scispace - formally typeset
Search or ask a question
Journal ArticleDOI

Roles of NF-κB in Cancer and Inflammatory Diseases and Their Therapeutic Approaches.

29 Mar 2016-Cells (Multidisciplinary Digital Publishing Institute)-Vol. 5, Iss: 2, pp 15
TL;DR: In this article, the authors proposed to inhibit NF-κB signaling in cancer and inflammatory diseases, such as rheumatoid arthritis, atherosclerosis, inflammatory bowel diseases, multiple sclerosis and malignant tumors.
Abstract: Nuclear factor-κB (NF-κB) is a transcription factor that plays a crucial role in various biological processes, including immune response, inflammation, cell growth and survival, and development. NF-κB is critical for human health, and aberrant NF-κB activation contributes to development of various autoimmune, inflammatory and malignant disorders including rheumatoid arthritis, atherosclerosis, inflammatory bowel diseases, multiple sclerosis and malignant tumors. Thus, inhibiting NF-κB signaling has potential therapeutic applications in cancer and inflammatory diseases.

Content maybe subject to copyright    Report

cells
Review
Roles of NF-κB in Cancer and Inflammatory Diseases
and Their Therapeutic Approaches
Mi Hee Park and Jin Tae Hong *
College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31,
Osongsaengmyeong 1-ro, Osong-eup, Cheongwon-gun, Chungbuk 28160, Korea; pmh5205@hanmail.net
* Correspondence: jinthong@chungbuk.ac.kr; Tel.: +82-43-261-2813; Fax: +82-43-268-2732
Academic Editor: Ruaidhri Carmody
Received: 22 February 2016; Accepted: 24 March 2016; Published: 29 March 2016
Abstract:
Nuclear factor-
κ
B (NF-
κ
B) is a transcription factor that plays a crucial role in various
biological processes, including immune response, inflammation, cell growth and survival, and
development. NF-
κ
B is critical for human health, and aberrant NF-
κ
B activation contributes
to development of various autoimmune, inflammatory and malignant disorders including
rheumatoid arthritis, atherosclerosis, inflammatory bowel diseases, multiple sclerosis and malignant
tumors. Thus, inhibiting NF-
κ
B signaling has potential therapeutic applications in cancer and
inflammatory diseases.
Keywords:
NF-
κ
B; canonical pathway; non-canonical pathway; cancer; inflammatory disease;
therapeutic approaches
1. Introduction
The nuclear factor-
κ
B (NF-
κ
B) family of transcription factors control the expression of genes
involved in many critical physiological responses such as inflammatory responses, proliferation,
differentiation, cell adhesion and apoptosis [
1
]. NF-
κ
B transcription complexes have a variety of
homo- and heterodimers consisting of the subunits p50, p52, c-Rel, RelA (p65) and RelB [
2
]. NF-
κ
B
signaling pathways can be divided into canonical and noncanonical pathways. In the canonical
pathway, I kappa B kinase (IKK) phosphorylates I
κ
B
α
at two N-terminal serines, triggering its
ubiquitination and proteasomal degradation; this leads to the nuclear translocation of NF-
κ
B
complexes, predominantly p50/RelA and p50/c-Rel dimers [
3
]. The noncanonical NF-
κ
B pathway
involves different signaling molecules and leads to the activation of the p52/RelB dimer [4].
NF-
κ
B is able to induce several of these cellular alterations and has been shown to be constitutively
activated in some types of cancer cells. Constitutively activated NF-
κ
B transcription factors have
been associated with several aspects of tumorigenesis, including promoting cancer-cell proliferation,
preventing apoptosis, and increasing a tumor
'
s angiogenic and metastatic potential. Activation of the
NF-
κ
B/Rel by nuclear translocation plays a role in inflammation through induction of transcription
of several proinflammatory genes [
5
]. Recent data indicate that activation of IKK-
β
, rather than
IKK-
α
, participates in the primary pathway of proinflammatory genes [
6
]. IKK-
β
is expressed
in fibroblast-like synoviocytes and plays a central role in TNF-
α
–mediated NF-
κ
B activation and
expression of proinflammatory genes [
7
]. IKK-
β
also activates NF-
κ
B and inflammatory gene
transcription in monocytes and CD4+ T lymphocytes [
7
]. Many natural products and drugs that have
been involved in anti-cancer and anti-inflammatory activity have also been shown to inhibit NF-κB.
This review provides the signaling mechanisms and biological functions of the NF-
κ
B pathway,
and the role of NF-
κ
B in cancer and inflammatory diseases, and the multitude of NF-
κ
B inhibitors that
have been reported.
Cells 2016, 5, 15; doi:10.3390/cells5020015 www.mdpi.com/journal/cells

