Open Access

Essential role of bromodomain proteins in renal cell carcinoma (Review)

  • Authors:
    • Qianghai Wen
    • Haicheng Liu
    • Kecheng Lou
    • Xing Zhang
    • Wei Chao
    • Jianhui Xin
    • Jiaxiang Gong
    • Junrong Zou
    • Xiaofeng Zou
  • View Affiliations

  • Published online on: June 8, 2023     https://doi.org/10.3892/mmr.2023.13026
  • Article Number: 139
  • Copyright: © Wen et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Histone alterations are a hallmark of kidney cancer. Histone acetylation modification mediated by bromodomain proteins (BRD) has been indicated to be related to a variety of cancer types and several targeted inhibitors have been proven to be promising modalities for cancer adjuvant therapy. As renal cell carcinoma (RCC) is not sensitive to radiotherapy or chemotherapy, the exploration of effective adjuvant therapies remains an important research direction for advanced RCC. At present, studies on bromodomain family proteins in RCC are limited and the roles of bromodomain family proteins in RCC have remained to be fully elucidated. The present review discussed the role of bromodomain family proteins in RCC, aiming to explore possible potential therapeutic targets of BRD‑related drugs in this type of cancer.

Introduction

According to the Global Cancer Statistics 2020, the number of new cases of renal cell carcinoma (RCC) has increased by ~100,000 over the past decade (1,2). Among cancers of the urinary system, RCC ranks third in terms of prevalence (3). Organ metastasis of advanced RCC is the main cause of a poor prognosis. Since RCC is not sensitive to conventional radiotherapy and chemotherapy (4), the search for specific biomarkers and individualized treatment options for RCC is an important direction.

Epigenetic alterations are among the hallmarks of cancer. Epigenetics has a critical role in RCC, including DNA methylation, chromatin remodeling and histone acetylation. The main function of the bromodomain proteins (BRD) is the acetylation modification of histones (5). The first BRD was discovered in 1992 (6). Although the BRD is highly conserved structurally, BRD family proteins may be divided into eight distinct subfamilies (I–VIII) based on their secondary structure (5,7). Research has indicated that targeting these BRDs may provide a novel strategy for the treatment of metastatic RCC. The present review summarizes the roles of existing BRD families in related pathways in renal cancer and discusses the efficacy of existing BRD inhibitors and their prospects for clinical application.

Acetylation of BRD

Histone modification in transcriptional regulation

In eukaryotic cells, DNA is packaged into chromatin in three processes: The binding of DNA to core histones, the formation of nucleosome core particles and chromosome core particles by core histones (H2A, H2B, H3 and H4) and junctional histones (H1) (8). The functions of histones are divided into the regulation of DNA transcription, DNA replication and DNA repair. Of these, core histones have a critical role in transcriptional regulation mainly through their post-translational modifications (PTMs) (9,10).

Histone modification was the first confirmed PTM. It was first hypothesized by Allfrey et al (11) that there was a ‘switch’ that regulates cellular gene expression. The subsequent discovery of histone acetyltransferase (HAT), histone deacetylase (HDAC) and the mechanisms that control histone acetylation activity support the hypothesis put forth by Allfrey et al (11) on the role of histone acetylation in gene expression (1214).

However, whether lysine acetylation controls histone activity and how it controls gene activation and repression remain to be fully elucidated. Until the discovery that BRD was the first chromatin ‘reader’, it was known to exist in the nucleus as an acetyl-lysine (Kac) binding domain and to be associated with numerous transcriptional proteins (6,15). Thus, histone acetylation was established as a fundamental mechanism for regulating gene transcription. However, BRD does not have a role in histone modification alone. Such ‘large-scale engineering’ may require multiple histone combination modifications or sequential initiation, with BRD acting on one or more histone tails to activate target genes and downstream functions (16,17). BRD-mediated lysine acetylation modifications are a major epigenetic modification in RCC tumor metabolism. BRDs are involved in histone modifications, transcription factor recruitment and transcriptional regulation, and DNA damage repair in RCC as chromatin remodeling factors (5). Furthermore, BRDs have unique structural properties (7).

Structure and function of BRD

BRD consists of 110 amino acids and includes four α helices (αZ, αA, αB and αC) with two distinct interhelical αZ-αA (ZA) and αB-αC (BC) loop regions. The two loops in turn produce a hydrophobic pocket that functions as a module for the recognition of the acetyl-lysine modification (Fig. 1) (15,18). Different BRDs have BRD modules with varying lengths of the ZA and BC loop regions and may contain other types of action modules (19).

BRDs also include other domains, which combine with BRD to perform specific functions (Fig. 2). Certain BRDs contain ATPase structures that enhance the ability to bind Kac, contribute to the assembly of its associated complex, facilitate the movement of the complex along chromatin and coordinate the function of multiple protein docking in chromatin remodeling (20). For instance, ATPase family AAA domain containing 2 (ATAD2) of the ATPase family is directly involved in DNA replication by being recruited to the DNA replication site (21). In addition, ATAD2 has been identified as a chromatin modifier that promotes melanoma (22). Although ATAD2 has not been extensively studied in RCC, studies have indicated an increased risk of developing RCC in patients with melanoma and vice versa. Both may have the same type of genomic mutation (23). Coactivators containing both HAT and BRD structural domains assist substrate recruitment by enabling the HAT-mediated acetylation of multiple lysine residues on histones and transcription factors, thereby promoting transcriptional activation (24). ATPase-containing structures of BRDs enhance the ability to bind Kac, contribute to the assembly of its associated complexes, facilitate the movement of the complexes along chromatin and coordinate multiple protein-protein functions in chromatin remodeling. Unlike other BRDs, BRD and extraterminal (BET) proteins have two tightly packed BRDs that specifically bind diacetylated lysine residues (25,26), promote chromatin opening, recruit transcription factors and coactivators to target gene promoters and enhancers, and activate the RNA polymerase II (Pol II) complex to promote transcriptional elongation (27). Similarly, tandem plant homeodomain (PHD) fingers with BRDs contribute to the assembly and activity of their associated complexes in nucleosomes and act on DNA replication in chromosome segregation (28,29). BRDs are widely involved in the transcription of cancer-related genes, and depending on the subtle differences in BRDs and various combinations of functional groups, highly selective inhibitors may be designed with potential clinical applications for the treatment of metastatic cancers (30).

Figure 2.

Domain of BRD. The name of the selected protein and the number of amino acids are provided at either end of the graphic presentation. Software used: BioRender (https://app.biorender.com/). GCN5L2, general control of amino acid synthesis protein 5-like 2; PCAF-N, PCAF N-terminal domain; HAT, acetyl transferase; BD, bromodomain; PCAF, P300/CBP-associated factor; EP300, E1A binding protein p300; NRID, nuclear receptor interaction domain; KIX, kinase-inducible domain of CREB-interacting domain; RING, really interesting new gene; PHD, plant homeodomain; CH1, cysteine-histidine-rich region 1; NRID, nuclear receptor interaction domain; CREBBP, CREB Binding protein; BRD9, bromodomain-containing protein 9; DUF3512, domain of unknown function; PBRM1, polybromo 1; BAH, bromo-adjacent homology; HMG, high-mobility group; SMARCA2, SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily a, member 2; QLQ, glutamine-leucine-glutamine domain; HSA, small helicase/SANT associated domain; BRK, brahma and kismet domain; BPTF, bromodomain PHD finger transcription factor; DDT, DNA-binding homeobox and different transcription factors domain; Q-rich, glutamine-rich domain; ATAD2, ATPase family AAA domain containing 2; ATPase, ATPase domain; BRD4, bromodomain-containing protein 4; NPS, N-terminal cluster of phosphorylation sites; BID, basic residue enriched interaction domain; ET, extra-terminal domain; BRD2, bromodomain-containing protein 2; BRDT, bromodomain testis associated; TRIM24, tripartite motif containing 24; B-Box, B-box-type zinc finger domain; BBC, B-box, C-terminal domain; TRIM33, tripartite motif-containing 33.

Functions of BRDs in RCC

The development of RCC mainly includes the following biological behaviors: Angiogenesis, proliferation and immune regulation. In the pathogenesis of RCC, 3p deletion and von Hippel-Lindau (VHL) gene inactivation are the most frequently-occurring mutations (31,32). Their co-mutation has a decisive role in the initiation of RCC. Inactivation of VHL leads to the uncontrolled activation of hypoxia-inducible factor (HIF) target genes that regulate angiogenesis, glycolysis and apoptosis (4). The deletion of these tumor suppressor genes and the activation of oncogenes eventually lead to the development of RCC.

