Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2009 Apr 13.
Published in final edited form as: Cancer Metastasis Rev. 2005 Jun;24(2):301–313. doi: 10.1007/s10555-005-1579-7

Role of nuclear factor-κB in melanoma

Katayoun I Amiri 1, Ann Richmond 1,*
PMCID: PMC2668255  NIHMSID: NIHMS49417  PMID: 15986139

Summary

Nuclear Factor-kappa B (NF-κB) is an inducible transcription factor that regulates the expression of many genes involved in the immune response. Recently, NF-κB activity has been shown to be upregulated in many cancers, including melanoma. Data indicate that the enhanced activation of NF-κB may be due to deregulations in upstream signaling pathways such as Ras/Raf, PI3K/Akt, and NIK. Multiple studies have shown that NF-κB is involved in the regulation of apoptosis, angiogenesis, and tumor cell invasion, all of which indicate the important role of NF-κB in tumorigenesis. Thus, understanding the molecular mechanism of melanoma progression will aid in designing new therapeutic approaches for melanoma. In this review, the association between NF-κB and melanoma tumorigenesis are discussed. Additionally, the potential of emerging selective NF-κB inhibitors for the treatment of melanoma is reviewed.

Keywords: nuclear factor-κB, melanoma, apoptosis, tumorigenesis, NF-κB inhibitors

1. Introduction

Melanoma is the most aggressive form of skin cancer and its incidence has increased over 6-fold over the past 50 years. Metastatic melanoma is estimated to have caused 7600 deaths in 2003 and this disease is the second greatest cause of lost productive years among cancers [1,2]. Although causality has been difficult to link in melanoma, sun exposure and genetic susceptibility are considered important predisposing factors. Traditionally, melanoma development and progression is viewed in five distinct stages. The common acquired nevus is the first step followed by dysplastic nevus showing an increased level of structural and architectural atypia. The third step is the first recognizable malignant stage, defined as the radial growth phase (RGP) primary melanoma. The cells in this phase are locally invasive but they lack metastatic capacity. Radial growth phase cells can progress to vertical growth phase (VGP) primary melanoma lesions, the fourth step in progression. In this step, melanoma cells infiltrate and invade the dermis as large clusters of cells and exhibit metastatic potential. Metastasis to distant organs followed by overgrowth of tumor cells at these sites is the final step in the progression process [3].

Although this model depicts melanoma progression as result of a series of genetic defects intrinsic to melanocytes, our current knowledge about the importance of the tumor microenvironment in control of growth, differentiation, invasion and metastasis adds to the complexity of the disease, hence the challenges faced both in clinics and research laboratories in the fight against melanoma. Due to the complex nature of the disease, melanoma has proven to be highly resistant to conventional chemotherapy with dacarbazine (DTIC) or its derivative Temozolomide (TMZ) having the best single agent activity with a response rate of only 15–20% and a short 4 month median response duration [4,5]. Patients at high risk for recurrence (stage III) are frequently given IFN-α as adjuvant treatment. Its effectiveness is widely debated, but even supporters acknowledge its benefit as small, accompanied by a large cost in toxicity [6]. Patients with metastatic disease (stage IV) have a median survival of 6–10 months with a 5-year survival of <5% [5]. Thus far, effective treatment options have been limited at best and it is imperative to investigate new therapeutic targets for the treatment of melanoma in order to improve the dismal prognosis for this disease. Recent advances in the understanding of the underlying biology in progression of melanoma have identified key signaling pathways that are important in promoting melanoma tumorigenesis, thus providing dynamic targets for therapy. One such important target identified in melanoma tumor progression is the nuclear factor-kappa B (NF-κB) pathway [7,8]. In this review, the role of NF-κB in melanoma tumorigenesis will be discussed and potential anti-tumor mechanisms of NF-κB inhibitors will be evaluated.

2. Constitutive activation of NF-κB in melanoma

Constitutive activation of NF-κB is an emerging hallmark of various types of tumors including breast, colon, pancreatic, ovarian, and melanoma [914]. In the healthy human, NF-κB regulates the expression of genes involved in normal immunologic reactions (e.g. generation of immunoregulatory molecules such as antibody light chains) in response to proinflammatory cytokines and by-products of microbial and viral infections [1517]. NF-κB also modulates the expression of factors responsible for growth as well as apoptosis. However, increased activation of NF-κB results in enhanced expression of proinflammatory mediators, leading to acute inflammatory injury to lungs and other organs, and development of multiple organ dysfunctions as well as cancer.

The NF-κB proteins constitute a family of proteins with homology to the chicken oncogene, rel. There are five known mammalian NF-κB subunits, each characterized by ankyrin repeat elements: p65 (RelA), RelB, Rel (c-Rel), p50/p105, and p52/p100. The NF-κB proteins share an approximately 300 amino acid N-terminal domain called the Rel homology (RH) domain containing sequences important for DNA binding, inhibitor of NF-κB (IκB) binding and dimerization. However, they differ in their C-terminal domain in that RelA, RelB, and cRel exhibit transactivating functions, while p100 and p105 contain inhibitory domains. Upon various stimulations, the latter are processed to shortened, active forms called p52 and p50, respectively [18]. The NF-κB protein is composed of two subunits, which may vary, affecting the transcriptional activity of the protein. For example, p50 homodimers lack strong transactivation domains and can actually inhibit gene expression by competing with p65/p50 or other transactivating complexes for the κB-sites. The most common Rel/NF-κB dimer in mammals contains p50-RelA and is specifically called NF-κB. For the purposes of this review, NF-κB will be used to refer any induced complex that can be translocated from the cytoplasm to the nucleus and can bind to κB-sites.

NF-κB proteins are normally sequestered in the cytoplasm, due to the binding of IκB to the nuclear localization sequence of NF-κB complex. With activating signals there is phosphorylation, ubiquitination, and degradation of IκB by the 26S proteasome, thus allowing the NF-κB complex to translocate into the nucleus and bind specific DNA promoter sequences [19]. In vitro studies have shown that NF-κB binding is constitutively elevated in human melanoma cultures compared to normal melanocytes [20,21]. This observation correlates with data from clinical specimens as well, where RelA expression is significantly elevated in human nevi and melanomas relative to normal skin. This elevation in expression also correlated with increased phosphorylation and nuclear translocation of RelA [21,22]. Given that these NF-κB aberrations are detected at early stage of melanoma tumorigenesis and are sustained throughout tumor progression, one could conclude that NF-κB may play a pivotal role in many aspects of melanoma tumorigenesis.

3. Role of NF-κB in melanoma tumor progression

Given that melanoma is a disorder that manifests diverse sets of clinical presentation and aggressiveness, NF-κB is an ideal target since its constitutive activation may contribute to multiple steps in processes such as transformation, initiation, promotion, angiogenesis, invasion, and metastasis. The role of NF-κB in these processes is discussed below.

3.1. Apoptosis resistance and cell proliferation

In processes such as tumor initiation and promotion where prolonged survival of cells is a crucial event, NF-κB plays an important role as a mediator of inhibition of apoptosis. In melanoma, NF-κB has been shown to activate expression of anti-apoptotic proteins such as tumor necrosis factor receptor-associated factor 1 (TRAF1), TRAF2, and the inhibitor-of-apoptosis (IAP) proteins c-IAP1, c-IAP2, and melanoma inhibitor of apoptosis (ML-IAP), survivin as well as Bcl-2 like proteins [2328]. Indeed, expression of the IAP family members survivin and ML-IAP becomes detectable in melanocytic nevi and is further increased as melanoma progresses to invasive and metastatic phase [2931]. Furthermore, tumour necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis is suppressed by NF-κB -dependent transcription in tumor cell lines [27], including melanoma [32]. Alternatively, suppression of NF-κB activity switches the prevailing death pathway in melanoma from Fas ligand (FasL) to TNF-mediated apoptosis [33]. In support of this concept, adenoviral transfer of IκB to melanoma cells renders them sensitive to TNF induced cytotoxicity [34]. Using a cell culture system we have shown that delivery of the super-repressor form of IκB to melanoma cells blocks focus formation and targets tumor cells to undergo apoptosis [35]. In addition to inhibition of apoptosis, NF-κB activation may regulate cell cycle progression by controlling expression of important cell cycle regulatory proteins such as cyclin D1 and cyclin dependent kinase 2 (CDK2), further contributing to tumor growth [3638]. These cell cycle regulatory proteins have been shown to be critical in the control of the G1/S transition and by overexpressing cyclin D1 and CDK2 among others, melanoma cells escape the cell-cycle control mechanisms and are able to initiate cell proliferation.

