fosmire S, et al: Super elongation complex as a targetable dependency in diffuse midline glioma. Cell Rep. 31:1074852020. View Article : Google Scholar : PubMed/NCBI

80 

Yu D, Liu R, Yang G and Zhou Q: The PARP1-Siah1 axis controls HIV-1 transcription and expression of Siah1 substrates. Cell Rep. 23:3741–3749. 2018. View Article : Google Scholar : PubMed/NCBI

81 

Wu J, Xue Y, Gao X and Zhou Q: Host cell factors stimulate HIV-1 transcription by antagonizing substrate-binding function of Siah1 ubiquitin ligase to stabilize transcription elongation factor ELL2. Nucleic Acids Res. 48:7321–7332. 2020.PubMed/NCBI

82 

Nagel CH, Albrecht N, Milovic-Holm K, Mariyanna L, Keyser B, Abel B, Weseloh B, Hofmann TG, Eibl MM and Hauber J: Herpes simplex virus immediate-early protein ICP0 is targeted by SIAH-1 for proteasomal degradation. J Virol. 85:7644–7657. 2011. View Article : Google Scholar : PubMed/NCBI

83 

Abe T, Umeki I, Kanno SI, Inoue SI, Niihori T and Aoki Y: LZTR1 facilitates polyubiquitination and degradation of RAS-GTPases. Cell Death Differ. 27:1023–1035. 2020. View Article : Google Scholar : PubMed/NCBI

84 

Frattini V, Trifonov V, Chan JM, Castano A, Lia M, Abate F, Keir ST, Ji AX, Zoppoli P, Niola F, et al: The integrated landscape of driver genomic alterations in glioblastoma. Nat Genet. 45:1141–1149. 2013. View Article : Google Scholar : PubMed/NCBI

85 

Wirsching HG, Galanis E and Weller M: Glioblastoma. Handb Clin Neurol. 134:381–397. 2016. View Article : Google Scholar : PubMed/NCBI

86 

He Y, Roos WP, Wu Q, Hofmann TG and Kaina B: The SIAH1-HIPK2-p53ser46 damage response pathway is involved in temozolomide-induced glioblastoma cell death. Mol Cancer Res. 17:1129–1141. 2019. View Article : Google Scholar : PubMed/NCBI

87 

Fan Z, Li Z, Yang Y, Liu S, Guo J and Xu Y: HIF-1α coordinates epigenetic activation of SIAH1 in hepatocytes in response to nutritional stress. Biochim Biophys Acta Gene Regul Mech. 1860:1037–1046. 2017. View Article : Google Scholar : PubMed/NCBI

88 

Matsui-Hasumi A, Sato Y, Uto-Konomi A, Yamashita S, Uehori J, Yoshimura A, Yamashita M, Asahara H, Suzuki S and Kubo M: E3 ubiquitin ligases SIAH1/2 regulate hypoxia-inducible factor-1 (HIF-1)-mediated Th17 cell differentiation. Int Immunol. 29:133–143. 2017. View Article : Google Scholar : PubMed/NCBI

89 

Ke Q and Costa M: Hypoxia-inducible factor-1 (HIF-1). Mol Pharmacol. 70:1469–1480. 2006. View Article : Google Scholar : PubMed/NCBI

90 

Semenza GL: Targeting HIF-1 for cancer therapy. Nat Rev Cancer. 3:721–732. 2003. View Article : Google Scholar : PubMed/NCBI

91 

You L, Wu W, Wang X, Fang L, Adam V, Nepovimova E, Wu Q and Kuca K: The role of hypoxia-inducible factor 1 in tumor immune evasion. Med Res Rev. 41:1622–1643. 2021. View Article : Google Scholar : PubMed/NCBI

92 

Peng S, Zhang J, Tan X, Huang Y, Xu J, Silk N, Zhang D, Liu Q and Jiang J: The VHL/HIF axis in the development and treatment of pheochromocytoma/paraganglioma. Front Endocrinol (Lausanne). 11:5868572020. View Article : Google Scholar : PubMed/NCBI