Different BRDs have different roles, which has led to the consideration of different BRDs in RCC (Table I). Angiogenesis is a typical feature of RCC and BRDs may be involved in RCC angiogenesis from different signaling pathways (33), and may even promote angiogenesis in RCC with a low HIF expression (34). Furthermore, the majority of the BRDs mainly function as oncogenic proteins, promoting the proliferation of RCC cells (3541), and are associated with influencing the occurrence of renal cancer recurrence and metastasis (42). According to the ‘braided river’ model to interpret RCC (43), the gene mutation of RCC has an evolutionary process. Certain BRDs appear in RCC of more advanced stages and are more likely to cause distant metastasis (39,44,45). Finally, the formation of tumors is also inseparable from changes in immune regulation. The immune-killing effect on the tumor is the main error correction method of the human system. The killing of RCC may be achieved by upregulating IFNα expression (46) and by the induction of CD4+ T-cells (47); polybromo 1 (PBRM1), as a protective factor, may enhance these effects. However, E1A binding protein P300 (EP300)/CREB binding protein (CBP) also leads to the depletion of peripheral lymphocytes (48), and the inhibition of peripheral inflammatory factors by BRD4 diminishes its killing effect on tumor cells (49). On the one hand, different or opposing roles between BRDs and mutations in BRDs are evolutionary in RCC. These generally increase the difficulty of RCC-specific biomarker research. On the other hand, differential mutations in RCC allow patients to select therapeutics with high specificity. In the following sections, the progress of BRDs in RCC is discussed after being classified according to their main roles.

Table I.

BRDs in RCC.

Table I.

BRDs in RCC.

ClassGene nameAliasFunctionRole in RCC(Refs.)
Histone acetyltransferaseGCN5L2KAT2A, GCN5, PCAF-BHistone acetyltransferaseCarcinogenic(99)
PCAFKAT2BHistone acetyltransferaseCancer inhibition(102)
EP300p300, KAT3B, MKHK2, RSTS2Transcriptional coactivatorCarcinogenic(35)
CREBBPCBP, KAT3A, MKHK1, RSTS1Chromatin remodeling factor, transcriptional coactivatorCarcinogenic(50)
Subunits of the SWI/SNF complexBRD9FLJ13441, LAVS3040, PRO9856, SMARCI2Transcriptional regulatorCarcinogenic(39)
PBRM1PB1, HPB1, BAF180,Chromatin remodelingCancer inhibition(90)
SMARCH1, RCCfactor (91)
SMARCA (A/B)BRM, HBRM, SNF2L2, BAF190, SNF2A, HSNF2a, NCBRSChromatin remodeling factorCancer inhibition(95)
ISWI family of chromatin remodeling factorsBPTFFAC1, FALZ, NURF301Chromatin remodeling factorCarcinogenic(44)
ATPase familyATAD2ANCCA, CT137, PRO2000, MGC5254Transcriptional coactivatorCarcinogenic(38)
BET family proteinsBRD4CAP, MCAP, HUNK1Transcriptional regulatorCarcinogenic(49)
BRD2FSH, RING3, KIAA9001, D6S113E, FSRG1, NAT, RNF3Transcriptional regulatorCarcinogenic(37)
BRDTBRD6, CT9, SPGF21Transcriptional coactivatorCarcinogenic(41)
TRIM family proteinsTRIM24TIF1α, PTC6, RNF82, TIF1ATranscriptional activatorCarcinogenic(40)
TRIM3 (A/B)TIF1-γ, PTC7, RFG7, KIAA1113, TIF1G, FLJ11429, ECTOE3 ubiquitin protein ligase, transcriptional inhibitorCancer inhibition(103)

[i] BRD, bromodomain protein; GCN5L2, general control of amino acid synthesis protein 5-like 2; PCAF, EP300/CBP-associated factor; CREBBP, CREB binding protein; PBRM1, polybromo 1; BPTF, BRD PHD finger transcription factor; ATAD2, ATPase family AAA domain containing 2; TRIM, tripartite motif-containing; EP300, E1A binding protein P300; SMARCA2, SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily A, member 2; RCC, renal cell carcinoma.

Possible functions of oncogenic BRD and their inhibitors in RCC

EP300/CBP

EP300 and CBP, two homologous lysine acetyltransferases (Fig. 2), are homologous transcriptional adapters targeted to the E1A oncoprotein and have a core role in the transcriptional regulation of hypoxia-responsive genes (50). EP300/CBP interacts with EP300/CBP-associated factor (PCAF) to function in the normal cell cycle. However, EP300/CBP is overexpressed in RCC, while PCAF is frequently demonstrated to be absent as a tumor suppressor gene (51).

EP300/CBP promotes cell growth and angiogenesis in RCC through acetylation modification. EP300/CBP functions as a co-transcription factor and forms a transcriptional complex with proteins such as HIF1α to recruit to the promoter of VEGF (52,53), and HIF-1α transcriptional activation or transcriptional repression is dependent on EP300/CBP (54). Consistent with HIF1α, HIF2α induces EP300/CBP recruitment so that it not only binds specifically to the enhancer of HIF2α to promote HIF2α expression for RCC cell growth, but also promotes RCC angiogenesis by enhancing VEGF transcription (55). In addition, HIF1α and HIF2α induce the recruitment of EP300/CBP at the promoter of telomerase reverse transcriptase, promoting the immortalization and transformation of renal cancer cells (56). Furthermore, EP300/CBP binding to H3AcK18 enhances RCC cell viability, adhesion and invasiveness (35).

EP300/CBP also promotes RCC progression by promoting macrophage infiltration through the RNA-binding motif protein 15 (RBM15)-C-X-C motif chemokine ligand 11 signaling axis (57). In addition, EP300/CBP upregulation is also associated with T-cell dysfunction. EP300/CBP reduces tumor-infiltrating lymphocytes by downregulating immune checkpoint gene expression via binding to serine and arginine-rich splicing factor 2 and cause immune escape of RCC cells (48).

EP300/CBP is structurally and functionally complex; however, it currently only has two related inhibitors in RCC. Inhibitors targeting the BRD binding of EP300/CBP include C646, which effectively inhibits EP300/CBP (57). HBS1, a high-affinity ligand of cysteine-histidine-rich region 1, functions as an antibody to HIF1α, but is able to block the binding of HIF1α to the CH300 structural domain of EP300/CBP without affecting normal cell function (58).

The BET family: BRD4, BRD2 and BRD testis associated (BRDT)

BRD4

BRD4 is highly expressed in RCC (36). BRD4 is a transcriptional and epigenetic regulator that has a crucial role in transcription (59,60). BRD4 has multiple substrate binding sites and a common kinase structural domain that phosphorylates Pol II, the proto-oncogene C-MYC and the transcription factors TATA-box binding protein associated factor 7 (a component of the TFIID complex, controls the first steps of transcription) and cyclin-dependent kinase (CDK)9. BRD4 is also a scaffolding protein that interacts with chromatin modifiers and transcription factors, including recruiting transcription factors and transcription elongation factor b (61).

Normally, BRD4 is inhibited by caspase-3, leading to the pyroptosis of RCC cells. When BRD4 is activated, it inhibits the production of the peripheral inflammatory factors, IL-1β and IL-18, by cells. This allows immune evasion of RCC from IL-1β and IL-18 (49), promotes RCC cell proliferation and epithelial-mesenchymal transition (EMT) and inhibits tumor killing by peripheral T-cells (62). Another study demonstrated that BRD4 acetylation modified the histones of B-cell lymphoma-2 (BCL2) and C-MYC to increase their expression (36). MYC is a heterogeneous gene fragment frequently activated in cancer, and targeting BRD4 may inhibit its expression (63). This suggests that BRD4 upregulation is closely related to RCC; the inhibition of BRD4 gene expression and the enhancement of peripheral inflammatory factors may become a novel treatment strategy for metastatic RCC.

BRD4-related drugs have been tested in vivo and in vitro. JQ1, a BET inhibitor, has been demonstrated to inhibit RCC (49). JQ1 not only acts on ordinary RCC (36), but also exerts a notable inhibitory effect on sunitinib-resistant RCC (64). There are other BET inhibitors, such as physachenolide C, which enhances T-cell-mediated tumor cell killing through the targeted inhibition of BRD4 (62). High selectivity is a prominent indicator of the inhibitory effect: BDF-1253 leads to a ~4-fold greater inhibition of BRD4 compared to the prototype, nitroxibenzylidine, and exhibits suitable selectivity for BET proteins over other BRDs or epigenetic modifiers (37). The effective blockade of BRD4 downstream gene expression by inhibiting BRD4 induces cell cycle arrest and apoptosis, impairs RCC cell viability and reduces RCC cell growth (36).