The constitutive nuclear activation of NF-κB could play a major role in the endogenous expression of chemokines, interleukin (IL)-1 and IL-6, vascular endothelial growth factor (VEGF), as well as other factors reported to impact melanoma growth. We have observed that once CXCL1 and CXCL8 are induced, these secreted chemokines feed back on the activation of NF-κB and further perpetuate the cycle. Endogenous activation of NF-κB in melanoma tumor cells can be inhibited ~50% with antibody to CXCL1, suggesting that additional cytokines or alterations in the signal transduction pathway contribute to the disregulation of NF-κB in melanoma [39]. Furthermore, CXCL8 expression in human melanoma cells correlates with the level of anoxia and the aggressiveness of the melanoma. The induction of transcription of CXCL8 in these necrotic/anoxic areas of the tumor is dependent upon NF-κB and AP1 activity [40]. Transfection of nonmetastatic and CXCL8-negative melanoma cells with the CXCL8 gene render them highly tumorigenic and increases their metastatic potential through upregulation of MMP-2 expression and activity, as demonstrated by increased invasiveness through Matrigel-coated filters [41]. In addition to tumor growth and invasion, these chemokines along with VEGF are involved in promotion of angiogenesis in melanoma as well and may actually work in concert with each other to promote neovascularization in tumors [42]. It was recently reported that tumor vascularity represents an early requirement for melanoma progression and that NF-κB acts as a mediator between melanoma cells and tumor vasculature [43]. Taken together, these data support a role for endogenous activation of NF-κB in association with progression and enhanced metastatic potential of malignant melanoma cells and suggest that targeting NF-κB may have significant therapeutic effect in clinical trials.

3.2. Invasion and metastasis

In invasion and metastasis of melanoma, NF-κB may regulate the production of prostaglandins via cyclooxygenase-2 (COX-2), which has been shown to be overexpressed in melanoma [44,45]. It was shown that COX-2 is expressed in the majority of primary malignant melanoma, as well as in five human malignant melanoma cell lines. These cell lines produced PGE2, which could be inhibited by the specific COX-2 inhibitor, NS-398. Although, this inhibition of COX-2 did not change proliferation of malignant melanoma, it reduced matrigel invasion in all cell lines. These data prompt for further investigation in the function of COX-2 in malignant melanoma tumors in determining the indications for chemoprevention.

NF-κB may also facilitate invasion and metastasis by inducing expression of molecules such as intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), endothelial-leukocyte adhesion molecule-1 (ELAM-1), and matrix metalloproteinases (MMPs) [4650]. These molecules are mediators of migration of cancerous cells and crossing of vessel walls and invasion at sites of metastasis. Screening of progressive melanoma lesions has demonstrated that the percentage of melanoma cells expressing ICAM-1 increases directly with tumor thickness and is higher in metastasis than in primary lesions. It has been postulated that ICAM-1 may facilitate the interaction between melanoma cells and tumor infiltrating leukocytes, thereby enhancing tumor cell adhesion to migratory and invasive leukocytes, enabling individual cells to dissociate from the primary tumor [5153].

VCAM-1 appears to contribute to melanoma metastasis by aiding in cell adhesion at sites of metastasis. Langley et al. show that upregulation of VCAM-1 is not a prerequisite for the formation of pulmonary metastasis during spontaneous melanoma metastasis, but exhibits organ-specific enhancement once lung metastasis become well established. Furthermore, these organ-specific increases in VCAM-1 expression correspond with documented clinical patterns of melanoma metastasis and are independent of systemic levels of TNF-alpha and IL-1alpha. However, increased VCAM-1 may be the result of melanoma-induced alterations at the local level, based upon evidence of melanoma cell occupation in heart, brain, and liver in mice with pulmonary metastasis [54].

The expression of ELAM-1 (E-selectin) plays a critical role in facilitating leukocyte adhesion to and subsequent transmigration of endothelium. On this basis, E-selectin has been suggested to promote tumor cell attachment to endothelium, thereby facilitating metastasis of certain types of tumors. Expression of E-selectin was shown to redirect metastasis of tumor cells expressing appropriate ligands in vivo [55]. The role of MMPs in melanoma progression, however, appears to be at an early stage, evidenced in a study by van den Oord et al. malignant melanoma lesions with thickness <1.6 mm, 63% expressed gelatinase B (MMP-9), whereas in melanoma lesions with >1.6 mm thickness, only 10% expressed MMP-9, indicating that early invasion of malignant melanoma is associated with de novo expression of MMP-9 by neoplastic melanocytes and this expression of MMP-9 may be partly responsible for the stromal changes observed in thin malignant melanoma. The absence of MMP-9 in the vertical growth phase and in metastatic lesions suggests that other factors are involved in tissue degradation and remodeling during later stages of tumor progression in malignant melanoma [56]. Thus, NF-κB may be a central regulator at multiple stages in the progression of malignant melanoma.

In addition to its role in protection against apoptosis, NF-κB may also play an critical role in resistance to conventional chemotherapy [57] by inducing expression of ATP-binding cassette (ABC) transporters [58,59]. Drug resistance is the major reason for melanoma therapy failure and melanoma cells often develop multiple mechanisms of drug resistance during tumor progression. Overexpression of multidrug resistance genes and their encoded P-glycoproteins is a major mechanism for the development of multidrug resistance in cancer cells. NF-κB has been shown to be a transcriptional regulator of the expression of these genes [5860]. Recent data indicate that NF-κB activity and NF-κB p65 subunit level were constitutively higher in MDR human leukemia and mouse mammary carcinoma cells than in drug-sensitive parental cells. Inhibition of NF-κB resulted in increased cytotoxicity of anticancer drugs, with consequent reversal of the drug-resistance of MDR cells [61]. These findings suggest yet another means by which NF-κB may facilitate melanoma tumor protection and as such further establish the importance of NF-κB as a target for the treatment of the disease.

4. Mechanism for deregulation of NF-κB

The disregulation of NF-κB in hematopoietic tumors as well as solid tumors has gradually become established as a paradigm [19]. Increase in p50 or p100 (the precursor of p52) was reported in non-small cell lung carcinoma and human breast carcinoma biopsies as well as head and neck cancer cells [10,12,62,63]. Nuclear localization of NF-κB associated with breast cancer may be related to the escape of these tumor cells from apoptosis [11]. This alteration in NF-κB is often accompanied by increased expression of anti-apoptotic cytokines and chemokines, growth factors, and oncogenes [35,6470], adhesion proteins and proteases [71], as well as inhibitors of apoptosis [72].

Inducible pathways for regulation of these events involve activation of the IκB kinase complex (comprised of IKKα, IKKβ and the adaptor molecule known as IKKγ or NEMO), leading to the phosphorylation of specific serine residues of IκB (S32 and S36 of IκBα, or S19 and S23 of IκBβ). In addition, there are non-inducible pathways for ubiquitination and degradation of IκB, which result in the targeting of this protein for degradation in the proteasome [73].

Malignant melanoma cells exhibit a constitutive activation of IKK, resulting in more rapid degradation of IκB, nuclear localization of NF-κB, and enhanced transactivating capacity of the NF-κB complex [21,39,74]. Embryonic fibroblasts from both IKKβ and IKKα null mice are unable to induce NF-κB transactivation. However, IKKβ restoration in IKKαβ null mice will not restore cytokine-stimulated activation of NF-κB. Both IKKα and IKK β are required for induction of the transactivating activity of NF-κB [75]. Since activation of NF-κB induces the transcription of inhibitors of apoptosis as well as factors associated with tumor angiogenesis, metastasis, and growth, we have come to understand that NF-κB could be an important potential therapeutic target in cancer, particularly if we could identify which upstream kinases affecting NF-κB become disregulated during tumorigenesis (Figure 1).

Figure 1.

Figure 1

Signaling Pathways activating NF-κB and potential mechanisms by which NF-κB activation promotes melanoma development and progression.

4.1. NIK modulation of NF-κB

Several recent studies have suggested that mitogen-activated kinases (MAPKs) such as NF-κB- inducing kinase (NIK) and MAP-kinase-kinase 1(MEKK1) can participate in the activation of NF-κB in the cytoplasm as well as in the modulation of its transactivation potential in the nucleus. NIK was initially thought to preferentially phosphorylate and activate IKKα, while MEKK1 was proposed to preferentially phosphorylate and activate IKKβ [76,77]. Interestingly, recent work with the NIK and MEKK1 −/− mice suggest that NIK and MEKK1 are not essential for TNFα induction of NF-κB, but are essential for activation of NF-κB by other factors. For example, NIK is required for lymphotoxin-β activation of NF-κB [78].

NIK was first identified on the basis of its association with the TRAF2 [79]. NIK associates with and is reported to activate the phosphorylation of IKKα and IKK β. When over-expressed, NIK enhances NF-κB activity and expression of kinase-defective NIK blocks NF-κB activation in response to most inducers. NIK is involved in the processing of p100 to the NF-κB/p52 subunit [80]. This p52 subunit will either homodimerize, interact with p65 to form the NF-κB p65/p52 heterodimer or form a heterodimer with RelB. Thus, the enhanced NIK activity in tumors [21] could be associated with the enhanced level of nuclear p52, which was reported to be present in breast cancer [81].