93 

Gopalsamy A, Hagen T and Swaminathan K: Investigating the molecular basis of Siah1 and Siah2 E3 ubiquitin ligase substrate specificity. PLoS One. 9:e1065472014. View Article : Google Scholar : PubMed/NCBI

94 

Kim SY, Choi DW, Kim EA and Choi CY: Stabilization of HIPK2 by escape from proteasomal degradation mediated by the E3 ubiquitin ligase Siah1. Cancer Lett. 279:177–184. 2009. View Article : Google Scholar : PubMed/NCBI

95 

Choi DW and Choi CY: HIPK2 modification code for cell death and survival. Mol Cell Oncol. 1:e9559992014. View Article : Google Scholar : PubMed/NCBI

96 

Feng Y, Zhou L, Sun X and Li Q: Homeodomain-interacting protein kinase 2 (HIPK2): A promising target for anti-cancer therapies. Oncotarget. 8:20452–20461. 2017. View Article : Google Scholar : PubMed/NCBI

97 

Moll UM and Petrenko O: The MDM2-p53 interaction. Mol Cancer Res. 1:1001–1008. 2003.PubMed/NCBI

98 

Krastev DB and Buchholz F: Ribosome biogenesis and p53: Who is regulating whom? Cell Cycle. 10:3417–3418. 2011. View Article : Google Scholar : PubMed/NCBI

99 

Chao CC: Mechanisms of p53 degradation. Clin Chim Acta. 438:139–147. 2015. View Article : Google Scholar : PubMed/NCBI

100 

Kanapathipillai M: Treating p53 mutant aggregation-associated cancer. Cancers (Basel). 10:1542018. View Article : Google Scholar : PubMed/NCBI

101 

Feng L, Hollstein M and Xu Y: Ser46 phosphorylation regulates p53-dependent apoptosis and replicative senescence. Cell Cycle. 5:2812–2819. 2006. View Article : Google Scholar : PubMed/NCBI

102 

Liebl MC and Hofmann TG: Cell fate regulation upon DNA damage: p53 serine 46 kinases pave the cell death road. Bioessays. 41:e19001272019. View Article : Google Scholar : PubMed/NCBI

103 

Winter M, Sombroek D, Dauth I, Moehlenbrink J, Scheuermann K, Crone J and Hofmann TG: Control of HIPK2 stability by ubiquitin ligase Siah-1 and checkpoint kinases ATM and ATR. Nat Cell Biol. 10:812–824. 2008. View Article : Google Scholar : PubMed/NCBI

104 

Moehlenbrink J, Bitomsky N and Hofmann TG: Hypoxia suppresses chemotherapeutic drug-induced p53 Serine 46 phosphorylation by triggering HIPK2 degradation. Cancer Lett. 292:119–124. 2010. View Article : Google Scholar : PubMed/NCBI

105 

Chen D, Li M, Luo J and Gu W: Direct interactions between HIF-1 alpha and Mdm2 modulate p53 function. J Biol Chem. 278:13595–13598. 2003. View Article : Google Scholar : PubMed/NCBI

106 

Wang P, Guan D, Zhang XP, Liu F and Wang W: Modeling the regulation of p53 activation by HIF-1 upon hypoxia. FEBS Lett. 593:2596–2611. 2019. View Article : Google Scholar : PubMed/NCBI

107 

Fukuba H, Yamashita H, Nagano Y, Jin HG, Hiji M, Ohtsuki T, Takahashi T, Kohriyama T and Matsumoto M: Siah-1 facilitates ubiquitination and degradation of factor inhibiting HIF-1alpha (FIH). Biochem Biophys Res Commun. 353:324–329. 2007. View Article : Google Scholar : PubMed/NCBI

108 

Xiao Z, Wei Z, Deng D, Zheng Z, Zhao Y, Jiang S, Zhang D, Zhang LJ, Fan M, Chen S, et al: Downregulation of Siah1 promotes colorectal cancer cell proliferation and migration by regulating AKT and YAP ubiquitylation and proteasome degradation. Cancer Cell Int. 20:502020. View Article : Google Scholar : PubMed/NCBI