Pharmacological studies on kidney cancer cells have revealed that BRD4 and the PI3K/mTOR pathway complement each other to promote the proliferation of kidney cancer cells. VS-5584, a dual inhibitor of PI3K/mTOR, may effectively inhibit the proliferation and survival of RCC cells (65), but leads to increased BRD4 expression, resulting in enhanced tumor drug resistance. By contrast, the simultaneous inhibition of BRD4 and PI3K/mTOR significantly inhibits tumor cell survival and does not increase tumor drug resistance. Furthermore, the dual inhibitor of mTORC1/2 (Palomid 529) has been found to be more effective in BRD4-negative RCC cells than normal RCC cells (66), demonstrating that BRD4 and the PI3K/mTOR pathway have a complementary effect on each other. By contrast, SF2523 is a dual inhibitor of BRD4 and PI3K-AKT; SF2523 is more effective than the PI3K inhibitor and JQ1 in killing RCC cells (67). Furthermore, the BET inhibitor OTX015 exerts a therapeutic effect on cancer patients with deletion of BRCA1-associated protein 1 (BAP1), a ubiquitin carboxy-terminal hydrolase (68); patients with a BAP1 deletion in RCC tend to have poor prognosis (69).

The aforementioned inhibitors exert potent effects by inhibiting BRD4 or when BRD4 is inhibited (Fig. 3). The inhibition of BET family members has great therapeutic potential in the treatment of RCC. Currently, clinical resistance to BET inhibitors limits their application; however, synergistic antitumor effects have been observed when used in combination with other tumor suppressors (70). Therefore, the design of BET BRDs dual-target inhibitors and their combination is a reasonable strategy, which may be used to enhance the efficacy of cancer therapy and reduce drug resistance. However, extensive animal studies are still required to verify the efficacy and toxicology prior to clinical application.

Figure 3.

BRD regulate important carcinogenic factors through histone modification. The EP300/CBP complex promotes the expression of RBM15 and downstream CXCL11 and SRSF2 expression. Furthermore, EP300/CBP recruits STAT3 and promotes MYC expression. EP300/CBP binds to HIF1α and HIF2α and promotes angiogenesis. In PBRM1-inactivated RCC cells, the increase in HIF1α/HIF2α expression promotes tumor tissue angiogenesis. The inactivation of PBRM1 also results in uninhibited STAT3 and PI3K pathways, promoting RCC development. In RCC, RBPJ promotes the production of DAPK3, which competes with PKA for the binding site of UBE3A, ultimately degrading PBRM1 and promoting RCC proliferation. BRD4/BRD2 promotes PI3K/mTOR pathways and the expression of BCL2 and MYC. It also inhibits T-cell infiltration in tumor tissues by inhibiting NLPR3. Software used: BioRender (https://app.biorender.com/). EP300, E1A binding protein P300; CBP, CREB binding protein; RBM15, RNA-binding motif protein 15; CXCL11, C-X-C motif chemokine ligand 11; SRSF2, serine and arginine-rich splicing factor 2; STAT3, signal transducer and activator of transcription 3; HIF, hypoxia-inducible factor; BRD, bromodomain protein; RCC, renal cell carcinoma; RBPJ, recombination signal binding protein for immunoglobulin kappa J region; DAPK3, death associated protein kinase 3; PKA, protein kinase A; UBE3A, ubiquitin protein ligase E3A; p21, one of the downstream target genes of p53; NLRP3, NOD-like receptor thermal protein domain associated protein 3.

BRD2

BRD2 may be a surrogate for BRD4 (37). JQ1, a BET inhibitor, is also partially selective for BRD2 and BRD3 (64). Researchers have found overexpression of both BRD2 and BRD4 BET family proteins in RCC. It has also been indicated that knockdown of BRD2 or BRD4 only moderately inhibits RCC cell proliferation. However, co-knockdown of BRD2 and BRD4 results in significant inhibition of BCL2 and C-MYC oncoprotein expression, thereby reducing the proliferation of RCC cells. This indicates the compensatory effect between members of the BET family (37).

BRDT

BRDT functions as a chromatin remodeling factor in recognizing acetylated histones and recruiting transcriptional complexes (71). It interacts with eukaryotic translation initiation factor 4E-binding protein 1 (eIF4EBP1) to promote C-MYC transcription and RCC progression. BRDT inhibitor PLX51107 exerts an inhibitory effect on RCC cells; however, eIF4EBP1 overexpression hinders its inhibitory effect (72). Therefore, dual inhibitors of both proteins or a combination of both inhibitors may be required for clinical application in RCC. The initiation of BET family proteins in RCC does not appear to proceed via a sole mechanism, i.e. not only in terms of the binding of BETs to other proteins, but also among BETs (64), which complement each other while interacting.

Other oncogenic BRDs
BRD PHD finger transcription factor (BPTF)

BPTF contains two PHD fingers and a BRD that regulates gene transcription by interacting with the MYC-associated zinc finger protein, ZF87/MAZ (73). BPTF is a subunit of the ISWI chromatin remodeling complex, NURF, and has a crucial role in chromatin remodeling as a transcription factor (74). The increased expression of BPTF causes glycolytic reprogramming and distant lung metastasis in RCC. Its inhibitor, AU1, is effective in inhibiting the metastatic potential of patient-derived cells, metastatic RCC-derived organoids, as well as in situ xenograft models of metastatic RCC, suggesting that BPTF may be applied as an initial therapeutic marker for metastatic RCC (44).

ATPase family AAA domain containing 2 (ATAD2)

There are two isoforms of human ATAD2, ATAD2A and ATAD2B (75). However, the majority of published functional studies have been performed on ATAD2A (21,75,76). ATAD2 is an emerging oncoprotein, a potential biomarker and a potent cancer drug target. In RCC, ATAD2 has been found to be a novel gene that is associated with prognosis, and its high expression increases the risk of RCC progression (77). By knocking down and overexpressing ATAD2, it has been found that its overexpression promotes RCC progression (38).

BRD9

BRD9 is highly expressed in HIF2α-deficient RCC. SOX17 recruits BRD9 to upregulate genes in RCC pathogenesis, including VEGFR2 (34). There is a negative correlation between the expression of BRD9 and HIF2α, and the expression of related genes is upregulated by BRD9 to target the Notch1-Hes1 signaling pathway to promote the proliferation, migration and invasion of RCC cells (39). A previous study demonstrated that BRD9 inhibitor (I-BRD9, also known as GSK602) effectively inhibited tumor growth in vitro and in vivo. It also prolonged the survival of RCC mice compared to sunitinib (34). Due to HIF activation in RCC caused by VHL inactivation, targeting HIF2α and applying anti-angiogenic targeted drugs are considered mainstream therapeutic approaches for metastatic RCC (78). A lower expression of HIF2α in RCC was indicated to be associated with a lower survival rate and prognosis. Hence, the search for novel drug targets in HIF2α-deficient RCC may be promising. BRD9 is expected to be a valuable target for the treatment of HIF2α-deficient RCC.

Tumor suppressor BRD in RCC

PBRM1 regulates multiple biological processes of RCC

PBRM1 is frequently mutated in human tumors and PRBM1 comprises 1,689 amino acids. PBRM1 is characterized by a C-terminal high migration pattern, including two bromine-associated homologous structural domains and six bromine structural domains (79). These bromine structural domains combine with acetylated residues at the histone tail (80), and each bromine domain has a different affinity for the specific acetylated peptides on the histones and may coordinate with the exact pattern of acetylated lysine residues in the nucleosome (81,82). Previous studies suggested that RCC is characterized by partial loss of chromosome 3p, while PBRM1 resides on 3p and has deletion mutations at a relatively high frequency, just second only to VHL in RCC, accounting for ~30–40% of RCC cases (79,80,83). As PBRM1 is a tumor suppressor, the deletion mutation of PBRM1 combined with VHL deletion produces stable tumor models (84,85).