Disregulation of NF-κB inducing kinase (NIK) has been shown to play a role in the constitutive NF-κB activity in melanoma. Data suggest that IKK-associated NIK activity is enhanced in melanoma cell lines compared with normal human epidermal melanocytes (NHEMs) [21]. Kinase dead NIK blocks constitutive NF-κB or CXCL1 promoter activity in melanoma cells, but not in control NHEMs, while transient overexpression of wild type NIK results in increased phosphorylation of extracellular signal-regulated kinases 1 and 2 (ERK1/2). This phosphorylation is inhibited in cells expressing the kinase-dead NIK. Moreover, the NF-κB promoter activity in melanoma cells decreased with over expression of dominant negative ERK expression constructs [21]. These data support the hypothesis that NIK mediated ERK activation contributes to the NF-κB activity.

4.2. AKT modulates NF-κB transactivation

Once IκB is degraded by the proteasome, the nuclear localization sequence of the p50/p65 or p52/p65 NF-κB complex is exposed and consequently this complex can move to the nucleus and bind to the NF-κB element of the promoter of responsive genes. However, simply binding to the DNA is not sufficient. The p65 needs to be phosphorylated in the transactivation domain to be fully active. It has recently been suggested that a serine/threonine protein kinase isolated from the AKR mouse thymoma (AKT) may facilitate the phosphorylation of p65 on serines 529 and 536 [8285]. In response to activation of PI3 kinase, AKT is recruited to the membrane by phosphatidyl inositol (3,4,5) trisphosphate or phosphatidyl inositol (3,4) bisphosphate, and becomes phosphorylated by 3′-phosphoinositide-dependent-kinase-1 (PDK-1) on Threonine-308 in the kinase domain and by phosphoinositide-dependent-kinase-2 (PDK-2) on Serine-473. AKT phosphorylates a number of substrates at a RXRXXS/T motif, some of which mediate escape from apoptosis and contribute to cell survival. One such substrate identified is IKKα, which has a perfect consensus sequence for AKT, and Li and Stark have suggested that AKT activation of IKKα could be upstream of the phosphorylation of p65 by IKK [82]. However, work from Delhase and Karin does not support this possibility [83]. The authors argue that since IKK-β can be fully activated by TNF-α or IL-1 in IKK-α deficient cells, IKK-α phosphorylation by Akt or any other kinase is not essential for IKK activation and subsequent NF-κB induction. However, one should be careful interpreting data from knock-out systems, since compensation phenomenon in these cells can not be excluded. Other work in the field showed that the phosphorylation of the p50 NF-κB subunit by the PI3K/Akt pathway increases the binding of the NF-κB complex to DNA [84]. Moreover, inhibitors of PI3K blocked the endogenous NF-κB luciferase activity of malignant melanoma cells, though these inhibitors did not block IKK phosphorylation of the IκBα substrate [86], thus indicating Akt-mediated NF-κB activation is downstream of IKK or separate from the IKKα mediated phosphorylation of IκBα.

One critical regulatory molecule in the PI3K/Akt pathway is the lipid phosphatase known as phosphatase and tensin homolog (PTEN), which is known to shut off AKT activation by dephosphorylating phosphatidylinositol-3,4,5-trisphosphate (PIP3) and blocking AKT membrane localization. PTEN is also reported to block NF-κB activation without altering the IκB degradation pathway [84]. However, PTEN is often deleted or mutated in melanoma tumors [87,88]. The activation of AKT, which we observe in some human melanoma cells in vitro is accompanied by loss or reduction in PTEN [86]. Furthermore, absence of PTEN was observed in a significant proportion of primary cutaneous melanoma supporting a role for PTEN loss in the pathogenesis of melanoma [89]. Loss of PTEN with consequent enhanced activation of AKT has also been reported for a number of other tumor types, suggesting this is a frequent step associated with transformation.

4.3. Ras/Raf activation of NF-κB

The activation of NF-κB can suppress apoptosis induced by oncogenic ras [90]. Paradoxically, when cells are transformed by Ha-Ras, NF-κB is activated [91]. Ras has been shown to utilize Raf-dependent and independent pathways to activate NF-κB transcriptional activity and this NF-κB activity is required for oncogenic Ras to transform NIH 3T3 cells [91,92]. Cells expressing H-Ras has been shown to exhibit NF-κB activity that correlates with sustained IKK activation and lower steady-state levels of IκB in the cytosol. The activation of NF-κB in these cells was able to impair staurosporine-induced apoptosis, thus indicating that Ras activation of NF-κB contributes to protection of cells against apoptosis [93].

Whereas about 20% of melanoma appear to carry mutation in the N-Ras oncogene [94,95], over 60% were recently reported to contain mutation within the B-Raf gene [96,97]. Mutation in B-Raf resulting in the V600E amino-acid substitution was found in 41 of 60 (68%) melanoma metastases, 4 of 5 (80%) primary melanomas and, unexpectedly, in 63 of 77 (82%) nevi, suggesting that mutational activation of the RAS/RAF/MAPK pathway in nevi is a critical step in the initiation of melanocytic neoplasia [96]. The mutations in Ras and B-Raf render both kinases constitutively active, thereby eliciting constant activation of down-stream signaling components and the corresponding transcription factor substrates, including NF-κB, c-Jun and activating transcription factor 2 (ATF2). Since pathways such as Ras/Raf may impinge on the NF-κB pathway to further activate NF-κB, targeting the Ras/Raf pathway may offer a potential mechanism for therapeutic targeting of NF-κB in melanoma.

4.4. Other factors can also modulate NF-κB

While many kinases have been demonstrated to contribute to the activation of NF-κB, it has been difficult to clearly demonstrate the requirement for an activating kinase upstream of the IKK complex. NIK is a MAP3K, as are TAK1, MEKK1 and MEKK3 ([82], for review). Deletion of NIK and MEKK1 did not block TNFα or IL-1 mediated induction of IKK activity, leaving TAK1 and MEKK3 open as potential modulators of IL-1 and TNFα induction of IKK. In addition to the MAP3 kinases and AKT, a number of other indirect modulators of NF-κB activation have been described [92,98102]. Protein kinase C (PKC) has also been noted as a potential modulator of IKK activity and PKC activation of IKK is linked to tumorigenesis [103107]. In addition to IKK phosphorylation and activation, the transactivation potential of NF-κB can be induced by phosphorylation of p65. Inducible p65 phosphorylation can occur at serine 529 by casein kinase II (CKII) [108,109], serine 536 by the IKK complex [110] and at serine 276 by PKAc as well as MSK1 [111113]. This p65 phosphorylation event may also be mediated by IKKβ [110] or possibly by IKKα [75]. Thus, in trying to discern why there is disregulation of NF-κB in tumor cells, it is important to look at a number of upstream signal transduction pathways.

5. Inhibitors of NF-κB as melanoma therapy

With the slow progress in finding effective treatment of melanoma in recent years, the expectation is that novel treatment agents that target signaling pathways essential for melanoma growth and invasion may offer hope for an otherwise dismal disease. It is now clear that melanoma exhibit constitutive activation of NF-κB. This activation of NF-κB leads to endogenous expression of a number of factors associated with escape from apoptosis, tumorigenesis, and metastasis. Since the expression of many chemokines is modulated by NF-κB, inhibition of NF-κB offers the risk of inhibiting the migration of natural killer (NK) cells, tumor infiltrating lymphocytes (TILs) and dendritic cells into the developing tumor and may therefore abrogate the host immune response against the tumor. However, this negative factor is apparently offset by the enhanced apoptosis of the tumor cells due to the inhibition of NF-κB. Indeed, treatment of melanoma in mice with non-steroidal anti-inflammatory drugs (NSAIDs) such as sulindac has been shown to both inhibit NF-κB and tumor growth, without detrimental effects on the host [19]. Thus, reagents like sulindac or other NSAIDs offer potential therapeutic reagents for human tumors [113116]. Another class of NF-κB pathway inhibitors identified is the proteasome inhibitors. Initial preclinical and clinical data with the proteasome inhibitor VELCADE (formerly known as PS-341) indicate it will provide a powerful means of killing tumor cells by targeting them toward apoptosis [117119]. Indeed, combination treatment with VELCADE and TMZ resulted in complete response and remission of human melanoma tumor xenografts in nude mice [120]. Among natural inhibitors of NF-κB pathway, Curcumin, the active ingredient from the spice turmeric (Curcuma longa Linn), has been shown to possess anti-oxidant and anti-inflammatory qualities. It has recently been demonstrated to possess anti-angiogenic effect and anti-metastatic property in mouse melanoma tumor models. Treatment of B16F10 tumors with Curcumin significantly inhibited MMP-2 activity, resulting in reduced tumor growth in this model [121,122].

