SciELO - Scientific Electronic Library Online

 
vol.25 issue3The National Cancer Institute of Colombia in an exceptional moment of the "History of present times"Acral lentiginous melanoma: Review of one of the most frequent melanomas in Latin America author indexsubject indexarticles search
Home Pagealphabetic serial listing  

Services on Demand

Journal

Article

Indicators

Related links

  • On index processCited by Google
  • Have no similar articlesSimilars in SciELO
  • On index processSimilars in Google

Share


Revista Colombiana de Cancerología

Print version ISSN 0123-9015

rev.colomb.cancerol. vol.25 no.3 Bogotá July/Sept. 2021  Epub May 10, 2022

https://doi.org/10.35509/01239015.717 

Artículos de revisión

Speckle type POZ adaptor protein (SPOP) and its role in cancer

La proteína adaptadora Speckle-type POZ (SPOP) y su papel en cáncer

Wendy Montero-Ovallea 
http://orcid.org/0000-0001-5617-2686

Maria Carolina Sanabria-Salasb 
http://orcid.org/0000-0002-7946-2026

Martha Lucia Serranoa  b  * 
http://orcid.org/0000-0003-4060-4418

a. Grupo de Investigación en Biología del Cáncer, Instituto Nacional de Cancerología, ESE. Bogotá, Colombia.

b. Departamento de Química, Facultad de Ciencias, Universidad Nacional de Colombia. Bogotá, Colombia.


Abstract

Proteasomal degradation is an essential regulatory mechanism for cellular homeostasis maintenance. The speckle-type POZ adaptor protein (SPOP) is part of the ubiquitin ligase E3 cullin-3 RING-box1 complex, responsible for the ubiquitination and proteasomal degradation of biomolecules involved in cell cycle control, proliferation, response to DNA damage, epigenetic control, and hormone signaling, among others. Changes in SPOP have been associated with the development of different types of cancer, since it can act as a tumor suppressor mainly in prostate, breast, colorectal, lung cancer and liver cancer, due to point mutations and/or reduced expression, or as an oncogene in kidney cancer by protein overexpression. In endometrial cancer it has a dual role, since it can act as a tumor suppressor or as an oncogene. SPOP is a potential prognostic biomarker and a promising therapeutic target.

Keywords: SPOP; Ubiquitin ligase; Cancer; Biomarker; Oncogene; Tumor suppressor gene; proteasomal degradation

Resumen

La degradación proteosómica es un mecanismo de regulación esencial para el mantenimiento de la homeostasis celular. La proteína adaptadora Speckle-type POZ (SPOP) hace parte del complejo ubiquitin ligasa E3 cullin-3 RING-box1, encargado de la ubiquitinación y degradación proteosomal de biomoléculas involucradas en el control del ciclo celular, proliferación, respuesta al daño de ADN, control epigenético, señalización hormonal, entre otros. Las alteraciones en SPOP han sido asociadas al desarrollo de diferentes tipos de cáncer, ya que puede actuar como supresor tumoral principalmente en cáncer de próstata, mama, colorrectal y pulmón, debido a mutaciones puntuales y/o expresión reducida o como oncogén en cáncer riñón por sobreexpresión de la proteína. En cáncer endometrial tiene un rol dual, ya que puede actuar como supresor tumoral o como oncogén. SPOP es considerado como un potencial biomarcador pronóstico y un objetivo terapéutico prometedor.

Palabras clave: SPOP; Ubiquitin Ligasa; Cáncer; Biomarcador; Oncogen; Gen supresor tumoral; degradación proteasomal

Introduction

Degradation of biomolecules is an especially important regulatory mechanism for maintaining cell balance. It may be mediated by the lysosomal or the ubiquitin-proteasome pathways, the latter being responsible for maintaining intracellular levels of proteins involved in many cellular processes such as cell cycle regulation, proliferation, apoptosis, response to DNA damage, and transcriptional activation, among others. Many of the proteins involved in the process of ubiquitination and proteolysis have gained attention because of the effects they can have on the development of cancer 1. One of them is the Speckle-type POZ protein (SPOP), which acts as an adaptor protein in the ubiquitin ligase E3 cullin-3 RING-box1 (Cul3-RING-box1) complex, recruiting substrates for ubiquitination and subsequent degradation in the 26S proteasome 2. Among the SPOP substrates, there are the androgen receptor (AR) 3,4, the estrogen receptor (ER) 5,6, the steroid receptor coactivator 3 (SRC3) 4, the bromodomain and extra-terminal BET domain proteins 7,8, the cell-division cycle protein 20 (Cdc20) 9 and the proteins associated with DAXX and FADD death domains (10-12), among other effectors, which show its importance in cell development and growth.

SPOP has recently been found to act as a tumor suppressor gene (TSG) or oncogene in different types of cancer, as shown by findings of mutations, loss in the number of copies and/or reduced expression, gain of function or protein overexpression, and it has been associated with prognosis, which has suggested SPOP as a prognostic biomarker and promising therapeutic goal (13-18). In this review, we will describe the structure of the adaptor protein, its main functions and molecular mechanisms, its relationship with cancer, its usefulness as a prognostic biomarker, and therapeutic advances.

2. Methodology

2.1 Eligible studies

We conducted a search of the literature from the last 16 years (2004 - 2020) using Pubmed (NIH) and Scielo 19,20. The eligibility criteria included original articles that addressed different roles of the SPOP protein, its main functions and molecular mechanisms related to the development of different types of cancer, as well as its usefulness as a prognostic biomarker and cancer treatment. Relevant review articles were consulted as well. Studies written in languages other than English or Spanish, studies that did not describe the topics mentioned and studies whose publications were not accessible were excluded.

2.2 Publication search

An initial search was performed in the PubMed database combining the Medical Subject Headings (MeSH): "SPOP", "SPOP mutation", "SPOP in cancer", "Speckle-type POZ protein" and retrieve 130 publications from which 106 articles were eligible. The last search was carried out on July 29, 2020. We revised manuscripts cited within those studies to identify additional publications that fulfilled our eligibility criteria. A total of 112 unique publications were included in our review.

3. Structure of the SPOP protein

The SPOP protein was first identified in 1997 as a nuclear protein with a speckle-type pattern 21. This protein is encoded by the SPOP gene which is located in locus 17q21.33 and has 16 exons (79280 bp). It has an isoform called SPOPL (SPOP like) that is encoded in locus 2q22.1 and is formed by 12 exons and 71778 bp (Figure 1 A) 22. SPOPL shares an overall 85% protein sequence identity with SPOP but has 18 more amino acid residues, suggesting that the isoform could perform similar functions; though, as we will see below, it also seems to play a role in modulating the SPOP function (2,23).

Figure 1 Structure and function of the SPOP protein. A. SPOP and SPOPL locus. B. SPOP domains: MATH (light blue), BTB (green), BACK (dark blue), and nuclear localization sequence (NLS) C. Dimeric and oligomeric association of SPOP. D. Cul3-Rbx1 E3 ubiquitin ligase complex, showing the adapter role of a SPOP oligomer for substrate ubiquitination (gray wavy line) and subsequent degradation in the 26S proteasome. E. In the nucleus, SPOP is observed in higher-order oligomers as small green nuclear speckles, and the substrate available to be ubiquitinated is shown in red. Increase in substrate concentration generates SPOP-substrate localization foci that form membraneless liquid organelles (yellow), which favors SPOP oligomerization and location of the mono or multivalent substrate for its ubiquitination.  

The SPOP protein is made up of 374 amino acid, has a molecular weight of 43.13 kDa, and its secondary structure consists of 18 a-helices and 14 6 sheets, which form three main domains: MATH, BTB, and BACK (Figure 1B). Followed by the BACK domain, in the C-terminal region is the NLS, which allows its location in the nucleus 24. It has been shown that SPOP can be found in a dimeric or oligomeric conformation, which increases binding to substrates (Figure 1C).

MATH (Meprin and TRAF-C homology) is the N-terminal domain, which function is to recruit substrates to the ubiquitin ligase complex by forming a cleft surrounding the hydrophobic side chain of the substrate (Figure 1D) 2,24-26.