RCC develops its mutations differently and at different time-points. In certain RCC cell lines, PBRM1 may remain present following the deletion of VHL. The deletion mutation or suppressed state of PBRM1 may be caused by other factors. For instance, recombination signal binding protein for immunoglobulin kappa J region (RBPJ) promotes death-associated protein kinase 3 binding to ubiquitin-protein ligase E3A (UBE3A), destabilizing PBRM1 in RCC cells and thereby reducing the protective effect of PBRM1 on normal renal tissue. UBE3A knockdown increased the sensitivity of CDK inhibitors, and RBPJ inhibitors modulate CDK4/6 inhibitor drug sensitivity to improve tumor suppression (86). By inhibiting PBRM1 upstream as described above, the presence of PBRM1 has been found to exert protective effects on normal kidney tissue, suggesting that it inhibits the action of common oncogenic factors in RRC. However, following the knockdown of PBRM1, it has been found that PBRM1 deletion did not allow for the inhibition of the HIF1/STAT3 signaling pathway in the kidney, and PBRM1 deletion attenuated the activity of the negative regulator of mTORC1, TSC1, facilitating mTORC1 activation and leading to advanced RCC (84,87). In addition, deletion of PBRM1 enhanced the hypoxic response, leading to increased induction of HIF1α and HIF2α, which promoted angiogenesis and cell proliferation in kidney cancer (88,89). The simultaneous knockdown of both PBRM1 and VHL genes in RCC cell lines also resulted in the activation of HIF and in the upregulation of PI3K signaling, promoting glucose uptake and adhesion in RCC cells (Fig. 3) (90,91).

PBRM1 deficiency is not only involved in angiogenesis following the induction of hypoxia, but also allows tumor cells to acquire the related functions of immune evasion. PBRM1 recruits lysine demethylase 5C to upregulate IFNα gene expression and acts on IFN-stimulated gene factor 3 to inhibit RCC progression (46). Furthermore, PBRM1 has been found to bind directly to the RRM1 and RRM2 sites and promote the infiltration of CD4 T-cells in the peripheral tissues of RCC (47). As with anti-vascular targeting agents, anti-programmed cell death 1 agents are associated with improved clinical prognosis in patients with PBRM1-deficient mutated RCC (92). This suggests that PBRM1-deficient RCC is sensitive to targeted drugs and immune agents.

Other tumor suppressor BRDs in RCC
SMARCA2

SWI/SNF-related, matrix-associated, actin-dependent regulator of chromatin subfamily A member 2 (SMARCA2)S mutations are highly associated with VHL inactivation, an elevated tumor grade and a relatively poorer prognosis (93,94). RCC cells in the G2/M phase are promoted to undergo apoptosis and cell cycle arrest by SMARCA2 mutations, not only by SMARCA2 deletion but also by epigenetic silencing of SMARCA2. Therefore, the inhibition of SMARCA2 transcription has a carcinogenic role (95), and the restoration of SMARCA2 expression by a HDAC3 inhibitor (RGFP966) effectively inhibited tumor progression in RCC (96). The restoration of the expression of tumor suppressor genes may thus be an effective strategy for the treatment of RCC.

Different functions of homologous BRD

General control of amino acid synthesis protein 5-like 2 (GCN5L2) and PCAF

GCN5L2 was the first histone acetyltransferase identified with an N-terminal structural domain, a conserved HAT structural domain and a C-terminal bromine structural domain (97). The aforementioned three structural domains are also found in PCAF, with only one molecular weight difference between the two (Fig. 2). The two proteins have approximately the same function, although PCAF can be methylated (98). Furthermore, both have opposite effects on RCC outcomes. The overexpression of GCN5L2 upregulates monocarboxylate transporter 1 (MCT1), thus promoting glycolysis in RCC cells. The proliferation of RCC cells with a high GCN5L2 expression may be significantly inhibited by an MCT1 inhibitor (AZD3965) (99).

PCAF is known as GCN5-related N-acetyltransferase in the lysine acetyltransferase family (100). As previously mentioned, PCAF (the EP300/CBP-associated factor) is a tumor suppressor gene in RCC (101). NADPH oxidase (NOX)4 functions as a mitochondrial energy sensor and the derived reactive oxygen species inhibit the acetylation of PCAF, promote metabolic reprogramming and enhance drug resistance in RCC cells (102). Promoting PACF expression or inhibiting NOX to reduce drug resistance in RCC may be a novel therapeutic approach.

Tripartite motif-containing (TRIM)24 and TRIM33

The TRIM family is a class of proteins with E3 ubiquitin ligase activity. Its members include a number of key biological processes, including autophagy, carcinogenic, intracellular signaling, protein ubiquitination and innate immunity (103). TRIM24 and TRIM33 form chromatin remodeling complexes with heterochromatin protein 1 and HDAC, with chromatin remodeling, mainly of different molecular weights (Fig. 2) (104). However, the two have distinct roles in the progression of RCC. In RCC, the transcription of TRIM24 is regulated by bone morphogenetic protein (BMP)8A, and the upregulation of TRIM24 by BMP8A enhances the Wnt-regulated Wnt signaling pathway, thus promoting proliferation, invasion, and metastasis and drug resistance (40,45).

In contrast to TRIM24, TRIM33 functions as a tumor suppressor in RCC and its overexpression inhibits β-linked proteins on the Wnt signaling pathway, which reduces the expression of cyclin D1 and C-MYC. Consequently, TRIM33 inhibits the growth of RCC and leads to the upregulation of E-calmodulin expression and the downregulation of N-calmodulin to reduce the EMT potential of RCC cells (103). In addition, the overexpression of TRIM33 significantly inhibits TGFβ-induced Smad activation, inhibiting RCC progression (105).

Clinical value of BRD

Large-scale studies have identified mutations in BRDs that cause dysfunction and lead to cancer development. BRDs are intimately involved in the regulation of gene transcription; therefore, the inhibition of BRD structural proteins is considered a strategy for targeting oncogenic transcription factors that have long been considered attractive drug targets, but cannot be directly regulated with small molecule inhibitors. This can also be tailored to the nature of the tumor, with different transcriptional repression. For instance, BET inhibition has been shown to block the association of MYC with BRD4 and MYC transcription (61,63). In anti-angiogenesis, the transcriptional expression of HIF, VEGF and VEGFA can be reduced by targeted inhibition of BRDs (33,34). In immunotherapy, immune checkpoints can be suppressed by targeting and inhibiting BRDs or activating BRDs to restore the transcriptional activity of antitumor factors (4648). As presented in Table II, a large number of related drugs (Fig. S1) have been applied to basic studies on RCC and these have exhibited improved tumor-suppressive effects by directly inhibiting BRD or its downstream functional molecules. Targeting BRD structural proteins holds promise in clinical practice; however, to date, at least to the best of our knowledge, no clinical trials have been performed on RCC.

Table II.

Inhibitors.

Table II.

Inhibitors.

InhibitorTarget proteinType of experiment(Refs.)
CPTH2EP300/CBPCell experiments(35)
C646EP300/CBPCell and animal experiments(57)
HBS1EP300/CBPCell and animal experiments(58)
SF2523BRD4, P13KCell and animal experiments(67)
JQ1BETCell and animal experiments(36)
BDF1253BETCell and animal experiments(37)
PCCBETCell experiments(62)
OTX015BRD2/3/4Cell and animal experiments(68)
AU1BPTFCell and animal experiments(44)
AZD3965MCT1 (downstream signaling of GCN5L2 is blocked)Cell and animal experiments(99)
RGFP966HDAC3 (expression of SMARCA2 returned to normal)Cell and animal experiments(96)
PLX51107BRDTCell and animal experiments(72)

[i] CPTH2, a histone acetyltransferase inhibitor; C646, a selective small molecule inhibitor of EP300/CBP; AU1, GSK1379725A, a BRD PHD finger transcription factor antagonist; HBS1, a high-affinity ligand of cysteine-histidine-rich region 1, an EP300/CBP inhibitor; RGFP966, an inhibitor of histone deacetylase 3; SF2523, a PI3K/BRD4 inhibitor; JQ1, a BET inhibitor; BDF1253, a BET inhibitor; PLX51107, a BET inhibitor; PPC, a BET inhibitor; OTX015, a BET inhibitor; AZD3965, an inhibitor of monocarboxylate transporter 1. CPTH2, cyclopentylidene-[4-(4-chlorophenyl)thiazol-2-yl]hydrazone; PPC, physachenolide C; C646, ChemBridge#5838646; EP300, E1A binding protein P300; CBP, CREB binding protein; BET, BRD and extraterminal; BRD, bromodomain protein.

