Hostname: page-component-7bb8b95d7b-s9k8s Total loading time: 0 Render date: 2024-10-04T22:13:26.692Z Has data issue: false hasContentIssue false

NF-κB: a mediator that promotes or inhibits angiogenesis in human diseases?

Published online by Cambridge University Press:  28 July 2023

Yijing Jiang
Affiliation:
Department of Pathophysiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, Jiangsu, People's Republic of China
Jie Zhang
Affiliation:
Department of Oncology, Affiliated Tumor Hospital of Nantong University, 30Tongyang North Road, Pingchao Town, Nantong 226361, Jiangsu, People's Republic of China
Conglin Shi
Affiliation:
Department of Pathogenic Biology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, Jiangsu, People's Republic of China
Xingjuan Li
Affiliation:
Department of Pathophysiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, Jiangsu, People's Republic of China
Yongying Jiang
Affiliation:
Department of Pathophysiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, Jiangsu, People's Republic of China
Renfang Mao*
Affiliation:
Department of Pathophysiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, Jiangsu, People's Republic of China
*
Corresponding author: Renfang Mao; Email: maorenfang@ntu.edu.cn
Rights & Permissions [Opens in a new window]

Abstract

The nuclear factor of κ-light chain of enhancer-activated B cells (NF-κB) signaling pathway, which is conserved in invertebrates, plays a significant role in human diseases such as inflammation-related diseases and carcinogenesis. Angiogenesis refers to the growth of new capillary vessels derived from already existing capillaries and postcapillary venules. Maintaining normal angiogenesis and effective vascular function is a prerequisite for the stability of organ tissue function, and abnormal angiogenesis often leads to a variety of diseases. It has been suggested that NK-κB signalling molecules under pathological conditions play an important role in vascular differentiation, proliferation, apoptosis and tumourigenesis by regulating the transcription of multiple target genes. Many NF-κB inhibitors are being tested in clinical trials for cancer treatment and their effect on angiogenesis is summarised. In this review, we will summarise the role of NF-κB signalling in various neovascular diseases, especially in tumours, and explore whether NF-κB can be used as an attack target or activation medium to inhibit tumour angiogenesis.

Type
Review
Copyright
Copyright © The Author(s), 2023. Published by Cambridge University Press

Introduction

Nuclear factor-κB (NF-κB) is a highly conserved multifunctional transcription factor family, which has been widely studied due to its different roles in the development of tumours. NF-κB includes five members, namely REL (c-REL), RELA (p65), RELB, NFκB1 (p50) and NFκB2 (p52) (Ref. Reference Sun, Chang and Jin1). They form different homodimers or heterodimers involved in transcriptional regulation of many genes and play roles in immune, inflammation, oxidative stress, angiogenesis, tumour formation, cell proliferation and apoptosis (Refs Reference Bottex-Gauthier2, Reference Zhang, Lenardo and Baltimore3, Reference Taniguchi and Karin4). Angiogenesis is the formation of new blood vessels from the existing vascular system, which is an integral part of normal and pathological processes (Refs Reference Delgado5, Reference Geudens and Gerhardt6). Blood vessels are stationary or dormant in mature organisms, but still maintain important homeostasis. Only under certain physiological or pathological stimuli can they be reactivated. Angiogenesis is a key process that leads to neovascular diseases and required by tumour growth and metastasis. This is a complex and highly ordered process that relies on a broad signal transduction network between and within endothelial cells (ECs), related parietal cells (vascular smooth muscle cells and pericytes) and other cell types (such as immune cells) (Refs Reference Polyak, Haviv and Campbell7, Reference Polyak and Weinberg8, Reference Quail and Joyce9). In recent years, the molecular and cellular mechanisms involved in the regulation of angiogenesis and vascular homeostasis have been extensively studied. In addition to the common Notch signalling (Refs Reference Rehman and Wang10, Reference Fernández-Chacón11), studies in the past decades have revealed that NF-κB signalling pathway is involved in regulating angiogenesis and intravascular homeostasis under pathological conditions (Refs Reference Zhou12, Reference Noort13, Reference Ashander14). However, the mechanism and treatment strategy of NF-κB in human vascular diseases, especially cancer, are still controversial. This review covers the general theme of the biological role and mechanism of NF-κB in vascular-related diseases, and explores how to use NF-κB as a therapeutic target or mediator for diseases.

Structures of NF-κB and IκB proteins

NF-κB is a protein with multiple transcriptional regulation. NF-κB can be found in almost all mammalian cells, which regulates the production of a variety of encoding cytokines, growth factors, cell adhesion molecules and some acute protein factors, and plays an important role in inflammatory response, stress response, activation, proliferation, differentiation, apoptosis and tumour formation of immune cells (Refs Reference Bottex-Gauthier2, Reference Zhang, Lenardo and Baltimore3, Reference Taniguchi and Karin4). In mammals, NF-κB superfamily consists of five transcription factors: REL (c-REL), RELA (p65), RELB, NFκB1 (p50) and NFκB2 (p52) (Ref. Reference Sun, Chang and Jin1). Their N-terminal region has a highly conserved Rel homology domain (RHD). RHD is composed of N-terminal domain and C-terminal domain (CTD), and there is a nuclear-localisation sequence (NLS) on CTD, which is responsible for DNA binding, dimerisation and nuclear translocation. According to different structures, functions and synthesis methods, NF-κB superfamily is usually divided into two subfamilies: NF-κB subfamily (p50 and p52) and REL subfamily (c-REL, RELA and RELB). NF-κB subfamily is formed by the cleavage of precursor proteins p105 and p100 containing C-terminal ankyrin repeat domains (ARDs), respectively. These proteins contain RHD, but lack transcriptional activity region and unable to independently activate gene transcription unless they form heterodimeric complexes with REL subfamily proteins (Refs Reference Ghosh15, Reference Smale16). In addition to the N-terminal RHD, the C-terminal of REL subfamily contains transcriptional activation domain, rendering them capable of activating gene transcription directly. Except for RELB, other members of NF-κB can form all combinations of heterodimers and homodimers (Ref. Reference Neumann and Naumann17). However, RELB can only form heterodimers composed of p50 or p52. Whether an NF-κB dimer activates or inhibits transcription depends on the binding DNA region and the interaction with other transcription factors. NF-κB transcription factors can also play an indirect role in DNA regulatory elements when interacting with other DNA-binding transcription factors (Refs Reference Mulero18, Reference Ji19).

IκB is an important protein regulating the activity of NF-κB dimer, with 5–8 ARDs composed of 30 amino acids. The activation of NF-κB depends on the degradation of IκB protein. The IκB protein family has nine members including IκBα, IκBβ, IκBɛ, IκBζ, IκBη, Bcl-3, IκBNS, p100 and p105 (Ref. Reference Schuster20). The ARD at the N-terminal of IκB can bind to the RHD at the C-terminal of NF-κB dimer, covering the NLS of NF-κB to prevent NF-κB from moving into the nucleus (Refs Reference Sun21, Reference Karin and Greten22). The proline-glutamate-serine-threonine-rich motif at the C-terminal of IκB mainly maintains its structural stability and inhibits NF-κB binding to DNA (Refs Reference Majdalawieh and Ro23, Reference Ferreiro and Komives24). IκB kinase (IKK) complex phosphorylates the inhibitory protein IκBα, then IκBα is ubiquitinised and degrades by proteasome-dependent pathway. The released NF-κB is transcribed into the nucleus to activate the target gene (Refs Reference Karin and Greten22, Reference Chen and Greene25, Reference Israël26).

Activation of NF-κB

A large number of experiments have shown that a variety of factors could cause the activation of NF-κB signal transduction pathway. There are two widely accepted pathways: the canonical and non-canonical pathway, which are driven by the degradation of IκB or the induction of NF-κB-induced kinase (NIK), respectively (Fig. 1).

Figure 1. Activation of canonical and non-canonical NF-κB pathways. The canonical NF-κB pathway (left) is triggered by a variety of inflammatory factors and mediated by activation of the IKK complex, leading to transient nuclear translocation of NF-κB heterodimers. The non-canonical NF-κB pathway (right) is stimulated by specific receptors, dependent on NIK and IKKα, and mediates the sustained activation of the RelB/p52 complex.

IKK is composed of a large protein kinase complex with a size of 700–900 kDa, including three subunits, namely, two homologous catalytic subunits IKKα (IKK1) and IKKβ (IKK2) and an essential auxiliary subunit IKKγ (NEMO) with regulatory function (Ref. Reference Vallabhapurapu and Karin27). These two catalytic kinases, IKKα and IKKβ, are involved in different signalling pathways of NF-κB. The canonical NF-κB activation pathway is mainly regulated by IKKβ, while the regulation of the non-canonical NF-κB activation pathway is completely dependent on IKKα. Actually, the oligomers formed by IKKα/β/γ are found to affect the activity of IKKβ (Refs Reference Israël26, Reference Ruland28). IKKα can phosphorylate Ser32 and Ser36 on IκBα, while IKKβ not only phosphorylates Ser32 and Ser36 on IκBα, but also phosphorylates Ser19 and Ser23 on IκBβ. IKKγ with regulatory function has two extended α-helix regions and a zinc finger domain. Although IKKγ has no catalytic activity, the activity of IKK in canonical NF-κB pathway depends on the integrity of IKKγ subunits (Refs Reference Rothwarf and Karin29, Reference May, Marienfeld and Ghosh30, Reference Karin and Delhase31).

In the resting state of cells, p50/p65 or p50/c-REL in the cytoplasm binds to IκB to form a trimer, so that NF-κB dimer cannot be translocated. In the canonical NF-κB pathway, after the cells are stimulated by extracellular signals such as pro-inflammatory cytokines, lipopolysaccharide (LPS), growth factors and antigen receptors, IKKβ subunit is phosphorylated and activated, and then the Ser32 and Ser36 sites of IκBα are phosphorylated. The phosphorylated IκBα is further ubiquitinised and degraded in the 26S protein hydrolase complex. The released p50/p65 or p50/c-REL is transported into the nucleus to induce the expression of target genes (Ref. Reference Zhang32). The activation of canonical NF-κB pathway usually occurs in the early and transient process of immune activation (Ref. Reference Pires33).

Compared with the canonical NF-κB pathway, the non-canonical NF-κB pathway activators are relatively less, mainly including ligands for some members of the tumour necrosis factor (TNF) receptor family, such as lymphotoxinβ receptor (LTβR), nuclear factor receptor activator B receptor activator (RANK), CD40 and B-cell activating factor family (Refs Reference Cildir, Low and Tergaonkar34, Reference Maubach35, Reference Sun36, Reference Bonizzi and Karin37, Reference Vallabhapurapu38). In resting cells, TRAF2/3 recruits NIK into the complex, and subsequently cIAP1/2 ubiquitinates NIK through the ubiquitination of K48 connection, leading to the degradation of NIK protein. After extracellular signal stimulation, the TRAF2/TRAF3/cIAP1/cIAP2 complex is degraded, which prevents NIK protein degradation. The phosphorylation of IKKα complex is induced by NIK, thereby further activating p100, resulting in phosphorylation-dependent cleavage of p100. Subsequently, the active p52/RelB complex enters the nucleus and binds to the target gene to regulate gene expression (Ref. Reference Senftleben39). The activation of NIK is the key to initiate the downstream pathway, so the activation of non-canonical NF-κB pathway is slower but lasts longer. The non-canonical NF-κB pathway is mainly involved in long-term cellular processes, such as chronic inflammation (Refs Reference Vallabhapurapu and Karin27, Reference Sun36).

NF-κB-related microRNA in inflammation and angiogenesis

Due to the wide range of biological functions of this protein, the role of NF-κB in the development of human diseases has received increasing attention. Many miRNAs have been reported to be involved in inflammation and angiogenesis. MiRNAs are a class of non-coding single-stranded RNA molecules with a length of about 22 nucleotides, which play a regulatory role in target mRNA by damaging the stability of target mRNA and inhibiting the translation of target mRNA. Some of the miRNAs related to NF-κB have been implicated in inflammation and angiogenesis (Fig. 2). One group of miRNAs such as miR-17 and miR-31 in response to TNF-α inhibit intercellular adhesion molecule-1 (ICAM-1) expression and regulate angiogenesis by inhibiting leukocyte aggregation (Ref. Reference Doebele40). In addition, miR-31 was shown to promote the migration of ECs induced by Kaposi's sarcoma-associated herpes virus, while miR-17 had dual effects on angiogenesis in different tumours (Refs Reference Liu41, Reference Tsoyi42, Reference Sabatel43, Reference Zhu44). Another group of non-coding RNA upregulated by TNF-α, including miR-21, miR-221 and miR222, were proved to have anti-angiogenesis effect. In addition to inhibitory effect, miR-21 can also indirectly upregulate the expression of vascular cell adhesion molecule-1(VCAM-1) via targeting PTEN and promote inflammatory cell infiltration. In prostate cancer, miR-21 might be thought to promote angiogenesis (Refs Reference Ueda45, Reference Hu46). However, the increase of pre-miR-21 in the vitreous reduces the angiogenic response in the choroidal neovascularisation mouse model (Ref. Reference Suárez47). On the other hand, the expression of VCAM-1 stimulated by angiotensin II is partially downregulated by overexpressed miR-221 and miR-222, suggesting that these miR RNAs were involved in anti-inflammatory responses (Ref. Reference Poliseno48). They also target ICAM-1 and participate in the regulation of inflammatory cell infiltration (Refs Reference Zhu49, Reference Cheng50). Moreover, miR-221 and miR-222 were evidenced to inhibit EC proliferation and migration and downregulate endothelial nitric oxide synthases (eNOS) and c-kit expression (Refs Reference Sun51, Reference Fasanaro52). Similarly, miR-146, upregulated in ECs in response to NF-κB, represses endothelial activation by negatively regulating the expression of ICAM1, VCAM1 and E-selectin. However, there is opposite evidence that miR-146 also promotes angiogenesis by upregulating eNOS (Refs Reference Qi53, Reference Maijer54). Contrary to miR-21, miR-146, miR-221 and miR-222, the expression of miR-181b in ECs is downregulated by TNF-α treatment. MiR-181b inhibits NF-κB response genes such as VCAM-1 and E-selectin by targeting importin-α3 required for NF-κB nuclear translocation (Ref. Reference Madge55). Available data convinced that decreased oxygen partial pressure could induce miR-210 expression in ECs through HIF-1α-dependent pathway (Refs Reference Brown, Claudio and Siebenlist56, Reference Tak and Firestein57). MiR-210, known as master hypoxamiR, can be induced by HIF-1α to regulate a series of intracellular functions in response to hypoxic stress, thereby promoting cell survival and differentiation. On one hand, overexpression of MIR-210 promotes the migration of EC and increased the formation of capillary-like structure under hypoxia. On the other hand, miR-210 inhibits p50 protein and negatively regulated the expression of angiogenesis-promoting cytokines induced by NF-κB, such as IL-6 and TNF-α.