109 

Wu W, Liu X, Wei L, Li T, Zang Y, Qian Y, Bai T, Li J, Xie M, Zhu Y, et al: Tp53 mutation inhibits ubiquitination and degradation of WISP1 via down-regulation of siah1 in pancreatic carcinogenesis. Front Pharmacol. 9:8572018. View Article : Google Scholar : PubMed/NCBI

110 

Ascherio A and Schwarzschild M: The epidemiology of Parkinson's disease: Risk factors and prevention. Lancet Neurol. 15:1257–1272. 2016. View Article : Google Scholar : PubMed/NCBI

111 

Bloem BR, Okun MS and Klein C: Parkinson's disease. Lancet. 397:2284–2303. 2021. View Article : Google Scholar : PubMed/NCBI

112 

Lees A, Hardy J and Revesz T: Parkinson's disease. Lancet. 373:2055–2066. 2009. View Article : Google Scholar : PubMed/NCBI

113 

Franck T, Krueger R, Woitalla D, Müller T, Engelender S and Riess O: Mutation analysis of the seven in absentia homolog 1 (SIAH1) gene in Parkinson's disease. J Neural Transm (Vienna). 113:1903–1908. 2006. View Article : Google Scholar : PubMed/NCBI

114 

Nagano Y, Yamashita H, Takahashi T, Kishida S, Nakamura T, Iseki E, Hattori N, Mizuno Y, Kikuchi A and Matsumoto M: Siah-1 facilitates ubiquitination and degradation of synphilin-1. J Biol Chem. 278:51504–51514. 2003. View Article : Google Scholar : PubMed/NCBI

115 

Liani E, Eyal A, Avraham E, Shemer R, Szargel R, Berg D, Bornemann A, Riess O, Ross CA, Rott R and Engelender S: Ubiquitylation of synphilin-1 and alpha-synuclein by SIAH and its presence in cellular inclusions and Lewy bodies imply a role in Parkinson's disease. Proc Natl Acad Sci USA. 101:5500–5505. 2004. View Article : Google Scholar : PubMed/NCBI

116 

Burchell VS, Nelson DE, Sanchez-Martinez A, Delgado-Camprubi M, Ivatt RM, Pogson JH, Randle SJ, Wray S, Lewis PA, Houlden H, et al: The Parkinson's disease-linked proteins Fbxo7 and Parkin interact to mediate mitophagy. Nat Neurosci. 16:1257–1265. 2013. View Article : Google Scholar : PubMed/NCBI

117 

Yamamura Y, Kuzuhara S, Kondo K, Yanagi T, Uchida M, Matsumine H and Mizuno Y: Clinical, pathologic and genetic studies on autosomal recessive early-onset parkinsonism with diurnal fluctuation. Parkinsonism Relat Disord. 4:65–72. 1998. View Article : Google Scholar : PubMed/NCBI

118 

Shahnawaz M, Mukherjee A, Pritzkow S, Mendez N, Rabadia P, Liu X, Hu B, Schmeichel A, Singer W, Wu G, et al: Discriminating α-synuclein strains in Parkinson's disease and multiple system atrophy. Nature. 578:273–277. 2020. View Article : Google Scholar : PubMed/NCBI

119 

Lövestam S, Schweighauser M, Matsubara T, Murayama S, Tomita T, Ando T, Hasegawa K, Yoshida M, Tarutani A, Hasegawa M, et al: Seeded assembly in vitro does not replicate the structures of α-synuclein filaments from multiple system atrophy. FEBS Open Bio. 11:999–1013. 2021. View Article : Google Scholar : PubMed/NCBI

120 

Schweighauser M, Shi Y, Tarutani A, Kametani F, Murzin AG, Ghetti B, Matsubara T, Tomita T, Ando T, Hasegawa K, et al: Structures of α-synuclein filaments from multiple system atrophy. Nature. 585:464–469. 2020. View Article : Google Scholar : PubMed/NCBI

121 

Szargel R, Shani V, Elghani FA, Mekies LN, Liani E, Rott R and Engelender S: The PINK1, synphilin-1 and SIAH-1 complex constitutes a novel mitophagy pathway. Hum Mol Genet. 25:3476–3490. 2016. View Article : Google Scholar : PubMed/NCBI