Conclusions and future perspectives

RCC is a common disease of the urinary system; however, its development process is highly complex, while research on BRDs has provided certain insight. BRDs are fully involved in the proliferation, angiogenesis and immune regulation of RCC, and supplement the signaling pathways of these cellular behaviors. Inhibiting the expression of these oncogenes or restoring the expression of those tumor suppressor genes may exert a notable inhibitory effect on RCC and may provide a potential solution for RCC resistance to VEGF-targeted drugs. Of note, the aforementioned studies on BRD molecules have revealed information regarding the promotion or inhibition of RCC; however, the specific underlying mechanisms remain to be fully elucidated and further in-depth research is required, as clinical trials on related inhibitors are lacking.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (grant no. 81860456).

Availability of data and materials

Data sharing is not applicable to this article, as no new data were generated or analyzed during the current study.

Authors' contributions

QW and HL wrote the manuscript. KL, WC and XZ acquired the data. JX and JG interpreted the data. JZ and XZ made substantial contributions to conception and design. All authors contributed to the article and approved the submitted version. All authors have read and agreed to the published version of the 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 

Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J and Jemal A: Global cancer statistics, 2012. CA Cancer J Clin. 65:87–108. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021. View Article : Google Scholar : PubMed/NCBI

3 

Siegel RL, Miller KD, Fuchs HE and Jemal A: Cancer statistics, 2021. CA Cancer J Clin. 71:7–33. 2021. View Article : Google Scholar : PubMed/NCBI

4 

Hsieh JJ, Purdue MP, Signoretti S, Swanton C, Albiges L, Schmidinger M, Heng DY, Larkin J and Ficarra V: Renal cell carcinoma. Nat Rev Dis Primers. 3:170092017. View Article : Google Scholar : PubMed/NCBI

5 

Fujisawa T and Filippakopoulos P: Functions of bromodomain-containing proteins and their roles in homeostasis and cancer. Nat Rev Mol Cell Biol. 18:246–262. 2017. View Article : Google Scholar : PubMed/NCBI

6 

Haynes SR, Dollard C, Winston F, Beck S, Trowsdale J and Dawid IB: The bromodomain: A conserved sequence found in human, Drosophila and yeast proteins. Nucleic Acids Res. 20:26031992. View Article : Google Scholar : PubMed/NCBI

7 

Boyson SP, Gao C, Quinn K, Boyd J, Paculova H, Frietze S and Glass KC: Functional roles of bromodomain proteins in cancer. Cancers (Basel). 13:36062021. View Article : Google Scholar : PubMed/NCBI

8 

Fyodorov DV, Zhou BR, Skoultchi AI and Bai Y: Emerging roles of linker histones in regulating chromatin structure and function. Nat Rev Mol Cell Biol. 19:192–206. 2018. View Article : Google Scholar : PubMed/NCBI

9 

Allis CD and Jenuwein T: The molecular hallmarks of epigenetic control. Nat Rev Genet. 17:487–500. 2016. View Article : Google Scholar : PubMed/NCBI

10 

Izzo A and Schneider R: The role of linker histone H1 modifications in the regulation of gene expression and chromatin dynamics. Biochim Biophys Acta. 1859:486–495. 2016. View Article : Google Scholar : PubMed/NCBI

11 

Allfrey VG, Faulkner R and Mirsky AE: Acetylation and methylation of histones and their possible role in the regulation of rna synthesis. Proc Natl Acad Sci USA. 51:786–794. 1964. View Article : Google Scholar : PubMed/NCBI

12 

Brownell JE, Zhou J, Ranalli T, Kobayashi R, Edmondson DG, Roth SY and Allis CD: Tetrahymena histone acetyltransferase A: A homolog to yeast Gcn5p linking histone acetylation to gene activation. Cell. 84:843–851. 1996. View Article : Google Scholar : PubMed/NCBI

13 

Kuo MH, Brownell JE, Sobel RE, Ranalli TA, Cook RG, Edmondson DG, Roth SY and Allis CD: Transcription-linked acetylation by Gcn5p of histones H3 and H4 at specific lysines. Nature. 383:269–272. 1996. View Article : Google Scholar : PubMed/NCBI

14 

Taunton J, Hassig CA and Schreiber SL: A mammalian histone deacetylase related to the yeast transcriptional regulator Rpd3p. Science. 272:408–411. 1996. View Article : Google Scholar : PubMed/NCBI

15 

Dhalluin C, Carlson JE, Zeng L, He C, Aggarwal AK and Zhou MM: Structure and ligand of a histone acetyltransferase bromodomain. Nature. 399:491–496. 1999. View Article : Google Scholar : PubMed/NCBI

16 

Strahl BD and Allis CD: The language of covalent histone modifications. Nature. 403:41–45. 2000. View Article : Google Scholar : PubMed/NCBI

17 

Tan M, Luo H, Lee S, Jin F, Yang JS, Montellier E, Buchou T, Cheng Z, Rousseaux S, Rajagopal N, et al: Identification of 67 histone marks and histone lysine crotonylation as a new type of histone modification. Cell. 146:1016–1028. 2011. View Article : Google Scholar : PubMed/NCBI

18 

Jeanmougin F, Wurtz JM, Le Douarin B, Chambon P and Losson R: The bromodomain revisited. Trends Biochem Sci. 22:151–153. 1997. View Article : Google Scholar : PubMed/NCBI

19 

Romero FA, Taylor AM, Crawford TD, Tsui V, Côté A and Magnuson S: Disrupting acetyl-lysine recognition: Progress in the development of bromodomain inhibitors. J Med Chem. 59:1271–1298. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Mashtalir N, D'Avino AR, Michel BC, Luo J, Pan J, Otto JE, Zullow HJ, McKenzie ZM, Kubiak RL, St Pierre R, et al: Modular organization and assembly of SWI/SNF family chromatin remodeling complexes. Cell. 175:1272–1288.e20. 2018. View Article : Google Scholar : PubMed/NCBI

21 

Koo SJ, Fernández-Montalván AE, Badock V, Holton SJ, von Ahsen O, Toedling J, Vittori S, Bradner JE and Gorjánácz M: ATAD2 is an epigenetic reader of newly synthesized histone marks during DNA replication. Oncotarget. 7:70323–70335. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Baggiolini A, Callahan SJ, Montal E, Weiss JM, Trieu T, Tagore MM, Tischfield SE, Walsh RM, Suresh S, Fan Y, et al: Developmental chromatin programs determine oncogenic competence in melanoma. Science. 373:eabc10482021. View Article : Google Scholar : PubMed/NCBI

23 

Maubec E, Chaudru V, Mohamdi H, Grange F, Patard JJ, Dalle S, Crickx B, Paillerets BB, Demenais F and Avril MF: Characteristics of the coexistence of melanoma and renal cell carcinoma. Cancer. 116:5716–5724. 2010. View Article : Google Scholar : PubMed/NCBI

24 

Dancy BM and Cole PA: Protein lysine acetylation by p300/CBP. Chem Rev. 115:2419–2452. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Morinière J, Rousseaux S, Steuerwald U, Soler-López M, Curtet S, Vitte AL, Govin J, Gaucher J, Sadoul K, Hart DJ, et al: Cooperative binding of two acetylation marks on a histone tail by a single bromodomain. Nature. 461:664–668. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Shi J, Wang Y, Zeng L, Wu Y, Deng J, Zhang Q, Lin Y, Li J, Kang T, Tao M, et al: Disrupting the interaction of BRD4 with diacetylated Twist suppresses tumorigenesis in basal-like breast cancer. Cancer Cell. 25:210–225. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Hnisz D, Abraham BJ, Lee TI, Lau A, Saint-André V, Sigova AA, Hoke HA and Young RA: Super-enhancers in the control of cell identity and disease. Cell. 155:934–947. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Thompson PJ, Norton KA, Niri FH, Dawe CE and McDermid HE: CECR2 is involved in spermatogenesis and forms a complex with SNF2H in the testis. J Mol Biol. 415:793–806. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Xiao A, Li H, Shechter D, Ahn SH, Fabrizio LA, Erdjument-Bromage H, Ishibe-Murakami S, Wang B, Tempst P, Hofmann K, et al: WSTF regulates the H2A.X DNA damage response via a novel tyrosine kinase activity. Nature. 457:57–62. 2009. View Article : Google Scholar : PubMed/NCBI

30 

Filippakopoulos P, Qi J, Picaud S, Shen Y, Smith WB, Fedorov O, Morse EM, Keates T, Hickman TT, Felletar I, et al: Selective inhibition of BET bromodomains. Nature. 468:1067–1073. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Szücs S, Müller-Brechlin R, DeRiese W and Kovacs G: Deletion 3p: The only chromosome loss in a primary renal cell carcinoma. Cancer Genet Cytogenet. 26:369–373. 1987. View Article : Google Scholar : PubMed/NCBI