Figure 2. Multiple mediators are shared in the molecular pathways of pro-inflammatory response and angiogenesis. TNF-α induces NF-κB signalling pathway to initiate inflammatory response and HIF-1α transcription factor involved in angiogenesis is regulated by NF-κB under hypoxic condition. MicroRNAs regulate inflammation-related angiogenesis by acting on the downstream signalling molecules in NF-κB-mediated pro-inflammatory responses. In addition, the Nrf2/HO-1 axis that interacts with HIF-1α can regulate hypoxia-induced angiogenesis by targeting VEGF.

Apart from the microRNA in inflammation-related angiogenesis, the transcription factors in NF-κB are also closely related to inflammatory angiogenesis. Noort et al. reported that non-canonical NF-κB signal could regulate inflammation-induced angiogenesis (Refs Reference Noort13, Reference Suárez58). They demonstrated that NIK was highly expressed in inflammatory synovial ECs of rheumatoid arthritis (RA). In synovial ECs, NIK upregulates the expression of C-X-C motif chemokine 12 (CXCL12) through non-canonical NF-κB signalling pathway, which promoted pathological angiogenesis (Ref. Reference Tsai59). Thus, NIK is considered to be one of the key regulators of angiogenesis induced by synovitis in RA (Refs Reference Wu60, Reference Shan61).

Mechanisms of the constitutive activation of NF-κB in different cancers

A proper understanding of NF-κB regulatory mechanisms would certainly help to clarify its role in diseases, especially in tumoural progression. In most cancers, activation of NF-κB is enhanced and this structural activation occurs in a large number of different cancers through several mechanisms. Firstly, NF-κB activity can be activated by mutations in tumour-associated genes, a form generally associated with haematological malignancies (Refs Reference Vrábel, Pour and Ševčíková62, Reference Eluard63, Reference Lim, Yang and Staudt64). These mutations, which affect canonical or non-canonical NF-κB signalling, can occur in both core NF-κB pathway proteins and upstream regulatory proteins of NF-κB. On the one hand, the NF-κB transcription factor itself is the target of mutation, among them REL, NFκB1 and NFκB2 are more frequently mutated. Oncogenic mutations in NF-κB signalling proteins result in C-terminal truncations or single amino acid mutations, which enhance NF-κB activity (Ref. Reference Courtois and Gilmore65). Complete loss-of-function mutation of the IκB protein results in chronic nuclear localisation of NF-κB in glioblastoma, diffuse large B-cell lymphoma (DLBCL), Hodgkin's lymphoma and nasopharyngeal carcinoma (Refs Reference Lake66, Reference Kalaitzidis67, Reference Bredel68, Reference Zheng69). On the other hand, the most common mutations that enhance NF-κB signalling in DLBCL and multiple myeloma may occur in upstream molecules or downstream target genes of specific signalling pathways. Continued activation of canonical or non-canonical NF-κB driven by these molecular mutations is required for the proliferation and survival of malignant B cells (Refs Reference Vrábel, Pour and Ševčíková62, Reference Matthews70, Reference Young71). Secondly, cytokines or other stimulus-dependent activations of the NF-κB pathway can increase the constitutive activation of NF-κB via an autocrine mechanism. For example, TNF-α, encoded by NF-κB target gene TNFA, is a strong activator of NF-κB and acts in an autocrine manner on some tumour cells with constitutive activation of NF-κB (Ref. Reference Schmitz72). Thirdly, epigenetic status or epigenetic regulatory factors interact with the chronically active NF-κB found in many cancers, which makes the induced NF-κB preferentially bind to the genomic position that initiates epigenetics. NF-κB can also recruit chromatin modification coactivators such as histone acetyltransferase and histone deacetylase to further modify local chromatin (Refs Reference Mulero18, Reference Bhatt and Ghosh73, Reference Dawson and Kouzarides74). In addition, mutations in non-coding sequences cause carcinogenesis by affecting transcription and gene expression (Ref. Reference Patel and Wang75). For example, promoter sequence mutations affecting the expression of NF-κB and its target genes have been found in oral cancer, breast cancer and bladder cancer (Refs Reference Lin76, Reference Kang77). Finally, several oncogenic human viruses also affect NF-κB signal transduction. These viruses replicate and cause cancer by chronic NF-κB-dependent inflammation or the sustained activity NF-κB viral activators (Refs Reference Costa RM78, Reference Charostad79, Reference Harhaj and Giam80, Reference Shokri81).

The effect of NF-κB in different aspects of cancer progression

Tumour cell apoptosis progression affected by the activation of NF-κB

In tumour cells, activation of NF-κB usually results in inhibition of apoptosis. NF-κB target genes are known to encode anti-apoptotic molecules, such as BCL2, BCLXL and apoptosis suppressors. Increased expression of anti-apoptotic target genes is detected in a variety of tumour cells with constitutive NF-κB activity (Ref. Reference Gilmore82). Furthermore, the activated NF-κB increases the number of DNA damage cells that accumulate carcinogenic mutations by preventing P53-dependent apoptosis (Ref. Reference Gudkov, Gurova and Komarova83). In addition to anti-apoptosis, there are upregulated target genes related to cell proliferation in NF-κB signalling pathway, such as cyclin D1/D2/D3. Moreover, NF-κB also affects cell proliferation by regulating key enzymes. A variety of studies have shown that the activation of NF-κB in inflammatory immune cells stimulates the expression of inflammatory cytokines such as TNF-α and IL-6, which makes tumour cells and tumour stromal cells proliferate (Refs Reference Cerhan84, Reference Becker85). Additionally, it has been proved that NF-κB is a necessary downstream pathway for RAS oncogene activation that causes cell proliferation (Refs Reference Bassères86, Reference Finco87).

Cancer stem cells regulated by NF-κB in various cancers

NF-κB is involved in the functional regulation of cancer stem cells (CSCs). Studies have found that NF-κB is structurally activated in leukaemia, glioblastoma, breast cancer, ovarian cancer, colon cancer and other CSCs, which mediates tumour inflammation, cell proliferation, survival and maintenance (Ref. Reference Vazquez-Santillan88). In breast cancer, NF-κB activation mediated by Notch signalling pathway induces CSC proliferation, and the highly expressed NIK can induce the activation of the non-canonical NF-κB pathway to regulate CSC self-renewal and metastasis (Refs Reference Yamamoto89, Reference Vazquez-Santillan90). In ovarian cancer, CD44+ CSCs enhance the activity of p50/RelA dimer by upregulating core NF-κB signalling protein, thereby promoting self-renewal and metastasis of tumour cells (Ref. Reference Gonzalez-Torres91). In pancreatic cancer, the expression of transcription factor Sry-related HMG box 9 is enhanced by activating the typical NF-κB pathway, resulting in increased CSCs and enhanced invasiveness (Ref. Reference Sun92). In colorectal cancer, inflammatory mediator prostaglandin E2 promotes the formation, survival, proliferation and metastasis of colorectal CSCs by activating NF-κB pathway. The transcription factor Foxp3, however, inhibits the expression of the NF-κB target gene, which is associated with decreased colorectal CSCs and invasiveness (Refs Reference Liu93, Reference Wang94).

NF-κB and tumour microenvironment involved in cancer progression

The development and metastasis of tumours are inseparable from the tumour microenvironment composed of multiple stromal cells, tumour cells, cytokines, chemokines and so on. TNF-α is one of the most common tumour-promoting cytokines, mainly produced by activated neutrophils and macrophages, inducing the expression of other proinflammatory cytokines and promoting tumour progression (Refs Reference Balkwill95, Reference Grivennikov and Karin96). IL-6 is a cytokine dependent on NF-κB in tumour microenvironment, which is mainly produced by malignant tumour cells and activated immune cells. The activation of IL-6 is related to inflammatory tumour and stem cell amplification (Refs Reference Taniguchi and Karin97, Reference Chang, Daly and Bromberg98). Another important cytokine is transforming growth factor-β (TGF-β), which can induce naive T cells to differentiate into Treg and TH17 cells, and enhance tumour metastasis and invasion (Refs Reference Meulmeester and Ten Dijke99, Reference Grivennikov, Greten and Karin100). Besides cytokines, tumourigenesis is also associated with NF-κB-driven chemokine in the tumour microenvironment. The chemokines induced by NF-κB recruit more immune cells, thereby stimulating the inflammatory response, and play an important role in promoting tumour growth and metastasis, synthesising growth factors and inducing angiogenesis. For instance, in human and mouse pancreatic tumour models, CCR5/CCL5 signal interference reduces the migration of Treg cells to tumour microenvironment, indicating that chemokine chemokine (C-C motif) ligand 5 (CCL5) is necessary for Treg cell migration (Ref. Reference Tan101). CCL5 induces the formation of eukaryotic initiation factor 4F translation initiation complex and upregulates the expression of cyclin D1, c-Myc and anti-cell death defence protein-1, thereby promoting the proliferation of breast cancer cells (Ref. Reference Murooka, Rahbar and Fish102). Meanwhile, macrophages are widely enriched in tumour microenvironment and activate cytokines by producing inflammatory factors, chemokines and proteases such as cysteine cathepsins, thereby promoting tumour progression (Refs Reference Quail and Joyce9, Reference Grivennikov, Greten and Karin100). Since M1 macrophages polarized to M2 macrophages through IL-1R and MyD88 requires the participation of the canonical NF-κB signalling pathway, inhibition of the NF-κB can increase the production of M1 macrophages with anti-tumour cytotoxicity (Ref. Reference Hagemann103). Dendritic cell (DC) is known to be the most powerful antigen-presenting cell and plays an important role in anti-tumour immunity. The experiment showed that PD-1 inhibited the anti-tumour function of DCs by inactivating NF-κB in infiltrating DCs of ovarian tumours (Refs Reference Karyampudi104, Reference Antonangeli105). In natural killer cells with antitumor activity, the expression of cytotoxicity effector is also affected by NF-κB (Ref. Reference Vivier106). In tumour immunity, Treg cells can secrete a variety of inhibitory cytokines, limiting the ability of the immune system to kill tumours and allowing tumour cells to escape immune surveillance. The maturation and maintenance of immune tolerance of Tregs require the involvement of p65 and Rel (Refs Reference Oh107, Reference Isomura108, Reference Evaristo109). It ss well known that T-cell receptor and B-cell receptor are one of the important stimuli that induce the activation of NF-κB. T and B cells regulated by NF-κB pathway play a pro-tumour or anti-tumour role in the tumour microenvironment (Ref. Reference Ward, Gubin and Schreiber110). Cancer-associated fibroblasts (CAFs) are one of the most important components in the tumour microenvironment and play an essential role in the development of tumours. They have multiple functions, including extracellular matrix (ECM) remodelling, metabolism, angiogenesis regulation and interactions with cancer cells and invasive immune cells through the production of growth factors, cytokines and chemokines (Refs Reference Kalluri111, Reference Koliaraki112). In the early stage of tumourigenesis, tumour-derived IL-1β induces CAF activation and promotes tumour inflammation in a NF-κB-dependent manner (Ref. Reference Erez113). Through the study of pancreatic cancer, it was found that CAF promoted the production of cytokines such as GM-κ and IL-6 with the participation of NF-κB, which induced extracellular metastasis and paracrine of tumour cells (Ref. Reference Santolla114).

The relationship between inflammation and tumour has been confirmed in cancer research. Chronic inflammation leads to the occurrence and development of malignant tumours by producing a large number of cytokines and recruiting a variety of immunosuppressive cells (Ref. Reference Taniguchi and Karin4). The activation of NF-κB signalling pathway is crucial in regulating inflammatory response. Early studies have shown that NF-κB plays a key role in inflammation-driven colitis-related cancer and hepatocellular carcinoma (Refs Reference Greten115, Reference Pikarsky116). In the mouse model of colitis-related cancer, the loss of IKKβ inhibits the occurrence of colon inflammation and colitis-related cancer. Some of the viruses, such as hepatitis B virus, Kaposi sarcoma herpesvirus and human papillomavirus, increase cell stress response and recruit inflammatory immune cells through chronic NF-κB-dependent inflammatory reactions, thereby leading to malignant tumours. There are also some viruses that directly encode proteins that activate the NF-κB signalling pathway (Refs Reference Costa RM78, Reference Charostad79, Reference Karin117). For example, in B-cell lymphoma and nasopharyngeal carcinoma, EB virus through the activation of NF-κB encodes the protein of LMP1 as a CD40 homologue to prevent the apoptosis of B cells infected by EB virus (Ref. Reference Charostad79). Another mechanism by which NF-κB stimulates tumourigenesis is to induce DNA damage. Chronic inflammation and NF-κB can not only cause DNA damage and carcinogenic mutations by promoting the production of reactive oxygen species and reactive nitrogen, but also lead to chromosomal instability, aneuploidy and epigenetic changes (Refs Reference DiDonato, Mercurio and Karin118, Reference Joyce119, Reference Kiraly120).