The anti-tumor activity of nitrosylcobalamin (NO-Cbl) was recently demonstrated in melanoma by Chawla-Sarkar et al. NO-Cbl is an analog of vitamin B12 that delivers nitric oxide (NO) and increases the expression of tumor necrosis factor-related apoptosis-inducing ligand (Apo2L/TRAIL) and its receptors in targeted cells. The authors showed that NO-Cbl suppressed Apo2L/TRAIL- and TNF-α-mediated activation of a transfected NF-κB-driven luciferase reporter. In addition, XIAP, an inhibitor of apoptosis, was inactivated by NO-Cbl and NO-Cbl treatment, rendering Apo2L/TRAIL-resistant malignancies sensitive to the anti-tumor effects of Apo2L/TRAIL in vitro and in vivo [123]. Thus, the use of NO-Cbl and Apo2L/TRAIL exploits the tumor-specific properties of both agents and represents another promising anti-cancer combination in the treatment of melanoma.

A major concern about utilizing the above mentioned inhibitors of NF-κB is the lack of selectivity. Most of these agents target other key regulatory molecules along with NF-κB, rendering it difficult to discern the specific role of NF-κB in the progression of the disease and its attribute as a therapeutic target. Recent progress in developing specific inhibitors of NF-κB, however, has resulted in designing inhibitors of IKKs. We demonstrated that treatment of melanoma cells with the specific IKK inhibitor, BMS-345541, reduced constitutive IKK activity in a reversible manner and blocked nuclear translocation of NF-κB/p65. BMS-345541 treatment also resulted in effective blockage of hyper-proliferation of melanoma cells in vitro and melanoma tumorigenesis in vivo. BMS-345541 was capable of inducing melanoma apoptosis through down-regulation of IKK activity and suppression of NF-κB-dependent transcription in melanoma BMS-345541 treatment also contributed to the apoptotic process. However, the apoptotic effect of BMS-345541 was not mediated through the ERK1/2 signal transduction pathway. Alternate methods of inhibiting NF-κB activity by over-expressing a dominant negative IKKβ or a super-repressor form of IκBα also induced melanoma cell apoptosis (Yang et al.- submitted manuscript). These data argue for the powerful role of NF-κB in the future of melanoma therapy targets and support the notion of the recent trend observed in the therapeutics development field to use biologically based target.

6. Conclusions

Constitutive activation of NF-κB is an emerging hallmark of various types of tumors and many experimental models in vitro and in animals indicate the role of this transcription factor in the regulation of apoptosis, tumor angiogenesis and proliferation, as well as tumor cell invasion and metastasis. The enhanced activation of NF-κB in tumors appears to be partially due to deregulation of upstream kinases such as Ras, Raf, NIK, and AKT that impinge on the NF-κB pathway. Thus, NF-κB may prove to be a key effector molecule executing the commands of top officials in above-mentioned signaling pathways. The importance of NF-κB in melanoma tumor progression is evident in many recent studies utilizing various inhibitors of NF-κB for the treatment of melanoma. The use of NF-κB inhibitors has resulted in significant anti-tumor effects in melanoma tumor xenograft models and some have led to ongoing clinical trials. As we are eagerly awaiting the results of these studies, it’s important to note that NF-κB targeting for treatment of cancers such as melanoma is a rather new field and in need of optimization in both drug design as well as drug delivery in patients.

Acknowledgments

We are appreciative of the support for the work described herein from the Department of Veterans Affairs for a SRCS award and Merit Award to Ann Richmond, and the NIH for CA56704 and CA34590. We would also like to thank Dr. Yukiko Ueda for critically reviewing the manuscript and providing helpful suggestions.