BTB (Bric-a-brac/Tamtrack/Broad), is an internal domain known as POZ (zinc finger and poxvirus protein) that allows SPOP binding to the ubiquitin ligase complex via Cullin3 (Figure 1D), and also allows the formation of SPOP-SPOP homodimers or SPOP-SPOPL heterodimers 2.

BACK (BTB with Kelch repeats) is a carboxyterminal domain that allows a greater interaction surface between BTB and Cul3, besides the formation of oligomers through interaction with other SPOP BACK domains (Figure 1C); this allows recruiting multivalent substrates, and significantly increases their degradation (Figure 1D) 2,14. The interaction between SPOP oligomers and substrates generates membraneless "organelles" formed by liquid-liquid phase separation (LLPS), as nuclear speckles (Figure 1E) 14,27. The creation of these complexes is dependent on the concentration of SPOPL since, by lacking 18 residues in the BACK domain, it is not possible to form oligomers, suggesting SPOPL as a regulator in the ubiquitination process 28. Formation of these LLPS enhances binding of enzymes and substrates, and it is suggested that the organelles that are created could be involved in cell proteostasis processes such as location, control, and balance of different proteins. Perturbations in proteostasis, leads to an accumulation of damaged and misfolded proteins 14.

4. Functions of SPOP

4.1 Regulation of substrates by proteasomal degradation

Approximately 20% of protein degradation in the cell is mediated by Ubiquitin ligases E3 CRL-RING complexes. Specifically, Cul3 uses proprietary substrate adaptors such as SPOP 29. Figure 1D shows the interaction of a SPOP oligomer with the Ubiquitin ligase E3 Cul3-RING-box1 complex. The ubiquitinated substrate is targeted to the 26S proteasome for degradation 29.

More than 33 substrates of SPOP have been described, whose functions vary and involve regulating important processes at the cellular level (Table 1) 3,4,5,7,8,9 12,14,22,28-36,37-46,47-56,57. It is interesting to see that most of these substrates have been associated with a particular type of cancer, and this may be due to the impact that a substrate can have on a given tissue, and causes different SPOP mutations to affect the degradation of said substrates, therefore being associated with a specific type of cancer.

Table 1 SPOP substrates classified by function and type of cancer. 

Type of cancer Function Substrate Reference
SPOP as a tumor suppressor gene (TSG)
Mutations in the MATH domain with loss-of-function: Increased effect on the substrate
Prostate Steroid hormone receptor Androgen receptor (AR) 3,4
Transcriptional regulation Tripartite motif-containing 24 (TRIM24) 38,39
Steroid receptor coactivator 3 (SRC3) 4,14
ETS transcription factor (ERG) 40,41
Activating transcription factor-2 (ATF2) 28
SR-related CTD associated factor 1 (SCAF1) 30
Proto-oncogene c-Myc 8,29
Homeobox transcription factor NANOG 32-34
Hedgehog pathway proteins: glioma-associated oncogene Gli2 25
Cell cycle regulation Cell division cycle protein 20 (Cdc20) 9
Cyclin E1 44
Cell cycle associated protein 1 (CAPRIN1) 45
Cell cycle regulation, transcription, and apoptosis Death domain-associated protein (DAXX) 10,11, 14
Epigenetic regulation / Chromatin remodeling and transcription regulation Proto-oncogene DEK 40
Zinc finger protein WIZ 30
Bromodomain and extra-terminal (BET) protein family (BRD2/3/4) 7,8,23) (78,103, 107
Immune system suppression Programmed death-ligand 1 (PDL1) 50
SUMO removal Sentrin-specific protease 7 (SENP7) 18
Regulation of actin polymerization, maintenance of Golgi structure, and mitochondrial fission Inverted formin 2 (INF2) 109
Lung Cell cycle regulation and apoptosis FAS-associated protein with death domain (FADD) 12
DNA repair and transcription regulation NAD-dependent deacetylase sirtuin 2 (SIRT2) 49
Endometrial Steroid hormone receptor Estrogen receptor (ER) 5
Transcription regulation Zinc finger and BTB domain containing 3 (ZBTB3 43
Steroid receptor coactivator 3 (SRC3) 5
Low SPOP expression levels: Increased effect on the substrate
Breast Steroid hormone receptor Progesterone receptor (PR) 37
Epigenetic and transcriptional regulation Breast cancer metastasis suppressor 1 (BRMS1) 31
Cell cycle regulation, transcription, and apoptosis Death domain-associated protein (DAXX) 10,11, 14
Colorectal Transcription regulation Interleukin enhancer binding factor 3 (ILF3) 110
Hedgehog pathway proteins: glioma-associated oncogene Gli2 86
Cell cycle regulation, transcription, and apoptosis Death domain-associated protein (DAXX) 10,11, 14
Liver SUMO removal Sentrin-specific protease 7 (SENP7) ( 51
SPOP overexpression: Decreasing effect on the substrate
Gastric Transcription regulation Hedgehog pathway proteins: glioma-associated oncogenes Gli2/Gli3 35,42
SPOP as an oncogene
Mutations in the MATH domain with gain-of-function: Decreasing effect on the substrate
Endometrial Chromatin remodeling and transcription regulation Bromodomain and extra-terminal (BET) protein family (BRD2/3/4) 7,78
SPOP overexpression: Decreasing effect on the substrate
Kidney Regulation of signaling pathways with phosphatase activity Dual-specificity phosphatase 6/7 (DUSP6/7) 93,102
Phosphatase, tumor suppressor *Phosphatidylinositol 3,4,5-trisphosphate 3-phosphatase and dual-specificity protein phosphatase (PTEN) 102
Cell cycle regulation, transcription, and apoptosis Death domain-associated protein (DAXX) 10,11, 14
Transcription regulation Hedgehog pathway proteins: glioma-associated oncogene Gli2 93
Chromatin remodeling SET domain containing 2, histone lysine methyltransferase (SETD2) 48
Cell cycle regulation and apoptosis Large tumor suppressor kinase 1 (LATS1) 56
CL myeloid (K562-HPC7) and animal model (mouse) Signal transduction to IL-1R-associated kinases (IRAK) Myeloid differentiation primary response 88 (MYD88) 54,55, 57
CL osteosarcoma Epigenetic regulation /Chromatin **B Lymphoma Mo-MLV insertion region 1 homolog (BMI1) 53
(U2OS) remodeling **Histone variant macroH2 47,53

CL: Cell line. *PTEN phosphatase is a SPOP substrate only in kidney cancer, in which SPOP is in the cytoplasm. ** Substrates with poor location.

4.2 Role in the response to DNA damage

It has been proposed that SPOP modulates transcriptional repression activities and participates in the repair of double-stranded DNA ruptures (DSB) 60, where it is recruited forming nuclear foci, which depend on the kinase activity of the ataxia-telangiectasia (ATM) protein, and are colocalized with histone y-H2AX foci in response to damage. Depletion or decrease in SPOP has been shown to generate impaired DNA damage response (DDR) and high sensitivity to ionizing radiation; however, the mechanism has not yet been determined, but it could be associated with the regulation it exerts on the levels of expression of Rad51 and Ku80 factors, which are important in DSB repair by homologous recombination (HR) 17,61. In addition, an interactome analysis showed that SPOP is associated with multiple proteins involved in the transcription, splicing, and export of the mRNA molecules 62.

4.3. Function of SPOPL

Besides being a regulator of SPOP, one study found that SPOPL is involved in the regulation of endosome maturation and traffic of endosome load to lysosomes and that this depended on ubiquitination and degradation of the EPS15 endocytic adapter, which according to the study, is a SPOPL substrate, not a SPOP substrate 63.

5. Relationship of SPOP with cancer: a view from protein structure and function

SPOP plays a key role in maintaining cell development and growth by controlling the degradation of proteins that regulate important processes for cell homeostasis (Table 1). However, it is striking that, apart from cancer, just a few functions of SPOP have been described, such as regulation of fetal hemoglobin expression 64, regulation of insulin and glucose homeostasis and having the pancreatic and duodenal homeobox transcription factor (PDX1) as a substrate 54,55. SPOP is also believed to have a role in the development of neurological disorders, as "de novo" genetic variants have been found in one study 65.