The potential tumour suppressor function of NF-κB

Despite the overwhelming evidence that NF-κB is oncogenic, studies have revealed its potential for tumour suppressor function. Since NF-κB can induce the expression of a variety of proapoptotic molecules, such as Bcl-xs, Bax, Fas or FasL, NF-κB is considered to promote apoptosis and may be a tumour suppressor gene (Ref. Reference Dutta121). In the epithelial tissue, downregulated NF-κB by overexpression of IκBα leads to skin epidermis hyperplasia in mice, indicating that NF-κB activation is important for cell growth inhibition. RelA-deficient mouse cell lines have a weak transformation phenotype, and RelA-deficient fibroblasts in immunodeficient mice have the ability to form tumours, indicating that RelA has tumour inhibitory activity in some cases, and RelA complex is involved in the control of various cell characteristics related to tumourigenesis (Ref. Reference Gapuzan, Yufit and Gilmore122). The anti-tumour function of NF-κB can also be achieved by participating in the activation of conventional T cells via the canonical pathway, which is necessary for CD8+T cell proliferation and anti-tumour immune response (Ref. Reference Gerondakis123). Studies have shown that LTβR-mediated non-canonical NF-κB activation induces the expression of adhesion molecules and chemokines, and ultimately forms tertiary lymphoid tissues. It is worth noting that the presence of tertiary lymphoid tissues in the tumour microenvironment generally suggests a better prognosis (Refs Reference Lin124, Reference Furtado125, Reference Luther126, Reference Sautès-Fridman127). Moreover, there is a complicate cross-signalling between NF-κB and wild-type/mutant tumour suppressor gene p53, which indicates that the NF-κB pathway is possibly involved in the mechanism of tumour suppression (Ref. Reference Carrà128). Although the above studies have shown that NF-κB may inhibit tumours by promoting apoptosis, enhancing immune function and regulating tumour microenvironment, there is still no report clearly indicating the inhibitory effect of NF-κB on different cancers in clinical research. Based on the current research, the activation of NF-κB has the potential to promote the occurrence and development of haematological tumours and most solid tumours including glioblastoma, nasopharyngeal carcinoma, breast cancer, ovarian cancer, colon cancer and pancreatic cancer.

NF-κB signalling pathway promotes pathological angiogenesis by activating endothelial cells and stimulating basement membrane degradation

Combined with the above reports, NF-κB signalling pathway plays an important role in the regulation of pathological angiogenesis, especially tumour angiogenesis (Fig. 3). Kei et al. found that in colorectal cancer, the abnormal activation of NF-κB signal was positively correlated with the increased number of pathological angiogenesis (Ref. Reference Sakamoto129). In fact, the positive effect of NF-κB on tumour angiogenesis is closely related to inflammatory response. NF-κB activated by inflammatory cytokines such as TNF, IL-1 and IL-6 can induce the expression of various pro-inflammatory genes and adhesion molecules in ECs, which in turn promoted the accumulation of inflammatory cells in perivascular matrix and thus enhanced the activation of NF-κB (Ref. Reference Ashander14). Meanwhile, in ECs activated by inflammatory factors, NF-κB can be phosphorylated and nuclear translocated, which promotes the transcription of many angiogenesis genes. Vascular endothelial growth factor (VEGF) is considered as a key medium for angiogenesis in tumours. Under the regulation of activated NF-κB, ECs, macrophages and tumour cells were shown to secrete VEGF, which was also one of the NF-κB-regulated target genes (Refs Reference Lennikov130, Reference Shen131). VEGFR2 is the main receptor of VEGF and the signal pathway activated by VEGF/VEGFR2 interaction plays a leading role in angiogenesis. It was found that NF-kB binds directly to the upstream promoter of VEGFR2 and activated its transcription. Once activated VEGFR2 binds to VEGF, VEGFR2 would activate a series of downstream mediators to promote angiogenesis (Ref. Reference Dong132).

Figure 3. The positive regulation mechanism of NF-κB on angiogenesis by transcription target gene. Under the action of inflammatory cytokines, NF-κB induces the expression of various pro-inflammatory genes and adhesion molecules in endothelial cells, which in turn promoted the accumulation of inflammatory cells in perivascular matrix and enhanced the activation of NF-κB. Activated NF-κB can not only make endothelial cells directly secrete VEGF, but also bind to the upstream promoter of VEGFR2 and activate its transcription. Once VEGFR2 binds to vascular endothelial growth factor, VEGFR2 activates a series of downstream mediators to promote angiogenesis. In addition, activated NF-κB can stimulate the expression of matrix metalloproteinases, promote the degradation of basement membrane and extracellular matrix. Nuclear transcription of NF-κB also upregulates inflammatory chemokines IL-6 and IL-8, thereby stimulating the proliferation and migration of tumour endothelial cells. Meanwhile, activated NF-κB can promote tumour angiogenesis by upregulating MCP-1 and COX-2. In non-canonical NF-κB pathway, NIK upregulates the expression of chemokine CXCL12, which plays an important role in the homing of endothelial progenitor cells.

It is well known that one of the key steps of tumour angiogenesis is the degradation of vascular basement membrane and ECM. The activated NF-κB can stimulate the expression of multiple matrix metalloproteinases, including MMP2, MMP3 and MMP9, thereby promoting the degradation of basement membrane and ECM (Refs Reference Mountain133, Reference Popov134, Reference Ko135). In addition, the study also found that the nuclear transcription of NF-κB upregulates inflammatory chemokine IL-8, which can activate ECs of tumour vessels and induce tumour-associated macrophages to secrete additional growth factors to promote tumour invasion and migration. Interestingly, IL-8 also induces NF-κB activation by binding to its homologous receptor C-X-C motif chemokine receptor 2 (CXCR2), which upregulates VEGF gene and protein levels in ECs, leading to autocrine activation of VEGFR2 (Refs Reference Martin, Galisteo and Gutkind136, Reference Passaro137). In addition, the activation of NF-κB can also upregulate the expression levels of angiogenesis factors, including MCP-1 and COX-2. MCP-1 stimulates tumour infiltrating immune cells to secrete multiple angiogenesis-related factors by recruiting macrophages into tumour microenvironment (Ref. Reference Chen138). COX-2 promotes tumour growth by upregulating the production of multiple angiogenesis-related proteins (Ref. Reference Chen and Liu139). Wang et al. found that in colorectal cancer, B7-H3 could upregulate the expression of VEGF-A and promote angiogenesis by activating the NF-κB pathway. However, NF-κB interacting long non-coding RNA inhibits EC proliferation, migration and angiogenesis by inhibiting the secretion of IL-6 via NF-κB signalling pathway in breast cancer (Ref. Reference Luo140). In prostate cancer, NF-κB inactivation significantly inhibits the expression of proangiogenic molecules, including VEGF, IL-8 and MMP-9 in vitro and in vivo, thereby reducing tumour angiogenesis (Ref. Reference Huang141). NF-κB has also been implicated in epithelial-mesenchymal transition (EMT) in tumour cells. NF-κB can regulate EMT transcription factors, mainly including Twist, zinc finger transcription factor Snail2 (Slug) and Smad interaction protein 1, which initiate EMT and exert the ability of self-renewal by enhancing the stemness and invasiveness of cancer cells (Refs Reference Pires142, Reference Scheel and Weinberg143). In addition to EMT transcription factors, inhibition of NF-κB activity leads to decreased activity of various substrate degradation enzymes that promote EMT. In the progression model of breast cancer, IKK-β/IκBα/NF-κB pathway is necessary to induce and maintain EMT, so the activation of NF-κB is crucial to the metastasis potential of epithelial cells in breast cancer (Ref. Reference Huber144). Since MMPs induced by NF-κB stimulate the production of TGF-β, which promotes EMT process and tissue hypoxia, and ultimately lead to tumour cell metastasis and proliferation, TGF-β-induced EMT is at least partially dependent on NF-κB (Refs Reference Malki145, Reference Balkwill146). Additionally, NF-κB combined with some cytokines can directly target metastasis-related genes to control tumour cell motility.

It is noteworthy that under hypoxic environment, hypoxia-inducible factor-1α (HIF-1α) pathway plays an important auxiliary role in the regulation of inflammation-related angiogenesis genes (Refs Reference Cummins147, Reference Oliver, Taylor and Cummins148). In standard conditions, prolyl hydroxylases (PHDs), the main protease that degrades HIF in vivo, hydroxylates IKKβ to make it inactive, resulting in the inactivation of NF-κB in cytoplasm. In hypoxic tumour microenvironment, however, hypoxia causes PHDs to fail, and IKKβ activity cannot be inhibited, which leads to NF-κB activation and released to the nucleus. Since the NF-κB binding site is located in the promoter region of HIF-1α, activated NF-κB can directly regulate the expression of HIF-1α (Fig. 4) (Refs Reference Bonello149, Reference Frede150, Reference Jung151). In the ECs of renal ischaemia/reperfusion model, NF-κB induced by LPS regulates the expression of HIF-2α, which increases the production of NO, thereby improving renal microvascular perfusion and reducing ischaemic renal tubular injury (Fig. 4) (Ref. Reference He152). Another important mechanism involved in the hypoxia-related angiogenesis is NF-E2-related factor 2 (Nrf2)/haeme oxygenase-1 (HO-1) pathway. As a key transcription factor involved in cell response to oxidative and electrophilic stress, Nrf2 plays an important role in inducing antioxidant response (Ref. Reference Bellezza153). HO-1 is one of the most important downstream regulatory products of Nrf2, which can catalyse haeme to produce biliverdin, carbon monoxide and iron. Biliverdin is converted into bilirubin by biliverdin reductase, both of which are powerful free radical scavengers in the body (Ref. Reference Maines154). Under hypoxic conditions, HIF-1α and Nrf2 stress response pathways exist in a complex, interacting signalling network. HIF-1α is a transcriptional activator of oxygen-sensitive genes and is involved in hypoxia-induced HO-1 expression. In recent years, the cascade reaction of Nrf2 and HO-1 is considered to be one of the most important intracellular antioxidant stress mechanisms (Ref. Reference Rubiolo, Mithieux and Vega155). HO-1 has been found to mediate the synthesis and activity of the important pro-angiogenic factor VEGF, forming a positive feedback mechanism (Ref. Reference Józkowicz156) (Fig. 2).

Figure 4. NF-κB promotes angiogenesis under different pathological conditions. In the hypoxic tumour microenvironment, hypoxia renders PHDs that inhibit IKKβ activity ineffective. Activated NF-κB is transported to the nucleus and induces HIF-1α transcription, which promotes the expression of angiogenic factors. In endothelial cells of renal ischaemia/reperfusion model, LPS-induced NF-κB regulates HIF-2α expression and increases NO production, thereby improving renal microvascular perfusion.

In addition to canonical NF-κB signalling pathway, non-canonical NF-κB pathway is also involved in tumour progression (Fig. 3), but its effect on angiogenesis is still not comprehensive. Unlike canonical pathway, non-canonical pathway does not require the participation of IKKβ, but NIK and IKKα are indispensable. Previous studies have shown that NIK-mediated non-canonical NF-κB signalling pathway regulated inflammation-induced and tumour-related angiogenesis. NIK has been confirmed to be expressed in human melanoma, renal cell carcinoma, breast cancer, colorectal cancer and pancreatic cancer, and with the deletion of NIK gene, tumour-related angiogenesis was reduced (Ref. Reference Noort13). This effect is due to NIK involved in non-canonical NF-κB pathway can upregulate CXCL12, which is a chemokine that promotes angiogenesis and plays an important role in CXCR4 homing of endothelial progenitor cells. Of note, gene deletion of NIK only inhibits pathological angiogenesis, and does not affect the angiogenesis induced by TNF-α or VEGF during normal development.

NF-κB signalling pathway inhibits pathological angiogenesis by negatively regulating ECM degradation and endothelial cell activity

Paradoxically, the activation of NF-κB signalling pathway also inhibits angiogenesis (Fig. 5). In 2006, Kisseleva et al. reported that the transplanted tumours grew faster and the tumour vessels increased significantly in Tie2 promoter-IκBα transgenic mice, leading to the inhibition of NF-κB activity in ECs (Ref. Reference Kisseleva157). This study first demonstrated the inhibitory effect of NF-κB on tumour angiogenesis. Actually, it is not clear what mechanism NF-κB inhibits tumour angiogenesis and how NF-κB is activated in ECs. In general, angiogenesis requires a series of coordinated steps, in which the degradation of vascular endothelial matrix is a necessary process. NF-κB upregulates tissue inhibitors of metalloproteinase-1 (TIMP-1), a matrix metalloproteinases inhibitor that can prevent EC migration, in astrocytes by secreting IL-1. Similarly, in ECs, reactive oxygen species and TNF-α induce the expression of plasminogen activator inhibitor-1 (PAI-1) through NF-κB, and inhibit the activation of plasminogen to hydrolyse endothelial matrix (Ref. Reference Ricke-Hoch158). TNF-α can also inhibit the expression of tissue plasminogen activator (tPA) through NF-κB, which leads to the decrease of ECM degradation ability and inhibition of angiogenesis (Ref. Reference Ulfhammer159). In addition to basement membrane degradation, ECM remodelling is also a necessary condition for angiogenesis. Integrin is a cell adhesion and signalling protein that mediates the interaction between cells and ECM. The interaction between αVβ3-integrin and ECM has been shown to activate canonical NF-κB signal pathway (Refs Reference Rice160, Reference Jiang161, Reference Liu162). However, the role of the activated signal in ECM remodelling needs to be further explored. The binding site of NF-κB is located in the promoter regions of thrombin-sensitive proteins-1 and 2 (TSP-1 and 2), which were the earliest discovered natural vascular inhibitors (Ref. Reference Yang163). Later, vascular endothelial growth inhibitor (VEGI), which inhibits vascular EC proliferation and angiogenesis, has also been found to be induced by NF-κB (Ref. Reference Xiao164).

