Skip to main content
Cellular and Molecular Life Sciences: CMLS logoLink to Cellular and Molecular Life Sciences: CMLS
. 2016 Aug 4;74(2):231–243. doi: 10.1007/s00018-016-2320-0

BET bromodomain proteins and epigenetic regulation of inflammation: implications for type 2 diabetes and breast cancer

Dequina A Nicholas 1,2, Guillaume Andrieu 1, Katherine J Strissel 1, Barbara S Nikolajczyk 2, Gerald V Denis 1,3,
PMCID: PMC5222701  NIHMSID: NIHMS808451  PMID: 27491296

Abstract

Chronic inflammation drives pathologies associated with type 2 diabetes (T2D) and breast cancer. Obesity-driven inflammation may explain increased risk and mortality of breast cancer with T2D reported in the epidemiology literature. Therapeutic approaches to target inflammation in both T2D and cancer have so far fallen short of the expected improvements in disease pathogenesis or outcomes. The targeting of epigenetic regulators of cytokine transcription and cytokine signaling offers one promising, untapped approach to treating diseases driven by inflammation. Recent work has deeply implicated the Bromodomain and Extra-Terminal domain (BET) proteins, which are acetylated histone “readers”, in epigenetic regulation of inflammation. This review focuses on inflammation associated with T2D and breast cancer, and the possibility of targeting BET proteins as an approach to regulating inflammation in the clinic. Understanding inflammation in the context of BET protein regulation may provide a basis for designing promising therapeutics for T2D and breast cancer.

Keywords: Chromatin reader, Metabolism

Introduction

The increasing prevalence of obesity, not only in the US, but worldwide, has become an alarming public health concern. Approximately one-third of adults in the US are obese, and the rise in obesity is paralleled by the increasing prevalence of T2D and obesity-associated cancers, such as breast cancer [1]. It is imperative to understand mechanisms that link obesity to increased risk of T2D and co-morbid cancer to develop approaches that specifically address this risk. Unequivocal epidemiological data associate obesity with both diabetes and cancer [24]. Several studies have provided evidence that obesity per se is not the driving factor. Rather, the metabolic and inflammatory status of individuals is proposed to be the most important factor for disease risk [3, 5]. Based on the literature, chronic inflammation is clearly implicated in the pathogenesis of type 2 diabetes (T2D) and cancer [613]. Nevertheless, targeting this inflammation in T2D has not been effective [1417] and targeting inflammation in cancer progression is yet to be thoroughly explored. Novel approaches to characterize and target inflammation in these disease processes will be necessary to overcome prior limitations. The previous approaches to reduce inflammation in T2D include neutralizing antibodies to individual cytokines thought to be important for disease progression [1417]. However, obesity-induced inflammation is not likely driven by a single cytokine [18]. In cancer, epidemiological studies suggest that non-steroidal anti-inflammatory drugs (NSAIDs), which inhibit cyclooxygenase (COX), reduce breast cancer risk [19]. The anti-inflammatory COX-2 inhibitor celecoxib (Celebrex) shows an anti-tumor transcriptional response in primary breast cancer as well as preliminary efficacy in clinical trials for advanced breast cancer [19, 20].

One under-utilized approach to treating chronic inflammation in disease is to target epigenetic regulators of cytokine transcription and signaling. The BET family of “histone reader” proteins plays critical roles in inflammation [5, 2123], suggesting that the three somatic proteins of this understudied family have potential to become useful targets for therapeutic regulation of inflammation in obesity-associated T2D, and subsequent T2D-associated cancers. Not only do BET proteins regulate inflammation, they also control cell cycle, a useful direct target in cancer (reviewed in [5]). The purpose of this review is to focus on inflammation associated with T2D and breast cancer, and the possibility of targeting BET proteins as a clinical approach to regulating unresolved, chronic inflammation.

Inflammation in obesity-associated type 2 diabetes

Understanding pro-inflammatory immune cells and their corresponding cytokine networks as mediators of T2D provides a basis to determine promising therapeutic targets. Obesity is the leading risk factor associated with the development of T2D. Obesity results in multiple inflammatory events. Some of these events include adipose tissue hypoxia [24, 25], leptin secretion and unfolded protein responses that are activated by endoplasmic reticulum stress [26]. Secretion of cytokines and increased lipolysis are also characteristic of obesity-driven inflammation [27]. Visceral adipose tissue (VAT) has proven to be a key site for the inflammation that is associated with T2D [28]. Infiltration of VAT by immune cells results in low-grade chronic inflammation and secretion of pro-inflammatory cytokines that can induce insulin resistance (IR), both locally and systemically (reviewed in [29]).

Immune cells, including macrophages, B cells and T cells, contribute to cytokine networks associated with diabetogenic inflammation and the development of T2D pathology [6, 28, 3036]. Macrophages are, by far, the best studied immune cells in diabetogenic inflammation. Macrophages are antigen-presenting cells (APCs) deemed responsible for activating T cells and secreting the bulk of cytokines during chronic inflammation in T2D [3739]. The infiltration (or in situ differentiation) of the so-called ‘M1’ pro-inflammatory macrophages into adipose tissue during obesity contributes to cytokine networks through secretion of tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, IL-12, resistin, chemokine ligand 5 (CCL5), and CCL2/MCP-1 [40, 41]. Similarly, B cells contribute to inflammation in T2D, although B cells are significantly less well studied than macrophages (reviewed in [42]). Much of what is known about the role of B cells in T2D inflammation is derived from diet-induced obese (DIO) mice studies. During obesity in mice, the total number of B cells increases in VAT [43], although in human obesity, few B cells are found in VAT depots [44]. Transfer of B cells from DIO mice to DIO B cell-null mice deteriorates the metabolic health of the recipient. Conversely, B-cell-deficient DIO mice have increased insulin sensitivity [43, 45]. However, B-cell induction of IR seems to be largely T-cell-dependent; studies show that transfer of splenic B cells from DIO mice to DIO RAG1null mice, which lack both T and B cells, has little effect on glucose tolerance [8]. Winer et al. demonstrated that B-cell-deficient mice have reduced levels of CD8+ T cells that produce interferon (IFN)γ in the fat [43]. T cells from humans with T2D only produce disease-associated levels of IL-17 in the presence of B cells [8]. Studies, such as these, identify both T cells and B cells as important for diabetogenic inflammation.

T cells and their roles in diabetogenic inflammation have also been extensively studied in mice (reviewed in [46]). In general, DIO mice have three times more T cells in VAT than mice fed a control diet which suggests increased T-cell recruitment [47]. CD4+ T cells co-localize with clusters of macrophages and dying (apoptotic/necrotic) adipocytes called “crown-like” structures (CLS) [48], which are also found in inflamed human breast adipose tissue [10, 25, 40, 49]. T-cell depletion reduces VAT inflammation, glucose intolerance, and IR in DIO mice [50]. The importance of T-cell infiltration in obesity-driven IR is highlighted by the fact that inhibition of T-cell migration to VAT prevents IR in DIO mice [51]. The pro-inflammatory T cell subsets most commonly implicated in T2D inflammation in mice are Th1 cells and CD8+ T cells [5255]. However, multiple investigations indicate that Th17 cells may be the central T-cell subset in human diabetogenic inflammation [56, 57]. Th17 cells are a pro-inflammatory subset of T helper cells (characterized by secretion of IL-17A, IL-17F, and IL-6) that frequently associate with autoimmunity and inflammatory diseases [58, 59]. Only monocytes from obese individuals with T2D (as opposed to lean individuals with T2D) support IL-17 production from T cells in vitro [60]. The disproportionate importance of Th17 mechanisms in human T2D was most convincingly shown by recent work that identified a pro-inflammatory Th17 cytokine signature, which differentiates subjects with T2D from metabolically healthy subjects, irrespective of BMI [18], and predicts T2D status with higher than random fidelity.

Although analysis of inflammation in obesity-driven T2D has generally relied on the analysis of individual cytokines, our recent work highlights the importance of analyzing cytokine networks. Analyses focused on circulating T cells, as sources of T2D-associated inflammation, identify a dominant Th17 cell cytokine profile, not just one cytokine, as important to differentiate equally obese subjects without diabetes (ND) and T2D subjects [18]. The existence of a cytokine network, as opposed to a single dominant cytokine, that drives inflammation in T2D may be the reason clinical trials that neutralize single pro-inflammatory “diabetogenic” cytokines, such as TNF-α or IL-1β [9, 11, 6169], have yielded disappointing results [1417]. We propose that, to target effects of a pathogenic cytokine network, therapies that simultaneously target leading regulatory pathways are necessary, for example, epigenetic regulation of cytokine gene transcription.

Defining new therapeutic targets for inflammation-mediated development of T2D in humans requires the ability to study diabetogenic inflammation in large cohorts. Although adipose tissue is an important site of inflammation in subjects with T2D, large-scale analysis with adipose tissue is both limiting and limited. Several studies have demonstrated that circulating immune cells is a valid alternative to adipose immune cells to study diabetogenic inflammation [7, 43, 7073]. One example of similarities between human adipose tissue and circulating immune cells is that the ratio of inflammatory Th1 and Th17 cells to anti-inflammatory regulatory T cells is higher in obese/IR compared with obese/insulin sensitive subjects [7, 7073]. A clear definition of diabetogenic inflammation will allow for the design of effective anti-inflammatory approaches that could also be useful for diabetes-associated cancers, such as breast cancer.

Inflammation as a risk factor in breast cancer

Epidemiological studies have associated increased risk and mortality in breast cancer with T2D [7481]. Women with T2D have ~20 % increased risk of breast cancer compared with women without T2D [74]. The Cancer Prevention Study II, among others [7477, 82], found that the incidence of T2D was associated with increased risk of mortality in women [hazard ratio (HR) (1.16) (95 % confidence interval (CI) (1.03–1.29)] [77]. Men with diabetes have an even greater risk of mortality [4.20 (2.20–8.04)] from breast cancer than females [77]. One obvious risk factor that may link T2D and breast cancer is obesity-induced diabetogenic inflammation [911, 6169]. Because inflammation in blood and adipose tissue depots is correlated [10, 69, 83], cytokine networks identified in obesity-associated T2D likely reflect local cytokine networks present in breast adipose tissue and could potentially explain increased mortality from breast cancer in patients with T2D. Although a causal relationship between T2D and breast cancer has yet to be established in humans, specific mouse models have begun to solidify this connection. Santander et al. demonstrated that DIO mice with breast tumors have larger tumors and increased numbers of CLS in breast adipose tissue compared to lean mice with breast tumors [84]. In obesity resistant BALB/c female mice, high fat diet is pro-tumorigenic for ER-breast cancer cell lines. The high fat diet increases breast cancer cell proliferation, lung and liver metastasis, angiogenesis, and circulation levels of pro-tumorigenic factors, such as VEGF, IL-6, and leptin [85]. Two other mouse studies have been able to link metabolic changes to inflammation and breast cancer progression. Feeding conjugated linoleic acid (CLA) to mice increases pro-inflammatory macrophage infiltration into adipose tissue [86]. When fed to ovariectomized PyMT mice, CLA increases mammary ductal hyperplasia and is associated with hyperinsulinemia [87]. In humans, breast adipose tissue inflammation and T2D are associated with shorter disease-free breast cancer survival [10, 7481]. Thus, if obesity and T2D increase breast adipose tissue inflammation similar to VAT inflammation, it is possible that diabetogenic inflammation could contribute directly to breast cancer progression via the inflammatory microenvironment (Fig. 1). It will be important for future studies to determine the mechanistic role of obesity and T2D in human breast cancer progression. In summary, targeting inflammation in subjects with both T2D and breast cancer to ameliorate breast cancer aggressiveness is an important avenue to explore for the development of therapeutics.