Given its functions, SPOP can act as a tumor suppressor gene (TSG), by regulating processes of cell division, repair and apoptosis, or as an oncogene, inducing proliferation, through different mechanisms as shown by findings of reduced expression or overexpression of the protein, loss of copy number, or mutations in the gene in different types of cancer 13-18.

5.1 SPOP as a tumor suppressor gene

5.1.1 Mutations in the MATH domain with loss-of-function

Missense mutations with loss of function have been found in prostate cancer (PCa), endometrial cancer (EC), and lung cancer (LC) (Figure 2 A); however, mutation hot spots are different. In PCa, they are located in the substrate-binding cleft, in EC they are outside the cleft, and in LC they are both inside and outside this site (Figure 2 B); these differences could be related to the role of SPOP in each of these malignancies, since SPOP substrates in PCa and LC are overexpressed, whereas in EC some of the substrates are overexpressed but others are underexpressed 38,66-71.

SPOP mutation in PCa is frequent and is taken as one of the molecular subtypes identified by the TCGA. Although a germline mutation in the BTB domain (N296I), associated with hereditary PCa has been described in a family of European ancestry 72, the remaining mutations found are somatic in the MATH domain (Figure 2). In localized PCa, the mutation frequency is between 6% and 15%, and between 15% and 25% in advanced PCa. A frequency of 20% has been reported in patients with African ancestry, while in those with European ancestry it ranges between 6% and 10%, between 7% and 12% in Asians, and between 5% and 14% in North Americans 24,38,69,73-76.

The frequency of SPOP mutations described in EC ranges between 6% and 10%, mainly in serous tumors and clear cell carcinoma 5,66,70,77,78, and 6% in non-small cell LC (NSCLC) (Figure 2) 70,71. Not all SPOP mutations in these cancers have been associated with an effect on protein function.

Figure 2 Point mutations of the MATH domain in SPOP in prostate, endometrial, and lung cancer. A. Mutation frequency, mutation hot spots, and number of mutations reported in prostate cancer (top), and endometrial and lung cancer (bottom) with clinical impact on the disease. B. Crystal structure of the MATH domain with missense point mutations, relevant in different types of cancer. Residues are marked with different colors depending on the type of cancer: prostate cancer (red), endometrial cancer (blue), and lung cancer (green). Endometrial cancer mutations include loss and gain of function. 

These loss-of-function mutations prevent SPOP from binding to its substrates, increasing their quantity. Figure 3 shows some SPOP substrates, by type of cancer, that participate in signaling pathways that could be deregulated and associated with tumor progression processes. In PCa, the overexpression of AR is associated with increased proliferation and survival 4,14. Another SPOP substrate found in EC and PCa is SRC3, which is found overexpressed in these tumors and activates the PI3K/mTOR and AKT signaling pathways (Figure 3), causing a higher cell growth and proliferation rate 3,4,66,79.

Figure 3 Signaling pathways compromised by SPOP alterations in different types of cancer. The signaling pathways presented here involve proteins that are SPOP substrates and are associated with increased or decreased growth, survival, migration, and invasion. The arrows next to these proteins indicate the following: upward arrows show overexpression and downward arrows show low expression. Pathway components are marked in colors according to the type of cancer in which their involvement has been demonstrated. Prostate cancer: AR pathway and SRC3/PI3K/Akt pathway. Breast cancer: PR pathway and metastasis suppressor 1 (BRMS1) pathway. Colorectal cancer: PI3K/Akt/Sp1 and Hedgehog pathways. Lung cancer: FAS-associatedprotein with death domain (FADD)/NF-kfi pathway. Liver cancer: SENP7pathway. Endometrial cancer: Pathway associated with ZBTB3 and increase in SRC3. Gastric cancer: Hedgehog pathway. Kidney cancer: Pathway associated with ZEB1. T (testosterone), DHT (dihydrotestosterone), SRC3 (steroid receptor coactivator 3), IGF-1 (insulin growth factor type 1), P (progesterone), MAPK (mitogen-activated protein kinase), uPA (urokinase-type plasminogen activator), OPN (osteopontin), MMP2/7 (metalloproteinases 2/7), SHH (Sonic hedgehog protein), Ptch (Patched), Smo (Smoothened), Gli2 (glioma-associated oncogene 2), SUMO (small ubiquitin-like modifier), HP1-a (heterochromatin protein 1 alpha), TCF4 (transcription factor 4). 

Other substrates are also increased in EC, such as the estrogen receptor alpha (ERa), which increases its transactivation 5, and the expression of the transcription factor ZBTB3, a substrate that accumulates and generates a positive regulation on Sonic hedgehog (SHH), which in turn promotes tumor proliferation and survival (Figure 3) 45.

Proteins from the bromodomain family and the extraterminal BET domain, such as BDR2, BDR3, and BDR4, have also been related to PCa and EC. These proteins increase their stability by mutations in some SPOP residues in PCa, while in EC their amount decreases due to the role of SPOP as an oncogene, which will be discussed later 80.

In the case of NSCLC, an increase in two substrates has been observed: NAD-dependent deacetylase sirtuin 2 (SIRT2), which favors tumor proliferation 51 and FADD, which regulates the NF-k/3 activity and is associated with an unfavorable prognosis (Figure 3) 12.

There are other genetic changes in PCa related to mutated SPOP, such as somatic deletions in 5q21 and 6q21 60,81. Locus 5q21 contains the CHD1 gene, which encodes for helicase with DNA-binding chromodomain, involved in regulating gene transcription through chromatin interaction and remodeling 38, and in genomic stability 82. Moreover, locus 6q21 contains the forkhead box O3 gene (FOXO3), which acts as a regulatory transcription factor of genes necessary for cell death, so its loss is related to PCa carcinogenesis and progression 38. This could mean that the effects of SPOP mutations are synergetic with those of these genes or others located in these regions.

Loss of SPOP function also affects DNA repair 62. In LC and PCa, a high genomic instability has been found, related to a deficiency in HR, a decrease in the recruitment of RAD51, and an increase in NHEJ 60-62. In LC, the decrease in SPOP expression also affects DDR, increases apoptosis, and activates cell cycle control points induced by ionizing radiation 61, and in PCa leads to replicative stress and cell cycle deregulation. In PCa, the F133V mutant generates a deficiency in the expression of BRCA2, ATR, CHK1, RAD51, TDP1, TDP2, and MRE11, which are essential enzymes for DNA repair 62,83,84.

5.1.2 Low levels of SPOP expression

Low SPOP expression has been found in breast cancer (BC), colorectal cancer (CRC) and liver cancer (HCC), associated with increased substrate expression (Figure 3), and even though point mutations have also been found, these have not been associated with loss of function or changes in SPOP expression 33,39,53,70,85,86.

In BC, low SPOP levels cause overexpression of the progesterone receptor (PR), favoring cell cycle progression and activation of ERK1/2 39 and breast cancer metastasis suppressor 1 (BRMS1), which inhibits activation of urokinase-type plasminogen activator (uPA) and osteopontin (OPN), promoting the onset and progression of this type of cancer (Figure 3) 33.

In CRC, the PI3K/Akt pathway that increases the levels of metalloproteinases is activated, promoting tumor proliferation and migration 86,87. Furthermore, Gli2 is also overexpressed causing an increase in the levels of the anti-apoptotic protein Bcl-2, which favors the inhibition of cell death (Figure 3) 88.

In HCC, SENP7 is increased, which in turn, induced by HP1- «, rises vimentin levels, and this triggers processes of migration, invasion, and metastasis (Figure 3) 53.

SENP7 is responsible for mediating epigenetic silencing in PCa 18, and invasion in BC 89.

5.1.3 SPOP overexpression

SPOP protein has been found highly expressed in gastric cancer (GC), and in this model acts as a TSG, since it has been related to the inhibition of the Hedgehog signaling pathway through the accelerated degradation of its Gli2 substrate (Figure 3), decreasing the processes of tumor proliferation, migration, and invasion 37; in addition, it has been found that SPOP suppresses the growth of cancer stem cells and reduces their power 90.