References

  • 1.Jemal A, Murray T, Samuels A, Ghafoor A, Ward E, Thun MJ. Cancer statistics, 2003. CA Cancer J Clin. 2003;53:5–26. doi: 10.3322/canjclin.53.1.5. [DOI] [PubMed] [Google Scholar]
  • 2.Oliveria S, Dusza S, Berwick M. Issues in the epidemiology of melanoma. Expert Rev Anticancer Ther. 2001;1:453–459. doi: 10.1586/14737140.1.3.453. [DOI] [PubMed] [Google Scholar]
  • 3.Clark WH. Tumour progression and the nature of cancer. Br J Cancer. 1991;64:631–644. doi: 10.1038/bjc.1991.375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Sun W, Schuchter LM. Metastatic melanoma. Curr Treat Options Oncol. 2001;2:193–202. doi: 10.1007/s11864-001-0033-5. [DOI] [PubMed] [Google Scholar]
  • 5.Balch CM, Buzaid AC, Soong SJ, Atkins MB, Cascinelli N, Coit DG, Fleming ID, Gershenwald JE, Houghton A, Jr, Kirkwood JM, McMasters KM, Mihm MF, Morton DL, Reintgen DS, Ross MI, Sober A, Thompson JA, Thompson JF. Final version of the American Joint Committee on Cancer staging system for cutaneous melanoma. J Clin Oncol. 2001;19:3635–3648. doi: 10.1200/JCO.2001.19.16.3635. [DOI] [PubMed] [Google Scholar]
  • 6.Punt CJ, Eggermont AM. Adjuvant interferon-alpha for melanoma revisited: News from old and new studies. Ann Oncol. 2001;12:1663–1666. doi: 10.1023/a:1013592219007. [DOI] [PubMed] [Google Scholar]
  • 7.Ivanov VN, Bhoumik A, Ronai Z. Death receptors and melanoma resistance to apoptosis. Oncogene. 2003;22:3152–3161. doi: 10.1038/sj.onc.1206456. [DOI] [PubMed] [Google Scholar]
  • 8.Nyormoi O, Bar-Eli M. Transcriptional regulation of metastasis-related genes in human melanoma. Clin Exp Metastasis. 2003;20:251–263. doi: 10.1023/a:1022991302172. [DOI] [PubMed] [Google Scholar]
  • 9.Wang W, Abbruzzese JL, Evans DB, Larry L, Cleary KR, Chiao PJ. The nuclear factor-kappa B RelA transcription factor is constitutively activated in human pancreatic adenocarcinoma cells. Clin Cancer Res. 1999;5:119–127. [PubMed] [Google Scholar]
  • 10.Bours V, Dejardin E, Goujon-Letawe F, Merville MP, Castronovo V. The NF-kappa B transcription factor and cancer: High expression of NF-kappa B- and I kappa B-related proteins in tumor cell lines. Biochem Pharmacol. 1994;47:145–149. doi: 10.1016/0006-2952(94)90448-0. [DOI] [PubMed] [Google Scholar]
  • 11.Sovak MA, Bellas RE, Kim DW, Zanieski GJ, Rogers AE, Traish AM, Sonenshein GE. Aberrant nuclear factor-kappaB/Rel expression and the pathogenesis of breast cancer. J Clin Invest. 1997;100:2952–2960. doi: 10.1172/JCI119848. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Dejardin E, Bonizzi G, Bellahcene A, Castronovo V, Merville MP, Bours V. Highly-expressed p100/p52 (NFKB2) sequesters other NF-kappa B-related proteins in the cytoplasm of human breast cancer cells. Oncogene. 1995;11:1835–1841. [PubMed] [Google Scholar]
  • 13.Duffey DC, Chen Z, Dong G, Ondrey FG, Wolf JS, Brown K, Siebenlist U, Van Waes C. Expression of a dominant-negative mutant inhibitor-kappaBalpha of nuclear factor-kappaB in human head and neck squamous cell carcinoma inhibits survival, proinflammatory cytokine expression, and tumor growth in vivo. Cancer Res. 1999;59:3468–3474. [PubMed] [Google Scholar]
  • 14.Nakshatri H, Bhat-Nakshatri P, Martin DA, Goulet RJ, Jr, Sledge GW., Jr Constitutive activation of NF-kappaB during progression of breast cancer to hormone-independent growth. Mol Cell Biol. 1997;17:3629–3639. doi: 10.1128/mcb.17.7.3629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Abraham E. NF-kappaB activation. Crit Care Med. 2000;28:N100–104. doi: 10.1097/00003246-200004001-00012. [DOI] [PubMed] [Google Scholar]
  • 16.Beg AA, Ruben SM, Scheinman RI, Haskill S, Rosen CA, Baldwin AS., Jr I kappa B interacts with the nuclear localization sequences of the subunits of NF-kappa B: A mechanism for cytoplasmic retention. Genes Dev. 1992;6:1899–1913. doi: 10.1101/gad.6.10.1899. [DOI] [PubMed] [Google Scholar]
  • 17.Makarov SS. NF-kappaB as a therapeutic *target in chronic inflammation: Recent advances. Mol Med Today. 2000;6:441–448. doi: 10.1016/s1357-4310(00)01814-1. [DOI] [PubMed] [Google Scholar]
  • 18.Chen FE, Ghosh G. Regulation of DNA binding by Rel/NF-kappaB transcription factors: Structural views. Oncogene. 1999;18:6845–6852. doi: 10.1038/sj.onc.1203224. [DOI] [PubMed] [Google Scholar]
  • 19.Baldwin AS. Control of oncogenesis and cancer therapy resistance by the transcription factor NF-kappaB. J Clin Invest. 2001;107:241–246. doi: 10.1172/JCI11991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.McNulty SE, Tohidian NB, Meyskens FL., Jr RelA, p50 and inhibitor of kappa B alpha are elevated in human metastatic melanoma cells and respond aberrantly to ultraviolet light B. Pigment Cell Res. 2001;14:456–465. doi: 10.1034/j.1600-0749.2001.140606.x. [DOI] [PubMed] [Google Scholar]
  • 21.Dhawan P, Richmond A. A novel NF-kappa B-inducing kinase-MAPK signaling pathway up-regulates NF-kappa B activity in melanoma cells. J Biol Chem. 2002;277:7920–7928. doi: 10.1074/jbc.M112210200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.McNulty SE, del Rosario R, Cen D, Meyskens FL, Jr, Yang S. Comparative expression of NF-kappaB proteins in melanocytes of normal skin vs. benign intradermal naevus and human metastatic melanoma biopsies. Pigment Cell Res. 2004;17:173–180. doi: 10.1111/j.1600-0749.2004.00128.x. [DOI] [PubMed] [Google Scholar]
  • 23.Deveraux QL, Roy N, Stennicke HR, Van Arsdale T, Zhou Q, Srinivasula SM, Alnemri ES, Salvesen GS, Reed JC. IAPs block apoptotic events induced by caspase-8 and cytochrome c by direct inhibition of distinct caspases. Embo J. 1998;17:2215–2223. doi: 10.1093/emboj/17.8.2215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Wang CY, Mayo MW, Korneluk RG, Goeddel DV, Baldwin AS., Jr NF-kappaB antiapoptosis: Induction of TRAF1 and TRAF2 and c-IAP1 and c-IAP2 to suppress caspase-8 activation. Science. 1998;281:1680–1683. doi: 10.1126/science.281.5383.1680. [DOI] [PubMed] [Google Scholar]
  • 25.Baldwin AS., Jr The NF-kappa B and I kappa B proteins: New discoveries and insights. Annu Rev Immunol. 1996;14:649–683. doi: 10.1146/annurev.immunol.14.1.649. [DOI] [PubMed] [Google Scholar]
  • 26.Micheau O, Lens S, Gaide O, Alevizopoulos K, Tschopp J. NF-kappaB signals induce the expression of c-FLIP. Mol Cell Biol. 2001;21:5299–5305. doi: 10.1128/MCB.21.16.5299-5305.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Ravi R, Bedi GC, Engstrom LW, Zeng Q, Mookerjee B, Gelinas C, Fuchs EJ, Bedi A. Regulation of death receptor expression and TRAIL/Apo2L-induced apoptosis by NF-kappaB. Nat Cell Biol. 2001;3:409–416. doi: 10.1038/35070096. [DOI] [PubMed] [Google Scholar]
  • 28.Catz SD, Johnson JL. Transcriptional regulation of bcl-2 by nuclear factor kappa B and its significance in prostate cancer. Oncogene. 2001;20:7342–7351. doi: 10.1038/sj.onc.1204926. [DOI] [PubMed] [Google Scholar]
  • 29.Grossman D, McNiff JM, Li F, Altieri DC. Expression and targeting of the apoptosis inhibitor, survivin, in human melanoma. J Invest Dermatol. 1999;113:1076–1081. doi: 10.1046/j.1523-1747.1999.00776.x. [DOI] [PubMed] [Google Scholar]
  • 30.Vucic D, Stennicke HR, Pisabarro MT, Salvesen GS, Dixit VM. ML-IAP, a novel inhibitor of apoptosis that is preferentially expressed in human melanomas. Curr Biol. 2000;10:1359–1366. doi: 10.1016/s0960-9822(00)00781-8. [DOI] [PubMed] [Google Scholar]
  • 31.Alonso SR, Ortiz P, Pollan M, Perez-Gomez B, Sanchez L, Acuna MJ, Pajares R, Martinez-Tello FJ, Hortelano CM, Piris MA, Rodriguez-Peralto JL. Progression in cutaneous malignant melanoma is associated with distinct expression profiles: A tissue microarray-based study. Am J Pathol. 2004;164:193–203. doi: 10.1016/s0002-9440(10)63110-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Franco AV, Zhang XD, Van Berkel E, Sanders JE, Zhang XY, Thomas WD, Nguyen T, Hersey P. The role of NF-kappa B in TNF-related apoptosis-inducing ligand (TRAIL)-induced apoptosis of melanoma cells. J Immunol. 2001;166:5337–5345. doi: 10.4049/jimmunol.166.9.5337. [DOI] [PubMed] [Google Scholar]