Figure 5. The negative regulation mechanism of NF-κB on angiogenesis by transcription target gene. On the one hand, NF-κB activates the expression of TIMP-1, PAI-1 and TSP-1, 2, resulting in decreased extracellular matrix degradation and hindering angiogenesis. On the other hand, NF-κB nuclear transcription can activate apoptosis signalling pathway, induce the expression of downstream pro-apoptotic genes such as JNK, Fas, FasL, caspase-3, thus promoting endothelial cell apoptosis. In addition, activated NF-κB induces the upregulation of VEGI, thereby inhibiting endothelial cell proliferation and growth.

A large number of evidences show that the activation of NF-κB is able to promote EC apoptosis. Under high glucose stimulation, PI3K/Akt/eNOS/NO pathway in HUVECs is activated to protect ECs from apoptosis at early stage. However, long-term stimulation with high concentration of glucose causes ECs to produce continuous and excessive reactive oxygen species, resulting in the continuous activation of NF-κB, and the downstream c-jun amino-terminal kinase (JNK) and caspase3 are also sequentially activated, which promotes EC apoptosis (Refs Reference Ho165, Reference Zhazykbayeva166). Furthermore, NO-mediated inhibition of NF-κB signal was reported to reverse cell apoptosis. IL-18 can induce the expression of proapoptotic genes by activating the NF-κB signalling pathway, including Bcl-Xs, Fas and FasL, leading to the apoptosis of endogenous and exogenous human cardiac microvascular endothelial cells (Ref. Reference Chandrasekar167). In addition, angiopoietin-1 was revealed to activate A20 binding inhibitor of NF-κB activation-2, an endogenous inhibitor of NF-κB, to protect ECs from apoptosis (Refs Reference Tadros168, Reference Chng169). Indeed, negative regulators that inhibit NF-κB activation were also shown to significantly reduce tube formation in HUVECs. Sprouty1 (SPRY1) is an inhibitor of MAPK pathway, and the regulation of SPRY1 expression is dependent of NF-κB signalling pathway. Some scholars have shown that SPRY1 promoted EC apoptosis, reduced EC proliferation, migration and adhesion as well as inhibited endogenous angiogenesis (Ref. Reference Sabatel170). Although silencing NF-κB can reverse the anti-apoptosis effect of apoptotic exosome-like vesicles (ApoExo) on ECs, inhibition of NF-κB restores the reduction of tube formation and CD31 expression in ECs induced by ApoExo (Ref. Reference Migneault171). Tumour angiogenesis is associated with uncontrolled gene expression of vascular ECs. Bromide domain containing 7 (BRD7) is considered to be a tumour suppressor gene, and the expression of BRD7 is negatively correlated with the activation of ECs (Refs Reference Park172, Reference Peng173). In ECs, the overexpression of BRD7 enhances NF-κB activity stimulated by bromine-dependent domain and upregulates the expression of NF-κB-dependent adhesion molecules (Ref. Reference van Beijnum174). However, the complex mechanism of BRD7 in NF-κB-mediated process has not been fully clarified. In addition to inhibiting tumour angiogenesis, the activation of NF-κB may also play an anti-tumour role through reversing endothelial anergy to inflammatory signals. Compared with normal ECs, tumour ECs are exposed to angiogenic growth factors and show reduced expression of adhesion molecules such as ICAM-1, ICAM-2 and VCAM-1. NF-κB has been found to upregulate multiple adhesion molecules in tumour ECs and restore leukocyte–endothelial interaction, resulting in anti-tumour immune effects. However, angiogenesis factors inhibit NF-κB pathway and the transcription of related adhesion molecules (Refs Reference Tromp175, Reference Luo176, Reference Dirkx177, Reference Yu178). These results suggest that NF-κB activated by vascular inhibitors not only directly negatively regulates ECs, but also indirectly exerts anti-tumour effect by upregulating the expression of adhesion molecules and reversing the inhibited inflammatory response.

NF-κB: a target for angiostatic therapy or angiogenic therapy?

It is well known that early activation of angiogenesis is a necessary condition for maintaining tumour cell proliferation. In addition to serving as a provider of nutrition, oxygen and waste transport, blood vessels also permit distant metastasis of tumour cells. The highly invasive process in tumours initiates with vasodilation and vascular permeability, causing extravasation of proteins that destroys the endothelial basement membrane (Ref. Reference Ghosh15). After the endothelial basement membrane of the vascular wall is destroyed, ECs, pericytes and vascular smooth muscle cells migrate to the direction of angiogenesis stimulation produced by tumour cells or host cells (Ref. Reference Armulik, Genové and Betsholtz179). The activated proliferating ECs adhere to each other to form tubular structures, accompanied by the formation of basement membrane and pericyte attachment. With the further maturation of the vascular network, capillaries are transformed into larger blood vessels, arteries and veins so that a new circulatory system is established (Ref. Reference Zuazo-Gaztelu and Casanovas180). In the tumour ecosystem, tumour cells and their microenvironment stimulate angiogenesis by regulating the expression of pro-angiogenic factors and/or anti-angiogenic factors to promote further tumour growth (Refs Reference Fraisl181, Reference Goel182).

Considering the important role of activated NF-κB in tumour angiogenesis, a lot of efforts have been made to explore strategies for NF-κB as a target to assist cancer prevention and treatment. Some NF-κB compound inhibitors that block the formation of new blood vessels in tumours have been reported. Since the cytokine IL-6 produced by NF-κB plays an important role in the formation of new blood vessels in tumour, NF-κB/IL-6 signal is considered as a promising target in tumour treatment. In triple-negative breast cancer (TNBC), the specific agonist of G protein-coupled oestrogen receptor activates ERK1/2 and PI3K/Akt in TNBC cells, and suppresses the phosphorylation of GSK-3β, leading to inhibit the transcription activity of NF-κB. The inactivated NF-κB reduces its binding to the IL-6 promoter, which allows tumour cell migration and angiogenesis to be hindered (Ref. Reference Chen183). STAT3 and HIF-1α are considered to be the main signal transduction molecules downstream of IL-6 signalling, and inactivation of HIF-1α and STAT3 can inhibit the occurrence and development of tumours. Liang et al. confirmed that inhibition of NF-kB/IL-6 was a prerequisite for inactivation of STAT3 and HIF-1α pathways induced by G protein-coupled oestrogen receptor-specific agonists. It is noteworthy that the activation of NF-κB/IL-6 seems also to play a positive role in cerebrovascular formation. Unlike anti-angiogenesis therapy in tumours, Zhuang et al. found that autophagy-related protein ATG7 homologous to ubiquitin-activated enzyme E1 mediated the production of IL-6 in cerebral microvascular ECs in a NF-κB-dependent manner and promoted angiogenesis in cerebral vessels (Ref. Reference Zhuang184).

Generally, many targets of NF-κB inhibitors ranging from cell surface receptors to nuclear signaling have been identified (Fig. 6) (Ref. Reference Ramadass, Vaiyapuri and Tergaonkar185). Among them, lots of inhibitors are extensively studied and undergoing clinic trials for treatment of cancer. Their effects on angiogenesis are partially examined. For example, the IKKa/b inhibitors including CHS-828, IMD-1041 and EB-1627 have been shown to reduce angiogenesis by downregulating CCL2, CXCL5, Cxcr2 and TNF-α (Refs Reference Lennikov130, Reference Huang186). BTK inhibitors such as ibrutinib, zanubrutinib and acalabrutinib are promising drugs for cancer treatment. A recent study shows that ibrutinib inhibits angiogenesis. Interestingly, the effect of ibrutinib on angiogenesis is independent on BTK but via stimulating more BMP4 expression (Ref. Reference Liu187). Another group of NF-κB inhibitors are inhibitors of proteasome, such as bortezomib and disulfiram. Both bortezomib and disulfiram have been shown to inhibit angiogenesis via reducing the production of VEGF, HGF and bFGF or weakening EGFR phosphorylation (Refs Reference Wang, Zhang and Yao188, Reference Li189). Selinexor is a potent and selective exportin-1 inhibitor that affects nuclear translocation of NF-κB. It also inhibits angiogenesis in tumour mouse models (Ref. Reference Gravina190). In nucleus, the activity of NF-κB is modulated by deacetylase and a group of deacetylase inhibitors including azacitidine, vorinostat, romidepsin, belinostat, panobinostat and decitabine can inhibit angiogenesis via downregulating VEGF and nitric oxide synthase gene (Ref. Reference Singh, Bishayee and Pandey191). Thus, small molecules among NF-κB inhibitors generally show inhibitory effect on angiogenesis.

Figure 6. Many NF-κB inhibitors show inhibitory effect on angiogenesis. The list of NFkB inhibitors of which the effects on angiogenesis have been examined in vitro and in vivo. The inhibitors including IKK inhibitors (CHS-828, IMD-1041 and EB-1627), BTK inhibitors (zanubrutinib, acalabrutinib and ibrutinib), proteasome inhibitors (disulfiram and bortezomib), nuclear translocation inhibitor (selinexor), histone deacetylase inhibitors (vorinostat, romidepsin, belinostat, panobinostat, azacitidine and decitabine) and natural products (curcumin, quercetin and neovastat).

Besides small molecules, there are also natural products that inhibit NF-κB (Fig. 6). As the most common natural NF-kB inhibitor, curcumin primarily induces tumour cell apoptosis and inhibits angiogenesis. The inhibitory effect of curcumin on angiogenesis is mainly through NF-κB-mediated downregulation of COX-2 and VEGF and other angiogenesis-related genes. This effect is reflected in the colon cancer xenograft model that curcumin significantly reduces tumour growth by inhibiting the expression of COX-2 and VGGF (Ref. Reference Kunnumakkara192). One study indicated that the inhibitory effect of curcumin on VEGF could inhibit the binding ability of NF-κB to DNA in human tongue squamous tumour cells transfected with luciferase gene, and enhance its sensitisation effect on treatment (Ref. Reference Chiang193). In addition, curcumin can increase the expression of apoptosis mediators such as cytochrome C, caspase-3 and caspase-8 in tumour cells. Curcumin inhibits p65 mediated by ERK1/2 and SAPK/JNK to limit the growth of androgen-independent prostate cancer cells, and curcumin also has the ability to effectively restrain stemness features of liver cancer cells (Refs Reference Li194, Reference Marquardt195). The substantial clinical trials on curcumin are underway or have been completed. A large number of clinical trials support the potential of curcumin for the treatment of various cancers (Ref. Reference Li and Zhang196). Considering that the bioavailability of curcumin is still generally low, the research and development of targeted formulations based on various carrier types has attracted increasing attention (Refs Reference Dhillon197, Reference Anand198).

Quercetin, a kind of flavonoids with chemical therapy and chemical prevention, can be found in some plants such as apples, grapes, onions, nuts and kale. This plant flavanol plays an important role in inhibiting tumour progression by regulating multiple signalling pathways. In serval tumours, quercetin can not only negatively regulate ECs and inhibit tumour angiogenesis, but also induce tumour cell apoptosis by inhibiting NF-κB and its downstream genes (Refs Reference Long199, Reference Lei200, Reference Huang201, Reference Granado-Serrano202). Several clinical trials have confirmed the safety of quercetin administration and observed its antitumor activity. Clinical experiments have shown that quercetin can inhibit the growth of prostate cancer tumours and reduce the risk of prostate cancer (Refs Reference McCann203, Reference Nieman204). Another clinical study evaluated the synergistic effect of quercetin and sorafenib in hepatocellular carcinoma (Ref. Reference Brito205). It is known that ovarian cancer stem cells (OCSCs) are transformed into vascular precursor cells after activation by NF-κB signalling pathway, which is the unique ability of OCSC. IKKβ inhibitor BAY11-7082 was reported to prevent ovarian cancer angiogenesis; however, this inhibitor had no effect on vascular development of normal ECs. These results showed that canonical NF-κB inactivation could inhibit OCSC differentiation and the ability to produce ECs (Refs Reference Vazquez-Santillan88, Reference Alvero206). Furthermore, kallistatin is a serine protease inhibitor that inhibits TNF-α-induced degradation of IκBα, phosphorylation of IKK and p65 protein and nuclear translocation of p65/50. Studies have shown that kallistatin may reduce the expression of some angiogenesis-related genes including VEGF via inhibition of the NF-kB signalling pathway, leading to anti-tumour angiogenesis (Ref. Reference Huang207). However, the potential application of kallistatin in tumours needs to be further verified in preclinical and clinical trials.