5.2 SPOP as an oncogene

5.2.1 SPOP overexpression

In clear cell renal cell carcinoma (ccRCC), SPOP is overexpressed and accumulates in the cytoplasm and increases degradation of PTEN, DUSP6, DUSP7, DAXX, Gli2, and LATS1 proteins, leading to a higher proliferation, survival, invasion, and apoptosis processes (24,58,91) and it can also increase the expression of β-catenin and transcription Factor 4 (TCF4), which positively regulate transcription factor ZEB1, an inducer of mesenchymal-epithelial transition (MET), favoring migration and invasion (Figure 3) 92.

BET proteins are underexpressed in EC as a result of the gain of function of SPOP due to mutations in residues E47, E50, E78, S80, M117, and D140 (Figure 2) 80. On the other hand, BET proteins are overexpressed in PCa. This shows how complex the role of SPOP in carcinogenesis is. Moreover, there are reports about the dual role of SPOP in the endometrium, since both loss of function and gain of function mutations has been described.

5.3 Regulation of SPOP expression

The decrease in SPOP expression in CRC and LC has been associated with hypermethylation of the SPOP promoter or binding sites close to the SPOP promoter (Figures 4A and 4B) 88,93 and to the binding of SMAD2/3 to the same promoter (Figure 4 C) 94. In GC, regulation in SPOP expression appears to be mediated by long non-coding RNA (LncRNA) ADAMTS9-AS2 90. In ccRCC, SPOP regulation could be mediated by hypoxia-inducible factors 1 and 2 (HIF1/2), which increase SPOP levels, generating accumulation in cells (Figure 4 D) 91. It has also been identified that the miR-520/372/373 family joins a 3'-UTR region of SPOP by suppressing its expression; these miRNA are regulated downwards in ccRCC (Figure 4 E) 95. More studies are required to know the precise mechanisms of regulation in each of these processes.

Figure 4 Regulatory mechanisms of SPOP expression. A. Hypermethylation in the SPOP promoter region. B. Hypermethylation at C/EBP& binding sites close to the SPOP promoter. C. Binding of TGF-Ji to its receptor leads to the binding of phosphorylated SMAD2/3 and SMAD4 to the SPOP promoter. Mechanisms A, B, and C decrease or inhibit SPOP expression. D. Hypoxia causes an increase in hypoxia-inducible factors HIF1/2, which are attached to a hypoxia-response element (HRE) located in the SPOP sequence. E. The overexpressed E2F1 transcription factor binds to the promoter regions of miR-520/372/373, blocking their expression, preventing these miRNAs from binding to the 3'UTR SPOP sequence, preventing their negative regulation on the protein. Mechanisms D and E increase SPOP expression. 

5.4 Usefulness of SPOP as a prognostic biomarker in cancer

It has been proposed that SPOP could be a prognostic biomarker in some types of cancer. Its role in PCa is controversial, some studies reveal that patients with mutated SPOP or with low protein expression have shorter biochemical and clinical progression free survival, compared to patients without the mutation (27.69.96) ; however, in other studies mutated SPOP has been associated with a favorable prognosis, finding a lower frequency of positive margins, extraprostatic extension, and invasion of seminal vesicles in radical prostatectomy, in addition to higher metastatic-free survival, particularly in high preoperative PSA patients (67.68.97) . More PCa studies are needed to analyze the effect of other variables, such as age, pathological condition, Gleason score, or ethnicity, in the prognosis when the mutation is present.

In CRC, HCC, and NSCLC, the decrease in SPOP levels seems to be involved in a worse prognosis of the disease associated with poor cell differentiation, distant metastasis, advanced TNM and shorter overall survival 53,86-88,98. In the same sense, SPOP overexpression in patients with GC is associated with a good prognosis, given that less metastasis in lymph nodes, greater histological differentiation, and less advanced TNM staging are evident 37,99. In contrast, in ccRCC, SPOP overexpression is related to a high histological grade and tumors with local invasion or metastasis, as well as with a shorter recurrence-free survival and advanced stage of the disease 92,100,101.

5.5 SPOP as a potential therapeutic target

In ccRCC, characterized with an overexpression of SPOP, therapeutic modulators such as miR-520/372/373, could be implemented as pharmacological agents for patients with this type of cancer, since it inhibits SPOP translation and decrease tumor size and metastasis 91,95. Also, other small molecules has been proposed to bind to cytoplasmic SPOP, preventing its interaction with substrates such as PTEN or DUSP7 in ccRCC and increasing the ell viability (Figure 5 A) 102-104.

Figure 5 Possible therapeutic interventions in tumors with SPOP alterations, according to action mechanisms. A.Inhibition of SPOP expression by miRNAs or small molecules (6b) that bind to the MATH (light blue) domain of SPOP and prevent substrate degradation. B. Effect on SPOP substrates: Inhibitors of its function (JQ1 inhibits BET proteins) or molecules that promote its degradation (PROTAC ARV-771 and NEO2734 promote BET degradation). C. Inhibition of signaling pathways activated by alteration in SPOP (RUSKI-43 inhibits the Hedgehog pathway and the AKT/mTOR and AR pathways by using HDAC3 inhibitors. D. Because of the effect of SPOP on DNA damage, PARP inhibitors may be used to generate effects on DNA repair, inducing cell death 

.

In those cases, in which mutations or underexpressed SPOP leads to increased substrates levels, as in EC, inhibitors of BET proteins, such as JQ1 or small molecules called proteolysis targeting chimeras (PROTAC) has been used and may favor patients with overexpressed AR or BET proteins. The designed chimera, ARV-771, has been showing great efficacy against BRD4 with suppression of AR levels and its signaling, leading to tumor regression; also the agent NEO2734, has been proposed as a new BET inhibitor, but its anticancer efficacy on patients is still under study (Figure 5 B ) 105-109.

It is also possible to block signaling pathways by inhibiting effectors such as the SHH protein by using the RUSKI-43 inhibitor as a therapeutic agent in EC, which suppresses proliferation, migration, and invasion 45,80. In the case of patients with aberrant activation of the AKT/mTOR and AR pathways in PCa, it was found that inhibition of histone deacetylase 3 (HDAC3) can target these pathways, decreasing their activation and suppressing tumor growth in organoids of patients with these characteristics (Figure 5C) 110.

Therapies that affect DNA repair, such as poly (ADP-ribose) polymerase (PARP) inhibitors, could benefit patients with PCa and LC, in which repair has also been affected with SPOP mutations (Figure 5D) 60.

Another study showed that patients with metastatic castration-resistant PCa (mCRPC) with SPOP mutations and/or CHD1 deletions, have a higher response rate to treatment with abiraterone, an androgen synthesis inhibitor, as compared to cases lacking these alterations; however, prospective clinical trials are required to validate this response to treatment 76.

6. SPOPL and cancer

Association and clinical relevance of SPOPL in medulloblastoma have been found. The expression of this isoform is reduced in 75% of biopsies analyzed, and this expression can be related to the level of cell differentiation and the 5-year cumulative survival when compared to patients with increased expression. This suggests that SPOPL could function as a biomarker of poor prognosis in patients with medulloblastoma 23.