32 

Latif F, Tory K, Gnarra J, Yao M, Duh FM, Orcutt ML, Stackhouse T, Kuzmin I, Modi W, Geil L, et al: Identification of the von Hippel-Lindau disease tumor suppressor gene. Science. 260:1317–1320. 1993. View Article : Google Scholar : PubMed/NCBI

33 

Grabmaier K, A de Weijert MC, Verhaegh GW, Schalken JA and Oosterwijk E: Strict regulation of CAIX(G250/MN) by HIF-1alpha in clear cell renal cell carcinoma. Oncogene. 23:5624–5631. 2004. View Article : Google Scholar : PubMed/NCBI

34 

Zhang C, Chen L, Lou W, Su J, Huang J, Liu A, Xu Y, He H, Gao Y, Xu D and Li Q: Aberrant activation of m6A demethylase FTO renders HIF2αlow/− clear cell renal cell carcinoma sensitive to BRD9 inhibitors. Sci Transl Med. 13:eabf60452021. View Article : Google Scholar : PubMed/NCBI

35 

Cocco E, Leo M, Canzonetta C, Di Vito S, Mai A, Rotili D, Di Napoli A, Vecchione A, De Nunzio C, Filetici P and Stoppacciaro A: KAT3B-p300 and H3AcK18/H3AcK14 levels are prognostic markers for kidney ccRCC tumor aggressiveness and target of KAT inhibitor CPTH2. Clin Epigenetics. 10:442018. View Article : Google Scholar : PubMed/NCBI

36 

Wu X, Liu D, Gao X, Xie F, Tao D, Xiao X, Wang L, Jiang G and Zeng F: Inhibition of BRD4 suppresses cell proliferation and induces apoptosis in renal cell carcinoma. Cell Physiol Biochem. 41:1947–1956. 2017. View Article : Google Scholar : PubMed/NCBI

37 

Chen W, Zhang H, Chen Z, Jiang H, Liao L, Fan S, Xing J, Xie Y, Chen S, Ding H, et al: Development and evaluation of a novel series of Nitroxoline-derived BET inhibitors with antitumor activity in renal cell carcinoma. Oncogenesis. 7:832018. View Article : Google Scholar : PubMed/NCBI

38 

Ji S, Su X, Zhang H, Han Z, Zhao Y and Liu Q: MicroRNA-372 functions as a tumor suppressor in cell invasion, migration and epithelial-mesenchymal transition by targeting ATAD2 in renal cell carcinoma. Oncol Lett. 17:2400–2408. 2019.PubMed/NCBI

39 

Lou W, Gao K, Xu C and Li Q: Bromodomain-containing protein 9 is a prognostic biomarker associated with immune infiltrates and promotes tumor malignancy through activating notch signaling pathway in negative HIF-2α clear cell renal cell carcinoma. IUBMB Life. 73:1334–1347. 2021. View Article : Google Scholar : PubMed/NCBI

40 

Jiang T, Mao H, Chen Q, Cao L, He Y, Gao X, Chen W and Zhang H: Trim24 prompts tumor progression via inducing EMT in renal cell carcinoma. Open Med (Wars). 15:1153–1162. 2020. View Article : Google Scholar : PubMed/NCBI

41 

Jin W, Miao Q, Wang M, Zhang Y, Ma Y, Huang Y, Wu H, Lin Y, Hu B and Pan J: A rare case of adrenal gland abscess due to anaerobes detected by metagenomic next-generation sequencing. Ann Transl Med. 8:2472020. View Article : Google Scholar : PubMed/NCBI

42 

Aurilio G, Santoni M, Massari F, Cimadamore A, Rizzo A, Mollica V, Verri E, Battelli N and Montironi R: Metabolomic profiling in renal cell carcinoma patients: News and views. Cancers (Basel). 13:52292021. View Article : Google Scholar : PubMed/NCBI

43 

Hsieh JJ and Cheng EH: A braided cancer river connects tumor heterogeneity and precision medicine. Clin Transl Med. 5:422016. View Article : Google Scholar : PubMed/NCBI

44 

Zhang C, Chen L, Liu Y, Huang J, Liu A, Xu Y, Shen Y, He H and Xu D: Downregulated METTL14 accumulates BPTF that reinforces super-enhancers and distal lung metastasis via glycolytic reprogramming in renal cell carcinoma. Theranostics. 11:3676–3693. 2021. View Article : Google Scholar : PubMed/NCBI

45 

Yu YP, Cai LC, Wang XY, Cheng SY, Zhang DM, Jian WG, Wang TD, Yang JK, Yang KB and Zhang C: BMP8A promotes survival and drug resistance via Nrf2/TRIM24 signaling pathway in clear cell renal cell carcinoma. Cancer Sci. 111:1555–1566. 2020. View Article : Google Scholar : PubMed/NCBI

46 

Liao L, Liu ZZ, Langbein L, Cai W, Cho EA, Na J, Niu X, Jiang W, Zhong Z, Cai WL, et al: Multiple tumor suppressors regulate a HIF-dependent negative feedback loop via ISGF3 in human clear cell renal cancer. Elife. 7:e379252018. View Article : Google Scholar : PubMed/NCBI

47 

Szarkowska J, Cwiek P, Szymanski M, Rusetska N, Jancewicz I, Stachowiak M, Swiatek M, Luba M, Konopinski R, Kubala S, et al: RRM2 gene expression depends on BAF180 subunit of SWISNF chromatin remodeling complex and correlates with abundance of tumor infiltrating lymphocytes in ccRCC. Am J Cancer Res. 11:5965–5978. 2021.PubMed/NCBI

48 

Wang Z, Li K, Chen W, Wang X, Huang Y, Wang W, Wu W, Cai Z and Huang W: Modulation of SRSF2 expression reverses the exhaustion of TILs via the epigenetic regulation of immune checkpoint molecules. Cell Mol Life Sci. 77:3441–3452. 2020. View Article : Google Scholar : PubMed/NCBI

49 

Tan YF, Wang M, Chen ZY, Wang L and Liu XH: Inhibition of BRD4 prevents proliferation and epithelial-mesenchymal transition in renal cell carcinoma via NLRP3 inflammasome-induced pyroptosis. Cell Death Dis. 11:2392020. View Article : Google Scholar : PubMed/NCBI

50 

Arany Z, Huang LE, Eckner R, Bhattacharya S, Jiang C, Goldberg MA, Bunn HF and Livingston DM: An essential role for p300/CBP in the cellular response to hypoxia. Proc Natl Acad Sci USA. 93:12969–12973. 1996. View Article : Google Scholar : PubMed/NCBI

51 

Yang XJ, Ogryzko VV, Nishikawa J, Howard BH and Nakatani Y: A p300/CBP-associated factor that competes with the adenoviral oncoprotein E1A. Nature. 382:319–324. 1996. View Article : Google Scholar : PubMed/NCBI

52 

Pawlus MR, Wang L and Hu CJ: STAT3 and HIF1α cooperatively activate HIF1 target genes in MDA-MB-231 and RCC4 cells. Oncogene. 33:1670–1679. 2014. View Article : Google Scholar : PubMed/NCBI

53 

Jung JE, Lee HG, Cho IH, Chung DH, Yoon SH, Yang YM, Lee JW, Choi S, Park JW, Ye SK and Chung MH: STAT3 is a potential modulator of HIF-1-mediated VEGF expression in human renal carcinoma cells. FASEB J. 19:1296–1298. 2005. View Article : Google Scholar : PubMed/NCBI

54 

Lee SH, Kang JH, Ha JS, Lee JS, Oh SJ, Choi HJ, Song J and Kim SY: Transglutaminase 2-mediated p53 depletion promotes angiogenesis by increasing HIF-1α-p300 binding in renal cell carcinoma. Int J Mol Sci. 21:50422020. View Article : Google Scholar : PubMed/NCBI

55 

Datta K, Li J, Bhattacharya R, Gasparian L, Wang E and Mukhopadhyay D: Protein kinase C zeta transactivates hypoxia-inducible factor alpha by promoting its association with p300 in renal cancer. Cancer Res. 64:456–462. 2004. View Article : Google Scholar : PubMed/NCBI

56 

Lou F, Chen X, Jalink M, Zhu Q, Ge N, Zhao S, Fang X, Fan Y, Björkholm M, Liu Z and Xu D: The opposing effect of hypoxia-inducible factor-2alpha on expression of telomerase reverse transcriptase. Mol Cancer Res. 5:793–800. 2007. View Article : Google Scholar : PubMed/NCBI