  • 33.Ivanov VN, Fodstad O, Ronai Z. Expression of ring finger-deleted TRAF2 sensitizes metastatic melanoma cells to apoptosis via up-regulation of p38, TNFalpha and suppression of NF-kappaB activities. Oncogene. 2001;20:2243–2253. doi: 10.1038/sj.onc.1204314. [DOI] [PubMed] [Google Scholar]
  • 34.Bakker TR, Reed D, Renno T, Jongeneel CV. Efficient adenoviral transfer of NF-kappaB inhibitor sensitizes melanoma to tumor necrosis factor-mediated apoptosis. Int J Cancer. 1999;80:320–323. doi: 10.1002/(sici)1097-0215(19990118)80:2<320::aid-ijc24>3.0.co;2-k. [DOI] [PubMed] [Google Scholar]
  • 35.Wang D, Yang W, Du J, Devalaraja MN, Liang P, Matsumoto K, Tsubakimoto K, Endo T, Richmond A. MGSA/GRO-mediated melanocyte transformation involves induction of Ras expression. Oncogene. 2000;19:4647–4659. doi: 10.1038/sj.onc.1203820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Guttridge DC, Albanese C, Reuther JY, Pestell RG, Baldwin AS., Jr NF-kappaB controls cell growth and differentiation through transcriptional regulation of cyclin D1. Mol Cell Biol. 1999;19:5785–5799. doi: 10.1128/mcb.19.8.5785. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Hinz M, Krappmann D, Eichten A, Heder A, Scheidereit C, Strauss M. NF-kappaB function in growth control: Regulation of cyclin D1 expression and G0/G1-to-S-phase transition. Mol Cell Biol. 1999;19:2690–2698. doi: 10.1128/mcb.19.4.2690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Bash J, Zong WX, Gelinas C. c-Rel arrests the proliferation of HeLa cells and affects critical regulators of the G1/S-phase transition. Mol Cell Biol. 1997;17:6526–6536. doi: 10.1128/mcb.17.11.6526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Yang J, Richmond A. Constitutive IkappaB kinase activity correlates with nuclear factor-kappaB activation in human melanoma cells. Cancer Res. 2001;61:4901–4909. [PubMed] [Google Scholar]
  • 40.Kunz M, Hartmann A, Flory E, Toksoy A, Koczan D, Thiesen HJ, Mukaida N, Neumann M, Rapp UR, Brocker EB, Gillitzer R. Anoxia-induced up-regulation of interleukin-8 in human malignant melanoma. A potential mechanism for high tumor aggressiveness. Am J Pathol. 1999;155:753–763. doi: 10.1016/S0002-9440(10)65174-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Luca M, Huang S, Gershenwald JE, Singh RK, Reich R, Bar-Eli M. Expression of interleukin-8 by human melanoma cells up-regulates MMP-2 activity and increases tumor growth and metastasis. Am J Pathol. 1997;151:1105–1113. [PMC free article] [PubMed] [Google Scholar]
  • 42.Rofstad EK, Halsor EF. Vascular endothelial growth factor, interleukin 8, platelet-derived endothelial cell growth factor, and basic fibroblast growth factor promote angiogenesis and metastasis in human melanoma xenografts. Cancer Res. 2000;60:4932–4938. [PubMed] [Google Scholar]
  • 43.Kashani-Sabet M, Shaikh L, Miller JR, 3rd, Nosrati M, Ferreira CM, Debs RJ, Sagebiel RW. NF-kappa B in the vascular progression of melanoma. J Clin Oncol. 2004;22:617–623. doi: 10.1200/JCO.2004.06.047. [DOI] [PubMed] [Google Scholar]
  • 44.Denkert C, Kobel M, Berger S, Siegert A, Leclere A, Trefzer U, Hauptmann S. Expression of cyclooxygenase 2 in human malignant melanoma. Cancer Res. 2001;61:303–308. [PubMed] [Google Scholar]
  • 45.Goulet AC, Einsphar JG, Alberts DS, Beas A, Burk C, Bhattacharyya A, Bangert J, Harmon JM, Fujiwara H, Koki A, Nelson MA. Analysis of cyclooxygenase 2 (COX-2) expression during malignant melanoma progression. Cancer Biol Ther. 2003;2:713–718. [PubMed] [Google Scholar]
  • 46.van de Stolpe A, Caldenhoven E, Stade BG, Koenderman L, Raaijmakers JA, Johnson JP, van der Saag PT. 12-O-tetradecanoylphorbol-13-acetate- and tumor necrosis factor alpha-mediated induction of intercellular adhesion molecule-1 is inhibited by dexamethasone. Functional analysis of the human intercellular adhesion molecular-1 promoter. J Biol Chem. 1994;269:6185–6192. [PubMed] [Google Scholar]
  • 47.Iademarco MF, McQuillan JJ, Rosen GD, Dean DC. Characterization of the promoter for vascular cell adhesion molecule-1 (VCAM-1) J Biol Chem. 1992;267:16323–16329. [PubMed] [Google Scholar]
  • 48.Whelan J, Ghersa P, Hooft van Huijsduijnen R, Gray J, Chandra G, Talabot F, DeLamarter JF. An NF kappa B-like factor is essential but not sufficient for cytokine induction of endothelial leukocyte adhesion molecule 1 (ELAM-1) gene transcription. Nucleic Acids Res. 1991;19:2645–2653. doi: 10.1093/nar/19.10.2645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Borghaei RC, Rawlings PL, Jr, Javadi M, Woloshin J. NF-kappaB binds to a polymorphic repressor element in the MMP-3 promoter. Biochem Biophys Res Commun. 2004;316:182–188. doi: 10.1016/j.bbrc.2004.02.030. [DOI] [PubMed] [Google Scholar]
  • 50.Bond M, Fabunmi RP, Baker AH, Newby AC. Synergistic upregulation of metalloproteinase-9 by growth factors and inflammatory cytokines: An absolute requirement for transcription factor NF-kappa B. FEBS Lett. 1998;435:29–34. doi: 10.1016/s0014-5793(98)01034-5. [DOI] [PubMed] [Google Scholar]
  • 51.Johnson JP, Stade BG, Holzmann B, Schwable W, Riethmuller G. De novo expression of intercellular-adhesion molecule-1 in melanoma correlates with increased risk of metastasis. Proc Natl Acad Sci USA. 1989;86:641–644. doi: 10.1073/pnas.86.2.641. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Boyano MD, Garcia-Vazquez MD, Lopez-Michelena T, Gardeazabal J, Bilbao J, Canavate ML, Galdeano AG, Izu R, Diaz-Ramon L, Raton JA, Diaz-Perez JL. Soluble interleukin-2 receptor, intercellular adhesion molecule-1 and interleukin-10 serum levels in patients with melanoma. Br J Cancer. 2000;83:847–852. doi: 10.1054/bjoc.2000.1402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Hirai S, Kageshita T, Kimura T, Tsujisaki M, Imai K, Wakamatsu K, Ito S, Ono T. Serum levels of sICAM-1 and 5-S-cysteinyldopa as markers of melanoma progression. Melanoma Res. 1997;7:58–62. doi: 10.1097/00008390-199702000-00009. [DOI] [PubMed] [Google Scholar]
  • 54.Langley RR, Carlisle R, Ma L, Specian RD, Gerritsen ME, Granger DN. Endothelial expression of vascular cell adhesion molecule-1 correlates with metastatic pattern in spontaneous melanoma. Microcirculation. 2001;8:335–345. doi: 10.1038/sj/mn/7800098. [DOI] [PubMed] [Google Scholar]
  • 55.Biancone L, Araki M, Araki K, Vassalli P, Stamenkovic I. Redirection of tumor metastasis by expression of E-selectin in vivo. J Exp Med. 1996;183:581–587. doi: 10.1084/jem.183.2.581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.van den Oord JJ, Paemen L, Opdenakker G, de Wolf-Peeters C. Expression of gelatinase B and the extracellular matrix metalloproteinase inducer EMMPRIN in benign and malignant pigment cell lesions of the skin. Am J Pathol. 1997;151:665–670. [PMC free article] [PubMed] [Google Scholar]
  • 57.Wang CY, Cusack JC, Jr, Liu R, Baldwin AS., Jr Control of inducible chemoresistance: Enhanced anti-tumor therapy through increased apoptosis by inhibition of NF-kappaB. Nat Med. 1999;5:412–417. doi: 10.1038/7410. [DOI] [PubMed] [Google Scholar]
  • 58.Kuo MT, Liu Z, Wei Y, Lin-Lee YC, Tatebe S, Mills GB, Unate H. Induction of human MDR1 gene expression by 2-acetylaminofluorene is mediated by effectors of the phosphoinositide 3-kinase pathway that activate NF-kappaB signaling. Oncogene. 2002;21:1945–1954. doi: 10.1038/sj.onc.1205117. [DOI] [PubMed] [Google Scholar]
  • 59.Deng L, Lin-Lee YC, Claret FX, Kuo MT. 2-acetylaminofluorene up-regulates rat mdr1b expression through generating reactive oxygen species that activate NF-kappa B pathway. J Biol Chem. 2001;276:413–420. doi: 10.1074/jbc.M004551200. [DOI] [PubMed] [Google Scholar]
  • 60.Zhou G, Kuo MT. NF-kappaB-mediated induction of mdr1b expression by insulin in rat hepatoma cells. J Biol Chem. 1997;272:15174–15183. doi: 10.1074/jbc.272.24.15174. [DOI] [PubMed] [Google Scholar]
  • 61.Um JH, Kang CD, Lee BG, Kim DW, Chung BS, Kim SH. Increased and correlated nuclear factor-kappa B and Ku autoantigen activities are associated with development of multidrug resistance. Oncogene. 2001;20:6048–6056. doi: 10.1038/sj.onc.1204732. [DOI] [PubMed] [Google Scholar]