Conclusion

Evidence shows that SPOP is a multifunctional adaptor protein, which plays a very important role in maintaining and regulating cell cycle, cell proliferation, response to DNA damage, epigenetic control, and transcriptional control, among others, through ubiquitination and degradation of a large number of substrates. Mutations or changes in the expression of the protein, which trigger tumor progression are evident in different types of cancer, showing the role of SPOP as a tumor suppressor gene or oncogene, and this has been associated with clinicopathological characteristics, which allows considering SPOP as a prognostic biomarker with potential targeted therapies. Given that the adapter protein acts differently in different types of cancer, also depending on the substrates, it is important to do further research on the molecular and biochemical mechanisms involved in carcinogenesis generated by SPOP, to improve understanding of its role in each of these malignancies, aiming at a better classification and association with outcomes

Bibliography

1. Soave CL, Guerin T, Liu J, Dou QP. Targeting the ubiquitin-proteasome system for cancer treatment: discovering novel inhibitors from nature and drug repurposing. Cancer Metastasis Rev. 2017;36(4):717-36. [ Links ]

2. Errington WJ, Khan MQ, Bueler SA, Rubinstein JL, Chakrabartty A, Privé GG. Adaptor protein self-assembly drives the control of a cullin-RING ubiquitin ligase. Structure. 2012;20(7):1141-53. [ Links ]

3. Blattner M, Liu D, Robinson BD, Huang D, Poliakov A, Gao D, et al. SPOP Mutation Drives Prostate Tumorigenesis In Vivo through Coordinate Regulation of PI3K/mTOR and AR Signaling. Cancer Cell [Internet]. 2017;31(3):436-51. Available from: http://dx.doi.org/10.1016/j.ccell.2017.02.004. [ Links ]

4. Chuandong, Rajapakshe, Shrijal S. Androgen receptor is the key transcriptional mediator of the tumor suppressor SPOP in prostate cancer. Cancer. 2015;74(19):5631-43. [ Links ]

5. Zhang P, Gao K, Jin X, Ma J, Peng J, Wumaier R, et al. Endometrial cancer-associated mutants of SPOP are defective in regulating estrogen receptor-a protein turnover. Cell Death Dis. 2015;6:e1687. [ Links ]

6. Petrovics G, Price DK, Lou H, Chen Y, Garland L, Bass S, et al. Increased frequency of germline BRCA2 mutations associates with prostate cancer metastasis in a racially diverse patient population. Prostate Cancer Prostatic Dis [Internet]. 2018; Available from: http://dx.doi.org/10.1038/s41391-018-0114-1. [ Links ]

7. Janouskova H, El Tekle G, Bellini E, Udeshi ND, Rinaldi A, Ulbricht A, et al. Opposing effects of cancer-Type-specific SPOP mutants on BET protein degradation and sensitivity to BET inhibitors. Nat Med [Internet]. 2017;23(9):1046-54. Available from: http://dx.doi.org/10.1038/nm.4372Links ]

8. Dai X, Gan W, Li X, Wang S, Zhang W, Huang L, et al. Prostate cancer - associated SPOP mutations confer resistance to BET inhibitors through stabilization of. Nat Publ Gr. 2017 Sep;23(9):1063-1071. doi: 10.1038/nm.4378. [ Links ]

9. Wu F, Dai X, Gan W, Wan L, Li M, Mitsiades N, et al. Prostate cancer-associated mutation in SPOP impairs its ability to target Cdc20 for poly-ubiquitination and degradation. 2018;207-14. [ Links ]

10. Papers JBC, Doi M, Kwon JE, La M, Oh KH, Oh YM, et al. BTB Domain-containing Speckle-type POZ Protein ( SPOP ) Serves as an Adaptor of Daxx for Ubiquitination by Cul3-based Ubiquitin Ligase. 2006;281(18):12664-72. [ Links ]

11. Mahmud I, Liao D. DAXX in cancer: phenomena, processes, mechanisms and regulation. Nucleic Acids Res. 2019;47(15):7734-52. [ Links ]

12. Luo J, Chen B, Gao CX, Xie HK, Han CN, Zhou CC. SPOP promotes FADD degradation and inhibits NF-KB activity in non-small cell lung cancer. Biochem Biophys Res Commun [Internet]. 2018;504(1):289-94. Available from: https://doi.org/10.1016/j.bbrc.2018.08.176. [ Links ]

13. Baca SC, Prandi D, Lawrence MS, Mosquera JM, Romanel A, Drier Y, et al. Punctuated evolution of prostate cancer genomes. Cell. 2013;153(3):666-77. [ Links ]

14. Bouchard JJ, Otero JH, Scott DC, Salvatella X, Schulman BA, Mittag T, et al. Cancer Mutations of the Tumor Suppressor SPOP Disrupt the Formation of Active , Phase-Separated Article Cancer Mutations of the Tumor Suppressor SPOP Disrupt the Formation of Active , Phase-Separated Compartments. Mol Cell [Internet]. 2018;72(1):19-36.e8. Available from: https://doi.org/10.1016/j.molcel.2018.08.027. [ Links ]

15. Cheng F, Zeng C, Zeng L, Wu C, Chen Y. The association of speckle-type POZ protein with lymph node metastasis and prognosis in cancer patients: A meta-analysis. Med (United States). 2019;98(40). [ Links ]

16. Hernández-Llodrà S, Segalés L, Safont A, Juanpere N, Lorenzo M, Fumadó L, et al. SPOP and FOXA1 mutations are associated with PSA recurrence in ERG wt tumors, and SPOP downregulation with ERG-rearranged prostate cancer. Prostate. 2019;79(10):1156-65. [ Links ]

17. Zhang D, Wang H, Sun M, Yang J, Zhang W, Han S, et al. Speckle-type POZ protein, SPOP, is involved in the DNA damage response. Carcinogenesis. 2014;35(8):1691-7. [ Links ]

18. Zhu H, Ren S, Bitler BG, Aird KM, Tu Z, Skordalakes E, et al. SPOP E3 Ubiquitin Ligase Adaptor Promotes Cellular Senescence by Degrading the SENP7 deSUMOylase. Cell Rep. 2015;13(6):1183-93. [ Links ]

19. (NIH) NL of M. PubMed [Internet]. 2020. Available from: https://pubmed.ncbi.nlm.nih.gov/. [ Links ]

20. SciELO. Scientific Electronic Library Online (SciELO) [Internet]. 2020. Available from: https://scielo.org/es/. [ Links ]

21. Nagai Y, Kojima T, Muro Y, Hachiya T, Nishizawa Y, Wakabayashi T, et al. Identification of a novel nuclear speckle-type protein, SPOP. FEBS Lett [Internet]. 1997;418(1-2):23-6. Available from: http://dx.doi.org/10.1016/S0014-5793(97)01340-9. [ Links ]

22. NCBI. SPOP speckle type BTB/POZ protein ; Homo sapiens (human) 2019. [ Links ]

23. Hu Y, Yang L, Zhang M, Huang Z, Lin J, Zhang N. Expression and clinical relevance of SPOPL in medulloblastoma. Oncol Lett. 2017;14(3):3051-6. [ Links ]

24. Zhuang M, Calabrese MF, Liu J, Waddell MB, Hammel M, Miller DJ, et al. Structure s of SPOP-Substrate Complexes: Insights into Molecular Architectures of BTB-Cul3 Ubiquitin Ligases. MolCell . 2010;36(1):39-50. [ Links ]

25. Zhang P, Wang D, Zhao Y, Ren S, Gao K, Ye Z, et al. Intrinsic BET inhibitor resistance in SPOP -mutated prostate cancer is mediated by BET protein stabilization and AKT - mTORC1 activation. Aug 2017; 23(9):1055-1062. [ Links ]

26. Nicole Jung-Eun Kim, Victoria Breckwich Vásquezc, Elizabeth Torrese, R. M., Bud Nicola and CK. Multiple weak linear motifs enhance recruitment and processivity in SPOP-mediated substrate ubiquitination. Physiol Behav. 2017;176(3):139-48. [ Links ]

27. Marzahn MR, Marada S, Lee J, Nourse A, Kenrick S, Zhao H, et al. Higher-order oligomerization promotes localization of SPOP to liquid nuclear speckles . EMBO J. 2016;35(12):1254-75. [ Links ]

28. Cuneo MJ, Mittag T. The ubiquitin ligase adaptor SPOP in cancer. FEBS J. 2019;286(20):3946-58. [ Links ]

29. Bulatov E, Ciulli A. Targeting Cullin-RING E3 ubiquitin ligases for drug discovery: structure, assembly and small-molecule modulation. Biochem J [Internet]. 2015;467(3):365-86. Available from: http://biochemj.org/lookup/doi/10.1042/ BJ20141450. [ Links ]

30. Ma J, Chang K, Peng J, Shi Q, Gan H, Gao K, et al. SPOP promotes ATF2 ubiquitination and degradation to suppress prostate cancer progression. J Exp Clin Cancer Res. 2018;37(1):1-13. [ Links ]

31. Geng C, Kaochar S, Li M, Rajapakshe K, Dong J, Foley C, et al. SPOP regulates prostate epithelial cell proliferation and promotes ubiquitination and turnover of cMYC oncoprotein. 2018;36(33):4767-77. [ Links ]