Fig. 1.

Fig. 1

BET proteins regulate inflammatory processes implicated in obesity-driven type 2 diabetes and breast cancer progression. Several inflammatory processes are co-regulated by BET proteins. Some processes include cytokine gene transcription and transcription of NF-κB and STAT target genes [109, 112, 120, 129, 130]. The cytokines IL-17, IL-6, and IL-1β are implicated in both obesity-driven T2D and breast cancer, and are transcriptionally regulated by BET proteins [107110]. Inflammation has been demonstrated to contribute to breast cancer aggressiveness and metastasis [5, 8285]. Inflammation is also central to the pathogenesis of T2D, although the reciprocal relationship of T2D and inflammation is not completely understood [1, 24, 2632]. 1 T2D increases the risk of breast cancer incidence and mortality. 2 Due to the role of inflammation in breast cancer, it is likely that diabetogenic inflammation contributes to the increase in breast cancer risk in T2D

Because T2D associates with worse breast cancer outcomes [77], we suspect that diabetogenic inflammation is a critical factor that contributes to these outcomes. Inflammation associates with breast cancer progression and mortality in several human studies [10, 8891]. Chronic inflammation is accepted to be carcinogenic and T2D in women associates with increased mortality, as well as adipose tissue inflammation in subjects who develop breast cancer [10]. This research suggests that diabetogenic inflammation may produce an inflammatory microenvironment in breast adipose tissue that could promote breast cancer, and likely exacerbates tumor progression. Various cytokines have been implicated in breast cancer aggressiveness (reviewed in [9294]). One such class is the Th17-associated cytokines IL-17A and IL-22. Increased intratumoral IL-17 mRNA in humans is associated with increased aggressiveness of invasive ductal carcinoma, reduced disease-free survival, and worse prognosis [95]. IL-17+ cells are identified in the microenvironment of breast cancer sections [96] and tumor-infiltrating T cells in mice produce increasing amounts of IL-17 as the tumor advances [95]. Interestingly, IL-17 may play multiple roles during cancer progression by acting directly on breast cancer cells, but also by remodeling the breast cancer microenvironment, to elicit tumor-associated angiogenesis, immune system evasion, and metastasis [97, 98]. IL-17A induces tumor growth in vivo and suppresses apoptosis [95]. The in vitro addition of IL-17 to the cell lines MDA-MB-231 and MDA-MB-435 markedly increases Matrigel invasion [96]. In further support of a role for Th17-driven inflammation in breast cancer progression, the systemic neutralization of IL-17A reduces breast cancer metastasis to the bone and lungs in arthritic mice [99, 100]. In addition, IL-17-producing immune cells are associated with tumor progression in breast cancer patients [101, 102], as well as tumor growth and angiogenesis in a mouse model of breast cancer [103]. IL-17 promotes resistance to anti-angiogenic therapy, such as anti-VEGF antibody treatment [104]. Finally, IL-17 produced by γδ T cells drives neutrophil polarization into a ‘myeloid-derived suppressor cell-like’ phenotype, leading to inhibition of cytotoxic CD8+ T cells and eliciting metastasis [105]. These insights strongly support a mechanistic link between the Th17 cytokine profile in T2D and breast cancer outcomes.

BET protein regulation of inflammation

Inflammatory processes, such as those identified in T2D and breast cancer, are subject to epigenetic regulation. Transcriptional control through epigenetic mechanisms includes the activity of chromatin “reader”, “writer”, and “eraser” proteins (reviewed in [106, 107]). One such family of proteins, the Bromodomain and extra-terminal domain (BET) proteins, is widely implicated in epigenetic regulation of inflammation (reviewed in [5, 2123]). The BET proteins, comprised of three somatic proteins, bromodomain-containing protein (BRD)2, BRD3, BRD4; and BRDT, a testis-specific variant, contain two mutually exclusive bromodomains (reviewed in [21]). The bromodomain is a 110-amino acid structural motif that gives BET proteins the ability to bind N-ε-acetylated lysine on histones in chromatin, and, therefore, to “read” histone tails in nucleosomal chromatin. This interaction enables BET proteins to recruit transcription co-regulators to promoters, thereby to control important genes, such as genes that are critical for inflammation, cell death, cell cycle, and proliferation in both normal and disease states [108, 109]. The bromodomain structure is highly conserved among species [108]. Humans have about 42 bromodomain-containing proteins [110, 111], which include scaffolding proteins, histone acetyltransferases, and helicases. Hydrogen bonding, often at asparagine residues, is the primary biochemical interaction that bromodomains utilize to bind to acetylated histones [112]. Thus, these structural units are fundamental, ubiquitous, and critical for transcription as effectors of signal transduction and as regulators of inflammatory disease.

The multiple mechanisms by which BET proteins regulate gene transcription have been well studied for BRD4 (reviewed in [113]). BRD4 enhances gene transcription through RNA Polymerase II (Pol II) by both direct and indirect mechanisms. Although the biochemical mechanisms are not understood, BRD4 exhibits intrinsic kinase activity that phosphorylates Pol II at serine-2 [114]. The intrinsic kinase activity of BRD4 is also important for the function of topoisomerase I. Phosphorylation of the carboxyl terminal domain (CTD) of Pol II by BRD4 is necessary for the CTD stimulation of topoisomerase I to induce DNA relaxation during Pol II-driven transcription [115]. BRD4 also promotes Pol II elongation by recruiting active positive transcriptional elongation factor b (P-TEFb) to phosphorylate Pol II [116, 117]. P-TEFb, a heterodimer of cyclin-dependent kinase 9 (Cdk9) and one of its regulatory subunits, Cyclin T1, T2, or K, is normally associated with 7SK/HEXIM, a ribonucleoprotein complex that sequesters P-TEFb in an inactive state. BRD4 binding to P-TEFb interrupts this association and recruits P-TEFb to acetylated chromatin [116]. In addition to promoting elongation, BRD4 is known to recruit transcription factors (TFs) to regulate gene transcription. An in vitro biochemical screen performed by Wu et al. identified c-Jun, p53, YY1, AP2, C/EBPα, V/EBPβ, and the Myc/Max heterodimer as TFs that interact with BRD4 in an acetylation-independent manner [118]. Interestingly, BRD4 can also interact with acetyl groups on TFs, for example, TWIST [119]. Like BRD4, the two other somatic family members BRD2 and BRD3 also promote gene transcription via association with hyperacetylated chromatin. In addition, BRD2 has intrinsic histone chaperone activity [120]. Importantly for inflammation, BRD4 is a coactivator of NF-κB. BRD4 binds to acetylated RelA via its bromodomains and recruits CDK9 to phosphorylate Pol II. These bromodomain-dependent interactions enhance the transcription of NF-κB-dependent pro-inflammatory cytokine genes [121]. The ability of BET proteins to regulate NF-κB, a regulator of inflammatory pathways, makes this family of proteins an appealing target for design of epigenetically directed therapeutic interventions in diabetogenic inflammation and breast cancer.

BET protein control of cytokine production

NF-κB is a master regulator of inflammation through its ability to regulate transcription of numerous cytokine genes. BET protein inhibitors have been shown to regulate general cytokine production through inhibition of recruitment of transcriptional machinery to gene promoter regions, and interruption of NF-κB target gene transcription [122124]. Overall, pan-BET inhibitors are very effective at blocking NF-κB-driven secretion of pro-inflammatory cytokines through various mechanisms (Table 1). In general, BET protein inhibition blocks the expression of the NF-κB target genes (Table 1) induced by mediators, such as TNF-α and bacterial lipopolysaccharide (LPS) [122]. Not only does JQ1 block NF-κB target gene transcription, but JQ1 also prevents phosphorylation of IκB, the upstream negative regulator of NF-κB [122]. In human macrophages, LPS activation of toll-like receptor 4 (TLR4) results in increased association of BRD4 with gene promoter regions [125, 126], whereas treatment with I-BET ablates this association [125]. Inhibition of BET proteins with I-BET151 inhibits the recruitment of transcriptional machinery, such as CREB-binding protein (which interacts with the NF-κB complex), to the IL-6 promoter [127].

Table 1.

Summary of BET inhibitor anti-inflammatory effects in vitro and in vivo

BET inhibitor Inflammatory diseases ameliorated Cytokines down regulated in vitro Tissue types tested in vitro
JQ1 [110] Psoriasis [133] and arthritis [122, 127, 135] IL-1β, TNF-α, IL-6, IL-8, and IL-10 [122, 136138] [139] Human airway smooth muscle cells [138], primary epithelial cells [137], multiple myeloma [136], RAW264.7 [126, 139], mouse macrophages [139], and fibroblast-like synoviocytes [122]
I-BET762 [132] Neuroinflammation [140] IL-1β, IL-6, IL-10, IL-12, IL-17, IL-19, IL-23, IL-27, IL-33, and TNF-α, [132] Human airway smooth muscle cells [138] and CD4+ T cells [140]
I-BET151 [141] Multiple sclerosis [127] IL-1α, IL-1β IL-6, IL-8, IFN-β, TNF-α, IL-23, and IFN-β [71, 125, 127] Synovial fibroblasts [135] macrophages [125], and RAW264.7 [127]
OTX-015 N/A IL-6 [142] Primary human cell line -mantle cell lymphoma, multiple myeloma, splenic marginal zone lymphoma, and prolymphocytic leukemia [142]
CPI-203 [143] N/A N/A N/A
CPI-0610 [144] N/A N/A N/A
RVX-208 [145, 146] N/A N/A N/A
LY294002 [147]a N/A

aPI3 kinase inhibitor—not yet tested as a BET protein inhibitor targeted at inflammation

In addition to regulating NF-κB signaling, BET proteins also regulate the STAT signaling pathway [125, 127]. STATs are important cytokine signaling mediators utilized by various immune cell receptors [128]. Deacetylase inhibitors are known to repress STAT5-mediated transcription [103, 129]. However, the mechanism of this inhibition was only recently suggested to be via displacement of BET proteins from acetylated target genes. Global non-specific acetylation induced by deacetylase inhibitors may dilute the effect of BET proteins [130]. In support of this concept, these authors also demonstrated that BRD2 associates with transcriptionally active STAT5 target genes. Moreover, JQ1 inhibits the expression of STAT5 target genes [130]. The previous work demonstrating that JQ1 reduces phosphorylation of JAK2/STAT5 supports the interpretation that BET proteins regulate STAT5 signaling [131]. In addition to STAT5, studies performed in primary human macrophages implicate BET proteins in the regulation of STAT1-mediated transcription [125]. These authors established that the pan-BET bromodomain inhibitor I-BET151 inhibits LPS-induced transcription and, more interestingly, inhibits cytokine-induced transcription of STAT target genes in a gene-specific manner [125]. These data clearly demonstrate that BET protein regulation of inflammation is not an all-or-none phenomenon, but rather a pleiotropic process that regulates specific genes, depending on the stimuli and particular cell type.