57 

Zeng X, Chen K, Li L, Tian J, Ruan W, Hu Z, Peng D and Chen Z: Epigenetic activation of RBM15 promotes clear cell renal cell carcinoma growth, metastasis and macrophage infiltration by regulating the m6A modification of CXCL11. Free Radic Biol Med. 184:135–147. 2022. View Article : Google Scholar : PubMed/NCBI

58 

Kushal S, Lao BB, Henchey LK, Dubey R, Mesallati H, Traaseth NJ, Olenyuk BZ and Arora PS: Protein domain mimetics as in vivo modulators of hypoxia-inducible factor signaling. Proc Natl Acad Sci USA. 110:15602–15607. 2013. View Article : Google Scholar : PubMed/NCBI

59 

Latif AL, Newcombe A, Li S, Gilroy K, Robertson NA, Lei X, Stewart HJS, Cole J, Terradas MT, Rishi L, et al: BRD4-mediated repression of p53 is a target for combination therapy in AML. Nat Commun. 12:2412021. View Article : Google Scholar : PubMed/NCBI

60 

Civenni G, Bosotti R, Timpanaro A, Vàzquez R, Merulla J, Pandit S, Rossi S, Albino D, Allegrini S, Mitra A, et al: Epigenetic control of mitochondrial fission enables self-renewal of stem-like tumor cells in human prostate cancer. Cell Metab. 30:303–318.e6. 2019. View Article : Google Scholar : PubMed/NCBI

61 

Weissman JD, Singh AK, Devaiah BN, Schuck P, LaRue RC and Singer DS: The intrinsic kinase activity of BRD4 spans its BD2-B-BID domains. J Biol Chem. 297:1013262021. View Article : Google Scholar : PubMed/NCBI

62 

Tewary P, Brooks AD, Xu YM, Wijeratne EMK, Babyak AL, Back TC, Chari R, Evans CN, Henrich CJ, Meyer TJ, et al: Small-molecule natural product physachenolide C potentiates immunotherapy efficacy by targeting BET proteins. Cancer Res. 81:3374–3386. 2021. View Article : Google Scholar : PubMed/NCBI

63 

Delmore JE, Issa GC, Lemieux ME, Rahl PB, Shi J, Jacobs HM, Kastritis E, Gilpatrick T, Paranal RM, Qi J, et al: BET bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell. 146:904–917. 2011. View Article : Google Scholar : PubMed/NCBI

64 

Sakaguchi T, Yoshino H, Sugita S, Miyamoto K, Yonemori M, Osako Y, Meguro-Horike M, Horike SI, Nakagawa M and Enokida H: Bromodomain protein BRD4 inhibitor JQ1 regulates potential prognostic molecules in advanced renal cell carcinoma. Oncotarget. 9:23003–23017. 2018. View Article : Google Scholar : PubMed/NCBI

65 

Xu M, Xu L, Wang Y, Dai G, Xue B, Liu YY and Zhu J and Zhu J: BRD4 inhibition sensitizes renal cell carcinoma cells to the PI3K/mTOR dual inhibitor VS-5584. Aging (Albany NY). 12:19147–19158. 2020. View Article : Google Scholar : PubMed/NCBI

66 

Xing ZY, Wang Y, Cheng L, Chen J, He XZ and Xing W: Bromodomain-containing protein 4 (BRD4) inhibition sensitizes palomid 529-induced anti-renal cell carcinoma cell activity in vitro and in vivo. Cell Physiol Biochem. 50:640–653. 2018. View Article : Google Scholar : PubMed/NCBI

67 

Zhu H, Mao JH, Wang Y, Gu DH, Pan XD, Shan Y and Zheng B: Dual inhibition of BRD4 and PI3K-AKT by SF2523 suppresses human renal cell carcinoma cell growth. Oncotarget. 8:98471–98481. 2017. View Article : Google Scholar : PubMed/NCBI

68 

Xu YY, Ren ZL, Liu XL, Zhang GM, Huang SS, Shi WH, Ye LX, Luo X, Liu SW, Li YL and Yu L: BAP1 loss augments sensitivity to BET inhibitors in cancer cells. Acta Pharmacol Sin. 43:1803–1815. 2022. View Article : Google Scholar : PubMed/NCBI

69 

Hsieh JJ, Chen D, Wang PI, Marker M, Redzematovic A, Chen YB, Selcuklu SD, Weinhold N, Bouvier N, Huberman KH, et al: Genomic biomarkers of a randomized trial comparing first-line everolimus and sunitinib in patients with metastatic renal cell carcinoma. Eur Urol. 71:405–414. 2017. View Article : Google Scholar : PubMed/NCBI

70 

Feng L, Wang G, Chen Y, He G, Liu B, Liu J, Chiang CM and Ouyang L: Dual-target inhibitors of bromodomain and extra-terminal proteins in cancer: A review from medicinal chemistry perspectives. Med Res Rev. 42:710–743. 2022. View Article : Google Scholar : PubMed/NCBI

71 

Pivot-Pajot C, Caron C, Govin J, Vion A, Rousseaux S and Khochbin S: Acetylation-dependent chromatin reorganization by BRDT, a testis-specific bromodomain-containing protein. Mol Cell Biol. 23:5354–5365. 2003. View Article : Google Scholar : PubMed/NCBI

72 

Wan P, Chen Z, Zhong W, Jiang H, Huang Z, Peng D, He Q and Chen N: BRDT is a novel regulator of eIF4EBP1 in renal cell carcinoma. Oncol Rep. 44:2475–2486. 2020. View Article : Google Scholar : PubMed/NCBI

73 

Zahid H, Olson NM and Pomerantz WCK: Opportunity knocks for uncovering the new function of an understudied nucleosome remodeling complex member, the bromodomain PHD finger transcription factor, BPTF. Curr Opin Chem Biol. 63:57–67. 2021. View Article : Google Scholar : PubMed/NCBI

74 

Wysocka J, Swigut T, Xiao H, Milne TA, Kwon SY, Landry J, Kauer M, Tackett AJ, Chait BT, Badenhorst P, et al: A PHD finger of NURF couples histone H3 lysine 4 trimethylation with chromatin remodelling. Nature. 442:86–90. 2006. View Article : Google Scholar : PubMed/NCBI

75 

Nayak A, Dutta M and Roychowdhury A: Emerging oncogene ATAD2: Signaling cascades and therapeutic initiatives. Life Sci. 276:1193222021. View Article : Google Scholar : PubMed/NCBI

76 

Liu H, Wen Q, Yan S, Zeng W, Zou Y, Liu Q, Zhang G, Zou J and Zou X: Tumor-promoting ATAD2 and its preclinical challenges. Biomolecules. 12:10402022. View Article : Google Scholar : PubMed/NCBI

77 

Chen D, Maruschke M, Hakenberg O, Zimmermann W, Stief CG and Buchner A: TOP2A, HELLS, ATAD2, and TET3 are novel prognostic markers in renal cell carcinoma. Urology. 102:265.e1–265.e7. 2017. View Article : Google Scholar : PubMed/NCBI

78 

Hsieh JJ, Le VH, Oyama T, Ricketts CJ, Ho TH and Cheng EH: Chromosome 3p loss-orchestrated VHL, HIF, and epigenetic deregulation in clear cell renal cell carcinoma. J Clin Oncol. Oct 29–2018.(Epub ahead of print). View Article : Google Scholar : PubMed/NCBI

79 

Jonasch E, Walker CL and Rathmell WK: Clear cell renal cell carcinoma ontogeny and mechanisms of lethality. Nat Rev Nephrol. 17:245–261. 2021. View Article : Google Scholar : PubMed/NCBI

80 

Carril-Ajuria L, Santos M, Roldán-Romero JM, Rodriguez-Antona C and de Velasco G: Prognostic and predictive value of PBRM1 in clear cell renal cell carcinoma. Cancers (Basel). 12:162019. View Article : Google Scholar : PubMed/NCBI

81 

Thompson M: Polybromo-1: The chromatin targeting subunit of the PBAF complex. Biochimie. 91:309–319. 2009. View Article : Google Scholar : PubMed/NCBI

82 

Brownlee PM, Chambers AL, Oliver AW and Downs JA: Cancer and the bromodomains of BAF180. Biochem Soc Trans. 40:364–369. 2012. View Article : Google Scholar : PubMed/NCBI