Cells 2016, 5, 15 2 of 13
2. NF-κB
Nuclear factor-
κ
B (NF-
κ
B) is a transcription factor that plays a crucial role in various biological
processes, including immune response, inflammation, cell growth and survival, and development [
1
,
8
].
NF-
κ
B is activated by various inflammatory stimuli such as growth factors and infectious microbes.
NF-
κ
B controls expression of a number of genes that regulate immune responses, cell growth and
proliferation, survival and apoptosis, stress responses and embryogenesis and development of a
variety of stimuli [
7
,
9
]. NF-
κ
B is critical for human health, and aberrant NF-
κ
B activation contributes
to development of various autoimmune, inflammatory and malignant disorders including rheumatoid
arthritis, atherosclerosis, inflammatory bowel diseases, multiple sclerosis and malignant tumors [
10
,
11
].
2.1. NF-κB Subunits
The mammalian NF-
κ
B family is composed of five members, including RelA (p65), RelB, c-Rel,
NF-
κ
B1 p50, and NF-
κ
B2 p52, which form various dimeric complexes that transactivate numerous
target genes via binding to the
κ
B enhancer [
2
]. The NF-
κ
B proteins are normally sequestered in the
cytoplasm by a family of inhibitors, including I
κ
B
α
and other ankyrin repeat-containing proteins [
6
,
12
].
Proteasome-mediated processing of p105 and p100 produces the mature NF-
κ
B1 and NF-
κ
B2 proteins
(p50 and p52) and results in disruption of the IκB-like function of these precursor proteins [13].
The NF-
κ
B transcription factor family in mammals consists of five proteins including p65
(RelA), RelB, c-Rel, p105/p50 (NF-
κ
B1), and p100/52 (NF-
κ
B2) that associate with each other to form
distinct transcriptionally active homo- and heterodimeric complexes [
13
,
14
]. Through combinatorial
associations, the Rel protein family members can form up to 15 different dimers. Among them,
the p50/65 heterodimer clearly represents the most abundant of Rel dimers, being found in almost
all cell types [
15
]. In addition, dimeric complexes of p65/p65, p65/c-Rel, p65/p52, c-Rel/c-Rel,
p52/c-Rel, p50/c-Rel, p50/p50, RelB/p50, and RelB/p52 have been described, some of them only in
limited subsets of cells [
10
12
]. NF-
κ
B family shares a Rel homology domain in their N-terminus. A
subfamily of NF-
κ
B proteins, including RelA (p65), RelB and c-Rel has a transactivation domain in
their C-termini [
16
,
17
]. After processing of p105 and p100 by the ubiquitin/proteasome pathway that
involves selective degradation of their C-terminal region containing ankyrin repeats, mature NF-
κ
B
subunits such as p50 and p52 are generated [
16
,
17
]. Actually, the p50 and p52 proteins have no intrinsic
ability to activate transcription and act as transcriptional repressors when binding
κ
B elements as
homodimers [18].
2.2. NF-κB Signaling Pathway
The NF-
κ
B dimers are sequestered in the cytoplasm by a family of inhibitors, called I
κ
Bs (Inhibitor
of
κ
B), which are proteins that contain multiple copies of a sequence called ankyrin repeats, in
unstimulated cells [
5
,
14
]. The I
κ
B proteins mask the nuclear localization signals (NLS) of NF-
κ
B
proteins and keep them sequestered in an inactive state in the cytoplasm by virtue of their ankyrin
repeat domains [
11
,
16
]. Because the presence of ankyrin repeats in their C-terminal halves, p105
and p100 also function as I
κ
B proteins. The C-terminal half of p100, that is often referred to as I
κ
B
δ
,
also functions as an inhibitor [
19
]. I
κ
B
δ
degradation in response to developmental stimuli, such as
those transduced through LT
β
R, potentiate NF-
κ
B dimer activation in a NIK dependent non-canonical
pathway [20].
2.2.1. Canonical Pathway
Canonical NF-
κ
B pathway of NF-
κ
B is activated after degradation of I
κ
B
α
, which results in nuclear
translocation of various NF-
κ
B complexes, predominantly the p50/p65 dimer [
3
] (Figure 1). The
degradation of I
κ
B
α
is mediated by phosphorylation through the I
κ
B kinase (IKK), a trimeric complex
composed of two catalytic subunits, IKK
α
and IKK
β
, and a regulatory subunit, IKK
γ
(also termed
NF-
κ
B essential modulator or NEMO) [
21
]. When activated by signals, the I
κ
B kinase phosphorylates