There are multiple demonstrations that BET protein inhibition decreases cytokine production (Table 1), including IL-6 (pleiotrophic), IL-17, and IL-1β, which are cytokines implicated in both diabetogenic inflammation and breast cancer [122, 132134]. In 2010, the demonstration that the small molecule inhibitor JQ1 associates with the hydrophobic pocket of bromodomains to interrupt histone binding [110], and that I-BET762, a synthetic acetylated histone mimic, disrupts BET protein interaction with chromatin, suggested that JQ1 and I-BET762 might be useful tools to investigate the relationship between BET proteins and cytokine gene transcription [110, 132]. Disruption of BET protein binding to promoters in mouse bone marrow-derived macrophages results in suppression of 38 LPS-inducible genes, including Il6, Ifnb1, Il1b, Il12a, Cxcl9, and Ccl12 [132]. Since this seminal finding, a growing body of evidence has supported a role for BET proteins in the regulation of inflammatory responses.

Of particular interest, BET proteins have been implicated in the regulation of Th17 inflammation. JQ1 inhibits retinoic acid receptor-related orphan receptor C (RORC—the Th17 master regulatory transcription factor) gene and protein expression in psoriatic mice [133]. By diminishing expression of RORC, BET inhibition reduces differentiation of Th17 cells as well as production of IL-17 and IL-22, which play a central role in Th17 inflammation as discussed above. BET proteins regulate the generation of Th17 cells and the secretion of Th17 cytokines. The ability of BET inhibitors to block Th17 inflammation might make these inhibitors useful, in light of the emerging role of Th17 inflammation in T2D and breast cancer [18, 95, 103, 148, 149].

Several studies have more specifically explored the role of individual BET protein family member in regulation of transcription of cytokine genes. In primary epithelial cells, knockdown with siBRD2 had no effect on the secretion of IL-6, an effect which would be masked by pan-BET inhibitors [137]. Substantiating this role for BRD2, knockdown of BRD2 with siRNA inhibits LPS-induced secretion of pro-inflammatory cytokines from mouse bone marrow-derived macrophages. Although BRD4 is present, only BRD2 becomes enriched at the TNF-α promoter upon LPS stimulation [139]. For future research, it will be critical to consider the exclusive effects of individual BET family members when assessing BET protein regulation of cytokine networks.

BET inhibitors can also affect cytokine networks through indirect mechanisms. T-cell secretion of cytokines is dependent on stimuli from APCs, such as dendritic cells (DCs). JQ1 inhibits LPS-induced maturation of monocyte-derived dendritic cells (moDCs). JQ1-treated moDCs have a reduced ability to support Th1 differentiation, as well as CD4+ and CD8+ T cell proliferation and cytokine secretion in vitro [150]. Taken together, the current literature on BET protein regulation of inflammation provides a basis for further exploring epigenetic targets for the development of anti-inflammatory therapeutics.

BET inhibitors and clinical applications for inflammation

The discovery of inhibitors that are selective for BET protein bromodomains has not only advanced our knowledge of BET protein function, but has provoked a concerted effort to develop these inhibitors further [151], ultimately for clinical use. The first small molecules reported to disrupt BET bromodomain interactions with acetylated histones were I-BET and JQ1 [110, 132]. Past and recent development of BET bromodomain inhibitors has been focused on either increasing the affinity, specificity, and efficacy of current compounds, or on the development of new chemical families of inhibitors. Current approaches include optimizing BET bromodomain inhibitors based on the imidazo[1,2-a]pyrazine scaffold shared by both I-BET and JQ1, and synthesizing novel bromodomain inhibitors with dissociation constants in the nanomolar range [152, 153]. Efforts to develop novel BET inhibitors resulted in the development of the isoxazole azepine, the benzo[cd]indol-2(1H)-one, and the 5- and 6-isoxazolyl benzimidazole family of small molecules as “selective” inhibitors of the BET proteins [154156], OTX015, and the Benzoisoxazoloazepine inhibitor (CPI-0610) [144]. Rvx-208 was designated as an inhibitor of BET proteins in 2013, although it was discovered in 2010 as a potent small molecule inducer of ApoA1 with an unknown mechanism [157]. This agent likely works by inhibiting BET proteins that normally act as co-repressors of transcription, thus their displacement from chromatin induces transcription [5].

Clinical use of pan-BET bromodomain inhibitors has focused on hematological cancers but has also shown efficacy for solid tumors [158162]. In particular, BET proteins regulate multiple pro-tumorigenic pathways in breast cancer cells, such as drug resistance, cell survival, proliferation, and invasion (reviewed in [162]). In vivo, JQ1 treatment significantly inhibits SUM1315 tumor growth in a xenograft mouse model [119]. Emerging data that support the efficacy of BET protein inhibition in solid tumors, including breast cancer, have provided a rationale for several clinical trials to test efficacy in humans [162].

Because BET proteins directly modulate cancer progression and inflammation, BET protein inhibition would essentially be a “two hit” approach for treating breast cancer in subjects with obesity-induced inflammation. In justification of this approach, the evidence base in support of BET inhibition for the treatment of inflammatory diseases is expanding. Complementing in vitro studies, preclinical and animal models indicate that BET inhibitors are effective for the treatment of inflammatory diseases in vivo (Table 1). Administration of JQ1 alleviates inflammation in mouse models of psoriasis, rheumatoid, and osteoarthritis (RA and OA), and multiple sclerosis (MS) [122, 127, 133, 135]. The ability of BET inhibition to treat these inflammatory diseases effectively in mice implies that BET inhibitors may be useful for the treatment of chronic inflammation associated with obesity, T2D, and breast cancers through epigenetic disruption of cytokine networks.

In light of the ubiquitous nature and diverse functions of the BET family of proteins, the need for BET inhibitors that can selectively target individual BET proteins is essential for safe transition of these inhibitors to the clinic. Currently, the potential side effects of using pan-BET bromodomain inhibitors are still under investigation. In animal models, JQ1 bioavailability is extremely high. After single-dose pharmacokinetic studies, it is evident that JQ1 is blood–brain barrier permeable (AUCbrain/AUCplasma = 98 %) and also highly available in the testes (AUCtestis/AUCplasma = 259 %) [163]. Indications that JQ1 treatment inhibits transcription in neurons and blocks memory in mice prompt concern regarding the use of pan-BET inhibitors in the clinic [164]. Furthermore, side effects of current BRD inhibitors remain significant. JQ1 reactivates HIV in patients with latent infections [165], causes uncontrolled increases in blood insulin [166, 167], and causes temporary male infertility due to inhibition of the testis-specific variant BRDT [163]. Bolden et al. demonstrated in mice that long-term suppression of BRD4 results in dramatic effects on multiple tissues, such as alopecia, stem cell depletion, and epidermal hyperplasia [168], which raises concerns over specificity. Therefore, safety concerns dictate that clinical use of BET proteins must be geared toward inhibitors that are selective among specific BET family members, coupled with tissue-specific drug targeting, to minimize the known side effects of BET protein inhibition.

Design of family member-selective inhibitors has implications for disease treatment. Because BET proteins share the structural bromodomains responsible for their epigenetic action, design of such selective inhibitors is difficult. Most efforts to develop selective inhibitors have been focused on BRD4 due to the discovery of a gene rearrangement that links BRD4 to aggressive carcinoma [169]. Additional efforts include the development of cell-based screening assays in BRD4-dependent cells [170], as well as designing small molecules that will target bromodomain 1 (BD1) of BRD4 [171, 172]. Taking the common approach to modify existing BET family inhibitors, I-BET and JQ1, Baud et al. developed an approach to introduce selectivity to BET bromodomain inhibitors [173], which involves Proteolysis Targeted Chimeras (PROTACs). These modifications tether JQ1 to a ligand for the E3 ubiquitin ligase, which results in degradation of the BET protein [174]. Surprisingly, this approach results in selective degradation of BRD4 over BRD2 and BRD3, indicating the need for more structure/function analyses as we move toward the clinical exploitation of BET protein inhibition. An important evidence base shows that BRD2 plays a definitive role in regulating responses of immune cells [139, 166, 175]. Selective BRD2 inhibitors, designed in the context of anti-cancer treatments, would be advantageous to alleviate inflammatory diseases, while minimizing the potentially serious side effects of pan-BET inhibitors currently in clinical trials.

Conclusions

Chronic, unresolved inflammation offers one logical, functional link between the increased incidence and mortality of breast cancer in persons with obesity-associated co-morbid T2D. The ability of BET proteins to regulate inflammation makes them an attractive therapeutic target. Although not yet directly demonstrated in humans, T2D is likely accompanied by an inflammatory microenvironment in breast adipose tissue that could exacerbate breast cancer risk. Common inflammatory mediators implicated in T2D and breast cancer, which are also regulated by BET proteins, include IL-6, TNF-α, IL-1β, and IL-17A [122, 132, 133]. The association of Th17 cytokines with both T2D and breast cancer, coupled with BET proteins’ unique ability to regulate Th17 inflammation via multiple mechanisms, makes BET bromodomain inhibitors potentially strong candidates for the treatment of Th17 inflammation [18, 133].

Although many gaps remain in our understanding of the role of BET proteins in T2D-associated inflammation, one study has shed light on potential avenues of research to pursue. Unexpectedly, hypomorphic Brd2 wt/lo mice, which express low levels of Brd2 compared with wild type, become obese without metabolic disease, thereby uncoupling obesity from T2D [51]. Surprisingly, in these mice, macrophage infiltration into the abundant adipose tissue is greatly attenuated, suggesting a role for Brd2 in obesity-induced macrophage responses. Although the mechanisms underlying this phenomenon remain unclear, the ability of an epigenetic regulator to couple obesity to T2D and inflammation indicates that targeting cytokine networks through BET protein inhibitors may be a viable option. Future research should focus on the role of BET proteins in T2D inflammation, and whether BET-regulated cytokines in T2D affect breast cancer progression. Developing the links between obesity-associated T2D, breast cancer, and BET proteins may facilitate the use of BET proteins inhibitors for the treatment of chronic inflammation believed to drive these diseases.