Paradoxically, the proliferation, migration and survival of ECs are found to be inhibited under the regulation of NF-κB, therefore the activation of NF-κB should have a dual effect on tumour angiogenesis. Several angiostatic compounds have been reported to activate NF-κB in ECs to prevent tumour growth. As an effective vascular inhibitor, the 16 kDa N-terminal fragment of prolactin (16 K PRL) inhibits the proliferation and migration of ECs. The16 K human PRL induces the activation of caspase-8, caspase-9 and subsequent cell apoptosis, which requires the activation of NF-kB (Refs Reference Tabruyn208, Reference Tabruyn209). In addition to the 16 kDa N-terminal fragment of prolactin, platelet factor 4 (PF4), a platelet-specific chemokine, inhibits EC proliferation, migration and angiogenesis in vitro and in vivo. Experiments and data showed that PF4 could increase the expression of E-selectin in ECs, and the cis-acting element sensitive to PF4 in E-selectin promoter was the binding site of NF-κB (Ref. Reference Yu178). These results suggested that PF4 induced E-selectin expression in ECs by activating NF-κB transcriptional activity. In order to analyse the changes of NF-κB-related gene expression after angiostatin treatment, researchers apply gene microarray analysis technology. Although there is no direct evidence that NF-κB is activated, angiostatin is associated with the increased expression of RELB and many NF-κB target genes, including E-selectin, cyclin D1, p21 and ICAM-1 (Ref. Reference Chen210). Furthermore, the use of statins has been reported to inhibit tumour growth and angiogenesis. Atorvastatin increases the production of reactive oxygen species through the activation of NF-κB, which leads to the loss of NO bioavailability, followed by the formation of strong oxidant peroxynitrite. The activation of NF-κB upregulates the expression of neuronal and inducible nitric oxide synthase capable of inhibiting angiogenesis and leading to vascular injury (Refs Reference Hindler211, Reference Nakata212). In addition, Gingras et al. demonstrated that neovastat, another angiogenesis inhibitor, suppresses angiogenesis by increasing tPA activity in a NF-κB-dependent manner (Refs Reference Gingras213, Reference Simard214). Clinical trials of neovastat in multiple myeloma, plasmacytoma, breast cancer, lung cancer, kidney cancer or colon cancer have been completed. Unfortunately, although neovastat can inhibit vascular growth in animal models, it has little or no benefit for cancer patients (Refs Reference Escudier215, Reference Lu216, Reference Loprinzi217). In summary, the above results indicate that the activation of NF-kB, especially in ECs, is probably an effective method for the treatment of cancer.

Conclusion

As a multi-effect transcription factor, NF-κB is involved in various biological processes such as inflammation, immune response, apoptosis and tumour. Stimulators that activate NF-κB signalling pathway include cytokines, chemokines, oxidative stress, bacterial LPS and so on. In the last few years, a large number of increasing research showed that NF-κB signalling molecules play an important role in regulating vascular-related diseases, especially tumours. In inflammation-related diseases, pathological angiogenesis is not only regulated by NIK-mediated non-canonical NF-κB signalling pathway, but also regulated by miR RNAs targeting to inhibit NF-κB-related mRNA expression. Moreover, the activation of NF-κB seems to have double-sided effects on tumourigenesis, progression and angiogenesis affecting tumour growth, invasion and migration. Based on the study of tumour growth and diffusion pathways, anti-angiogenesis treatment has become an important strategy that cannot be ignored. Although new insights have been provided into the biological function of NF-κB in angiogenesis and its role in some diseases such as cancer, there is still little work on NF-κB or IKK targeted drugs as angiogenesis regulators in clinical trials. More and more angiogenic inhibitors are required to explore the inhibition of tumour angiogenesis by regulating the NF-κB signalling pathway. In addition, the available literature to date mainly describes it as a tumour promotor, but this could not be the case in all tumour subtypes. These results indicate that more clinical samples need to be included in the study. Finally, the role of NF-κB pathway in tumour angiogenesis remains controversial, and whether NF-κB is used as a target or a medium for inhibiting angiogenesis needs to be further investigated.

Author contributions

R. M. contributed to research orientation, creativity and administrative support. Y. J., C. S. and X. L. contributed to the collection and collation of the literature. Y. J., J. Z. and C. S. contributed to the concept and design of this study and the editing of the manuscript. All authors have read and approved the final version of the manuscript.

Financial support

This work was supported by the National Natural Science Foundation of China (82070505); the Distinguished Professorship Program of Jiangsu Province to Renfang Mao; Jiangsu Postgraduate Research and Practice Innovation Project (KYCX22_3357); Nantong Science and Technology (JCZ2022072).

Competing interest

None.

Ethical standards

This study was approved by the ethics committee of Nantong University Medical School. Institutional approval was not required to publish this manuscript.

Footnotes

*

These authors made equal contributions to this work.