Cells 2016, 5, 15 3 of 13
two serine residues located in an I
κ
B regulatory domain [
19
,
20
]. When phosphorylated on these serines
(e.g., serines 32 and 36 in human I
κ
B
α
), the I
κ
B inhibitor molecules are processed by ubiquitination,
which then leads them to be degraded by a cell structure called the proteasome [
22
,
23
] . With the
degradation of I
κ
B, the NF-
κ
B complex then enters into the nucleus where it can
'
turn on
'
the expression
of several genes that have DNA-binding sites for NF-
κ
B [
22
,
23
]. The activation of these genes by
NF-
κ
B then leads to the given physiological response, for example, an inflammatory or immune
response, a cell survival response, or cellular proliferation [
24
]. NF-
κ
B turns on expression of its own
repressor, I
κ
B
α
. The newly synthesized I
κ
B
α
then re-inhibits NF-
κ
B and forms an auto feedback loop,
which results in oscillating levels of NF-
κ
B activity [
22
,
23
]. Genetic evidence suggests that this NF-
κ
B
pathway regulates important biological functions, such as lymphoid organogenesis, B-cell survival
and maturation, dendritic cell activation, and bone metabolism.
κ
κ α κ
κ κ
κ
κ
κ
κ α κ α κ
κ
κ
κ
β
β κ κ
κ κ
α
κ
κ
κ κ
κ α
κ
κ
Figure 1.
The canonical NF-
κ
B pathway (
left
) induced by signals including antigens, TLR ligands
and cytokines such as TNF uses a wide variety of signaling adaptors to engage and activate the
IKK
β
subunit of the IKK complex. IKK
β
phosphorylation of I
κ
B proteins bound to NF-
κ
B dimers
results in ubiquitination (Ub) of I
κ
B and proteasome-induced degradation. This allows NF-
κ
B to enter
the nucleus and be involved in controlling the transcription of gene encoding functions as diverse as
inflammation, cell survival and cell division. The noncanonical pathway (
right
) engaged in by members
of the TNF-like family of cytokines requires NIK to activate IKK
α
, which then phosphorylates p100
(NF-
κ
B2), triggering its proteosomal processing needed for the activation of p52-RelB dimers. Among its
functions, this specific NF-
κ
B heterodimer controls gene expression crucial for lymphoid organogenesis.
2.2.2. Non-Canonical Pathway
The non-canonical NF-
κ
B pathway activates the RelB/p52 NF-
κ
B complex using a mechanism that
relies on the inducible processing of p100 instead of degradation of I
κ
B
α
(Figure 1). The processing of
p100 serves to both generate p52 and induce the nuclear translocation of the RelB/p52 heterodimer [
4
].
The discovery of non-canonical NF-
κ
B signaling pathway came from the study of p100 processing [
25
].
In contrast to the constitutive and co-translational processing of p105, the processing of p100 is tightly
regulated. In most cell types, p100 is the predominant product of NF-
κ
B2 [
26
,
27
]. Overexpressed
p100 is barely converted to p52 in mammalian cells, as opposed to the constitutive production of p50
from p105 [
25
]. However, p52 is actively generated in specific cell types, such as B cells, leading to