Acknowledgments

This work was supported by Grants from the National Institutes of Health: R56 DK090455 and U01 CA182898 (GVD); R21 DK089270 (BSN); Hematology Training Program T32 HL007501; and Immunology Training Program T32 AI089673.

References

  • 1.Gallagher EJ, LeRoith D. Obesity and diabetes: the increased risk of cancer and cancer-related mortality. Physiol Rev. 2015;95(3):727–748. doi: 10.1152/physrev.00030.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Xu H. Obesity and metabolic inflammation. Drug Discov Today Dis Mech. 2013;10(1):e21–e25. doi: 10.1016/j.ddmec.2013.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Rose DP, Gracheck PJ, Vona-Davis L. The interactions of obesity, inflammation and Insulin resistance in breast cancer. Cancers (Basel) 2015;7(4):2147–2168. doi: 10.3390/cancers7040883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Goran MI, Alderete TL. Targeting adipose tissue inflammation to treat the underlying basis of the metabolic complications of obesity. Nestle Nutr Inst Workshop Ser. 2012;73:49–60. doi: 10.1159/000341287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Belkina AC, Denis GV. BET domain co-regulators in obesity, inflammation and cancer. Nat Rev Cancer. 2012;12(7):465–477. doi: 10.1038/nrc3256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Bastard JP, Maachi M, Lagathu C, Kim MJ, Caron M, Vidal H, et al. Recent advances in the relationship between obesity, inflammation, and insulin resistance. Eur Cytokine Netw. 2006;17(1):4–12. [PubMed] [Google Scholar]
  • 7.Dalmas E, Venteclef N, Caer C, Poitou C, Cremer I, Aron-Wisnewsky J, et al. T cell-derived IL-22 amplifies IL-1beta-driven inflammation in human adipose tissue: relevance to obesity and type 2 diabetes. Diabetes. 2014;63(6):1966–1977. doi: 10.2337/db13-1511. [DOI] [PubMed] [Google Scholar]
  • 8.DeFuria J, Belkina AC, Jagannathan-Bogdan M, Snyder-Cappione J, Carr JD, Nersesova YR, et al. B cells promote inflammation in obesity and type 2 diabetes through regulation of T-cell function and an inflammatory cytokine profile. Proc Natl Acad Sci USA. 2013;110(13):5133–5138. doi: 10.1073/pnas.1215840110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Duncan BB, Schmidt MI, Pankow JS, Ballantyne CM, Couper D, Vigo A, et al. Low-grade systemic inflammation and the development of type 2 diabetes: the atherosclerosis risk in communities study. Diabetes. 2003;52(7):1799–1805. doi: 10.2337/diabetes.52.7.1799. [DOI] [PubMed] [Google Scholar]
  • 10.Iyengar NM, Zhou XK, Gucalp A, Morris PG, Howe LR, Giri DD, et al. Systemic correlates of white adipose tissue inflammation in early-stage breast cancer. Clin Cancer Res. 2015;22(9):2283–2289. doi: 10.1158/1078-0432.CCR-15-2239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Spranger J, Kroke A, Mohlig M, Hoffmann K, Bergmann MM, Ristow M, et al. Inflammatory cytokines and the risk to develop type 2 diabetes: results of the prospective population-based European Prospective Investigation into Cancer and Nutrition (EPIC)-Potsdam Study. Diabetes. 2003;52(3):812–817. doi: 10.2337/diabetes.52.3.812. [DOI] [PubMed] [Google Scholar]
  • 12.Howe LR, Subbaramaiah K, Hudis CA, Dannenberg AJ. Molecular pathways: adipose inflammation as a mediator of obesity-associated cancer. Clin Cancer Res. 2013;19(22):6074–6083. doi: 10.1158/1078-0432.CCR-12-2603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Iyengar NM, Hudis CA, Dannenberg AJ (2013) Obesity and inflammation: new insights into breast cancer development and progression. Am Soc Clin Oncol Educ Book:46–51 [DOI] [PMC free article] [PubMed]
  • 14.Larsen CM, Faulenbach M, Vaag A, Volund A, Ehses JA, Seifert B, et al. Interleukin-1-receptor antagonist in type 2 diabetes mellitus. N Engl J Med. 2007;356(15):1517–1526. doi: 10.1056/NEJMoa065213. [DOI] [PubMed] [Google Scholar]
  • 15.Ofei F, Hurel S, Newkirk J, Sopwith M, Taylor R. Effects of an engineered human anti-TNF-alpha antibody (CDP571) on insulin sensitivity and glycemic control in patients with NIDDM. Diabetes. 1996;45(7):881–885. doi: 10.2337/diab.45.7.881. [DOI] [PubMed] [Google Scholar]
  • 16.Yazdani-Biuki B, Stelzl H, Brezinschek HP, Hermann J, Mueller T, Krippl P, et al. Improvement of insulin sensitivity in insulin resistant subjects during prolonged treatment with the anti-TNF-alpha antibody infliximab. Eur J Clin Invest. 2004;34(9):641–642. doi: 10.1111/j.1365-2362.2004.01390.x. [DOI] [PubMed] [Google Scholar]
  • 17.Dominguez H, Storgaard H, Rask-Madsen C, Steffen Hermann T, Ihlemann N, Baunbjerg Nielsen D, et al. Metabolic and vascular effects of tumor necrosis factor-alpha blockade with etanercept in obese patients with type 2 diabetes. J Vasc Res. 2005;42(6):517–525. doi: 10.1159/000088261. [DOI] [PubMed] [Google Scholar]
  • 18.Ip B, Cilfone NA, Belkina AC, DeFuria J, Jagannathan-Bogdan M, Zhu M, et al. Th17 cytokines differentiate obesity from obesity-associated type 2 diabetes and promote TNFalpha production. Obesity (Silver Spring) 2016;24(1):102–112. doi: 10.1002/oby.21243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Agrawal A, Fentiman IS. NSAIDs and breast cancer: a possible prevention and treatment strategy. Int J Clin Pract. 2008;62(3):444–449. doi: 10.1111/j.1742-1241.2007.01668.x. [DOI] [PubMed] [Google Scholar]
  • 20.Brandao RD, Veeck J, Van de Vijver KK, Lindsey P, de Vries B, van Elssen CH, et al. A randomised controlled phase II trial of pre-operative celecoxib treatment reveals anti-tumour transcriptional response in primary breast cancer. Breast Cancer Res. 2013;15(2):R29. doi: 10.1186/bcr3409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Denis GV. Bromodomain coactivators in cancer, obesity, type 2 diabetes, and inflammation. Discov Med. 2010;10(55):489–499. [PMC free article] [PubMed] [Google Scholar]
  • 22.Shi J, Vakoc CR. The mechanisms behind the therapeutic activity of BET bromodomain inhibition. Mol Cell. 2014;54(5):728–736. doi: 10.1016/j.molcel.2014.05.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Filippakopoulos P, Knapp S. Targeting bromodomains: epigenetic readers of lysine acetylation. Nat Rev Drug Discov. 2014;13(5):337–356. doi: 10.1038/nrd4286. [DOI] [PubMed] [Google Scholar]
  • 24.Hosogai N, Fukuhara A, Oshima K, Miyata Y, Tanaka S, Segawa K, et al. Adipose tissue hypoxia in obesity and its impact on adipocytokine dysregulation. Diabetes. 2007;56(4):901–911. doi: 10.2337/db06-0911. [DOI] [PubMed] [Google Scholar]
  • 25.Rausch ME, Weisberg S, Vardhana P, Tortoriello DV. Obesity in C57BL/6J mice is characterized by adipose tissue hypoxia and cytotoxic T-cell infiltration. Int J Obes (Lond) 2008;32(3):451–463. doi: 10.1038/sj.ijo.0803744. [DOI] [PubMed] [Google Scholar]
  • 26.Hummasti S, Hotamisligil GS. Endoplasmic reticulum stress and inflammation in obesity and diabetes. Circ Res. 2010;107(5):579–591. doi: 10.1161/CIRCRESAHA.110.225698. [DOI] [PubMed] [Google Scholar]
  • 27.Halberg N, Khan T, Trujillo ME, Wernstedt-Asterholm I, Attie AD, Sherwani S, et al. Hypoxia-inducible factor 1alpha induces fibrosis and insulin resistance in white adipose tissue. Mol Cell Biol. 2009;29(16):4467–4483. doi: 10.1128/MCB.00192-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Winer S, Winer DA. The adaptive immune system as a fundamental regulator of adipose tissue inflammation and insulin resistance. Immunol Cell Biol. 2012;90(8):755–762. doi: 10.1038/icb.2011.110. [DOI] [PubMed] [Google Scholar]
  • 29.Khodabandehloo H, Gorgani-Firuzjaee S, Panahi G, Meshkani R. Molecular and cellular mechanisms linking inflammation to insulin resistance and beta-cell dysfunction. Transl Res. 2016;167(1):228–256. doi: 10.1016/j.trsl.2015.08.011. [DOI] [PubMed] [Google Scholar]
  • 30.Song MJ, Kim KH, Yoon JM, Kim JB. Activation of Toll-like receptor 4 is associated with insulin resistance in adipocytes. Biochem Biophys Res Commun. 2006;346(3):739–745. doi: 10.1016/j.bbrc.2006.05.170. [DOI] [PubMed] [Google Scholar]
  • 31.Grimble RF. Inflammatory status and insulin resistance. Curr Opin Clin Nutr Metab Care. 2002;5(5):551–559. doi: 10.1097/00075197-200209000-00015. [DOI] [PubMed] [Google Scholar]
  • 32.Pickup JC, Crook MA. Is type II diabetes mellitus a disease of the innate immune system? Diabetologia. 1998;41(10):1241–1248. doi: 10.1007/s001250051058. [DOI] [PubMed] [Google Scholar]
  • 33.Moller DE. Potential role of TNF-alpha in the pathogenesis of insulin resistance and type 2 diabetes. Trends Endocrinol Metab. 2000;11(6):212–217. doi: 10.1016/S1043-2760(00)00272-1. [DOI] [PubMed] [Google Scholar]
  • 34.Solinas G, Vilcu C, Neels JG, Bandyopadhyay GK, Luo JL, Naugler W, et al. JNK1 in hematopoietically derived cells contributes to diet-induced inflammation and insulin resistance without affecting obesity. Cell Metab. 2007;6(5):386–397. doi: 10.1016/j.cmet.2007.09.011. [DOI] [PubMed] [Google Scholar]