32. Theurillat JP, Udeshi ND, Errington WJ, Baca SC, Pop M, Wild PJ, et al. Ubiquitylome analysis identifies dysregulation of effector substrates in SPOP-mutant prostate cancer. 2014;346(6205):85-9. [ Links ]

33. Kim B, Jin H, Eun K, Jung M, Soo I, Kim D, et al. Breast cancer metastasis suppressor 1 ( BRMS1 ) is destabilized by the Cul3 - SPOP E3 ubiquitin ligase complex. Biochem Biophys Res Commun [Internet]. 2011;415(4):720-6. Available from: http://dx.doi.org/10.1016/j.bbrc.2011.10.154. [ Links ]

34. Tan P, Xu Y, Du Y, Wu L, Guo B, Huang S, et al. SPOP suppresses pancreatic cancer progression by promoting the degradation of NANOG. Cell Death Dis . 2019;10(11). [ Links ]

35. Wozniak Ph.D. , Jeffrey R. , Riley Ph.D. , Edward P. , Michael E. Charness MD . SPOP Promotes Nanog Destruction to Suppress Stem Cell Traits and Prostate Cancer Progression. Physiol Behav . 2019;176(1):139-48. [ Links ]

36. Wang X, Jin J, Wan F, Zhao L, Chu H, Chen C, et al. AMPK Promotes SPOP-Mediated NANOG Degradation to Regulate Prostate Cancer Cell Stemness. Dev Cell [Internet]. 2019;48(3):345-360.e7. Available from: https://doi.org/10.1016/j.devcel.2018.11.033. [ Links ]

37. Zeng C, Wang Y, Lu Q, Chen J, Zhang J, Liu T, et al. SPOP suppresses tumorigenesis by regulating Hedgehog/Gli2 signaling pathway in gastric cancer. J Exp Clin Cancer Res. 2014;33(1):1-12. [ Links ]

38. Christopher E, Baca SC, Lawrence MS, Demichelis F, Blattner M, Theurillat J, et al. Exome sequencing identifies recurrent SPOP, FOXA1 and MED12 mutations in prostate cancer. 2014;44(6):685-9. [ Links ]

39. Gao K, Jin X, Tang Y, Ma J, Peng J, Yu L, et al. Tumor suppressor SPOP mediates the proteasomal degradation of progesterone receptors (PRs) in breast cancer cells. Am J Cancer Res. 2015;5(10):3210-20. [ Links ]

40. Fong K wing, Zhao JC, Song B, Zheng B, Yu J. TRIM28 protects TRIM24 from SPOP-mediated degradation and promotes prostate cancer progression. Nat Commun [Internet]. 2018;9(1). Available from: http://dx.doi.org/10.1038/s41467-018-07475-5. [ Links ]

41. Groner AC, Cato L, Tribolet-hardy J De, Bernasocchi T, Melchers D, Houtman R, et al. TRIM24 is an oncogenic transcriptional activator in prostate cancer. 2017;29(6):846-58. [ Links ]

42. An J, Ren S, Murphy SJ, Dalangood S, Chang C, Pang X, et al. Truncated ERG Oncoproteins from TMPRSS2-ERG Fusions Are Resistant to SPOP-Mediated Proteasome Degradation. Mol Cell [Internet]. 2015;59(6):904-16. Available from: http://dx.doi.org/10.1016/j.molcel.2015.07.025. [ Links ]

43. Gan W, Dai X, Lunardi A, Li Z, Inuzuka H, Liu P, et al. SPOP Promotes Ubiquitination and Degradation of the ERG Oncoprotein to Suppress Prostate Cancer Progression. Mol Cell [Internet]. 2015;59(6):917-30. Available from: http://dx.doi.org/10.1016/j.molcel.2015.07.026. [ Links ]

44. Coquenlorge S, Yin WC, Yung T, Pan J, Zhang X, Mo R, et al. GLI2 Modulated by SUFU and SPOP Induces Intestinal Stem Cell Niche Signals in Development and Tumorigenesis. Cell Rep [Internet]. 2019;27(10):3006-3018.e4. Available from: https://doi.org/10.1016/j.celrep.2019.05.016. [ Links ]

45. Jin X, Wang J, Li Q, Zhuang H, Yang J, Lin Z, et al. SPOP targets oncogenic protein ZBTB3 for destruction to suppress endometrial cancer. 2019;9(12):2797-812. [ Links ]

46. Yuan LJ, Long ZQ, Xiang XL, Tang LS, Yin L, Xiao Y, et al. SPOP suppresses prostate cancer through regulation of CYCLIN E1 stability. Cell Death Differ [Internet]. 2019;1156-68. Available from: http://dx.doi.org/10.1038/s41418-018-0198-0. [ Links ]

47. Shi Q, Zhu Y, Ma J, Chang K, Ding D, Bai Y, et al. Prostate Cancer -associated SPOP mutations enhance cancer cell survival and docetaxel resistance by upregulating Caprin1-dependent stress granule assembly. MolCancer . 2019;18(1):1-14. [ Links ]

48. Packer JR, Maitland NJ. The molecular and cellular origin of human prostate cancer. Biochim Biophys Acta - Mol Cell Res [Internet]. 2016 Jun 1 [cited 2018 Mar 1];1863(6):1238-60. Available from: Available from: https://www.sciencedirect.com/science/article/pii/S0167488916300416 . [ Links ]

49. Lund AH, Stoop P Van Der, Boutsma E, Muijrers I. Stable X chromosome inactivation involves the PRC1 Polycomb complex and requires histone MACROH2A1 and the CULLIN3 SPOP ubiquitin E3 ligase. 2005;102(21):7635-40. [ Links ]

50. Zhu K, Lei PJ, Ju LG, Wang X, Huang K, Yang B, et al. SPOP-containing complex regulates SETD2 stability and H3K36me3-coupled alternative splicing. Nucleic Acids Res . 2017;45(1):92-105. [ Links ]

51. Luo J, Bao Y chen, Ji X xiu, Chen B, Deng Q fang, Zhou S wen. SPOP promotes SIRT2 degradation and suppresses non-small cell lung cancer cell growth. Biochem Biophys Res Commun [Internet]. 2017;483(2):880-4. Available from: http://dx.doi.org/10.1016/j.bbrc.2017.01.027. [ Links ]

52. Zhang J, Bu X, Wang H, Zhu Y, Geng Y, Nihira NT, et al. Cyclin D-CDK4 kinase destabilizes PD-L1 via cullin 3-SPOP to control cancer immune surveillance. Vol. 553, Nature. 2018. 91-95 p. [ Links ]

53. Ji P, Liang S, Li P, Xie C, Li J, Zhang K, et al. Speckle-type POZ protein suppresses hepatocellular carcinoma cell migration and invasion via ubiquitin-dependent proteolysis of SUMO1/sentrin specific peptidase 7. Biochem Biophys Res Commun [Internet]. 2018;502(1):30-42. Available from: https://doi.org/10.1016/j. bbrc.2018.05.115. [ Links ]

54. Ostertag MS, Messias AC, Sattler M. The Structure of the SPOP-Pdx1 Interface Reveals Insights into the Phosphorylation-Dependent Binding Regulation. Struct Des [Internet]. 2019;27(2):327-334.e3. Available from: https://doi.org/10.1016/j.str.2018.10.005. [ Links ]

55. Claiborn KC, Sachdeva MM, Cannon CE, Groff DN, Singer JD, Stoffers DA. Pcif1 modulates Pdx1 protein stability and pancreatic 6 cell function and survival in mice. 2010;120(10):3713-21. [ Links ]

56. Li Q, Wang F, Wang Q, Zhang N, Zheng J, Zheng M, et al. SPOP promotes ubiquitination and degradation of MyD88 to suppress the innate immune response. PLOS Pathog [Internet]. 2020;16(5):e1008188. Available from: http://dx.doi.org/10.1371/journal.ppat.1008188. [ Links ]

57. Hu YH, Wang Y, Wang F, Dong YM, Jiang WL, Wang YP, et al. SPOP negatively regulates Toll-like receptor-induced inflammation by disrupting MyD88 self-association. Cell Mol Immunol [Internet]. 2020;(March) . Available from: http://dx.doi.org/10.1038/s41423-020-0411-1. [ Links ]