122 

Subramaniam SR and Chesselet MF: Mitochondrial dysfunction and oxidative stress in Parkinson's disease. Prog Neurobiol. 106–107. 17–32. 2013.PubMed/NCBI

123 

Pickrell AM and Youle RJ: The roles of PINK1, parkin, and mitochondrial fidelity in Parkinson's disease. Neuron. 85:257–273. 2015. View Article : Google Scholar : PubMed/NCBI

124 

Ganguly G, Chakrabarti S, Chatterjee U and Saso L: Proteinopathy, oxidative stress and mitochondrial dysfunction: Cross talk in Alzheimer's disease and Parkinson's disease. Drug Des Devel Ther. 11:797–810. 2017. View Article : Google Scholar : PubMed/NCBI

125 

Rocha EM, De Miranda B and Sanders LH: Alpha-synuclein: Pathology, mitochondrial dysfunction and neuroinflammation in Parkinson's disease. Neurobiol Dis. 109:249–257. 2018. View Article : Google Scholar : PubMed/NCBI

126 

Macdonald R, Barnes K, Hastings C and Mortiboys H: Mitochondrial abnormalities in Parkinson's disease and Alzheimer's disease: Can mitochondria be targeted therapeutically? Biochem Soc Trans. 46:891–909. 2018. View Article : Google Scholar : PubMed/NCBI

127 

Wang T, Zhang J, Hu M, Zhang Y, Cui P, Li X, Li J, Vestin E, Brännström M, Shao LR and Billig H: Differential expression patterns of glycolytic enzymes and mitochondria-dependent apoptosis in PCOS patients with endometrial hyperplasia, an early hallmark of endometrial cancer, in vivo and the impact of metformin in vitro. Int J Biol Sci. 15:714–725. 2019. View Article : Google Scholar : PubMed/NCBI

128 

Buratti J, Ji L, Keren B, Lee Y, Booke S, Erdin S, Kim SY, Palculict TB, Meiner V, Chae JH, et al: De novo variants in SIAH1, encoding an E3 ubiquitin ligase, are associated with developmental delay, hypotonia and dysmorphic features. J Med Genet. 58:205–212. 2021. View Article : Google Scholar : PubMed/NCBI

129 

Khan I and Leventhal BL: Developmental delay. StatPearls StatPearls Publishing. Copyright © 2021, StatPearls Publishing LLC.; Treasure Island, FL: 2021

130 

Oberklaid F and Efron D: Developmental delay - identification and management. Aust Fam Physician. 34:739–742. 2005.PubMed/NCBI

131 

Martiniuk AL, Vujovich-Dunn C, Park M, Yu W and Lucas BR: Plagiocephaly and developmental delay: A systematic review. J Dev Behav Pediatr. 38:67–78. 2017. View Article : Google Scholar : PubMed/NCBI

132 

Zhou T, Chen Y, Zhao B, Hu S, Li J, Liu M, Liang S, Bao Z and Wu X: Characterization and functional analysis of SIAH1 during skin and hair follicle development in the angora rabbit (Oryctolagus cuniculus). Hereditas. 157:102020. View Article : Google Scholar : PubMed/NCBI

133 

Farmanullah Hosseini SM, Liang A, Hua G, Rehman ZU, Talpur HS, Salim M, Ahmad S, Abulaiti A, Khan M, et al: Adaptive molecular evolution of AKT3 gene for positive diversifying selection in mammals. Biomed Res Int. 2020:25846272020. View Article : Google Scholar : PubMed/NCBI

134 

Alcantara D, Timms AE, Gripp K, Baker L, Park K, Collins S, Cheng C, Stewart F, Mehta SG, Saggar A, et al: Mutations of AKT3 are associated with a wide spectrum of developmental disorders including extreme megalencephaly. Brain. 140:2610–2622. 2017. View Article : Google Scholar : PubMed/NCBI