83 

Peña-Llopis S, Vega-Rubín-de-Celis S, Liao A, Leng N, Pavía-Jiménez A, Wang S, Yamasaki T, Zhrebker L, Sivanand S, Spence P, et al: BAP1 loss defines a new class of renal cell carcinoma. Nat Genet. 44:751–759. 2012. View Article : Google Scholar : PubMed/NCBI

84 

Espana-Agusti J, Warren A, Chew SK, Adams DJ and Matakidou A: Loss of PBRM1 rescues VHL dependent replication stress to promote renal carcinogenesis. Nat Commun. 8:20262017. View Article : Google Scholar : PubMed/NCBI

85 

Gu YF, Cohn S, Christie A, McKenzie T, Wolff N, Do QN, Madhuranthakam AJ, Pedrosa I, Wang T, Dey A, et al: Modeling renal cell carcinoma in mice: Bap1 and Pbrm1 inactivation drive tumor grade. Cancer Discov. 7:900–917. 2017. View Article : Google Scholar : PubMed/NCBI

86 

Liu W, Zhang B, Zhang D, Guo F, Ye K, Zhu L and Jin X: The RBPJ/DAPK3/UBE3A signaling axis induces PBRM1 degradation to modulate the sensitivity of renal cell carcinoma to CDK4/6 inhibitors. Cell Death Dis. 13:2952022. View Article : Google Scholar : PubMed/NCBI

87 

Nargund AM, Pham CG, Dong Y, Wang PI, Osmangeyoglu HU, Xie Y, Aras O, Han S, Oyama T, Takeda S, et al: The SWI/SNF protein PBRM1 restrains VHL-loss-driven clear cell renal cell carcinoma. Cell Rep. 18:2893–2906. 2017. View Article : Google Scholar : PubMed/NCBI

88 

Gao W, Li W, Xiao T, Liu XS and Kaelin WG Jr: Inactivation of the PBRM1 tumor suppressor gene amplifies the HIF-response in VHL-/- clear cell renal carcinoma. Proc Natl Acad Sci USA. 114:1027–1032. 2017. View Article : Google Scholar : PubMed/NCBI

89 

Zhou M, Leung JY, Gessner KH, Hepperla AJ, Simon JM, Davis IJ and Kim WY: PBRM1 inactivation promotes upregulation of human endogenous retroviruses in a HIF-dependent manner. Cancer Immunol Res. 10:285–290. 2022. View Article : Google Scholar : PubMed/NCBI

90 

Chowdhury B, Porter EG, Stewart JC, Ferreira CR, Schipma MJ and Dykhuizen EC: PBRM1 regulates the expression of genes involved in metabolism and cell adhesion in renal clear cell carcinoma. PLoS One. 11:e01537182016. View Article : Google Scholar : PubMed/NCBI

91 

Porter EG, Dhiman A, Chowdhury B, Carter BC, Lin H, Stewart JC, Kazemian M, Wendt MK and Dykhuizen EC: PBRM1 regulates stress response in epithelial cells. iScience. 15:196–210. 2019. View Article : Google Scholar : PubMed/NCBI

92 

Braun DA, Hou Y, Bakouny Z, Ficial M, Sant' Angelo M, Forman J, Ross-Macdonald P, Berger AC, Jegede OA, Elagina L, et al: Interplay of somatic alterations and immune infiltration modulates response to PD-1 blockade in advanced clear cell renal cell carcinoma. Nat Med. 26:909–918. 2020. View Article : Google Scholar : PubMed/NCBI

93 

Xia QY, Rao Q, Cheng L, Shen Q, Shi SS, Li L, Liu B, Zhang J, Wang YF, Shi QL, et al: Loss of BRM expression is a frequently observed event in poorly differentiated clear cell renal cell carcinoma. Histopathology. 64:847–862. 2014. View Article : Google Scholar : PubMed/NCBI

94 

Guerrero-Martinez JA and Reyes JC: High expression of SMARCA4 or SMARCA2 is frequently associated with an opposite prognosis in cancer. Sci Rep. 8:20432018. View Article : Google Scholar : PubMed/NCBI

95 

Fang R, Xia Q, Sun J, Ng HZ, Liang Y, Wang X, Wang X, Ma H, Zhou X, Cheng Y and Rao Q: Hypermethylation of BRM promoter plays oncogenic roles in development of clear cell renal cell carcinoma. J Cancer. 10:5256–5263. 2019. View Article : Google Scholar : PubMed/NCBI

96 

Fang R, Pan R, Wang X, Liang Y, Wang X, Ma H, Zhou X, Xia Q and Rao Q: Inactivation of BRM/SMARCA2 sensitizes clear cell renal cell carcinoma to histone deacetylase complex inhibitors. Pathol Res Pract. 216:1528672020. View Article : Google Scholar : PubMed/NCBI

97 

Schuetz A, Bernstein G, Dong A, Antoshenko T, Wu H, Loppnau P, Bochkarev A and Plotnikov AN: Crystal structure of a binary complex between human GCN5 histone acetyltransferase domain and acetyl coenzyme A. Proteins. 68:403–407. 2007. View Article : Google Scholar : PubMed/NCBI

98 

Ononye OE and Downey M: Posttranslational regulation of the GCN5 and PCAF acetyltransferases. PLoS Genet. 18:e10103522022. View Article : Google Scholar : PubMed/NCBI

99 

Guo Y, Liu B, Liu Y, Sun W, Gao W, Mao S and Chen L: Oncogenic chromatin modifier KAT2A activates MCT1 to drive the glycolytic process and tumor progression in renal cell carcinoma. Front Cell Dev Biol. 9:6907962021. View Article : Google Scholar : PubMed/NCBI

100 

Vetting MW, de Carvalho LP, Yu M, Hegde SS, Magnet S, Roderick SL and Blanchard JS: Structure and functions of the GNAT superfamily of acetyltransferases. Arch Biochem Biophys. 433:212–226. 2005. View Article : Google Scholar : PubMed/NCBI

101 

Lu J, Qian C, Ji Y, Bao Q and Lu B: Gene signature associated with bromodomain genes predicts the prognosis of kidney renal clear cell carcinoma. Front Genet. 12:6439352021. View Article : Google Scholar : PubMed/NCBI

102 

Shanmugasundaram K, Nayak BK, Friedrichs WE, Kaushik D, Rodriguez R and Block K: NOX4 functions as a mitochondrial energetic sensor coupling cancer metabolic reprogramming to drug resistance. Nat Commun. 8:9972017. View Article : Google Scholar : PubMed/NCBI

103 

Xu Y, Wu G, Zhang J, Li J, Ruan N, Zhang J, Zhang Z, Chen Y, Zhang Q and Xia Q: TRIM33 overexpression inhibits the progression of clear cell renal cell carcinoma in vivo and in vitro. Biomed Res Int. 2020:84092392020.PubMed/NCBI

104 

Lu K, Pan Y, Huang Z, Liang H, Ding ZY and Zhang B: TRIM proteins in hepatocellular carcinoma. J Biomed Sci. 29:692022. View Article : Google Scholar : PubMed/NCBI

105 

Jingushi K, Ueda Y, Kitae K, Hase H, Egawa H, Ohshio I, Kawakami R, Kashiwagi Y, Tsukada Y, Kobayashi T, et al: miR-629 targets TRIM33 to promote TGFβ/Smad signaling and metastatic phenotypes in ccRCC. Mol Cancer Res. 13:565–574. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2023
Volume 28 Issue 1

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wen Q, Liu H, Lou K, Zhang X, Chao W, Xin J, Gong J, Zou J and Zou X: Essential role of bromodomain proteins in renal cell carcinoma (Review). Mol Med Rep 28: 139, 2023
APA
Wen, Q., Liu, H., Lou, K., Zhang, X., Chao, W., Xin, J. ... Zou, X. (2023). Essential role of bromodomain proteins in renal cell carcinoma (Review). Molecular Medicine Reports, 28, 139. https://doi.org/10.3892/mmr.2023.13026
MLA
Wen, Q., Liu, H., Lou, K., Zhang, X., Chao, W., Xin, J., Gong, J., Zou, J., Zou, X."Essential role of bromodomain proteins in renal cell carcinoma (Review)". Molecular Medicine Reports 28.1 (2023): 139.
Chicago
Wen, Q., Liu, H., Lou, K., Zhang, X., Chao, W., Xin, J., Gong, J., Zou, J., Zou, X."Essential role of bromodomain proteins in renal cell carcinoma (Review)". Molecular Medicine Reports 28, no. 1 (2023): 139. https://doi.org/10.3892/mmr.2023.13026