Cells 2016, 5, 15 4 of 13
the idea that p100 processing might be a signal-regulated event [
25
,
28
]. Indeed, the NF-
κ
B-inducing
kinase (NIK) induces p100 processing and is required for p100 processing in splenocytes. Moreover,
endogenous p100 processing can be activated by various receptor signals in an NIK-dependent
manner [29,30].
In this pathway, activation of the NF-
κ
B inducing kinase (NIK) led to the phosphorylation and
subsequent proteasomal processing of the NF-
κ
B2 precursor protein p100 into mature p52 subunits
in an IKK1/IKKa dependent manner [
31
]. Then, p52 dimerizes with RelB to appear as a nuclear
RelB/p52 DNA binding activity and regulate a distinct class of genes [
32
]. In contrast to the canonical
signaling that relies upon NF-kB essential modulator (NEMO)-IKK2 mediated degradation of I
κ
B
α
, -
β
,
-
ε
, the non-canonical signaling critically depends on NIK mediated processing of p100 into p52 [
30
,
31
].
Recent studies showed that synthesis of the constituents of the non-canonical pathway, RelB and
p52, is controlled by the canonical IKK2-I
κ
B-RelA: p50 signaling [
30
,
31
]. These studies suggest that
an integrated NF-
κ
B system network underlies activation of both RelA and RelB containing dimer
and that a malfunctioning canonical pathway will lead to an aberrant cellular response also through
the non-canonical pathway [
30
,
31
]. Deregulated non-canonical NF-
κ
B signaling is associated with
lymphoid malignancies [
28
,
33
]. Cell-differentiating or developmental stimuli such as B-Cell activation
factor (BAFF), receptor activator of nuclear factor kappa-B ligand (RANKL) or lymphotoxin-
α
, activate
the non-canonical NF-κB pathway [34].
3. Role of NF-κB in Diseases
NF-
κ
B activation affects hallmarks of cancer and inflammatory diseases through the transcription
of genes involved in cell proliferation, survival, angiogenesis, inflammation and tumor promotion and
metastasis as shown in Figure 2.
κ
κ
κ
κ α
β ε
κ
κ
κ
α κ
κ
κ
κ
Figure 2.
NF-
κ
B activation affects hallmarks of cancer and inflammatory diseases through the
transcription of genes involved in cell proliferation, survival, angiogenesis, inflammation and tumor
promotion and metastasis. BCL2, B-cell lymphoma protein 2; BCL-XL, also known as BCL2-like 1; BFL1,
also known as BCL2A1; CDK2, cyclin-dependent kinase 2; COX2, cyclooxygenase 2; CXCL, chemokine
(C-X-C motif) ligand; DR, death receptor; ELAM1, endothelial adhesion molecule 1; FLIP, also
known as CASP8; GADD45beta, growth arrest and DNA-damage-inducible protein beta; HIF1alpha,
hypoxia-inducible factor 1alpha; ICAM1, intracellular adhesion molecule 1; IEX-1L, radiation-inducible
immediate early gene (also known as IER3); IL, interleukin; iNOS, inducible nitric oxide synthase;
KAL1, Kallmann syndrome 1 sequence; MCP1, monocyte chemoattractant protein 1 (also known
as CCL2); MIP2, macrophage inflammatory protein 2; MMP, matrix metalloproteinase; MnSOD,
manganese superoxide dismutase (also known as SOD2); TNF, tumour necrosis factor; TRAF, TNF
receptor-associated factor; uPA, urokinase plasminogen activator; VCAM1, vascular cell adhesion
molecule 1; VEGF, vascular endothelial growth factor; XIAP, X-linked inhibitor of apoptosis protein.