References

Sun, SC, Chang, JH and Jin, J (2013) Regulation of nuclear factor-κB in autoimmunity. Trends in Immunology 34, 282289.CrossRefGoogle ScholarPubMed
Bottex-Gauthier, C et al. (2002) [The Rel/NF-kappa-B transcription factors: complex role in cell regulation]. Pathologie-Biologie 50, 204211.CrossRefGoogle ScholarPubMed
Zhang, Q, Lenardo, MJ and Baltimore, D (2017) 30 Years of NF-κB: a blossoming of relevance to human pathobiology. Cell 168, 3757.CrossRefGoogle ScholarPubMed
Taniguchi, K and Karin, M (2018) NF-κB, inflammation, immunity and cancer: coming of age. Nature reviews. Immunology 18, 309324.CrossRefGoogle ScholarPubMed
Delgado, VM et al. (2011) Modulation of endothelial cell migration and angiogenesis: a novel function for the ‘tandem-repeat’ lectin galectin-8. FASEB Journal 25, 242254.CrossRefGoogle ScholarPubMed
Geudens, I and Gerhardt, H (2011) Coordinating cell behaviour during blood vessel formation. Development 138, 45694583.CrossRefGoogle ScholarPubMed
Polyak, K, Haviv, I and Campbell, IG (2009) Co-evolution of tumor cells and their microenvironment. Trends in Genetics: TIG 25, 3038.CrossRefGoogle ScholarPubMed
Polyak, K and Weinberg, RA (2009) Transitions between epithelial and mesenchymal states: acquisition of malignant and stem cell traits. Nature Reviews. Cancer 9, 265273.CrossRefGoogle ScholarPubMed
Quail, DF and Joyce, JA (2013) Microenvironmental regulation of tumor progression and metastasis. Nature Medicine 19, 14231437.CrossRefGoogle ScholarPubMed
Rehman, AO and Wang, CY (2006) Notch signaling in the regulation of tumor angiogenesis. Trends in Cell Biology 16, 293300.CrossRefGoogle ScholarPubMed
Fernández-Chacón, M et al. (2021) Role of Notch in endothelial biology. Angiogenesis 24, 237250.CrossRefGoogle ScholarPubMed
Zhou, W et al. (2021) Unraveling the molecular mechanisms between inflammation and tumor angiogenesis. American Journal of Cancer Research 11, 301317.Google ScholarPubMed
Noort, AR et al. (2014) NF-κB-inducing kinase is a key regulator of inflammation-induced and tumour-associated angiogenesis. The Journal of Pathology 234, 375385.CrossRefGoogle ScholarPubMed
Ashander, LM et al. (2016) Targeting endothelial adhesion molecule transcription for treatment of inflammatory disease: a proof-of-concept study. Mediators of Inflammation 2016, 7945848.CrossRefGoogle ScholarPubMed
Ghosh, G et al. (2012) NF-κB regulation: lessons from structures. Immunological Reviews 246, 3658.CrossRefGoogle ScholarPubMed
Smale, ST (2012) Dimer-specific regulatory mechanisms within the NF-κB family of transcription factors. Immunological Reviews 246, 193204.CrossRefGoogle ScholarPubMed
Neumann, M and Naumann, M (2007) Beyond IkappaBs: alternative regulation of NF-kappaB activity. FASEB Journal 21, 26422654.CrossRefGoogle ScholarPubMed
Mulero, MC et al. (2019) Genome reading by the NF-κB transcription factors. Nucleic Acids Research 47, 99679989.CrossRefGoogle ScholarPubMed
Ji, Z et al. (2019) Inflammatory regulatory network mediated by the joint action of NF-kB, STAT3, and AP-1 factors is involved in many human cancers. Proceedings of the National Academy of Sciences of the USA 116, 94539462.CrossRefGoogle ScholarPubMed
Schuster, M et al. (2013) Atypical IκB proteins – nuclear modulators of NF-κB signaling. Cell Communication and Signaling: CCS 11, 23.CrossRefGoogle ScholarPubMed
Sun, SC (2011) Non-canonical NF-κB signaling pathway. Cell Research 21, 7185.CrossRefGoogle ScholarPubMed
Karin, M and Greten, FR (2005) NF-kappaB: linking inflammation and immunity to cancer development and progression. Nature Reviews. Immunology 5, 749759.CrossRefGoogle ScholarPubMed
Majdalawieh, A and Ro, HS (2010) Regulation of IkappaBalpha function and NF-kappaB signaling: AEBP1 is a novel proinflammatory mediator in macrophages. Mediators of Inflammation 2010, 823821.CrossRefGoogle ScholarPubMed
Ferreiro, DU and Komives, EA (2010) Molecular mechanisms of system control of NF-kappaB signaling by IkappaBalpha. Biochemistry 49, 15601567.CrossRefGoogle ScholarPubMed
Chen, LF and Greene, WC (2004) Shaping the nuclear action of NF-kappaB. Nature Reviews. Molecular Cell Biology 5, 392401.CrossRefGoogle ScholarPubMed
Israël, A (2010) The IKK complex, a central regulator of NF-kappaB activation. Cold Spring Harbor Perspectives in Biology 2, a000158.CrossRefGoogle ScholarPubMed
Vallabhapurapu, S and Karin, M (2009) Regulation and function of NF-kappaB transcription factors in the immune system. Annual Review of Immunology 27, 693733.CrossRefGoogle ScholarPubMed
Ruland, J (2011) Return to homeostasis: downregulation of NF-κB responses. Nature Immunology 12, 709714.CrossRefGoogle ScholarPubMed
Rothwarf, DM and Karin, M (1999) The NF-kappa B activation pathway: a paradigm in information transfer from membrane to nucleus. Science's STKE: signal transduction knowledge environment 1999, RE1.CrossRefGoogle Scholar
May, MJ, Marienfeld, RB and Ghosh, S (2002) Characterization of the Ikappa B-kinase NEMO binding domain. The Journal of Biological Chemistry 277, 4599246000.CrossRefGoogle ScholarPubMed
Karin, M and Delhase, M (2000) The I kappa B kinase (IKK) and NF-kappa B: key elements of proinflammatory signalling. Seminars in Immunology 12, 8598.CrossRefGoogle ScholarPubMed
Zhang, T et al. (2021) NF-κB signaling in inflammation and cancer. MedComm 2, 618653.CrossRefGoogle ScholarPubMed
Pires, B et al. (2018) NF-kappaB: two sides of the same coin. Genes 9, 24.CrossRefGoogle ScholarPubMed
Cildir, G, Low, KC and Tergaonkar, V (2016) Noncanonical NF-κB signaling in health and disease. Trends in Molecular Medicine 22, 414429.CrossRefGoogle ScholarPubMed
Maubach, G et al. (2019) NF-kappaB-inducing kinase in cancer. Biochimica et biophysica acta. Reviews on Cancer 1871, 4049.CrossRefGoogle ScholarPubMed
Sun, SC (2012) The noncanonical NF-κB pathway. Immunological Reviews 246, 125140.CrossRefGoogle ScholarPubMed
Bonizzi, G and Karin, M (2004) The two NF-kappaB activation pathways and their role in innate and adaptive immunity. Trends in Immunology 25, 280288.CrossRefGoogle ScholarPubMed
Vallabhapurapu, S et al. (2008) Nonredundant and complementary functions of TRAF2 and TRAF3 in a ubiquitination cascade that activates NIK-dependent alternative NF-kappaB signaling. Nature Immunology 9, 13641370.CrossRefGoogle Scholar
Senftleben, U et al. (2001) Activation by IKKalpha of a second, evolutionary conserved, NF-kappa B signaling pathway. Science 293, 14951499.CrossRefGoogle ScholarPubMed
Doebele, C et al. (2010) Members of the microRNA-17-92 cluster exhibit a cell-intrinsic antiangiogenic function in endothelial cells. Blood 115, 49444950.CrossRefGoogle ScholarPubMed
Liu, LZ et al. (2011) MiR-21 induced angiogenesis through AKT and ERK activation and HIF-1α expression. PLoS ONE 6, e19139.CrossRefGoogle ScholarPubMed
Tsoyi, K et al. (2010) PTEN differentially regulates expressions of ICAM-1 and VCAM-1 through PI3K/Akt/GSK-3β/GATA-6 signaling pathways in TNF-α-activated human endothelial cells. Atherosclerosis 213, 115121.CrossRefGoogle ScholarPubMed
Sabatel, C et al. (2011) MicroRNA-21 exhibits antiangiogenic function by targeting RhoB expression in endothelial cells. PLoS ONE 6, e16979.CrossRefGoogle ScholarPubMed
Zhu, N et al. (2011) Endothelial enriched microRNAs regulate angiotensin II-induced endothelial inflammation and migration. Atherosclerosis 215, 286293.CrossRefGoogle ScholarPubMed
Ueda, R et al. (2009) Dicer-regulated microRNAs 222 and 339 promote resistance of cancer cells to cytotoxic T-lymphocytes by down-regulation of ICAM-1. Proceedings of the National Academy of Sciences of the USA 106, 1074610751.CrossRefGoogle ScholarPubMed
Hu, G et al. (2010) miR-221 suppresses ICAM-1 translation and regulates interferon-gamma-induced ICAM-1 expression in human cholangiocytes. American Journal of Physiology. Gastrointestinal and Liver Physiology 298, G542G550.CrossRefGoogle ScholarPubMed
Suárez, Y et al. (2007) Dicer dependent microRNAs regulate gene expression and functions in human endothelial cells. Circulation Research 100, 11641173.CrossRefGoogle ScholarPubMed
Poliseno, L et al. (2006) MicroRNAs modulate the angiogenic properties of HUVECs. Blood 108, 30683071.CrossRefGoogle ScholarPubMed
Zhu, K et al. (2013) MiR-146a enhances angiogenic activity of endothelial cells in hepatocellular carcinoma by promoting PDGFRA expression. Carcinogenesis 34, 20712079.CrossRefGoogle ScholarPubMed
Cheng, HS et al. (2013) MicroRNA-146 represses endothelial activation by inhibiting pro-inflammatory pathways. EMBO Molecular Medicine 5, 10171034.CrossRefGoogle ScholarPubMed
Sun, X et al. (2012) MicroRNA-181b regulates NF-κB-mediated vascular inflammation. The Journal of Clinical Investigation 122, 19731990.Google ScholarPubMed
Fasanaro, P et al. (2008) MicroRNA-210 modulates endothelial cell response to hypoxia and inhibits the receptor tyrosine kinase ligand Ephrin-A3. The Journal of Biological Chemistry 283, 1587815883.CrossRefGoogle ScholarPubMed
Qi, J et al. (2012) microRNA-210 negatively regulates LPS-induced production of proinflammatory cytokines by targeting NF-κB1 in murine macrophages. FEBS Letters 586, 12011207.CrossRefGoogle ScholarPubMed
Maijer, KI et al. (2015) Nuclear factor-κB-inducing kinase Is expressed in synovial endothelial cells in patients with early arthritis and correlates with markers of inflammation: a prospective cohort study. The Journal of Rheumatology 42, 15731581.CrossRefGoogle Scholar
Madge, LA et al. (2008) Lymphotoxin-alpha 1 beta 2 and LIGHT induce classical and noncanonical NF-kappa B-dependent proinflammatory gene expression in vascular endothelial cells. The Journal of Immunology 180, 34673477.CrossRefGoogle ScholarPubMed
Brown, KD, Claudio, E and Siebenlist, U (2008) The roles of the classical and alternative nuclear factor-kappaB pathways: potential implications for autoimmunity and rheumatoid arthritis. Arthritis Research & Therapy 10, 212.CrossRefGoogle ScholarPubMed
Tak, PP and Firestein, GS (2001) NF-kappaB: a key role in inflammatory diseases. The Journal of Clinical Investigation 107, 711.CrossRefGoogle ScholarPubMed
Suárez, Y et al. (2010) Cutting edge: TNF-induced microRNAs regulate TNF-induced expression of E-selectin and intercellular adhesion molecule-1 on human endothelial cells: feedback control of inflammation. The Journal of Immunology 184, 2125.CrossRefGoogle ScholarPubMed
Tsai, YH et al. (2009) The M type K15 protein of Kaposi's sarcoma-associated herpesvirus regulates microRNA expression via its SH2-binding motif to induce cell migration and invasion. Journal of Virology 83, 622632.CrossRefGoogle Scholar
Wu, YH et al. (2011) The manipulation of miRNA-gene regulatory networks by KSHV induces endothelial cell motility. Blood 118, 28962905.CrossRefGoogle ScholarPubMed
Shan, SW et al. (2013) Mature miR-17-5p and passenger miR-17-3p induce hepatocellular carcinoma by targeting PTEN, GalNT7 and vimentin in different signal pathways. Journal of Cell Science 126, 15171530.Google ScholarPubMed
Vrábel, D, Pour, L and Ševčíková, S (2019) The impact of NF-κB signaling on pathogenesis and current treatment strategies in multiple myeloma. Blood Reviews 34, 5666.CrossRefGoogle ScholarPubMed
Eluard, B et al. (2022) The alternative RelB NF-κB subunit is a novel critical player in diffuse large B-cell lymphoma. Blood 139, 384398.CrossRefGoogle ScholarPubMed
Lim, KH, Yang, Y and Staudt, LM (2012) Pathogenetic importance and therapeutic implications of NF-κB in lymphoid malignancies. Immunological Reviews 246, 359378.CrossRefGoogle ScholarPubMed
Courtois, G and Gilmore, TD (2006) Mutations in the NF-kappaB signaling pathway: implications for human disease. Oncogene 25, 68316843.CrossRefGoogle ScholarPubMed
Lake, A et al. (2009) Mutations of NFKBIA, encoding IkappaB alpha, are a recurrent finding in classical Hodgkin lymphoma but are not a unifying feature of non-EBV-associated cases. International Journal of Cancer 125, 13341342.CrossRefGoogle Scholar
Kalaitzidis, D et al. (2002) The human B-cell lymphoma cell line RC-K8 has multiple genetic alterations that dysregulate the Rel/NF-kappaB signal transduction pathway. Oncogene 21, 87598768.CrossRefGoogle ScholarPubMed
Bredel, M et al. (2011) NFKBIA deletion in glioblastomas. The New England Journal of Medicine 364, 627637.CrossRefGoogle ScholarPubMed
Zheng, H et al. (2016) Whole-exome sequencing identifies multiple loss-of-function mutations of NF-κB pathway regulators in nasopharyngeal carcinoma. Proceedings of the National Academy of Sciences of the USA 113, 1128311288.CrossRefGoogle ScholarPubMed
Matthews, GM et al. (2016) NF-κB dysregulation in multiple myeloma. Seminars in Cancer Biology 39, 6876.CrossRefGoogle ScholarPubMed
Young, RM et al. (2019) Pathogenic B-cell receptor signaling in lymphoid malignancies: new insights to improve treatment. Immunological Reviews 291, 190213.CrossRefGoogle ScholarPubMed
Schmitz, R et al. (2018) Genetics and pathogenesis of diffuse large B-cell lymphoma. The New England Journal of Medicine 378, 13961407.CrossRefGoogle ScholarPubMed
Bhatt, D and Ghosh, S (2014) Regulation of the NF-κB-mediated transcription of inflammatory genes. Frontiers in Immunology 5, 71.CrossRefGoogle ScholarPubMed
Dawson, MA and Kouzarides, T (2012) Cancer epigenetics: from mechanism to therapy. Cell 150, 1227.CrossRefGoogle ScholarPubMed
Patel, MB and Wang, J (2018) The identification and interpretation of cis-regulatory noncoding mutations in cancer. High-Throughput 8, 1.CrossRefGoogle ScholarPubMed
Lin, SC et al. (2006) Functional polymorphism in NFKB1 promoter is related to the risks of oral squamous cell carcinoma occurring on older male areca (betel) chewers. Cancer Letters 243, 4754.CrossRefGoogle Scholar
Kang, S et al. (2005) Polymorphism in the nuclear factor kappa-B binding promoter region of cyclooxygenase-2 is associated with an increased risk of bladder cancer. Cancer Letters 217, 1116.CrossRefGoogle ScholarPubMed
Costa RM, DA et al. (2016) The NFκB signaling pathway in papillomavirus-induced lesions: friend or foe. Anticancer Research 36, 20732083.Google ScholarPubMed
Charostad, J et al. (2020) The interplay between EBV and KSHV viral products and NF-κB pathway in oncogenesis. Infectious Agents and Cancer 15, 62.CrossRefGoogle ScholarPubMed
Harhaj, EW and Giam, CZ (2018) NF-κB signaling mechanisms in HTLV-1-induced adult T-cell leukemia/lymphoma. The FEBS Journal 285, 33243336.CrossRefGoogle ScholarPubMed
Shokri, S et al. (2019) Complexity on modulation of NF-κB pathways by hepatitis B and C: a double-edged sword in hepatocarcinogenesis. Journal of Cellular Physiology 234, 1473414742.CrossRefGoogle Scholar
Gilmore, TD (2021) NF-κB and human cancer: what have we learned over the past 35 years. Biomedicines 9, 889.CrossRefGoogle ScholarPubMed
Gudkov, AV, Gurova, KV and Komarova, EA (2011) Inflammation and p53: a tale of two stresses. Genes & Cancer 2, 503516.CrossRefGoogle ScholarPubMed
Cerhan, JR et al. (2003) Association of aspirin and other non-steroidal anti-inflammatory drug use with incidence of non-Hodgkin lymphoma. International Journal of Cancer 106, 784788.CrossRefGoogle ScholarPubMed
Becker, C et al. (2004) TGF-beta suppresses tumor progression in colon cancer by inhibition of IL-6 trans-signaling. Immunity 21, 491501.CrossRefGoogle ScholarPubMed
Bassères, DS et al. (2010) Requirement of the NF-kappaB subunit p65/RelA for K-Ras-induced lung tumorigenesis. Cancer Research 70, 35373546.CrossRefGoogle ScholarPubMed
Finco, TS et al. (1997) Oncogenic Ha-Ras-induced signaling activates NF-kappaB transcriptional activity, which is required for cellular transformation. The Journal of Biological Chemistry 272, 2411324116.CrossRefGoogle ScholarPubMed
Vazquez-Santillan, K et al. (2015) NF-κB signaling in cancer stem cells: a promising therapeutic target. Cellular Oncology 38, 327339.CrossRefGoogle ScholarPubMed
Yamamoto, M et al. (2013) NF-κB non-cell-autonomously regulates cancer stem cell populations in the basal-like breast cancer subtype. Nature Communications 4, 2299.CrossRefGoogle ScholarPubMed
Vazquez-Santillan, K et al. (2016) NF-kappaΒ-inducing kinase regulates stem cell phenotype in breast cancer. Scientific Reports 6, 37340.CrossRefGoogle ScholarPubMed
Gonzalez-Torres, C et al. (2017) NF-κB participates in the stem cell phenotype of ovarian cancer cells. Archives of Medical Research 48, 343351.CrossRefGoogle ScholarPubMed
Sun, L et al. (2013) Epigenetic regulation of SOX9 by the NF-κB signaling pathway in pancreatic cancer stem cells. Stem Cells 31, 14541466.CrossRefGoogle ScholarPubMed
Liu, S et al. (2017) FOXP3 inhibits cancer stem cell self-renewal via transcriptional repression of COX2 in colorectal cancer cells. Oncotarget 8, 4469444704.CrossRefGoogle ScholarPubMed
Wang, D et al. (2015) Prostaglandin E2 promotes colorectal cancer stem cell expansion and metastasis in mice. Gastroenterology 149, 18841895, e4.CrossRefGoogle ScholarPubMed
Balkwill, F (2009) Tumour necrosis factor and cancer. Nature Reviews. Cancer 9, 361371.CrossRefGoogle ScholarPubMed
Grivennikov, SI and Karin, M (2011) Inflammatory cytokines in cancer: tumour necrosis factor and interleukin 6 take the stage. Annals of the Rheumatic Diseases 70, i104i108.CrossRefGoogle ScholarPubMed
Taniguchi, K and Karin, M (2014) IL-6 and related cytokines as the critical lynchpins between inflammation and cancer. Seminars in Immunology 26, 5474.CrossRefGoogle ScholarPubMed
Chang, Q, Daly, L and Bromberg, J (2014) The IL-6 feed-forward loop: a driver of tumorigenesis. Seminars in Immunology 26, 4853.CrossRefGoogle Scholar
Meulmeester, E and Ten Dijke, P (2011) The dynamic roles of TGF-β in cancer. The Journal of Pathology 223, 205218.CrossRefGoogle ScholarPubMed
Grivennikov, SI, Greten, FR and Karin, M (2010) Immunity, inflammation, and cancer. Cell 140, 883899.CrossRefGoogle ScholarPubMed
Tan, MC et al. (2009) Disruption of CCR5-dependent homing of regulatory T cells inhibits tumor growth in a murine model of pancreatic cancer. The Journal of Immunology 182, 17461755.CrossRefGoogle Scholar
Murooka, TT, Rahbar, R and Fish, EN (2009) CCL5 promotes proliferation of MCF-7 cells through mTOR-dependent mRNA translation. Biochemical and Biophysical Research Communications 387, 381386.CrossRefGoogle ScholarPubMed
Hagemann, T et al. (2008) ‘Re-educating’ tumor-associated macrophages by targeting NF-kappaB. The Journal of Experimental Medicine 205, 12611268.CrossRefGoogle ScholarPubMed
Karyampudi, L et al. (2016) PD-1 blunts the function of ovarian tumor-infiltrating dendritic cells by inactivating NF-κB. Cancer Research 76, 239250.CrossRefGoogle ScholarPubMed
Antonangeli, F et al. (2020) Regulation of PD-L1 expression by NF-κB in cancer. Frontiers in Immunology 11, 584626.CrossRefGoogle ScholarPubMed
Vivier, E et al. (2008) Functions of natural killer cells. Nature Immunology 9, 503510.CrossRefGoogle ScholarPubMed
Oh, H et al. (2017) An NF-κB transcription-factor-dependent lineage-specific transcriptional program promotes regulatory T cell identity and function. Immunity 47, 450465, e5.CrossRefGoogle ScholarPubMed
Isomura, I et al. (2009) c-Rel is required for the development of thymic Foxp3 + CD4 regulatory T cells. The Journal of Experimental Medicine 206, 30013014.CrossRefGoogle ScholarPubMed
Evaristo, C et al. (2016) Cutting edge: engineering active IKKβ in T cells drives tumor rejection. The Journal of Immunology 196, 29332938.CrossRefGoogle Scholar
Ward, JP, Gubin, MM and Schreiber, RD (2016) The role of neoantigens in naturally occurring and therapeutically induced immune responses to cancer. Advances in Immunology 130, 2574.CrossRefGoogle ScholarPubMed
Kalluri, R (2016) The biology and function of fibroblasts in cancer. Nature Reviews. Cancer 16, 582598.CrossRefGoogle ScholarPubMed
Koliaraki, V et al. (2017) Mesenchymal cells in colon cancer. Gastroenterology 152, 964979.CrossRefGoogle ScholarPubMed
Erez, N et al. (2010) Cancer-associated fibroblasts are activated in incipient neoplasia to orchestrate tumor-promoting inflammation in an NF-kappaB-dependent manner. Cancer Cell 17, 135147.CrossRefGoogle Scholar
Santolla, MF et al. (2018) miR-221 stimulates breast cancer cells and cancer-associated fibroblasts (CAFs) through selective interference with the A20/c-Rel/CTGF signaling. Journal of Experimental & Clinical Cancer Research: CR 37, 94.CrossRefGoogle ScholarPubMed
Greten, FR et al. (2004) IKKbeta links inflammation and tumorigenesis in a mouse model of colitis-associated cancer. Cell 118, 285296.CrossRefGoogle Scholar