  • 62.Tamatani T, Azuma M, Aota K, Yamashita T, Bando T, Sato M. Enhanced IkappaB kinase activity is responsible for the augmented activity of NF-kappaB in human head and neck carcinoma cells. Cancer Lett. 2001;171:165–172. doi: 10.1016/s0304-3835(01)00611-5. [DOI] [PubMed] [Google Scholar]
  • 63.Mukhopadhyay T, Roth JA, Maxwell SA. Altered expression of the p50 subunit of the NF-kappa B transcription factor complex in non-small cell lung carcinoma. Oncogene. 1995;11:999–1003. [PubMed] [Google Scholar]
  • 64.Shattuck-Brandt RL, Richmond A. Enhanced degradation of I-kappaB alpha contributes to endogenous activation of NF-kappaB in Hs294T melanoma cells. Cancer Res. 1997;57:3032–3039. [PubMed] [Google Scholar]
  • 65.Shattuck RL, Wood LD, Jaffe GJ, Richmond A. MGSA/GRO transcription is differentially regulated in normal retinal pigment epithelial and melanoma cells. Mol Cell Biol. 1994;14:791–802. doi: 10.1128/mcb.14.1.791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Wood LD, Farmer AA, Richmond A. HMGI(Y) and Sp1 in addition to NF-kappa B regulate transcription of the MGSA/GRO alpha gene. Nucleic Acids Res. 1995;23:4210–4219. doi: 10.1093/nar/23.20.4210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Wood LD, Richmond A. Constitutive and cytokine-induced expression of the melanoma growth stimulatory activity/GRO alpha gene requires both NF-kappa B and novel constitutive factors. J Biol Chem. 1995;270:30619–30626. doi: 10.1074/jbc.270.51.30619. [DOI] [PubMed] [Google Scholar]
  • 68.Kondo S, Kono T, Sauder DN, McKenzie RC. IL-8 gene expression and production in human keratinocytes and their modulation by UVB. J Invest Dermatol. 1993;101:690–694. doi: 10.1111/1523-1747.ep12371677. [DOI] [PubMed] [Google Scholar]
  • 69.Wang Y, Becker D. Antisense targeting of basic fibroblast growth factor and fibroblast growth factor receptor-1 in human melanomas blocks intratumoral angiogenesis and tumor growth. Nat Med. 1997;3:887–893. doi: 10.1038/nm0897-887. [DOI] [PubMed] [Google Scholar]
  • 70.Shih IM, Herlyn M. Autocrine and paracrine roles for growth factors in melanoma. In Vivo. 1994;8:113–123. [PubMed] [Google Scholar]
  • 71.Gilmore TD, Koedood M, Piffat KA, White DW. Rel/NF-kappaB/IkappaB proteins and cancer. Oncogene. 1996;13:1367–1378. [PubMed] [Google Scholar]
  • 72.Karin M, Lin A. NF-kappaB at the crossroads of life and death. Nat Immunol. 2002;3:221–227. doi: 10.1038/ni0302-221. [DOI] [PubMed] [Google Scholar]
  • 73.Pando MP, Verma IM. Signal-dependent and -independent degradation of free and NF-kappa B-bound IkappaBalpha. J Biol Chem. 2000;275:21278–21286. doi: 10.1074/jbc.M002532200. [DOI] [PubMed] [Google Scholar]
  • 74.Devalaraja MN, Wang DZ, Ballard DW, Richmond A. Elevated constitutive IkappaB kinase activity and IkappaB-alpha phosphorylation in Hs294T melanoma cells lead to increased basal MGSA/GRO-alpha transcription. Cancer Res. 1999;59:1372–1377. [PubMed] [Google Scholar]
  • 75.Sizemore N, Lerner N, Dombrowski N, Sakurai H, Stark GR. Distinct roles of the Ikappa B kinase alpha and beta subunits in liberating nuclear factor kappa B (NF-kappa B) from Ikappa B and in phosphorylating the p65 subunit of NF-kappa B. J Biol Chem. 2002;277:3863–3869. doi: 10.1074/jbc.M110572200. [DOI] [PubMed] [Google Scholar]
  • 76.Ling L, Cao Z, Goeddel DV. NF-kappaB-inducing kinase activates IKK-alpha by phosphorylation of Ser-176. Proc Natl Acad Sci USA. 1998;95:3792–3797. doi: 10.1073/pnas.95.7.3792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Nakano H, Shindo M, Sakon S, Nishinaka S, Mihara M, Yagita H, Okumura K. Differential regulation of IkappaB kinase alpha and beta by two upstream kinases, NF-kappaB-inducing kinase and mitogen-activated protein kinase/ERK kinase kinase-1. Proc Natl Acad Sci USA. 1998;95:3537–3542. doi: 10.1073/pnas.95.7.3537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Smith C, Andreakos E, Crawley JB, Brennan FM, Feldmann M, Foxwell BM. NF-kappaB-inducing kinase is dispensable for activation of NF-kappaB in inflammatory settings but essential for lymphotoxin beta receptor activation of NF-kappaB in primary human fibroblasts. J Immunol. 2001;167:5895–5903. doi: 10.4049/jimmunol.167.10.5895. [DOI] [PubMed] [Google Scholar]
  • 79.Malinin NL, Boldin MP, Kovalenko AV, Wallach D. MAP3K-related kinase involved in NF-kappaB induction by TNF, CD95 and IL-1. Nature. 1997;385:540–544. doi: 10.1038/385540a0. [DOI] [PubMed] [Google Scholar]
  • 80.Xiao G, Harhaj EW, Sun SC. NF-kappaB-inducing kinase regulates the processing of NF-kappaB2 p100. Mol Cell. 2001;7:401–409. doi: 10.1016/s1097-2765(01)00187-3. [DOI] [PubMed] [Google Scholar]
  • 81.Cogswell PC, Guttridge DC, Funkhouser WK, Baldwin AS., Jr Selective activation of NF-kappa B subunits in human breast cancer: Potential roles for NF-kappa B2/p52 and for Bcl-3. Oncogene. 2000;19:1123–1131. doi: 10.1038/sj.onc.1203412. [DOI] [PubMed] [Google Scholar]
  • 82.Li X, Stark GR. NF-kappaB-dependent signaling pathways. Exp Hematol. 2002;30:285–296. doi: 10.1016/s0301-472x(02)00777-4. [DOI] [PubMed] [Google Scholar]
  • 83.Delhase M, Li N, Karin M. Kinase regulation in inflammatory response. Nature. 2000;406:367–368. doi: 10.1038/35019154. [DOI] [PubMed] [Google Scholar]
  • 84.Koul D, Yao Y, Abbruzzese JL, Yung WK, Reddy SA. Tumor suppressor MMAC/PTEN inhibits cytokine-induced NF-kappaB activation without interfering with the IkappaB degradation pathway. J Biol Chem. 2001;276:11402–11408. doi: 10.1074/jbc.M007806200. [DOI] [PubMed] [Google Scholar]
  • 85.Madrid LV, Mayo MW, Reuther JY, Baldwin AS., Jr Akt stimulates the transactivation potential of the RelA/p65 Subunit of NF-kappa B through utilization of the Ikappa B kinase and activation of the mitogen-activated protein kinase p38. J Biol Chem. 2001;276:18934–18940. doi: 10.1074/jbc.M101103200. [DOI] [PubMed] [Google Scholar]
  • 86.Dhawan P, Singh AB, Ellis DL, Richmond A. Constitutive activation of Akt/protein kinase B in melanoma leads to up-regulation of nuclear factor-kappaB and tumor progression. Cancer Res. 2002;62:7335–7342. [PubMed] [Google Scholar]
  • 87.Celebi JT, Shendrik I, Silvers DN, Peacocke M. Identification of PTEN mutations in metastatic melanoma specimens. J Med Genet. 2000;37:653–657. doi: 10.1136/jmg.37.9.653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Zhou XP, Gimm O, Hampel H, Niemann T, Walker MJ, Eng C. Epigenetic PTEN silencing in malignant melanomas without PTEN mutation. Am J Pathol. 2000;157:1123–1128. doi: 10.1016/S0002-9440(10)64627-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Tsao H, Mihm MC, Jr, Sheehan C. PTEN expression in normal skin, acquired melanocytic nevi, and cutaneous melanoma. J Am Acad Dermatol. 2003;49:865–872. doi: 10.1016/s0190-9622(03)02473-3. [DOI] [PubMed] [Google Scholar]
  • 90.Mayo MW, Wang CY, Cogswell PC, Rogers-Graham KS, Lowe SW, Der CJ, Baldwin AS., Jr Requirement of NF-kappaB activation to suppress p53-independent apoptosis induced by oncogenic Ras. Science. 1997;278:1812–1815. doi: 10.1126/science.278.5344.1812. [DOI] [PubMed] [Google Scholar]
  • 91.Finco TS, Westwick JK, Norris JL, Beg AA, Der CJ, Baldwin AS., Jr Oncogenic Ha-Ras-induced signaling activates NF-kappaB transcriptional activity, which is required for cellular transformation. J Biol Chem. 1997;272:24113–24116. doi: 10.1074/jbc.272.39.24113. [DOI] [PubMed] [Google Scholar]
  • 92.Norris JL, Baldwin AS., Jr Oncogenic Ras enhances NF-kappaB transcriptional activity through Raf-dependent and Raf-independent mitogen-activated protein kinase signaling pathways. J Biol Chem. 1999;274:13841–13846. doi: 10.1074/jbc.274.20.13841. [DOI] [PubMed] [Google Scholar]
  • 93.Millan O, Ballester A, Castrillo A, Oliva JL, Traves PG, Rojas JM, Bosca L. H-Ras-specific activation of NF-kappaB protects NIH 3T3 cells against stimulus-dependent apoptosis. Oncogene. 2003;22:477–483. doi: 10.1038/sj.onc.1206179. [DOI] [PubMed] [Google Scholar]
  • 94.Herlyn M, Satyamoorthy K. Activated ras. Yet another player in melanoma? Am J Pathol. 1996;149:739–744. [PMC free article] [PubMed] [Google Scholar]
  • 95.Castellano M, Parmiani G. Genes involved in melanoma: An overview of INK4a and other loci. Melanoma Res. 1999;9:421–432. [PubMed] [Google Scholar]