Cells 2016, 5, 15 5 of 13
3.1. NF-κB and Cancer
NF-
κ
B regulates the genes that control cell proliferation and cell survival [
35
]. Many different
types of human tumors have misregulated NF-
κ
B; that is, NF-
κ
B is constitutively active. Active
NF-
κ
B turns on the expression of genes that keeps the cell proliferating and protects the cell from
conditions that would otherwise cause it to die via apoptosis [
36
]. Cancer-associated chromosomal
translocations, deletions and mutations might also disrupt genes that encode NF-
κ
B and I
κ
B proteins,
uncoupling NF-
κ
B factors from their regulators and causing constitutive NF-
κ
B activation [
37
].
Constitutively activated NF-
κ
B transcription factors have been associated with several aspects of
tumorigenesis, including promoting cancer-cell proliferation, preventing apoptosis, and increasing a
tumor
'
s angiogenic and metastatic potential [
37
,
38
]. In tumor cells, NF-
κ
B is consequently activated
because mutations in genes encoding the NF-
κ
B transcription factors themselves or in genes that
control NF-
κ
B activity. In addition, some tumor cells secrete factors that cause NF-
κ
B to become
active [
39
,
40
]. Blocking NF-
κ
B can cause tumor cells to stop proliferating, to die, or to become more
sensitive to the action of anti-tumor agents [
41
]. NF-
κ
B stimulates the transcription of genes that
encode G1 cyclins [
1
,
40
]. A
κ
B site is present within the cyclin D1 promoter and there is strong
evidence that NF-
κ
B dependent cyclin D1 induction drives the proliferation of mammary epithelial
cells during pregnancy [
42
,
43
]. NF-
κ
B is also an inhibitor of programmed cell death [
44
,
45
]. This factor
activates the transcription of several target genes that block the induction of apoptosis by TNF-
α
and
other pro-apoptotic members of this family [
22
]. The anti-apoptotic factors that are induced by NF-
κ
B
include cellular inhibitors of apoptosis (cIAPs), caspase-8/FADD (FAS-associated death domain)-like
IL-1beta-converting enzyme (FLICE) inhibitory protein (c-FLIP) and members of the BCL2 family
(such as A1/BFL1 and BCL-XL) [
22
]. Cells with elevated NF-
κ
B activity deregulate production of
chemokines, which increases migratory activity [
1
]. At least one NF-
κ
B-regulated chemokine, IL-8, has
been shown to promote angiogenesis [
46
]. In addition,
κ
B sites were identified in the promoters of
genes that encode several matrix metalloproteinases (MMPs) that are proteolytic enzymes involved
in promoting tumor invasion of surrounding tissue [
47
]. NF-
κ
B contributes to extracellular matrix
destruction by cancer cells [
48
,
49
]. NF-
κ
B has also been shown to be involved in the development of
carcinomas—cancers of epithelial origin, such as breast cancer [
50
]. Several studies have documented
elevated or constitutive NF-
κ
B DNA-binding activity both in mammary carcinoma cell lines and
primary breast cancer cells [51,52].
In inflammatory cells, continuous NF-
κ
B activity could promote the production of reactive oxygen
species, thereby damaging DNA of surrounding epithelial cells [
53
]. Some of the best circumstantial
evidence that supports such a role for NF-
κ
B comes from various gastrointestinal cancers [
54
]. NF-
κ
B
activation is also associated with colorectal cancer. Colon cancer cell lines, human tumor samples, and
stromal macrophages in sporadic adenomatous polyps also have increased NF-
κ
B activity [
55
]. It has
been shown that canonical NF-
κ
B is a Fas transcription activator and the alternative NF-
κ
B is a Fas
transcription repressor [56].
3.2. NF-κB and Inflammatory Disease
NF-
κ
B is a major transcription factor that regulates genes responsible for both the innate and
adaptive immune response [
57
]. After activation of T- or B-cell receptors, NF-
κ
B is activated through
distinct signaling [
58
]. Upon ligation of the T-cell receptor, protein kinase Lck is recruited and
phosphorylates the immunoreceptor tyrosine-based activation motifs (ITAMs) of the CD3 cytoplasmic
tail [
59
]. ZAP70 is then recruited to the phosphorylated ITAMs and helps recruit Linker-for-activation
of T cells (LAT) and Phospholipase C (PLC)-
γ
, which causes activation of Protein kinase C (PKC) [
60
].
Through a cascade of phosphorylation, the kinase complex is activated and NF-
κ
B enter the nucleus
to upregulate genes involved in T-cell proliferation, maturation and development [
61
]. NF-
κ
B
is chronically activated in many inflammatory diseases, including inflammatory bowel disease,
arthritis, sepsis, gastritis, asthma, atherosclerosis and others [
62
]. It is important to note, though,
that elevation of some NF-κB activators, such as osteoprotegerin (OPG), are associated with elevated