135 

Ko HR, Jin E, Lee SB, Kim CK, Yun T, Cho SW, Park KW and Ahn JY: SIAH1 ubiquitin ligase mediates ubiquitination and degradation of Akt3 in neural development. J Biol Chem. 294:15435–15445. 2019. View Article : Google Scholar : PubMed/NCBI

136 

Taoufik E and Probert L: Ischemic neuronal damage. Curr Pharm Des. 14:3565–3573. 2008. View Article : Google Scholar : PubMed/NCBI

137 

Bruce-Keller AJ: Microglial-neuronal interactions in synaptic damage and recovery. J Neurosci Res. 58:191–201. 1999. View Article : Google Scholar : PubMed/NCBI

138 

Huo J, Zhu XL, Ma R, Dong HL and Su BX: GAPDH/Siah1 cascade is involved in traumatic spinal cord injury and could be attenuated by sivelestat sodium. Neuroscience. 330:171–180. 2016. View Article : Google Scholar : PubMed/NCBI

139 

Huo J, Ma R, Chai X, Liang HJ, Jiang P, Zhu XL, Chen X and Su BX: Inhibiting a spinal cord signaling pathway protects against ischemia injury in rats. J Thoracic Cardiovascular Surg. 157:494–503. 2019. View Article : Google Scholar : PubMed/NCBI

140 

Hara MR, Thomas B, Cascio MB, Bae BI, Hester LD, Dawson VL, Dawson TM, Sawa A and Snyder SH: Neuroprotection by pharmacologic blockade of the GAPDH death cascade. Proc Natl Acad Sci USA. 103:3887–3889. 2006. View Article : Google Scholar : PubMed/NCBI

141 

Bangaru ML, Park F, Hudmon A, McCallum JB and Hogan QH: Quantification of gene expression after painful nerve injury: Validation of optimal reference genes. J Mol Neurosci. 46:497–504. 2012. View Article : Google Scholar : PubMed/NCBI

142 

Chuang DM and Ishitani R: A role for GAPDH in apoptosis and neurodegeneration. Nat Med. 2:609–610. 1996. View Article : Google Scholar : PubMed/NCBI

143 

Su BX, Chen X, Huo J, Guo SY, Ma R and Liu YW: The synthetic cannabinoid WIN55212-2 ameliorates traumatic spinal cord injury via inhibition of GAPDH/Siah1 in a CB2-receptor dependent manner. Brain Res. 1671:85–92. 2017. View Article : Google Scholar : PubMed/NCBI

144 

Zhai D, Chin K, Wang M and Liu F: Disruption of the nuclear p53-GAPDH complex protects against ischemia-induced neuronal damage. Mol Brain. 7:202014. View Article : Google Scholar : PubMed/NCBI

145 

Li C, Feng JJ, Wu YP and Zhang GY: Cerebral ischemia-reperfusion induces GAPDH S-nitrosylation and nuclear translocation. Biochemistry (Mosc). 77:671–678. 2012. View Article : Google Scholar : PubMed/NCBI

146 

Jiang X and Shen X: Knockdown of miR-299-5p inhibits the progression of hepatocellular carcinoma by targeting SIAH1. Bull Cancer. 105:873–883. 2018. View Article : Google Scholar : PubMed/NCBI

147 

Hui Z, Zhijun Y, Yushan Y, Liping C, Yiying Z, Difan Z, Chunglit CT and Wei C: The combination of acyclovir and dexamethasone protects against Alzheimer's disease-related cognitive impairments in mice. Psychopharmacology (Berl). 237:1851–1860. 2020. View Article : Google Scholar : PubMed/NCBI

148 

Vogel J, Young A, Oxtoby N, Smith R, Ossenkoppele R, Strandberg OT, Joie RL, Aksman LM, Grothe MJ, Iturria-Medina Y, et al: Four distinct trajectories of tau deposition identified in Alzheimer's disease. Nat Med. 27:871–881. 2021. View Article : Google Scholar : PubMed/NCBI

149 

Knopman DS, Amieva H, Petersen RC, Chételat G, Holtzman DM, Hyman BT, Nixon RA and Jones DT: Alzheimer disease. Nat Rev Dis Primers. 7:332021. View Article : Google Scholar : PubMed/NCBI