  • 35.Saberi M, Woods NB, de Luca C, Schenk S, Lu JC, Bandyopadhyay G, et al. Hematopoietic cell-specific deletion of toll-like receptor 4 ameliorates hepatic and adipose tissue insulin resistance in high-fat-fed mice. Cell Metab. 2009;10(5):419–429. doi: 10.1016/j.cmet.2009.09.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Nikolajczyk BS, Jagannathan-Bogdan M, Denis GV. The outliers become a stampede as immunometabolism reaches a tipping point. Immunol Rev. 2012;249(1):253–275. doi: 10.1111/j.1600-065X.2012.01142.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Morris DL, Cho KW, Delproposto JL, Oatmen KE, Geletka LM, Martinez-Santibanez G, et al. Adipose tissue macrophages function as antigen-presenting cells and regulate adipose tissue CD4+ T cells in mice. Diabetes. 2013;62(8):2762–2772. doi: 10.2337/db12-1404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Oh DY, Morinaga H, Talukdar S, Bae EJ, Olefsky JM. Increased macrophage migration into adipose tissue in obese mice. Diabetes. 2012;61(2):346–354. doi: 10.2337/db11-0860. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Boutens L, Stienstra R. Adipose tissue macrophages: going off track during obesity. Diabetologia. 2016;59(5):879–894. doi: 10.1007/s00125-016-3904-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Olefsky JM, Glass CK. Macrophages, inflammation, and insulin resistance. Annu Rev Physiol. 2010;72:219–246. doi: 10.1146/annurev-physiol-021909-135846. [DOI] [PubMed] [Google Scholar]
  • 41.Mantovani A, Sozzani S, Locati M, Allavena P, Sica A. Macrophage polarization: tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends Immunol. 2002;23(11):549–555. doi: 10.1016/S1471-4906(02)02302-5. [DOI] [PubMed] [Google Scholar]
  • 42.Winer DA, Winer S, Shen L, Chng MH, Engleman EG. B lymphocytes as emerging mediators of insulin resistance. Int J Obes Suppl. 2012;2(Suppl 1):S4–S7. doi: 10.1038/ijosup.2012.2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Winer DA, Winer S, Shen L, Wadia PP, Yantha J, Paltser G, et al. B cells promote insulin resistance through modulation of T cells and production of pathogenic IgG antibodies. Nat Med. 2011;17(5):610–617. doi: 10.1038/nm.2353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.McDonnell ME, Ganley-Leal LM, Mehta A, Bigornia SJ, Mott M, Rehman Q, et al. B lymphocytes in human subcutaneous adipose crown-like structures. Obesity (Silver Spring) 2012;20(7):1372–1378. doi: 10.1038/oby.2012.54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Duffaut C, Galitzky J, Lafontan M, Bouloumie A. Unexpected trafficking of immune cells within the adipose tissue during the onset of obesity. Biochem Biophys Res Commun. 2009;384(4):482–485. doi: 10.1016/j.bbrc.2009.05.002. [DOI] [PubMed] [Google Scholar]
  • 46.Lee BC, Lee J. Cellular and molecular players in adipose tissue inflammation in the development of obesity-induced insulin resistance. Biochim Biophys Acta. 2014;1842(3):446–462. doi: 10.1016/j.bbadis.2013.05.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Wu H, Ghosh S, Perrard XD, Feng L, Garcia GE, Perrard JL, et al. T-cell accumulation and regulated on activation, normal T cell expressed and secreted upregulation in adipose tissue in obesity. Circulation. 2007;115(8):1029–1038. doi: 10.1161/CIRCULATIONAHA.106.638379. [DOI] [PubMed] [Google Scholar]
  • 48.Strissel KJ, Stancheva Z, Miyoshi H, Perfield JW, 2nd, DeFuria J, Jick Z, et al. Adipocyte death, adipose tissue remodeling, and obesity complications. Diabetes. 2007;56(12):2910–2918. doi: 10.2337/db07-0767. [DOI] [PubMed] [Google Scholar]
  • 49.Morris PG, Hudis CA, Giri D, Morrow M, Falcone DJ, Zhou XK, et al. Inflammation and increased aromatase expression occur in the breast tissue of obese women with breast cancer. Cancer Prev Res (Phila) 2011;4(7):1021–1029. doi: 10.1158/1940-6207.CAPR-11-0110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Nishimura S, Manabe I, Nagasaki M, Eto K, Yamashita H, Ohsugi M, et al. CD8+ effector T cells contribute to macrophage recruitment and adipose tissue inflammation in obesity. Nat Med. 2009;15(8):914–920. doi: 10.1038/nm.1964. [DOI] [PubMed] [Google Scholar]
  • 51.Kendall MR, Hupfeld CJ. FTY720, a sphingosine-1-phosphate receptor modulator, reverses high-fat diet-induced weight gain, insulin resistance and adipose tissue inflammation in C57BL/6 mice. Diabetes Obes Metab. 2008;10(9):802–805. doi: 10.1111/j.1463-1326.2008.00910.x. [DOI] [PubMed] [Google Scholar]
  • 52.Strissel KJ, DeFuria J, Shaul ME, Bennett G, Greenberg AS, Obin MS. T-cell recruitment and Th1 polarization in adipose tissue during diet-induced obesity in C57BL/6 mice. Obesity. 2010;18(10):1918–1925. doi: 10.1038/oby.2010.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Rocha VZ, Folco EJ, Sukhova G, Shimizu K, Gotsman I, Vernon AH, et al. Interferon-gamma, a Th1 cytokine, regulates fat inflammation: a role for adaptive immunity in obesity. Circ Res. 2008;103(5):467–476. doi: 10.1161/CIRCRESAHA.108.177105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Winer S, Chan Y, Paltser G, Truong D, Tsui H, Bahrami J, et al. Normalization of obesity-associated insulin resistance through immunotherapy. Nat Med. 2009;15(8):921–929. doi: 10.1038/nm.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.O’Rourke RW, Metcalf MD, White AE, Madala A, Winters BR, Maizlin II, et al. Depot-specific differences in inflammatory mediators and a role for NK cells and IFN-gamma in inflammation in human adipose tissue. Int J Obes (Lond) 2009;33(9):978–990. doi: 10.1038/ijo.2009.133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Eljaafari A, Robert M, Chehimi M, Chanon S, Durand C, Vial G, et al. Adipose tissue-derived stem cells from obese subjects contribute to inflammation and reduced insulin response in adipocytes through differential regulation of the Th1/Th17 balance and monocyte activation. Diabetes. 2015;64(7):2477–2488. doi: 10.2337/db15-0162. [DOI] [PubMed] [Google Scholar]
  • 57.Guo H, Xu BC, Yang XG, Peng D, Wang Y, Liu XB, et al. A high frequency of peripheral blood IL-22 CD4 T cells in patients with new onset type 2 diabetes mellitus. J Clin Lab Anal. 2014;30(2):95–102. doi: 10.1002/jcla.21821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Weaver CT, Harrington LE, Mangan PR, Gavrieli M, Murphy KM. Th17: an effector CD4 T cell lineage with regulatory T cell ties. Immunity. 2006;24(6):677–688. doi: 10.1016/j.immuni.2006.06.002. [DOI] [PubMed] [Google Scholar]
  • 59.Langrish CL, Chen Y, Blumenschein WM, Mattson J, Basham B, Sedgwick JD, et al. IL-23 drives a pathogenic T cell population that induces autoimmune inflammation. J Exp Med. 2005;201(2):233–240. doi: 10.1084/jem.20041257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Dai X, Zhan J, Demmy TA, Poordad FB, Fauceglia PL, Zhang H, et al. Monocytes play different roles in stimulating T cells in obese diabetic individuals. Int J Immunopathol Pharmacol. 2015;28(3):374–383. doi: 10.1177/0394632015598848. [DOI] [PubMed] [Google Scholar]
  • 61.Pradhan AD, Manson JE, Rifai N, Buring JE, Ridker PM. C-reactive protein, interleukin 6, and risk of developing type 2 diabetes mellitus. JAMA. 2001;286(3):327–334. doi: 10.1001/jama.286.3.327. [DOI] [PubMed] [Google Scholar]
  • 62.Barzilay JI, Abraham L, Heckbert SR, Cushman M, Kuller LH, Resnick HE, et al. The relation of markers of inflammation to the development of glucose disorders in the elderly: the Cardiovascular Health Study. Diabetes. 2001;50(10):2384–2389. doi: 10.2337/diabetes.50.10.2384. [DOI] [PubMed] [Google Scholar]
  • 63.Festa A, Hanley AJ, Tracy RP, D’Agostino R, Jr, Haffner SM. Inflammation in the prediabetic state is related to increased insulin resistance rather than decreased insulin secretion. Circulation. 2003;108(15):1822–1830. doi: 10.1161/01.CIR.0000091339.70120.53. [DOI] [PubMed] [Google Scholar]
  • 64.Schmidt MI, Duncan BB, Sharrett AR, Lindberg G, Savage PJ, Offenbacher S, et al. Markers of inflammation and prediction of diabetes mellitus in adults (Atherosclerosis Risk in Communities study): a cohort study. Lancet. 1999;353(9165):1649–1652. doi: 10.1016/S0140-6736(99)01046-6. [DOI] [PubMed] [Google Scholar]
  • 65.Vozarova B, Weyer C, Lindsay RS, Pratley RE, Bogardus C, Tataranni PA. High white blood cell count is associated with a worsening of insulin sensitivity and predicts the development of type 2 diabetes. Diabetes. 2002;51(2):455–461. doi: 10.2337/diabetes.51.2.455. [DOI] [PubMed] [Google Scholar]
  • 66.Bigornia SJ, Farb MG, Mott MM, Hess DT, Carmine B, Fiscale A, et al. Relation of depot-specific adipose inflammation to insulin resistance in human obesity. Nutr Diabetes. 2012;2:e30. doi: 10.1038/nutd.2012.3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Farb MG, Bigornia S, Mott M, Tanriverdi K, Morin KM, Freedman JE, et al. Reduced adipose tissue inflammation represents an intermediate cardiometabolic phenotype in obesity. J Am Coll Cardiol. 2011;58(3):232–237. doi: 10.1016/j.jacc.2011.01.051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Gauthier MS, O’Brien EL, Bigornia S, Mott M, Cacicedo JM, Xu XJ, et al. Decreased AMP-activated protein kinase activity is associated with increased inflammation in visceral adipose tissue and with whole-body insulin resistance in morbidly obese humans. Biochem Biophys Res Commun. 2011;404(1):382–387. doi: 10.1016/j.bbrc.2010.11.127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Apovian CM, Bigornia S, Mott M, Meyers MR, Ulloor J, Gagua M, et al. Adipose macrophage infiltration is associated with insulin resistance and vascular endothelial dysfunction in obese subjects. Arterioscler Thromb Vasc Biol. 2008;28(9):1654–1659. doi: 10.1161/ATVBAHA.108.170316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Zeng C, Shi X, Zhang B, Liu H, Zhang L, Ding W, et al. The imbalance of Th17/Th1/Tregs in patients with type 2 diabetes: relationship with metabolic factors and complications. J Mol Med (Berl) 2012;90(2):175–186. doi: 10.1007/s00109-011-0816-5. [DOI] [PubMed] [Google Scholar]