Pikarsky, E et al. (2004) NF-kappaB functions as a tumour promoter in inflammation-associated cancer. Nature 431, 461466.CrossRefGoogle ScholarPubMed
Karin, M (2006) Nuclear factor-kappaB in cancer development and progression. Nature 441, 431436.CrossRefGoogle ScholarPubMed
DiDonato, JA, Mercurio, F and Karin, M (2012) NF-κB and the link between inflammation and cancer. Immunological Reviews 246, 379400.CrossRefGoogle ScholarPubMed
Joyce, D et al. (2001) NF-kappaB and cell-cycle regulation: the cyclin connection. Cytokine & Growth Factor Reviews 12, 7390.CrossRefGoogle ScholarPubMed
Kiraly, O et al. (2015) Inflammation-induced cell proliferation potentiates DNA damage-induced mutations in vivo. PLoS Genetics 11, e1004901.CrossRefGoogle ScholarPubMed
Dutta, J et al. (2006) Current insights into the regulation of programmed cell death by NF-kappaB. Oncogene 25, 68006816.CrossRefGoogle ScholarPubMed
Gapuzan, ME, Yufit, PV and Gilmore, TD (2002) Immortalized embryonic mouse fibroblasts lacking the RelA subunit of transcription factor NF-kappaB have a malignantly transformed phenotype. Oncogene 21, 24842492.CrossRefGoogle ScholarPubMed
Gerondakis, S et al. (2014) NF-κB control of T cell development. Nature Immunology 15, 1525.CrossRefGoogle ScholarPubMed
Lin, L et al. (2019) Tertiary lymphoid organs in cancer immunology: mechanisms and the new strategy for immunotherapy. Frontiers in Immunology 10, 1398.CrossRefGoogle ScholarPubMed
Furtado, GC et al. (2007) Lymphotoxin beta receptor signaling is required for inflammatory lymphangiogenesis in the thyroid. Proceedings of the National Academy of Sciences of the USA 104, 50265031.CrossRefGoogle ScholarPubMed
Luther, SA et al. (2002) Differing activities of homeostatic chemokines CCL19, CCL21, and CXCL12 in lymphocyte and dendritic cell recruitment and lymphoid neogenesis. The Journal of Immunology 169, 424433.CrossRefGoogle ScholarPubMed
Sautès-Fridman, C et al. (2019) Tertiary lymphoid structures in the era of cancer immunotherapy. Nature Reviews. Cancer 19, 307325.CrossRefGoogle ScholarPubMed
Carrà, G et al. (2020) P53 vs NF-κB: the role of nuclear factor-kappa B in the regulation of p53 activity and vice versa. Cellular and Molecular Life Sciences: CMLS 77, 44494458.CrossRefGoogle ScholarPubMed
Sakamoto, K et al. (2009) Constitutive NF-kappaB activation in colorectal carcinoma plays a key role in angiogenesis, promoting tumor growth. Clinical Cancer Research 15, 22482258.CrossRefGoogle Scholar
Lennikov, A et al. (2018) Selective IKK2 inhibitor IMD0354 disrupts NF-κB signaling to suppress corneal inflammation and angiogenesis. Angiogenesis 21, 267285.CrossRefGoogle ScholarPubMed
Shen, J et al. (2018) IL-17 induces macrophages to M2-like phenotype via NF-κB. Cancer Management and Research 10, 42174228.CrossRefGoogle ScholarPubMed
Dong, F et al. (2014) Dihydroartemisinin targets VEGFR2 via the NF-κB pathway in endothelial cells to inhibit angiogenesis. Cancer Biology & Therapy 15, 14791488.CrossRefGoogle ScholarPubMed
Mountain, DJ et al. (2007) Interleukin-1beta increases expression and activity of matrix metalloproteinase-2 in cardiac microvascular endothelial cells: role of PKCalpha/beta1 and MAPKs. American Journal of Physiology. Cell Physiology 292, C867C875.CrossRefGoogle ScholarPubMed
Popov, Y et al. (2006) Halofuginone induces matrix metalloproteinases in rat hepatic stellate cells via activation of p38 and NFkappaB. The Journal of Biological Chemistry 281, 1509015098.CrossRefGoogle ScholarPubMed
Ko, HM et al. (2005) Platelet-activating factor induces matrix metalloproteinase-9 expression through Ca(2+)- or PI3K-dependent signaling pathway in a human vascular endothelial cell line. FEBS Letters 579, 64516458.CrossRefGoogle ScholarPubMed
Martin, D, Galisteo, R and Gutkind, JS (2009) CXCL8/IL8 stimulates vascular endothelial growth factor (VEGF) expression and the autocrine activation of VEGFR2 in endothelial cells by activating NFkappaB through the CBM (Carma3/Bcl10/Malt1) complex. The Journal of Biological Chemistry 284, 60386042.CrossRefGoogle ScholarPubMed
Passaro, C et al. (2016) The oncolytic virus dl922-947 reduces IL-8/CXCL8 and MCP-1/CCL2 expression and impairs angiogenesis and macrophage infiltration in anaplastic thyroid carcinoma. Oncotarget 7, 15001515.CrossRefGoogle ScholarPubMed
Chen, L et al. (2018) Monocyte chemoattractant protein 1 and fractalkine play opposite roles in angiogenesis via recruitment of different macrophage subtypes. International Journal of Ophthalmology 11, 216222.Google ScholarPubMed
Chen, Y and Liu, J (2019) The prognostic roles of cyclooxygenase-2 for patients with basal cell carcinoma. Artificial Cells, Nanomedicine, and Biotechnology 47, 30533057.CrossRefGoogle ScholarPubMed
Luo, LH et al. (2020) Long non-coding RNA NKILA inhibited angiogenesis of breast cancer through NF-κB/IL-6 signaling pathway. Microvascular Research 129, 103968.CrossRefGoogle ScholarPubMed
Huang, S et al. (2001) Blockade of NF-kappaB activity in human prostate cancer cells is associated with suppression of angiogenesis, invasion, and metastasis. Oncogene 20, 41884197.CrossRefGoogle ScholarPubMed
Pires, BR et al. (2017) NF-kappaB is involved in the regulation of EMT genes in breast cancer cells. PLoS ONE 12, e0169622.CrossRefGoogle ScholarPubMed
Scheel, C and Weinberg, RA (2012) Cancer stem cells and epithelial-mesenchymal transition: concepts and molecular links. Seminars in Cancer Biology 22, 396403.CrossRefGoogle ScholarPubMed
Huber, MA et al. (2004) NF-kappaB is essential for epithelial-mesenchymal transition and metastasis in a model of breast cancer progression. The Journal of Clinical Investigation 114, 569581.CrossRefGoogle Scholar
Malki, A et al. (2020) Molecular mechanisms of colon cancer progression and metastasis: recent insights and advancements. International Journal of Molecular Sciences 22, 130.CrossRefGoogle ScholarPubMed
Balkwill, F (2004) Cancer and the chemokine network. Nature Reviews. Cancer 4, 540550.CrossRefGoogle ScholarPubMed
Cummins, EP et al. (2006) Prolyl hydroxylase-1 negatively regulates IkappaB kinase-beta, giving insight into hypoxia-induced NFkappaB activity. Proceedings of the National Academy of Sciences of the USA 103, 1815418159.CrossRefGoogle ScholarPubMed
Oliver, KM, Taylor, CT and Cummins, EP (2009) Hypoxia. Regulation of NFkappaB signalling during inflammation: the role of hydroxylases. Arthritis Research & Therapy 11, 215.CrossRefGoogle ScholarPubMed
Bonello, S et al. (2007) Reactive oxygen species activate the HIF-1alpha promoter via a functional NFkappaB site. Arteriosclerosis, Thrombosis, and Vascular Biology 27, 755761.CrossRefGoogle Scholar
Frede, S et al. (2006) Bacterial lipopolysaccharide induces HIF-1 activation in human monocytes via p44/42 MAPK and NF-kappaB. The Biochemical Journal 396, 517527.CrossRefGoogle ScholarPubMed
Jung, Y et al. (2003) Hypoxia-inducible factor induction by tumour necrosis factor in normoxic cells requires receptor-interacting protein-dependent nuclear factor kappa B activation. The Biochemical Journal 370, 10111017.CrossRefGoogle ScholarPubMed
He, K et al. (2014) Lipopolysaccharide-induced cross-tolerance against renal ischemia-reperfusion injury is mediated by hypoxia-inducible factor-2α-regulated nitric oxide production. Kidney International 85, 276288.CrossRefGoogle ScholarPubMed
Bellezza, I et al. (2018) Nrf2-Keap1 signaling in oxidative and reductive stress. Biochimica et biophysica acta. Molecular cell research 1865, 721733.CrossRefGoogle ScholarPubMed
Maines, MD (1997) The heme oxygenase system: a regulator of second messenger gases. Annual Review of Pharmacology and Toxicology 37, 517554.CrossRefGoogle ScholarPubMed
Rubiolo, JA, Mithieux, G and Vega, FV (2008) Resveratrol protects primary rat hepatocytes against oxidative stress damage: activation of the Nrf2 transcription factor and augmented activities of antioxidant enzymes. European Journal of Pharmacology 591, 6672.CrossRefGoogle ScholarPubMed
Józkowicz, A et al. (2003) Heme oxygenase and angiogenic activity of endothelial cells: stimulation by carbon monoxide and inhibition by tin protoporphyrin-IX. Antioxidants & Redox Signaling 5, 155162.CrossRefGoogle ScholarPubMed
Kisseleva, T et al. (2006) NF-kappaB regulation of endothelial cell function during LPS-induced toxemia and cancer. The Journal of Clinical Investigation 116, 29552963.CrossRefGoogle ScholarPubMed
Ricke-Hoch, M et al. (2020) In peripartum cardiomyopathy plasminogen activator inhibitor-1 is a potential new biomarker with controversial roles. Cardiovascular Research 116, 18751886.CrossRefGoogle ScholarPubMed
Ulfhammer, E et al. (2006) TNF-alpha mediated suppression of tissue type plasminogen activator expression in vascular endothelial cells is NF-kappaB- and p38 MAPK-dependent. Journal of Thrombosis and Haemostasis: JTH 4, 17811789.CrossRefGoogle ScholarPubMed
Rice, J et al. (2006) Molecular mediators of alphavbeta3-induced endothelial cell survival. Journal of Vascular Research 43, 422436.CrossRefGoogle ScholarPubMed
Jiang, Q et al. (2016) Lunasin suppresses the migration and invasion of breast cancer cells by inhibiting matrix metalloproteinase-2/-9 via the FAK/Akt/ERK and NF-κB signaling pathways. Oncology Reports 36, 253262.CrossRefGoogle ScholarPubMed
Liu, JF et al. (2018) Thrombospondin 2 promotes tumor metastasis by inducing matrix metalloproteinase-13 production in lung cancer cells. Biochemical Pharmacology 155, 537546.CrossRefGoogle ScholarPubMed
Yang, YL et al. (2004) Thrombospondin-1 mediates distal tubule hypertrophy induced by glycated albumin. The Biochemical Journal 379, 8997.CrossRefGoogle ScholarPubMed
Xiao, Q et al. (2005) Characterization of cis-regulatory elements of the vascular endothelial growth inhibitor gene promoter. The Biochemical Journal 388, 913920.CrossRefGoogle ScholarPubMed
Ho, FM et al. (2000) High glucose-induced apoptosis in human endothelial cells is mediated by sequential activations of c-Jun NH(2)-terminal kinase and caspase-3. Circulation 101, 26182624.CrossRefGoogle ScholarPubMed
Zhazykbayeva, S et al. (2020) The molecular mechanisms associated with the physiological responses to inflammation and oxidative stress in cardiovascular diseases. Biophysical Reviews 12, 947968.CrossRefGoogle ScholarPubMed
Chandrasekar, B et al. (2004) Activation of intrinsic and extrinsic proapoptotic signaling pathways in interleukin-18-mediated human cardiac endothelial cell death. The Journal of Biological Chemistry 279, 2022120233.CrossRefGoogle ScholarPubMed
Tadros, A et al. (2003) ABIN-2 protects endothelial cells from death and has a role in the antiapoptotic effect of angiopoietin-1. Blood 102, 44074409.CrossRefGoogle Scholar
Chng, HW et al. (2006) A new role for the anti-apoptotic gene A20 in angiogenesis. Experimental Cell Research 312, 28972907.CrossRefGoogle ScholarPubMed
Sabatel, C et al. (2010) Sprouty1, a new target of the angiostatic agent 16 K prolactin, negatively regulates angiogenesis. Molecular Cancer 9, 231.CrossRefGoogle ScholarPubMed
Migneault, F et al. (2020) Apoptotic exosome-like vesicles regulate endothelial gene expression, inflammatory signaling, and function through the NF-κB signaling pathway. Scientific Reports 10, 12562.CrossRefGoogle ScholarPubMed
Park, YA et al. (2014) Tumor suppressive effects of bromodomain-containing protein 7 (BRD7) in epithelial ovarian carcinoma. Clinical Cancer Research 20, 565575.CrossRefGoogle ScholarPubMed
Peng, C et al. (2007) BRD7 suppresses the growth of nasopharyngeal carcinoma cells (HNE1) through negatively regulating beta-catenin and ERK pathways. Molecular and Cellular Biochemistry 303, 141149.CrossRefGoogle ScholarPubMed
van Beijnum, JR et al. (2017) A genomic screen for angiosuppressor genes in the tumor endothelium identifies a multifaceted angiostatic role for bromodomain containing 7 (BRD7). Angiogenesis 20, 641654.CrossRefGoogle ScholarPubMed
Tromp, SC et al. (2000) Tumor angiogenesis factors reduce leukocyte adhesion in vivo. International Immunology 12, 671676.CrossRefGoogle ScholarPubMed
Luo, J et al. (1998) Angiostatin upregulates E-selectin in proliferating endothelial cells. Biochemical and Biophysical Research Communications 245, 906911.CrossRefGoogle ScholarPubMed
Dirkx, AE et al. (2006) Anti-angiogenesis therapy can overcome endothelial cell anergy and promote leukocyte-endothelium interactions and infiltration in tumors. FASEB Journal 20, 621630.CrossRefGoogle ScholarPubMed
Yu, G et al. (2005) Endothelial expression of E-selectin is induced by the platelet-specific chemokine platelet factor 4 through LRP in an NF-kappaB-dependent manner. Blood 105, 35453551.CrossRefGoogle Scholar
Armulik, A, Genové, G and Betsholtz, C (2011) Pericytes: developmental, physiological, and pathological perspectives, problems, and promises. Developmental Cell 21, 193215.CrossRefGoogle ScholarPubMed
Zuazo-Gaztelu, I and Casanovas, O (2018) Unraveling the role of angiogenesis in cancer ecosystems. Frontiers in Oncology 8, 248.CrossRefGoogle ScholarPubMed
Fraisl, P et al. (2009) Regulation of angiogenesis by oxygen and metabolism. Developmental Cell 16, 167179.CrossRefGoogle ScholarPubMed
Goel, S et al. (2011) Normalization of the vasculature for treatment of cancer and other diseases. Physiological Reviews 91, 10711121.CrossRefGoogle ScholarPubMed
Chen, ZJ et al. (2016) Activation of GPER suppresses epithelial mesenchymal transition of triple negative breast cancer cells via NF-κB signals. Molecular Oncology 10, 775788.CrossRefGoogle ScholarPubMed
Zhuang, SF et al. (2017) Atg7 regulates brain angiogenesis via NF-κB-dependent IL-6 production. International Journal of Molecular Sciences 18, 968.CrossRefGoogle ScholarPubMed
Ramadass, V, Vaiyapuri, T and Tergaonkar, V (2020) Small molecule NF-κB pathway inhibitors in clinic. International Journal of Molecular Sciences 21, 5164.CrossRefGoogle ScholarPubMed
Huang, JJ et al. (2014) Recent advances in the structure-based and ligand-based design of IKKβ inhibitors as anti-inflammation and anti-cancer agents. Current Medicinal Chemistry 21, 38933917.CrossRefGoogle ScholarPubMed
Liu, J et al. (2022) Ibrutinib inhibits angiogenesis and tumorigenesis in a BTK-independent manner. Pharmaceutics 14, 1876.CrossRefGoogle Scholar
Wang, X, Zhang, Z and Yao, C (2012) Bortezomib inhibits the angiogenesis mediated by mesenchymal stem cells. Cancer Investigation 30, 657662.CrossRefGoogle ScholarPubMed
Li, Y et al. (2015) Copper improves the anti-angiogenic activity of disulfiram through the EGFR/Src/VEGF pathway in gliomas. Cancer Letters 369, 8696.CrossRefGoogle ScholarPubMed
Gravina, GL et al. (2015) KPT-330, a potent and selective exportin-1 (XPO-1) inhibitor, shows antitumor effects modulating the expression of cyclin D1 and survivin [corrected] in prostate cancer models. BMC Cancer 15, 941.CrossRefGoogle ScholarPubMed
Singh, AK, Bishayee, A and Pandey, AK (2018) Targeting histone deacetylases with natural and synthetic agents: an emerging anticancer strategy. Nutrients 10, 731.CrossRefGoogle ScholarPubMed
Kunnumakkara, AB et al. (2008) Curcumin sensitizes human colorectal cancer xenografts in nude mice to gamma-radiation by targeting nuclear factor-kappaB-regulated gene products. Clinical Cancer Research 14, 21282136.CrossRefGoogle ScholarPubMed
Chiang, IT et al. (2014) Curcumin synergistically enhances the radiosensitivity of human oral squamous cell carcinoma via suppression of radiation-induced NF-κB activity. Oncology Reports 31, 17291737.CrossRefGoogle ScholarPubMed
Li, J et al. (2015) Combination of curcumin and bicalutamide enhanced the growth inhibition of androgen-independent prostate cancer cells through SAPK/JNK and MEK/ERK1/2-mediated targeting NF-κB/p65 and MUC1-C. Journal of Experimental & Clinical Cancer Research: CR 34, 46.CrossRefGoogle ScholarPubMed
Marquardt, JU et al. (2015) Curcumin effectively inhibits oncogenic NF-κB signaling and restrains stemness features in liver cancer. Journal of Hepatology 63, 661669.CrossRefGoogle ScholarPubMed
Li, Y and Zhang, T (2014) Targeting cancer stem cells by curcumin and clinical applications. Cancer Letters 346, 197205.CrossRefGoogle ScholarPubMed
Dhillon, N et al. (2008) Phase II trial of curcumin in patients with advanced pancreatic cancer. Clinical Cancer Research 14, 44914499.CrossRefGoogle ScholarPubMed
Anand, P et al. (2007) Bioavailability of curcumin: problems and promises. Molecular Pharmaceutics 4, 807818.CrossRefGoogle ScholarPubMed
Long, Q et al. (2013) Induction of apoptosis and inhibition of angiogenesis by PEGylated liposomal quercetin in both cisplatin-sensitive and cisplatin-resistant ovarian cancers. Journal of Biomedical Nanotechnology 9, 965975.CrossRefGoogle ScholarPubMed
Lei, CS et al. (2018) Effects of quercetin combined with anticancer drugs on metastasis-associated factors of gastric cancer cells: in vitro and in vivo studies. The Journal of Nutritional Biochemistry 51, 105113.CrossRefGoogle ScholarPubMed
Huang, DY et al. (2018) Inhibition of EGF expression and NF-κB activity by treatment with quercetin leads to suppression of angiogenesis in nasopharyngeal carcinoma. Saudi Journal of Biological Sciences 25, 826831.CrossRefGoogle ScholarPubMed
Granado-Serrano, AB et al. (2010) Quercetin modulates NF-kappa B and AP-1/JNK pathways to induce cell death in human hepatoma cells. Nutrition and Cancer 62, 390401.CrossRefGoogle ScholarPubMed
McCann, SE et al. (2005) Intakes of selected nutrients, foods, and phytochemicals and prostate cancer risk in western New York. Nutrition and Cancer 53, 3341.CrossRefGoogle ScholarPubMed
Nieman, DC et al. (2010) Quercetin's influence on exercise performance and muscle mitochondrial biogenesis. Medicine and Science in Sports and Exercise 42, 338345.CrossRefGoogle ScholarPubMed
Brito, AF et al. (2016) New approach for treatment of primary liver tumors: the role of quercetin. Nutrition and Cancer 68, 250266.CrossRefGoogle ScholarPubMed
Alvero, AB et al. (2009) Stem-like ovarian cancer cells can serve as tumor vascular progenitors. Stem Cells 27, 24052413.CrossRefGoogle ScholarPubMed
Huang, KF et al. (2014) Kallistatin, a novel anti-angiogenesis agent, inhibits angiogenesis via inhibition of the NF-κB signaling pathway. Biomedicine & pharmacotherapy = Biomédecine & Pharmacothérapie 68, 455461.CrossRefGoogle ScholarPubMed
Tabruyn, SP et al. (2005) The antiangiogenic factor, 16-kDa human prolactin, induces endothelial cell cycle arrest by acting at both the G0-G1 and the G2-M phases. Molecular Endocrinology 19, 19321942.CrossRefGoogle ScholarPubMed
Tabruyn, SP et al. (2003) The antiangiogenic factor 16K human prolactin induces caspase-dependent apoptosis by a mechanism that requires activation of nuclear factor-kappaB. Molecular Endocrinology 17, 18151823.CrossRefGoogle ScholarPubMed
Chen, YH et al. (2006) Anti-angiogenesis mediated by angiostatin K1-3, K1-4 and K1-4.5. Involvement of p53, FasL, AKT and mRNA deregulation. Thrombosis and Haemostasis 95, 668677.CrossRefGoogle ScholarPubMed
Hindler, K et al. (2006) The role of statins in cancer therapy. The Oncologist 11, 306315.CrossRefGoogle ScholarPubMed
Nakata, S et al. (2007) Statin treatment upregulates vascular neuronal nitric oxide synthase through Akt/NF-kappaB pathway. Arteriosclerosis, Thrombosis, and Vascular Biology 27, 9298.CrossRefGoogle ScholarPubMed
Gingras, D et al. (2004) Activation of tissue plasminogen activator gene transcription by Neovastat, a multifunctional antiangiogenic agent. Biochemical and Biophysical Research Communications 320, 205212.CrossRefGoogle ScholarPubMed
Simard, B et al. (2013) Shark cartilage extract induces cytokines expression and release in endothelial cells and induces E-selectin, plasminogen and t-PA genes expression through an antioxidant-sensitive mechanism. Cytokine 61, 104111.CrossRefGoogle ScholarPubMed
Escudier, B et al. (2007) Prognostic factors of metastatic renal cell carcinoma after failure of immunotherapy: new paradigm from a large phase III trial with shark cartilage extract AE 941. The Journal of Urology 178, 19011905.CrossRefGoogle ScholarPubMed
Lu, C et al. (2010) Chemoradiotherapy with or without AE-941 in stage III non-small cell lung cancer: a randomized phase III trial. Journal of the National Cancer Institute 102, 859865.CrossRefGoogle ScholarPubMed
Loprinzi, CL et al. (2005) Evaluation of shark cartilage in patients with advanced cancer: a North Central Cancer Treatment Group trial. Cancer 104, 176182.CrossRefGoogle Scholar
Figure 0