150 

Wasik U, Schneider G, Mietelska-Porowska A, Mazurkiewicz M, Fabczak H, Weis S, Zabke C, Harrington CR, Filipek A and Niewiadomska G: Calcyclin binding protein and Siah-1 interacting protein in Alzheimer's disease pathology: Neuronal localization and possible function. Neurobiol Aging. 34:1380–1388. 2013. View Article : Google Scholar : PubMed/NCBI

151 

Chen Y, Lian YJ, Ma YQ, Wu CJ, Zheng YK and Xie NC: LncRNA SNHG1 promotes α-synuclein aggregation and toxicity by targeting miR-15b-5p to activate SIAH1 in human neuroblastoma SH-SY5Y cells. Neurotoxicology. 68:212–221. 2018. View Article : Google Scholar : PubMed/NCBI

152 

Pang RT, Liu WM, Leung CO, Ye TM, Kwan PC, Lee KF and Yeung WS: miR-135A regulates preimplantation embryo development through down-regulation of E3 ubiquitin ligase seven in absentia homolog 1A (SIAH1A) expression. PLoS One. 6:e278782011. View Article : Google Scholar : PubMed/NCBI

153 

Schafer AR, Smith JL, Pryke KM, DeFilippis VR and Hirsch AJ: The E3 ubiquitin ligase SIAH1 targets MyD88 for proteasomal degradation during dengue virus infection. Front Microbiol. 11:242020. View Article : Google Scholar : PubMed/NCBI

154 

Zhou Y, Li L, Liu Q, Xing G, Kuai X, Sun J, Yin X, Wang J, Zhang L and He F: E3 ubiquitin ligase SIAH1 mediates ubiquitination and degradation of TRB3. Cell Signal. 20:942–948. 2008. View Article : Google Scholar : PubMed/NCBI

155 

Nagano Y, Fukushima T, Okemoto K, Tanaka K, Bowtell DD, Ronai Z, Reed JC and Matsuzawa SI: Siah1/SIP regulates p27(kip1) stability and cell migration under metabolic stress. Cell Cycle. 10:2592–2602. 2011. View Article : Google Scholar : PubMed/NCBI

156 

Hara MR and Snyder SH: Nitric oxide-GAPDH-Siah: A novel cell death cascade. Cell Mol Neurobiol. 26:527–538. 2006. View Article : Google Scholar : PubMed/NCBI

157 

Song Y, Xu Y, Pan C, Yan L, Wang ZW and Zhu X: The emerging role of SPOP protein in tumorigenesis and cancer therapy. Mol Cancer. 19:22020. View Article : Google Scholar : PubMed/NCBI

158 

Shah M, Stebbins JL, Dewing A, Qi J, Pellecchia M and Ronai ZA: Inhibition of Siah2 ubiquitin ligase by vitamin K3 (menadione) attenuates hypoxia and MAPK signaling and blocks melanoma tumorigenesis. Pigment Cell Melanoma Res. 22:799–808. 2009. View Article : Google Scholar : PubMed/NCBI

159 

Zhang H, Cao X, Wang J, Li Q, Zhao Y and Jin X: LZTR1: A promising adaptor of the CUL3 family. Oncol Lett. 22:5642021. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2022
Volume 47 Issue 2

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang H, Wang J, Ge Y, Ye M and Jin X: Siah1 in cancer and nervous system diseases (Review). Oncol Rep 47: 35, 2022
APA
Zhang, H., Wang, J., Ge, Y., Ye, M., & Jin, X. (2022). Siah1 in cancer and nervous system diseases (Review). Oncology Reports, 47, 35. https://doi.org/10.3892/or.2021.8246
MLA
Zhang, H., Wang, J., Ge, Y., Ye, M., Jin, X."Siah1 in cancer and nervous system diseases (Review)". Oncology Reports 47.2 (2022): 35.
Chicago
Zhang, H., Wang, J., Ge, Y., Ye, M., Jin, X."Siah1 in cancer and nervous system diseases (Review)". Oncology Reports 47, no. 2 (2022): 35. https://doi.org/10.3892/or.2021.8246