  • 71.Fabbrini E, Cella M, McCartney SA, Fuchs A, Abumrad NA, Pietka TA, et al. Association between specific adipose tissue CD4+ T-cell populations and insulin resistance in obese individuals. Gastroenterology. 2013;145(2):366e1–3–374 e1–3. doi: 10.1053/j.gastro.2013.04.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Jagannathan-Bogdan M, McDonnell ME, Shin H, Rehman Q, Hasturk H, Apovian CM, et al. Elevated proinflammatory cytokine production by a skewed T cell compartment requires monocytes and promotes inflammation in type 2 diabetes. J Immunol. 2011;186(2):1162–1172. doi: 10.4049/jimmunol.1002615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Feuerer M, Herrero L, Cipolletta D, Naaz A, Wong J, Nayer A, et al. Lean, but not obese, fat is enriched for a unique population of regulatory T cells that affect metabolic parameters. Nat Med. 2009;15(8):930–939. doi: 10.1038/nm.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Larsson SC, Mantzoros CS, Wolk A. Diabetes mellitus and risk of breast cancer: a meta-analysis. Int J Cancer. 2007;121(4):856–862. doi: 10.1002/ijc.22717. [DOI] [PubMed] [Google Scholar]
  • 75.Barone BB, Yeh HC, Snyder CF, Peairs KS, Stein KB, Derr RL, et al. Long-term all-cause mortality in cancer patients with preexisting diabetes mellitus: a systematic review and meta-analysis. JAMA. 2008;300(23):2754–2764. doi: 10.1001/jama.2008.824. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Moore LL, Chadid S, Singer MR, Kreger BE, Denis GV. Metabolic health reduces risk of obesity-related cancer in framingham study adults. Cancer Epidemiol Biomark Prev. 2014;23(10):2057–2065. doi: 10.1158/1055-9965.EPI-14-0240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Campbell PT, Newton CC, Patel AV, Jacobs EJ, Gapstur SM. Diabetes and cause-specific mortality in a prospective cohort of one million US adults. Diabetes Care. 2012;35(9):1835–1844. doi: 10.2337/dc12-0002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Lipscombe LL, Goodwin PJ, Zinman B, McLaughlin JR, Hux JE. The impact of diabetes on survival following breast cancer. Breast Cancer Res Treat. 2008;109(2):389–395. doi: 10.1007/s10549-007-9654-0. [DOI] [PubMed] [Google Scholar]
  • 79.Currie CJ, Poole CD, Jenkins-Jones S, Gale EA, Johnson JA, Morgan CL. Mortality after incident cancer in people with and without type 2 diabetes: impact of metformin on survival. Diabetes Care. 2012;35(2):299–304. doi: 10.2337/dc11-1313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Chen JY, Chiou WK, Chou WY, Lin JD. The impact of type 2 diabetes mellitus on mortality in hospitalized female cancer patients in Taiwan. Asia Pac J Clin Oncol. 2016;12(1):e75–e81. doi: 10.1111/ajco.12084. [DOI] [PubMed] [Google Scholar]
  • 81.Cleveland RJ, North KE, Stevens J, Teitelbaum SL, Neugut AI, Gammon MD. The association of diabetes with breast cancer incidence and mortality in the Long Island Breast Cancer Study Project. Cancer Causes Control. 2012;23(7):1193–1203. doi: 10.1007/s10552-012-9989-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Calle EE, Rodriguez C, Walker-Thurmond K, Thun MJ. Overweight, obesity, and mortality from cancer in a prospectively studied cohort of US adults. N Engl J Med. 2003;348(17):1625–1638. doi: 10.1056/NEJMoa021423. [DOI] [PubMed] [Google Scholar]
  • 83.Subbaramaiah K, Howe LR, Bhardwaj P, Du B, Gravaghi C, Yantiss RK, et al. Obesity is associated with inflammation and elevated aromatase expression in the mouse mammary gland. Cancer Prev Res (Phila) 2011;4(3):329–346. doi: 10.1158/1940-6207.CAPR-10-0381. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 84.Santander AM, Lopez-Ocejo O, Casas O, Agostini T, Sanchez L, Lamas-Basulto E, et al. paracrine interactions between adipocytes and tumor cells recruit and modify macrophages to the mammary tumor microenvironment: the role of obesity and inflammation in breast adipose tissue. Cancers (Basel) 2015;7(1):143–178. doi: 10.3390/cancers7010143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Kim EJ, Choi MR, Park H, Kim M, Hong JE, Lee JY, et al. Dietary fat increases solid tumor growth and metastasis of 4T1 murine mammary carcinoma cells and mortality in obesity-resistant BALB/c mice. Breast Cancer Res. 2011;13(4):R78. doi: 10.1186/bcr2927. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Poirier H, Shapiro JS, Kim RJ, Lazar MA. Nutritional supplementation with trans-10, cis-12-conjugated linoleic acid induces inflammation of white adipose tissue. Diabetes. 2006;55(6):1634–1641. doi: 10.2337/db06-0036. [DOI] [PubMed] [Google Scholar]
  • 87.Berryhill GE, Gloviczki JM, Trott JF, Aimo L, Kraft J, Cardiff RD, et al. Diet-induced metabolic change induces estrogen-independent allometric mammary growth. Proc Natl Acad Sci USA. 2012;109(40):16294–16299. doi: 10.1073/pnas.1210527109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Frydenberg H, Thune I, Lofterod T, Mortensen ES, Eggen AE, Risberg T, et al. Pre-diagnostic high-sensitive C-reactive protein and breast cancer risk, recurrence, and survival. Breast Cancer Res Treat. 2016;155(2):345–354. doi: 10.1007/s10549-015-3671-1. [DOI] [PubMed] [Google Scholar]
  • 89.Lee SK, Choi MY, Bae SY, Lee JH, Lee HC, Kil WH, et al. Immediate postoperative inflammation is an important prognostic factor in breast cancer. Oncology. 2015;88(6):337–344. doi: 10.1159/000368985. [DOI] [PubMed] [Google Scholar]
  • 90.van Verschuer VM, Hooning MJ, van Baare-Georgieva RD, Hollestelle A, Timmermans AM, Koppert LB, et al. Tumor-associated inflammation as a potential prognostic tool in BRCA1/2-associated breast cancer. Hum Pathol. 2015;46(2):182–190. doi: 10.1016/j.humpath.2014.10.020. [DOI] [PubMed] [Google Scholar]
  • 91.Bertolini F, Orecchioni S, Petit JY, Kolonin MG. Obesity, proinflammatory mediators, adipose tissue progenitors, and breast cancer. Curr Opin Oncol. 2014;26(6):545–550. doi: 10.1097/CCO.0000000000000130. [DOI] [PubMed] [Google Scholar]
  • 92.Dethlefsen C, Hojfeldt G, Hojman P. The role of intratumoral and systemic IL-6 in breast cancer. Breast Cancer Res Treat. 2013;138(3):657–664. doi: 10.1007/s10549-013-2488-z. [DOI] [PubMed] [Google Scholar]
  • 93.Davison Z, de Blacquiere GE, Westley BR, May FE. Insulin-like growth factor-dependent proliferation and survival of triple-negative breast cancer cells: implications for therapy. Neoplasia. 2011;13(6):504–515. doi: 10.1593/neo.101590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, and cancer. Cell. 2010;140(6):883–899. doi: 10.1016/j.cell.2010.01.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Benevides L, da Fonseca DM, Donate PB, Tiezzi DG, De Carvalho DD, de Andrade JM, et al. IL17 promotes mammary tumor progression by changing the behavior of tumor cells and eliciting tumorigenic neutrophils recruitment. Cancer Res. 2015;75(18):3788–3799. doi: 10.1158/0008-5472.CAN-15-0054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Zhu X, Mulcahy LA, Mohammed RA, Lee AH, Franks HA, Kilpatrick L, et al. IL-17 expression by breast-cancer-associated macrophages: IL-17 promotes invasiveness of breast cancer cell lines. Breast Cancer Res. 2008;10(6):R95. doi: 10.1186/bcr2195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Nam JS, Terabe M, Kang MJ, Chae H, Voong N, Yang YA, et al. Transforming growth factor beta subverts the immune system into directly promoting tumor growth through interleukin-17. Cancer Res. 2008;68(10):3915–3923. doi: 10.1158/0008-5472.CAN-08-0206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Welte T, Zhang XH. Interleukin-17 could promote breast cancer progression at several stages of the disease. Mediators Inflamm. 2015;2015:804347. doi: 10.1155/2015/804347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Roy LD, Sahraei M, Schettini JL, Gruber HE, Besmer DM, Mukherjee P. Systemic neutralization of IL-17A significantly reduces breast cancer associated metastasis in arthritic mice by reducing CXCL12/SDF-1 expression in the metastatic niches. BMC Cancer. 2014;14:225. doi: 10.1186/1471-2407-14-225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Roy LD, Ghosh S, Pathangey LB, Tinder TL, Gruber HE, Mukherjee P. Collagen induced arthritis increases secondary metastasis in MMTV-PyV MT mouse model of mammary cancer. BMC Cancer. 2011;11:365. doi: 10.1186/1471-2407-11-365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Yang L, Qi Y, Hu J, Tang L, Zhao S, Shan B. Expression of Th17 cells in breast cancer tissue and its association with clinical parameters. Cell Biochem Biophys. 2012;62(1):153–159. doi: 10.1007/s12013-011-9276-3. [DOI] [PubMed] [Google Scholar]
  • 102.Faghih Z, Rezaeifard S, Safaei A, Ghaderi A, Erfani N. IL-17 and IL-4 producing CD8+ T cells in tumor draining lymph nodes of breast cancer patients: positive association with tumor progression. Iran J Immunol. 2013;10(4):193–204. [PubMed] [Google Scholar]
  • 103.Du JW, Xu KY, Fang LY, Qi XL. Interleukin-17, produced by lymphocytes, promotes tumor growth and angiogenesis in a mouse model of breast cancer. Mol Med Rep. 2012;6(5):1099–1102. doi: 10.3892/mmr.2012.1036. [DOI] [PubMed] [Google Scholar]
  • 104.Chung AS, Wu X, Zhuang G, Ngu H, Kasman I, Zhang J, et al. An interleukin-17-mediated paracrine network promotes tumor resistance to anti-angiogenic therapy. Nat Med. 2013;19(9):1114–1123. doi: 10.1038/nm.3291. [DOI] [PubMed] [Google Scholar]
  • 105.Coffelt SB, Kersten K, Doornebal CW, Weiden J, Vrijland K, Hau CS, et al. IL-17-producing gammadelta T cells and neutrophils conspire to promote breast cancer metastasis. Nature. 2015;522(7556):345–348. doi: 10.1038/nature14282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Yasmin R, Siraj S, Hassan A, Khan AR, Abbasi R, Ahmad N. Epigenetic regulation of inflammatory cytokines and associated genes in human malignancies. Mediators Inflamm. 2015;2015:201703. doi: 10.1155/2015/201703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Falkenberg KJ, Johnstone RW. Histone deacetylases and their inhibitors in cancer, neurological diseases and immune disorders. Nat Rev Drug Discov. 2014;13(9):673–691. doi: 10.1038/nrd4360. [DOI] [PubMed] [Google Scholar]