Figure 1. Activation of canonical and non-canonical NF-κB pathways. The canonical NF-κB pathway (left) is triggered by a variety of inflammatory factors and mediated by activation of the IKK complex, leading to transient nuclear translocation of NF-κB heterodimers. The non-canonical NF-κB pathway (right) is stimulated by specific receptors, dependent on NIK and IKKα, and mediates the sustained activation of the RelB/p52 complex.

Figure 1

Figure 2. Multiple mediators are shared in the molecular pathways of pro-inflammatory response and angiogenesis. TNF-α induces NF-κB signalling pathway to initiate inflammatory response and HIF-1α transcription factor involved in angiogenesis is regulated by NF-κB under hypoxic condition. MicroRNAs regulate inflammation-related angiogenesis by acting on the downstream signalling molecules in NF-κB-mediated pro-inflammatory responses. In addition, the Nrf2/HO-1 axis that interacts with HIF-1α can regulate hypoxia-induced angiogenesis by targeting VEGF.

Figure 2

Figure 3. The positive regulation mechanism of NF-κB on angiogenesis by transcription target gene. Under the action of inflammatory cytokines, NF-κB induces the expression of various pro-inflammatory genes and adhesion molecules in endothelial cells, which in turn promoted the accumulation of inflammatory cells in perivascular matrix and enhanced the activation of NF-κB. Activated NF-κB can not only make endothelial cells directly secrete VEGF, but also bind to the upstream promoter of VEGFR2 and activate its transcription. Once VEGFR2 binds to vascular endothelial growth factor, VEGFR2 activates a series of downstream mediators to promote angiogenesis. In addition, activated NF-κB can stimulate the expression of matrix metalloproteinases, promote the degradation of basement membrane and extracellular matrix. Nuclear transcription of NF-κB also upregulates inflammatory chemokines IL-6 and IL-8, thereby stimulating the proliferation and migration of tumour endothelial cells. Meanwhile, activated NF-κB can promote tumour angiogenesis by upregulating MCP-1 and COX-2. In non-canonical NF-κB pathway, NIK upregulates the expression of chemokine CXCL12, which plays an important role in the homing of endothelial progenitor cells.

Figure 3

Figure 4. NF-κB promotes angiogenesis under different pathological conditions. In the hypoxic tumour microenvironment, hypoxia renders PHDs that inhibit IKKβ activity ineffective. Activated NF-κB is transported to the nucleus and induces HIF-1α transcription, which promotes the expression of angiogenic factors. In endothelial cells of renal ischaemia/reperfusion model, LPS-induced NF-κB regulates HIF-2α expression and increases NO production, thereby improving renal microvascular perfusion.

Figure 4

Figure 5. The negative regulation mechanism of NF-κB on angiogenesis by transcription target gene. On the one hand, NF-κB activates the expression of TIMP-1, PAI-1 and TSP-1, 2, resulting in decreased extracellular matrix degradation and hindering angiogenesis. On the other hand, NF-κB nuclear transcription can activate apoptosis signalling pathway, induce the expression of downstream pro-apoptotic genes such as JNK, Fas, FasL, caspase-3, thus promoting endothelial cell apoptosis. In addition, activated NF-κB induces the upregulation of VEGI, thereby inhibiting endothelial cell proliferation and growth.

Figure 5

Figure 6. Many NF-κB inhibitors show inhibitory effect on angiogenesis. The list of NFkB inhibitors of which the effects on angiogenesis have been examined in vitro and in vivo. The inhibitors including IKK inhibitors (CHS-828, IMD-1041 and EB-1627), BTK inhibitors (zanubrutinib, acalabrutinib and ibrutinib), proteasome inhibitors (disulfiram and bortezomib), nuclear translocation inhibitor (selinexor), histone deacetylase inhibitors (vorinostat, romidepsin, belinostat, panobinostat, azacitidine and decitabine) and natural products (curcumin, quercetin and neovastat).