Citations
More filters
Journal ArticleDOI
TL;DR: A better understanding of the mechanisms regulating adipose tissue expansion in obesity is required for the development of future therapeutic approaches in obesity-associated metabolic complications.
Abstract: Obesity is a critical risk factor for the development of type 2 diabetes (T2D), and its prevalence is rising worldwide. White adipose tissue (WAT) has a crucial role in regulating systemic energy homeostasis. Adipose tissue expands by a combination of an increase in adipocyte size (hypertrophy) and number (hyperplasia). The recruitment and differentiation of adipose precursor cells in the subcutaneous adipose tissue (SAT), rather than merely inflating the cells, would be protective from the obesity-associated metabolic complications. In metabolically unhealthy obesity, the storage capacity of SAT, the largest WAT depot, is limited, and further caloric overload leads to the fat accumulation in ectopic tissues (e.g., liver, skeletal muscle, and heart) and in the visceral adipose depots, an event commonly defined as “lipotoxicity.” Excessive ectopic lipid accumulation leads to local inflammation and insulin resistance (IR). Indeed, overnutrition triggers uncontrolled inflammatory responses in WAT, leading to chronic low-grade inflammation, therefore fostering the progression of IR. This review summarizes the current knowledge on WAT dysfunction in obesity and its associated metabolic abnormalities, such as IR. A better understanding of the mechanisms regulating adipose tissue expansion in obesity is required for the development of future therapeutic approaches in obesity-associated metabolic complications.

708 citations

Journal ArticleDOI
Zhen Qiu1, Yuhong He1, Hao Ming1, Shaoqing Lei1, Yan Leng1, Zhongyuan Xia1 
TL;DR: LPS could increase the sensitivity of H9C2 cells to HG and H/R and aggravated HG- andH/R-induced H 9C2 cell injury by promoting ROS production to induce NLRP3 inflammasome-mediated pyroptosis.
Abstract: Diabetes aggravates myocardial ischemia-reperfusion (I/R) injury because of the combination effects of changes in glucose and lipid energy metabolism, oxidative stress, and systemic inflammatory response. Studies have indicated that myocardial I/R may coincide and interact with sepsis and inflammation. However, the role of LPS in hypoxia/reoxygenation (H/R) injury in cardiomyocytes under high glucose conditions is still unclear. Our objective was to examine whether lipopolysaccharide (LPS) could aggravate high glucose- (HG-) and hypoxia/reoxygenation- (H/R-) induced injury by upregulating ROS production to activate NLRP3 inflammasome-mediated pyroptosis in H9C2 cardiomyocytes. H9C2 cardiomyocytes were exposed to HG (30 mM) condition with or without LPS, along with caspase-1 inhibitor (Ac-YVAD-CMK), inflammasome inhibitor (BAY11-7082), ROS scavenger N-acetylcysteine (NAC), or not for 24 h, then subjected to 4 h of hypoxia followed by 2 h of reoxygenation (H/R). The cell viability, lactate dehydrogenase (LDH) release, caspase-1 activity, and intracellular ROS production were detected by using assay kits. The incidence of pyroptosis was detected by calcein-AM/propidium iodide (PI) double staining kit. The concentrations of IL-1β and IL-18 in the supernatants were assessed by ELISA. The mRNA levels of NLRP3, ASC, and caspase-1 were detected by qRT-PCR. The protein levels of NF-κB p65, NLRP3, ASC, cleaved caspase-1 (p10), IL-1β, and IL-18 were detected by western blot. The results indicated that pretreatment LPS with 1 μg/ml not 0.1 μg/ml could efficiently aggravate HG and H/R injury by activating NLRP3 inflammasome to mediate pyroptosis in H9C2 cells, as evidenced by increased LDH release and decreased cell viability in the cells, and increased expression of NLRP3, ASC, cleaved caspase-1 (p10), IL-1β, and IL-18. Meanwhile, Ac-YVAD-CMK, BAY11-7082, or NAC attenuated HG- and H/R-induced H9C2 cell injury with LPS stimulated by reversing the activation of NLRP3 inflammasome-mediated pyroptosis. In conclusion, LPS could increase the sensitivity of H9C2 cells to HG and H/R and aggravated HG- and H/R-induced H9C2 cell injury by promoting ROS production to induce NLRP3 inflammasome-mediated pyroptosis.

181 citations


Cites background from "Roles of NF-κB in Cancer and Inflam..."

  • ...ROS can activate the expression of NF-κB which is a crucial transcription factor in inflammation, stress response, and cell growth and survival [53]....

    [...]