  • 108.Dhalluin C, Carlson JE, Zeng L, He C, Aggarwal AK, Zhou MM. Structure and ligand of a histone acetyltransferase bromodomain. Nature. 1999;399(6735):491–496. doi: 10.1038/20974. [DOI] [PubMed] [Google Scholar]
  • 109.Haynes SR, Dollard C, Winston F, Beck S, Trowsdale J, Dawid IB. The bromodomain: a conserved sequence found in human, Drosophila and yeast proteins. Nucleic Acids Res. 1992;20(10):2603. doi: 10.1093/nar/20.10.2603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Filippakopoulos P, Qi J, Picaud S, Shen Y, Smith WB, Fedorov O, et al. Selective inhibition of BET bromodomains. Nature. 2010;468(7327):1067–1073. doi: 10.1038/nature09504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Sanchez R, Zhou MM. The role of human bromodomains in chromatin biology and gene transcription. Curr Opin Drug Discov Dev. 2009;12(5):659–665. [PMC free article] [PubMed] [Google Scholar]
  • 112.Owen DJ, Ornaghi P, Yang JC, Lowe N, Evans PR, Ballario P, et al. The structural basis for the recognition of acetylated histone H4 by the bromodomain of histone acetyltransferase gcn5p. EMBO J. 2000;19(22):6141–6149. doi: 10.1093/emboj/19.22.6141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Devaiah BN, Singer DS. Two faces of brd4: mitotic bookmark and transcriptional lynchpin. Transcription. 2013;4(1):13–17. doi: 10.4161/trns.22542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Devaiah BN, Lewis BA, Cherman N, Hewitt MC, Albrecht BK, Robey PG, et al. BRD4 is an atypical kinase that phosphorylates serine2 of the RNA polymerase II carboxy-terminal domain. Proc Natl Acad Sci USA. 2012;109(18):6927–6932. doi: 10.1073/pnas.1120422109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Baranello L, Wojtowicz D, Cui K, Devaiah BN, Chung HJ, Chan-Salis KY, et al. RNA polymerase II regulates topoisomerase 1 activity to favor efficient transcription. Cell. 2016;165(2):357–371. doi: 10.1016/j.cell.2016.02.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Yang Z, Yik JH, Chen R, He N, Jang MK, Ozato K, et al. Recruitment of P-TEFb for stimulation of transcriptional elongation by the bromodomain protein Brd4. Mol Cell. 2005;19(4):535–545. doi: 10.1016/j.molcel.2005.06.029. [DOI] [PubMed] [Google Scholar]
  • 117.Zhou Q, Li T, Price DH. RNA polymerase II elongation control. Annu Rev Biochem. 2012;81:119–143. doi: 10.1146/annurev-biochem-052610-095910. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Wu SY, Lee AY, Lai HT, Zhang H, Chiang CM. Phospho switch triggers Brd4 chromatin binding and activator recruitment for gene-specific targeting. Mol Cell. 2013;49(5):843–857. doi: 10.1016/j.molcel.2012.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Shi J, Wang Y, Zeng L, Wu Y, Deng J, Zhang Q, et al. Disrupting the interaction of BRD4 with diacetylated Twist suppresses tumorigenesis in basal-like breast cancer. Cancer Cell. 2014;25(2):210–225. doi: 10.1016/j.ccr.2014.01.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.LeRoy G, Rickards B, Flint SJ. The double bromodomain proteins Brd2 and Brd3 couple histone acetylation to transcription. Mol Cell. 2008;30(1):51–60. doi: 10.1016/j.molcel.2008.01.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Huang B, Yang XD, Zhou MM, Ozato K, Chen LF. Brd4 coactivates transcriptional activation of NF-kappaB via specific binding to acetylated RelA. Mol Cell Biol. 2009;29(5):1375–1387. doi: 10.1128/MCB.01365-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Xiao Y, Liang L, Huang M, Qiu Q, Zeng S, Shi M, et al. Bromodomain and extra-terminal domain bromodomain inhibition prevents synovial inflammation via blocking IkappaB kinase-dependent NF-kappaB activation in rheumatoid fibroblast-like synoviocytes. Rheumatology. 2015;55(1):173–184. doi: 10.1093/rheumatology/kev312. [DOI] [PubMed] [Google Scholar]
  • 123.Jung KH, Das A, Chai JC, Kim SH, Morya N, Park KS, et al. RNA sequencing reveals distinct mechanisms underlying BET inhibitor JQ1-mediated modulation of the LPS-induced activation of BV-2 microglial cells. J Neuroinflamm. 2015;12:36. doi: 10.1186/s12974-015-0260-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Choi CS, Hong SH, Sim S, Cho KS, Kim JW, Yang SM, et al. The epigenetic reader BRD2 as a specific modulator of PAI-1 expression in lipopolysaccharide-stimulated mouse primary astrocytes. Neurochem Res. 2015;40(11):2211–2219. doi: 10.1007/s11064-015-1710-2. [DOI] [PubMed] [Google Scholar]
  • 125.Chan CH, Fang C, Qiao Y, Yarilina A, Prinjha RK, Ivashkiv LB. BET bromodomain inhibition suppresses transcriptional responses to cytokine-Jak-STAT signaling in a gene-specific manner in human monocytes. Eur J Immunol. 2015;45(1):287–297. doi: 10.1002/eji.201444862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Meng S, Zhang L, Tang Y, Tu Q, Zheng L, Yu L, et al. BET inhibitor JQ1 blocks inflammation and bone destruction. J Dent Res. 2014;93(7):657–662. doi: 10.1177/0022034514534261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Barrett E, Brothers S, Wahlestedt C, Beurel E. I-BET151 selectively regulates IL-6 production. Biochim Biophys Acta. 2014;1842(9):1549–1555. doi: 10.1016/j.bbadis.2014.05.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Nosaka T, Kawashima T, Misawa K, Ikuta K, Mui AL, Kitamura T. STAT5 as a molecular regulator of proliferation, differentiation and apoptosis in hematopoietic cells. EMBO J. 1999;18(17):4754–4765. doi: 10.1093/emboj/18.17.4754. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Rascle A, Johnston JA, Amati B. Deacetylase activity is required for recruitment of the basal transcription machinery and transactivation by STAT5. Mol Cell Biol. 2003;23(12):4162–4173. doi: 10.1128/MCB.23.12.4162-4173.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Pinz S, Unser S, Buob D, Fischer P, Jobst B, Rascle A. Deacetylase inhibitors repress STAT5-mediated transcription by interfering with bromodomain and extra-terminal (BET) protein function. Nucleic Acids Res. 2015;43(7):3524–3545. doi: 10.1093/nar/gkv188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Ott CJ, Kopp N, Bird L, Paranal RM, Qi J, Bowman T, et al. BET bromodomain inhibition targets both c-Myc and IL7R in high-risk acute lymphoblastic leukemia. Blood. 2012;120(14):2843–2852. doi: 10.1182/blood-2012-02-413021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Nicodeme E, Jeffrey KL, Schaefer U, Beinke S, Dewell S, Chung CW, et al. Suppression of inflammation by a synthetic histone mimic. Nature. 2010;468(7327):1119–1123. doi: 10.1038/nature09589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Nadeem A, Al-Harbi NO, Al-Harbi MM, El-Sherbeeny AM, Ahmad SF, Siddiqui N, et al. Imiquimod-induced psoriasis-like skin inflammation is suppressed by BET bromodomain inhibitor in mice through RORC/IL-17A pathway modulation. Pharmacol Res. 2015;99:248–257. doi: 10.1016/j.phrs.2015.06.001. [DOI] [PubMed] [Google Scholar]
  • 134.Mele DA, Salmeron A, Ghosh S, Huang HR, Bryant BM, Lora JM. BET bromodomain inhibition suppresses TH17-mediated pathology. J Exp Med. 2013;210(11):2181–2190. doi: 10.1084/jem.20130376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Klein K, Kabala PA, Grabiec AM, Gay RE, Kolling C, Lin LL, et al. The bromodomain protein inhibitor I-BET151 suppresses expression of inflammatory genes and matrix degrading enzymes in rheumatoid arthritis synovial fibroblasts. Ann Rheum Dis. 2016;75(2):422–429. doi: 10.1136/annrheumdis-2014-205809. [DOI] [PubMed] [Google Scholar]
  • 136.Ghurye RR, Stewart HJ, Chevassut TJ. Bromodomain inhibition by JQ1 suppresses lipopolysaccharide-stimulated interleukin-6 secretion in multiple myeloma cells. Cytokine. 2015;71(2):415–417. doi: 10.1016/j.cyto.2014.11.013. [DOI] [PubMed] [Google Scholar]
  • 137.Khan YM, Kirkham P, Barnes PJ, Adcock IM. Brd4 is essential for IL-1beta-induced inflammation in human airway epithelial cells. PLoS One. 2014;9(4):e95051. doi: 10.1371/journal.pone.0095051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Perry MM, Durham AL, Austin PJ, Adcock IM, Chung KF. BET bromodomains regulate transforming growth factor-beta-induced proliferation and cytokine release in asthmatic airway smooth muscle. J Biol Chem. 2015;290(14):9111–9121. doi: 10.1074/jbc.M114.612671. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Belkina AC, Nikolajczyk BS, Denis GV. BET protein function is required for inflammation: Brd2 genetic disruption and BET inhibitor JQ1 impair mouse macrophage inflammatory responses. J Immunol. 2013;190(7):3670–3678. doi: 10.4049/jimmunol.1202838. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Bandukwala HS, Gagnon J, Togher S, Greenbaum JA, Lamperti ED, Parr NJ, et al. Selective inhibition of CD4+ T-cell cytokine production and autoimmunity by BET protein and c-Myc inhibitors. Proc Natl Acad Sci USA. 2012;109(36):14532–14537. doi: 10.1073/pnas.1212264109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Mirguet O, Lamotte Y, Donche F, Toum J, Gellibert F, Bouillot A, et al. From ApoA1 upregulation to BET family bromodomain inhibition: discovery of I-BET151. Bioorg Med Chem Lett. 2012;22(8):2963–2967. doi: 10.1016/j.bmcl.2012.01.125. [DOI] [PubMed] [Google Scholar]