58. Wang L, Lin M, Chu M, Liu Y, Ma J, He Y, et al. SPOP Promotes Ubiquitination and Degradation of LATS1 to Enhance Kidney Cancer Progression. EBioMedicine [Internet]. 2020;56:102795. Available from: https://doi.org/10.1016/j.ebiom.2020.102795. [ Links ]

59. Guillamot M, Ouazia D, Dolgalev I, Yeung ST, Kourtis N, Dai Y, et al. The E3 ubiquitin ligase SPOP controls resolution of systemic inflammation by triggering MYD88 degradation. Nat Immunol [Internet]. 2019;20(9):1196-207. Available from: http://dx.doi.org/10.1038/s41590-019-0454-6. [ Links ]

60. Boysen G, Barbieri CE, Prandi D, Blattner M, Chae SS, Dahija A, et al. SPOP mutation leads to genomic instability in prostate cancer. Elife. 2015;4(September):1-4. [ Links ]

61. Dong Y, Zhang D, Cai M, Luo Z, Zhu Y, Gong L, et al. SPOP regulates the DNA damage response and lung adenocarcinoma cell response to radiation. Am J Cancer Res [Internet]. 2019;9(7):1469-83. Available from: www.ajcr.us/. [ Links ]

62. Hjorth-Jensen K, Maya-Mendoza A, Dalgaard N, Sigurõsson JO, Bartek J, Iglesias-Gato D, et al. SPOP promotes transcriptional expression of DNA repair and replication factors to prevent replication stress and genomic instability. Nucleic Acids Res . 2018;46(18):9484-95. [ Links ]

63. Gschweitl M, Ulbricht A, Barnes CA, Enchev RI, Stoffel-Studer I, Meyer-Schaller N, et al. A SPOPL/cullin-3 ubiquitin ligase complex regulates endocytic trafficking by targeting EPS15 at endosomes. Elife . 2016;5(MARCH2016):1-26. [ Links ]

64. Lan X, Khandros E, Huang P, Peslak SA, Bhardwaj SK, Grevet JD, et al. The E3 ligase adaptor molecule SPOP regulates fetal hemoglobin levels in adult erythroid cells. Blood Adv. 2019;3(10):1586-97. [ Links ]

65. Nabais Sá MJ, El Tekle G, de Brouwer APM, Sawyer SL, del Gaudio D, Parker MJ, et al. De Novo Variants in SPOP Cause Two Clinically Distinct Neurodevelopmental Disorders. Am J Hum Genet. 2020;106(3):405-11. [ Links ]

66. Gallo M Le, Hara AJO, Rudd ML, Urick ME, Nancy F, Neil NJO, et al. Exome sequencing of serous endometrial tumors identifies recurrent somatic mutations in chromatin-remodeling and ubiquitin ligase complex genes. 2013;44(12):1310-5. [ Links ]

67. Shoag J, Liu D, Ma X, Oromendia C, Christos P, Ballman K, et al. Prognostic value of the SPOP mutant genomic subclass in prostate cancer. Urol Oncol Semin Orig Investig [Internet]. 2020;000:1-5. Available from: https://doi.org/10.1016/j.urolonc.2020.02.011. [ Links ]

68. Liu D, Takhar M, Alshalalfa M, Erho N, Shoag J, Jenkins RB, et al. Impact of the SPOP Mutant Subtype on the Interpretation of Clinical Parameters in ProstateCancer . JCO Precis Oncol. 2018;(2):1-13. [ Links ]

69. Blattner M, Lee DJ, Reilly CO, Park K, Macdonald TY, Khani F, et al. SPOP Mutations in Prostate Cancer across Demographically Diverse Patient Cohorts. 2014;16(1):14-20. [ Links ]

70. Memorial Sloan Kettering Cancer Center (MSK), Princess Margaret Cancer Centre in Toronto, Children's Hospital of Philadelphia TH in the N and BU in A. cBioPortal for Cancer Genomics [Internet]. 2020. Available from: https://www.cbioportal.org/.Links ]

71. Catalogue of Somatic Mutation in Cancer COSMIC. SPOP Gene [Internet]. 2020 [cited 2018 Mar 10]. Available from: Available from: https://cancer.sanger.ac.uk/cosmic/gene/analysis?coords=b-p%3AAA&wgs=off&id=6661&ln=SPOP&start=1&end=375 . [ Links ]

72. Lythgoe C, Dynda D, Alanee S. Identification of a novel germline SPOP mutation in a family with hereditary prostate cancer. Prostate Cancer Sci Clin Pract Second Ed. 2016;74(9):141-7. [ Links ]

73. Adam Abeshouse, Jaeil Ahn, Rehan Akbani AA. The Molecular Taxonomy of Primary ProstateCancer . Cell . 2015;163:1011-25. [ Links ]

74. Yuan J, Kensler KH, Hu Z, Zhang Y, Zhang T, Jiang J, et al. Integrative comparison of the genomic and transcriptomic landscape between prostate cancer patients of predominantly African or European genetic ancestry. PLoS Genet [Internet]. 2020;16(2):1-26. Available from: http://dx.doi.org/10.1371/journal.pgen.1008641. [ Links ]

75. Khani F, Mosquera JM, Park K, Blattner M, Reilly O, Macdonald TY, et al. Evidence for Molecular Differences in Prostate Cancer between African American and Caucasian Men. Clin Cancer Res. 2015;20(March 2012):4925-34. [ Links ]

76. Riisnaes R, Crespo M, Zafeiriou Z, Sumanasuriya S, Bianchini D, Hunt J, et al. SPOP-Mutated/CHD1-Deleted Lethal Prostate Cancer and Abiraterone Sensitivity. Clin Cancer Res . 2019;24(22):5585-93. [ Links ]

77. Zhao S, Choi M, Overton JD, Bellone S, Roque DM, Cocco E, et al. Landscape of somatic single-nucleotide and copy-number mutations in uterine serous carcinoma. Proc Natl Acad Sci U S A. 2013;110(8):2916-21. [ Links ]

78. Le Gallo M, Bell DW. The emerging genomic landscape of endometrial cancer. Clin Chem. 2014;60(1):98-110. [ Links ]

79. Torres-Arzayus MI, De Mora JF, Yuan J, Vazquez F, Bronson R, Rue M, et al. High tumor incidence and activation of the PI3K/ AKT pathway in transgenic mice define AIB1 as an oncogene. Cancer Cell . 2004;6(3):263-74. [ Links ]

80. Ostertag MS, Hutwelker W, Plettenburg O, Sattler M, Popowicz GM. Structural Insights into BET Client Recognition of Endometrial and Prostate Cancer -Associated SPOP Mutants. J Mol Biol [Internet]. 2019;431(11):2213-21. Available from: https://doi.org/10.1016/j.jmb.2019.04.017. [ Links ]

81. Cramer LR y S. SPOP the mutation. Elife . 2015;4:e11760:1-4. [ Links ]

82. Shenoy TR, Boysen G, Wang MY, Xu QZ, Guo W, Koh FM, et al. CHD1 loss sensitizes prostate cancer to DNA damaging therapy by promoting error-prone double-strand break repair. Ann Oncol Off J Eur Soc Med Oncol. 2017;28(7):1495-507. [ Links ]

83. Bezawy R El, Tripari M, Percio S, Cicchetti A, Tortoreto M, Stucchi C, et al. SPOP deregulation improves the radiation response of prostate cancer models by impairing DNA damage repair. Cancer s (Basel). 2020;12(6):1-15. [ Links ]

84. Watanabe R, Maekawa M, Hieda M, Taguchi T, Miura N, Kikugawa T, et al. SPOP is essential for DNA-protein cross-link repair in prostate cancer cells: SPOP-dependent removal of topoisomerase 2A from the topoisomerase 2A-DNA cleavage complex. Mol BiolCell . 2020;31(6):478-90. [ Links ]

85. Sizemore GM, Pitarresi JR, Balakrishnan S, Ostrowski MC. The ETS family of oncogenic transcription factors in solid tumours. Nat Rev Cancer [Internet]. 2017;17(6):337-51. Available from: http://dx.doi.org/10.1038/nrc.2017.20. [ Links ]