  • 96.Pollock PM, Harper UL, Hansen KS, Yudt LM, Stark M, Robbins CM, Moses TY, Hostetter G, Wagner U, Kakareka J, Salem G, Pohida T, Heenan P, Duray P, Kallioniemi O, Hayward NK, Trent JM, Meltzer PS. High frequency of BRAF mutations in nevi. Nat Genet. 2003;33:19–20. doi: 10.1038/ng1054. [DOI] [PubMed] [Google Scholar]
  • 97.Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, Teague J, Woffendin H, Garnett MJ, Bottomley W, Davis N, Dicks E, Ewing R, Floyd Y, Gray K, Hall S, Hawes R, Hughes J, Kosmidou V, Menzies A, Mould C, Parker A, Stevens C, Watt S, Hooper S, Wilson R, Jayatilake H, Gusterson BA, Cooper C, Shipley J, Hargrave D, Pritchard-Jones K, Maitland N, Chenevix-Trench G, Riggins GJ, Bigner DD, Palmieri G, Cossu A, Flanagan A, Nicholson A, Ho JW, Leung SY, Yuen ST, Weber BL, Seigler HF, Darrow TL, Paterson H, Marais R, Marshall CJ, Wooster R, Stratton MR, Futreal PA. Mutations of the BRAF gene in human cancer. Nature. 2002;417:949–954. doi: 10.1038/nature00766. [DOI] [PubMed] [Google Scholar]
  • 98.Troppmair J, Hartkamp J, Rapp UR. Activation of NF-kappa B by oncogenic Raf in HEK 293 cells occurs through autocrine recruitment of the stress kinase cascade. Oncogene. 1998;17:685–690. doi: 10.1038/sj.onc.1201981. [DOI] [PubMed] [Google Scholar]
  • 99.Yin MJ, Christerson LB, Yamamoto Y, Kwak YT, Xu S, Mercurio F, Barbosa M, Cobb MH, Gaynor RB. HTLV-I Tax protein binds to MEKK1 to stimulate IkappaB kinase activity and NF-kappaB activation. Cell. 1998;93:875–884. doi: 10.1016/s0092-8674(00)81447-6. [DOI] [PubMed] [Google Scholar]
  • 100.Chu ZL, DiDonato JA, Hawiger J, Ballard DW. The tax oncoprotein of human T-cell leukemia virus type 1 associates with and persistently activates IkappaB kinases containing IKKalpha and IKKbeta. J Biol Chem. 1998;273:15891–15894. doi: 10.1074/jbc.273.26.15891. [DOI] [PubMed] [Google Scholar]
  • 101.Geleziunas R, Ferrell S, Lin X, Mu Y, Cunningham ET, Jr, Grant M, Connelly MA, Hambor JE, Marcu KB, Greene WC. Human T-cell leukemia virus type 1 Tax induction of NF-kappaB involves activation of the IkappaB kinase alpha (IKKalpha) and IKKbeta cellular kinases. Mol Cell Biol. 1998;18:5157–5165. doi: 10.1128/mcb.18.9.5157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Mosialos G. The role of Rel/NF-kappa B proteins in viral oncogenesis and the regulation of viral transcription. Semin Cancer Biol. 1997;8:121–129. doi: 10.1006/scbi.1997.0063. [DOI] [PubMed] [Google Scholar]
  • 103.La Porta CA, Comolli R. PKC-dependent modulation of IkB alpha-NFkB pathway in low metastatic B16F1 murine melanoma cells and in highly metastatic BL6 cells. Anticancer Res. 1998;18:2591–2597. [PubMed] [Google Scholar]
  • 104.Biswas DK, Dai SC, Cruz A, Weiser B, Graner E, Pardee AB. The nuclear factor kappa B (NF-kappa B): A potential therapeutic target for estrogen receptor negative breast cancers. Proc Natl Acad Sci USA. 2001;98:10386–10391. doi: 10.1073/pnas.151257998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Wilson L, Szabo C, Salzman AL. Protein kinase C-dependent activation of NF-kappaB in enterocytes is independent of IkappaB degradation. Gastroenterology. 1999;117:106–114. doi: 10.1016/s0016-5085(99)70556-1. [DOI] [PubMed] [Google Scholar]
  • 106.Han Y, Meng T, Murray NR, Fields AP, Brasier AR. Interleukin-1-induced nuclear factor-kappaB-IkappaBalpha autoregulatory feedback loop in hepatocytes. A role for protein kinase calpha in post-transcriptional regulation of ikappabalpha resynthesis. J Biol Chem. 1999;274:939–947. doi: 10.1074/jbc.274.2.939. [DOI] [PubMed] [Google Scholar]
  • 107.Anrather J, Csizmadia V, Soares MP, Winkler H. Regulation of NF-kappaB RelA phosphorylation and transcriptional activity by p21(ras) and protein kinase Czeta in primary endothelial cells. J Biol Chem. 1999;274:13594–13603. doi: 10.1074/jbc.274.19.13594. [DOI] [PubMed] [Google Scholar]
  • 108.Wang D, Baldwin AS., Jr Activation of nuclear factor-kappaB-dependent transcription by tumor necrosis factor-alpha is mediated through phosphorylation of RelA/p65 on serine 529. J Biol Chem. 1998;273:29411–29416. doi: 10.1074/jbc.273.45.29411. [DOI] [PubMed] [Google Scholar]
  • 109.Wang D, Westerheide SD, Hanson JL, Baldwin AS., Jr Tumor necrosis factor alpha-induced phosphorylation of RelA/p65 on Ser529 is controlled by casein kinase II. J Biol Chem. 2000;275:32592–32597. doi: 10.1074/jbc.M001358200. [DOI] [PubMed] [Google Scholar]
  • 110.Sakurai H, Chiba H, Miyoshi H, Sugita T, Toriumi W. IkappaB kinases phosphorylate NF-kappaB p65 subunit on serine 536 in the transactivation domain. J Biol Chem. 1999;274:30353–30356. doi: 10.1074/jbc.274.43.30353. [DOI] [PubMed] [Google Scholar]
  • 111.Zhong H, SuYang H, Erdjument-Bromage H, Tempst P, Ghosh S. The transcriptional activity of NF-kappaB is regulated by the IkappaB-associated PKAc subunit through a cyclic AMP-independent mechanism. Cell. 1997;89:413–424. doi: 10.1016/s0092-8674(00)80222-6. [DOI] [PubMed] [Google Scholar]
  • 112.Zhong H, Voll RE, Ghosh S. Phosphorylation of NF-kappa B p65 by PKA stimulates transcriptional activity by promoting a novel bivalent interaction with the coactivator CBP/p300. Mol Cell. 1998;1:661–671. doi: 10.1016/s1097-2765(00)80066-0. [DOI] [PubMed] [Google Scholar]
  • 113.Vermeulen L, De Wilde G, Van Damme P, Vanden Berghe W, Haegeman G. Transcriptional activation of the NF-kappaB p65 subunit by mitogen- and stress-activated protein kinase-1 (MSK1) Embo J. 2003;22:1313–1324. doi: 10.1093/emboj/cdg139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Berman KS, Verma UN, Harburg G, Minna JD, Cobb MH, Gaynor RB. Sulindac enhances tumor necrosis factor-alpha-mediated apoptosis of lung cancer cell lines by inhibition of nuclear factor-kappaB. Clin Cancer Res. 2002;8:354–360. [PubMed] [Google Scholar]
  • 115.May MJ, D’Acquisto F, Madge LA, Glockner J, Pober JS, Ghosh S. Selective inhibition of NF-kappaB activation by a peptide that blocks the interaction of NEMO with the IkappaB kinase complex. Science. 2000;289:1550–1554. doi: 10.1126/science.289.5484.1550. [DOI] [PubMed] [Google Scholar]
  • 116.Yamamoto Y, Gaynor RB. Therapeutic potential of inhibition of the NF-kappaB pathway in the treatment of inflammation and cancer. J Clin Invest. 2001;107:135–142. doi: 10.1172/JCI11914. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Hideshima T, Chauhan D, Richardson P, Mitsiades C, Mitsiades N, Hayashi T, Munshi N, Dang L, Castro A, Palombella V, Adams J, Anderson KC. NF-kappa B as a therapeutic target in multiple myeloma. J Biol Chem. 2002;277:16639–16647. doi: 10.1074/jbc.M200360200. [DOI] [PubMed] [Google Scholar]
  • 118.Tan C, Waldmann TA. Proteasome inhibitor PS-341, a potential therapeutic agent for adult T-cell leukemia. Cancer Res. 2002;62:1083–1086. [PubMed] [Google Scholar]
  • 119.Adams J. Proteasome inhibition: A novel approach to cancer therapy. Trends Mol Med. 2002;8:S49–54. doi: 10.1016/s1471-4914(02)02315-8. [DOI] [PubMed] [Google Scholar]
  • 120.Amiri KI, Horton LW, LaFleur BJ, Sosman JA, Richmond A. Augmenting chemosensitivity of malignant melanoma tumors via proteasome inhibition: Implication for bortezomib (VELCADE, PS-341) as a therapeutic agent for malignant melanoma. Cancer Res. 2004;64:4912–4918. doi: 10.1158/0008-5472.CAN-04-0673. [DOI] [PubMed] [Google Scholar]
  • 121.Banerji A, Chakrabarti J, Mitra A, Chatterjee A. Effect of curcumin on gelatinase A (MMP-2) activity in B16F10 melanoma cells. Cancer Lett. 2004;211:235–242. doi: 10.1016/j.canlet.2004.02.007. [DOI] [PubMed] [Google Scholar]
  • 122.Odot J, Albert P, Carlier A, Tarpin M, Devy J, Madoulet C. In vitro and in vivo anti-tumoral effect of curcumin against melanoma cells. Int J Cancer. 2004;111:381–387. doi: 10.1002/ijc.20160. [DOI] [PubMed] [Google Scholar]
  • 123.Chawla-Sarkar M, Bauer JA, Lupica JA, Morrison BH, Tang Z, Oates RK, Almasan A, DiDonato JA, Borden EC, Lindner DJ. Suppression of NF-kappa B survival signaling by nitrosylcobalamin sensitizes neoplasms to the anti-tumor effects of Apo2L/TRAIL. J Biol Chem. 2003;278:39461–39469. doi: 10.1074/jbc.M306111200. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]

RESOURCES