  • 142.Boi M, Gaudio E, Bonetti P, Kwee I, Bernasconi E, Tarantelli C, et al. The BET bromodomain inhibitor OTX015 affects pathogenetic pathways in preclinical B-cell tumor models and synergizes with targeted drugs. Clin Cancer Res. 2015;21(7):1628–1638. doi: 10.1158/1078-0432.CCR-14-1561. [DOI] [PubMed] [Google Scholar]
  • 143.Moros A, Rodriguez V, Saborit-Villarroya I, Montraveta A, Balsas P, Sandy P, et al. Synergistic antitumor activity of lenalidomide with the BET bromodomain inhibitor CPI203 in bortezomib-resistant mantle cell lymphoma. Leukemia. 2014;28(10):2049–2059. doi: 10.1038/leu.2014.106. [DOI] [PubMed] [Google Scholar]
  • 144.Albrecht BK, Gehling VS, Hewitt MC, Vaswani RG, Cote A, Leblanc Y, et al. Identification of a benzoisoxazoloazepine inhibitor (CPI-0610) of the bromodomain and extra-terminal (BET) family as a candidate for human clinical trials. J Med Chem. 2016;59(4):1330–1339. doi: 10.1021/acs.jmedchem.5b01882. [DOI] [PubMed] [Google Scholar]
  • 145.Picaud S, Wells C, Felletar I, Brotherton D, Martin S, Savitsky P, et al. RVX-208, an inhibitor of BET transcriptional regulators with selectivity for the second bromodomain. Proc Natl Acad Sci USA. 2013;110(49):19754–19759. doi: 10.1073/pnas.1310658110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.McLure KG, Gesner EM, Tsujikawa L, Kharenko OA, Attwell S, Campeau E, et al. RVX-208, an inducer of ApoA-I in humans, is a BET bromodomain antagonist. PLoS One. 2013;8(12):e83190. doi: 10.1371/journal.pone.0083190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Dittmann A, Werner T, Chung CW, Savitski MM, Falth Savitski M, Grandi P, et al. The commonly used PI3-kinase probe LY294002 is an inhibitor of BET bromodomains. ACS Chem Biol. 2014;9(2):495–502. doi: 10.1021/cb400789e. [DOI] [PubMed] [Google Scholar]
  • 148.Chen WC, Lai YH, Chen HY, Guo HR, Su IJ, Chen HH. Interleukin-17-producing cell infiltration in the breast cancer tumour microenvironment is a poor prognostic factor. Histopathology. 2013;63(2):225–233. doi: 10.1111/his.12156. [DOI] [PubMed] [Google Scholar]
  • 149.Qian X, Gu L, Ning H, Zhang Y, Hsueh EC, Fu M, et al. Increased Th17 cells in the tumor microenvironment is mediated by IL-23 via tumor-secreted prostaglandin E2. J Immunol. 2013;190(11):5894–5902. doi: 10.4049/jimmunol.1203141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Toniolo PA, Liu S, Yeh JE, Moraes-Vieira PM, Walker SR, Vafaizadeh V, et al. Inhibiting STAT5 by the BET bromodomain inhibitor JQ1 disrupts human dendritic cell maturation. J Immunol. 2015;194(7):3180–3190. doi: 10.4049/jimmunol.1401635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Brand M, Measures AM, Wilson BG, Cortopassi WA, Alexander R, Hoss M, et al. Small molecule inhibitors of bromodomain-acetyl-lysine interactions. ACS Chem Biol. 2015;10(1):22–39. doi: 10.1021/cb500996u. [DOI] [PubMed] [Google Scholar]
  • 152.Baud MG, Lin-Shiao E, Zengerle M, Tallant C, Ciulli A. New synthetic routes to triazolo-benzodiazepine analogues: expanding the scope of the bump-and-hole approach for selective bromo and extra-terminal (BET) bromodomain inhibition. J Med Chem. 2015;59(4):1492–1500. doi: 10.1021/acs.jmedchem.5b01135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.McKeown MR, Shaw DL, Fu H, Liu S, Xu X, Marineau JJ, et al. Biased multicomponent reactions to develop novel bromodomain inhibitors. J Med Chem. 2014;57(21):9019–9027. doi: 10.1021/jm501120z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Xue X, Zhang Y, Liu Z, Song M, Xing Y, Xiang Q, et al. Discovery of benzo[cd]indol-2(1H)-ones as potent and specific BET bromodomain inhibitors: structure-based virtual screening, optimization, and biological evaluation. J Med Chem. 2016;59(4):1565–1579. doi: 10.1021/acs.jmedchem.5b01511. [DOI] [PubMed] [Google Scholar]
  • 155.Gehling VS, Hewitt MC, Vaswani RG, Leblanc Y, Cote A, Nasveschuk CG, et al. Discovery, design, and optimization of isoxazole Azepine BET inhibitors. ACS Med Chem Lett. 2013;4(9):835–840. doi: 10.1021/ml4001485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Hay D, Fedorov O, Filippakopoulos P, Martin S, Philpott M, Picaud S, et al. The design and synthesis of 5- and 6-isoxazolylbenzimidazoles as selective inhibitors of the BET bromodomains. Medchemcomm. 2013;4(1):140–144. doi: 10.1039/C2MD20189E. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Gosmini R, Nguyen VL, Toum J, Simon C, Brusq JM, Krysa G, et al. The discovery of I-BET726 (GSK1324726A), a potent tetrahydroquinoline ApoA1 up-regulator and selective BET bromodomain inhibitor. J Med Chem. 2014;57(19):8111–8131. doi: 10.1021/jm5010539. [DOI] [PubMed] [Google Scholar]
  • 158.Asangani IA, Wilder-Romans K, Dommeti VL, Krishnamurthy PM, Apel IJ, Escara-Wilke J, et al. BET bromodomain inhibitors enhance efficacy and disrupt resistance to AR antagonists in the treatment of prostate cancer. Mol Cancer Res. 2016;14(4):324–331. doi: 10.1158/1541-7786.MCR-15-0472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Basheer F, Huntly BJ. BET bromodomain inhibitors in leukemia. Exp Hematol. 2015;43(8):718–731. doi: 10.1016/j.exphem.2015.06.004. [DOI] [PubMed] [Google Scholar]
  • 160.Chaidos A, Caputo V, Karadimitris A. Inhibition of bromodomain and extra-terminal proteins (BET) as a potential therapeutic approach in haematological malignancies: emerging preclinical and clinical evidence. Ther Adv Hematol. 2015;6(3):128–141. doi: 10.1177/2040620715576662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Jung M, Gelato KA, Fernandez-Montalvan A, Siegel S, Haendler B. Targeting BET bromodomains for cancer treatment. Epigenomics. 2015;7(3):487–501. doi: 10.2217/epi.14.91. [DOI] [PubMed] [Google Scholar]
  • 162.Sahai V, Redig AJ, Collier KA, Eckerdt FD, Munshi HG. Targeting bet bromodomain proteins in solid tumors. Oncotarget. 2016 doi: 10.18632/oncotarget.9804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Matzuk MM, McKeown MR, Filippakopoulos P, Li Q, Ma L, Agno JE, et al. Small-molecule inhibition of BRDT for male contraception. Cell. 2012;150(4):673–684. doi: 10.1016/j.cell.2012.06.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Korb E, Herre M, Zucker-Scharff I, Darnell RB, Allis CD. BET protein Brd4 activates transcription in neurons and BET inhibitor Jq1 blocks memory in mice. Nat Neurosci. 2015;18(10):1464–1473. doi: 10.1038/nn.4095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Banerjee C, Archin N, Michaels D, Belkina AC, Denis GV, Bradner J, et al. BET bromodomain inhibition as a novel strategy for reactivation of HIV-1. J Leukoc Biol. 2012;92(6):1147–1154. doi: 10.1189/jlb.0312165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Wang F, Liu H, Blanton WP, Belkina A, Lebrasseur NK, Denis GV. Brd2 disruption in mice causes severe obesity without type 2 diabetes. Biochem J. 2010;425(1):71–83. doi: 10.1042/BJ20090928. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Deeney JT, Belkina AC, Shirihai OS, Corkey BE, Denis GV. BET bromodomain proteins Brd2, Brd3 and Brd4 selectively regulate metabolic Pathways in the pancreatic beta-cell. PLoS One. 2016;11(3):e0151329. doi: 10.1371/journal.pone.0151329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Bolden JE, Tasdemir N, Dow LE, van Es JH, Wilkinson JE, Zhao Z, et al. Inducible in vivo silencing of Brd4 identifies potential toxicities of sustained BET protein inhibition. Cell Rep. 2014;8(6):1919–1929. doi: 10.1016/j.celrep.2014.08.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.French CA, Miyoshi I, Aster JC, Kubonishi I, Kroll TG, Dal Cin P, et al. BRD4 bromodomain gene rearrangement in aggressive carcinoma with translocation t(15;19) Am J Pathol. 2001;159(6):1987–1992. doi: 10.1016/S0002-9440(10)63049-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Schulze J, Moosmayer D, Weiske J, Fernandez-Montalvan A, Herbst C, Jung M, et al. Cell-based protein stabilization assays for the detection of interactions between small-molecule inhibitors and BRD4. J Biomol Screen. 2015;20(2):180–189. doi: 10.1177/1087057114552398. [DOI] [PubMed] [Google Scholar]
  • 171.Hugle M, Lucas X, Weitzel G, Ostrovskyi D, Breit B, Gerhardt S, et al. 4-Acyl pyrrole derivatives yield novel vectors for designing inhibitors of the acetyl-lysine recognition site of BRD4(1) J Med Chem. 2016;59(4):1518–1530. doi: 10.1021/acs.jmedchem.5b01267. [DOI] [PubMed] [Google Scholar]
  • 172.Raux B, Voitovich Y, Derviaux C, Lugari A, Rebuffet E, Milhas S, et al. Exploring selective inhibition of the first bromodomain of the human bromodomain and extra-terminal domain (BET) proteins. J Med Chem. 2016;59(4):1634–1641. doi: 10.1021/acs.jmedchem.5b01708. [DOI] [PubMed] [Google Scholar]
  • 173.Baud MG, Lin-Shiao E, Cardote T, Tallant C, Pschibul A, Chan KH, et al. Chemical biology. A bump-and-hole approach to engineer controlled selectivity of BET bromodomain chemical probes. Science. 2014;346(6209):638–641. doi: 10.1126/science.1249830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Zengerle M, Chan KH, Ciulli A. Selective small molecule induced degradation of the BET bromodomain protein BRD4. ACS Chem Biol. 2015;10(8):1770–1777. doi: 10.1021/acschembio.5b00216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Belkina AC, Blanton WP, Nikolajczyk BS, Denis GV. The double bromodomain protein Brd2 promotes B cell expansion and mitogenesis. J Leukocyte Biol. 2014;95(3):451–460. doi: 10.1189/jlb.1112588. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Cellular and Molecular Life Sciences: CMLS are provided here courtesy of Springer

RESOURCES