86. Xu J, Wang F, Jiang H, Jiang Y, Chen J, Qin J. Properties and Clinical Relevance of Speckle-Type POZ Protein in Human ColorectalCancer . J Gastrointest Surg. 2015;19(8):1484-96. [ Links ]

87. Zhang S, Xiao J, Chai Y, Hong Z, Liu Z, Yuan R, et al. Speckle -Type POZ Protein Down - Regulates Matrix Metalloproteinase 2 Expression via Sp1 / PI3K / Akt Signaling Pathway in ColorectalCancer . Dig Dis Sci [Internet]. 2017;(0123456789). Available from: https://doi.org/10.1007/s10620-017-4884-4. [ Links ]

88. Zhi X, Tao J, Zhang L, Tao R, Ma L, Qin J. Silencing speckle-type POZ protein by promoter hypermethylation decreases cell apoptosis through upregulating hedgehog signaling pathway in colorectal cancer. Cell Death Dis [Internet]. 2016;7(12):1-11. Available from: http://dx.doi.org/10.1038/cddis.2016.435. [ Links ]

89. Bawa-Khalfe T, Lu LS, Zuo Y, Huang C, Dere R, Lin FM, et al. Differential expression of SUMO-specific protease 7 variants regulates epithelial-mesenchymal transition. Proc Natl Acad Sci U S A . 2012;109(43):17466-71. [ Links ]

90. Wang F, Tang C, Xu D, Tang Y, Jiang Y, Gao X, et al. LncRNA ADAMTS9-AS2 suppresses the proliferation of gastric cancer cells and the tumorigenicity of cancer stem cells through regulating SPOP. J Cell Mol Med. 2020;(August 2019):1-9. [ Links ]

91. Li G, Ci W, Karmakar S, Chen K, Dhar R, Fan Z, et al. SPOP Promotes Tumorigenesis by Acting as a Key Regulatory Hub in KidneyCancer . Cancer Cell [Internet]. 2014;25(4):455-68. Available from: http://dx.doi.org/10.1016/j.ccr.2014.02.007. [ Links ]

92. Zhao W, Zhou J, Deng Z, Gao Y, Cheng Y. SPOP promotes tumor progression via activation of catenin/ TCF4 complex in clear cell renal cell carcinoma. Int J Oncol. 2016;49(3):1001-8. [ Links ]

93. Yao S, Chen X, Chan J, Guan Y, Liu Y, Chen J, et al. Speckle-type POZ protein functions as a tumor suppressor in non-small cell lung cancer due to DNA methylation. Cancer Cell Int [Internet]. 2018;18(1):1-14. Available from: https://doi.org/10.1186/s12935-018-0711-z. [ Links ]

94. Jiao C, Meng T, Zhou C, Wang X, Wang P, Lu M, et al. TGF-6 s ignaling regulates SPOP expression and promotes prostate cancer cell stemness. 2020;12:1-14. [ Links ]

95. Ding M, Lu X, Wang C, Zhao Q, Ge J, Xia Q, et al. The E2F1-miR-520/372/373-SPOP axis modulates progression of renal carcinoma. Cancer Res. 2018;78(24):6771-84. [ Links ]

96. García-Flores M, Casanova-Salas I, Rubio-Briones J, Calatrava A, Domínguez-Escrig J, Rubio L, et al. Clinico-pathological significance of the molecular alterations of the SPOP gene in prostate cancer. Eur JCancer . 2014;50(17):2994-3002. [ Links ]

97. Acosta N, Varela R, Mesa JA, Serrano López ML, Cómbita AL, Sanabria-Salas MC. Biomarcadores de pronóstico en pacientes con cáncer de próstata localizado. Rev Colomb Cancerol. 2017;21(2):113-25. [ Links ]

98. Li JJ, Zhang JF, Yao SM, Huang H, Zhang S, Zhao M, et al. Decreased expression of speckle-type POZ protein for the prediction of poor prognosis in patients with non-small cell lung cancer. Oncol Lett . 2017;14(3):2743-8. [ Links ]

99. Xu J, Wang F, Wang X, He Z, Zhu X. MiRNA-543 promotes cell migration and invasion by targeting SPOP in gastric cancer. Onco Targets Ther. 2018;11:5075-82. [ Links ]

00. 1 Harb OA, Elfeky MA, El Shafaay BS, Taha HF, Osman G, Harera IS, et al. SPOP, ZEB-1 and E-cadherin expression in clear cell renal cell carcinoma (cc-RCC): Clinicopathological and prognostic significance. Pathophysiology [Internet]. 2018;25(4):335-45. Available from: https://doi.org/10.1016/j.pathophys.2018.05.004. [ Links ]

101. Ojha R, Mandal AK. Speckle - type POZ protein as a diagnostic biomarker in renal cell carcinoma. 2018;977-82. [ Links ]

102. Zheng T, Yang CG. Targeting SPOP with small molecules provides a novel strategy for kidney cancer therapy. Sci China Life Sci. 2017;60(1):91-3. [ Links ]

103. Guo ZQ, Zheng T, Chen B, Luo C, Ouyang S, Gong S, et al. Small-Molecule Targeting of E3 Ligase Adaptor SPOP in KidneyCancer . Cancer Cell . 2016;30(3):474-84. [ Links ]

104. Dong Z, Wang Z, Guo Z-Q, Gong S, Zhang T, Liu J, et al. Structure -activity Relationship of SPOP Inhibitors Against KidneyCancer . J Med Chem. 2020; 63(9):4849-4866. Available from: https://doi.org/10.1021/acs.jmedchem.0c00161. [ Links ]

105. Raina K, Lu J, Qian Y, Altieri M, Gordon D, Rossi AMK, et al. PROTAC-induced BET protein degradation as a therapy for castration-resistant prostate cancer. Proc Natl Acad Sci U S A . 2016;113(26):7124-9. [ Links ]

106. Salami J, Alabi S, Willard RR, Vitale NJ, Wang J, Dong H, et al. Androgen receptor degradation by the proteolysis-targeting chimera ARCC-4 outperforms enzalutamide in cellular models of prostate cancer drug resistance. Commun Biol [Internet]. 2018;1(1):1-9. Available from: http://dx.doi.org/10.1038/s42003-018-0105-8. [ Links ]

107. Han X, Wang C, Qin C, Xiang W, Fernandez-Salas E, Yang CY, et al. Discovery of ARD-69 as a Highly Potent Proteolysis Targeting Chimera (PROTAC) Degrader of Androgen Receptor (AR) for the Treatment of ProstateCancer . J Med Chem. 2019;62(2):941-64. [ Links ]

108. Neklesa TK, Winkler JD, Crews CM. Targeted protein degradation by PROTACs. Pharmacol Ther [Internet]. 2017;174:138-44. Available from: http://dx.doi.org/10.1016/j.pharmthera.2017.02.027. [ Links ]

109. Yan Y, Ma J, Wang D, Lin D, Pang X, Wang S, et al. The novel BET-CBP/p300 dual inhibitor NEO2734 is active in SPOP mutant and wild-type prostate cancer. EMBO Mol Med. 2019;11(11):1-19. [ Links ]

110. Yan Y, An J, Yang Y, Wu D, Bai Y, Cao W, et al. Dual inhibition of AKT -m TOR and AR signaling by targeting HDAC 3 in PTEN - or SPOP -mutated prostate cancer . EMBO Mol Med . 2018;10(4):1-20. [ Links ]

111. Jin X, Wang J, Gao K, Zhang P, Yao L, Tang Y, et al. Dysregulation of INF2-mediated mitochondrial fission in SPOP-mutated prostate cancer. PLoS Genet . 2017;13(4):1-24. [ Links ]

112. Li K, Wu J lin, Qin B, Fan Z, Tang Q, Lu W, et al. ILF3 is a substrate of SPOP for regulating serine biosynthesis in colorectal cancer. Cell Res [Internet]. 2019;(June) [ Links ]

Received: August 10, 2020; Accepted: September 30, 2020; Published: December 11, 2020

* Autor para correspondencia: Martha Lucía Serrano Correo electrónico:mlserranol@unal.edu.co

Creative Commons License This is an open-access article distributed under the terms of the Creative Commons Attribution License