Journal ArticleDOI
TL;DR: This review summarizes the current knowledge of ROS as atypical paracrine signaling molecules for field cancerization and metastasis and offers a model that placed these events with ROS as the focal instigating agent in a broader “seed-soil” hypothesis.
Abstract: Field cancerization and metastasis are the leading causes for cancer recurrence and mortality in cancer patients. The formation of primary, secondary tumors or metastasis is greatly influenced by multifaceted tumor-stroma interactions, in which stromal components of the tumor microenvironment (TME) can affect the behavior of the cancer cells. Many studies have identified cytokines and growth factors as cell signaling molecules that aid cell to cell communication. However, the functional contribution of reactive oxygen species (ROS), a family of volatile chemicals, as communication molecules are less understood. Cancer cells and various tumor-associated stromal cells produce and secrete a copious amount of ROS into the TME. Intracellular ROS modulate cell signaling cascades that aid in the acquisition of several hallmarks of cancers. Extracellular ROS help to propagate, amplify, and effectively create a mutagenic and oncogenic field which facilitate the formation of multifoci tumors and act as a springboard for metastatic tumor cells. In this review, we summarize our current knowledge of ROS as atypical paracrine signaling molecules for field cancerization and metastasis. Field cancerization and metastasis are often discussed separately; we offer a model that placed these events with ROS as the focal instigating agent in a broader “seed-soil” hypothesis.

168 citations

Journal ArticleDOI
TL;DR: An overview on chemo preventive of ITCs against several types of cancer cell lines and the molecular interaction(s) of the antagonistic effect of BITC, PEITC and SFN with Nrf2 and NF-κB to prevent cancer are discussed.
Abstract: Glucosinolates (GSL) are naturally occurring β-d-thioglucosides found across the cruciferous vegetables. Core structure formation and side-chain modifications lead to the synthesis of more than 200 types of GSLs in Brassicaceae. Isothiocyanates (ITCs) are chemoprotectives produced as the hydrolyzed product of GSLs by enzyme myrosinase. Benzyl isothiocyanate (BITC), phenethyl isothiocyanate (PEITC) and sulforaphane ([1-isothioyanato-4-(methyl-sulfinyl) butane], SFN) are potential ITCs with efficient therapeutic properties. Beneficial role of BITC, PEITC and SFN was widely studied against various cancers such as breast, brain, blood, bone, colon, gastric, liver, lung, oral, pancreatic, prostate and so forth. Nuclear factor-erythroid 2-related factor-2 (Nrf2) is a key transcription factor limits the tumor progression. Induction of ARE (antioxidant responsive element) and ROS (reactive oxygen species) mediated pathway by Nrf2 controls the activity of nuclear factor-kappaB (NF-κB). NF-κB has a double edged role in the immune system. NF-κB induced during inflammatory is essential for an acute immune process. Meanwhile, hyper activation of NF-κB transcription factors was witnessed in the tumor cells. Antagonistic activity of BITC, PEITC and SFN against cancer was related with the direct/indirect interaction with Nrf2 and NF-κB protein. All three ITCs able to disrupts Nrf2-Keap1 complex and translocate Nrf2 into the nucleus. BITC have the affinity to inhibit the NF-κB than SFN due to the presence of additional benzyl structure. This review will give the overview on chemo preventive of ITCs against several types of cancer cell lines. We have also discussed the molecular interaction(s) of the antagonistic effect of BITC, PEITC and SFN with Nrf2 and NF-κB to prevent cancer.

157 citations

Journal ArticleDOI
06 May 2019
TL;DR: Efforts have been made to bring together most of the concrete studies pertaining to the bioactive potential of fisetin and quercetin, especially in the modulation of a range of cancer signaling pathways, which could be helpful in designing effective treatment strategies.
Abstract: Despite advancements in healthcare facilities for diagnosis and treatment, cancer remains the leading cause of death worldwide. As prevention is always better than cure, efficient strategies are needed in order to deal with the menace of cancer. The use of phytochemicals as adjuvant chemotherapeutic agents in heterogeneous human carcinomas like breast, colon, lung, ovary, and prostate cancers has shown an upward trend during the last decade or so. Flavonoids are well-known products of plant derivatives that are reportedly documented to be therapeutically active phytochemicals against many diseases encompassing malignancies, inflammatory disorders (cardiovascular disease, neurodegenerative disorder), and oxidative stress. The current review focuses on two key flavonols, fisetin and quercetin, known for their potential pharmacological relevance. Also, efforts have been made to bring together most of the concrete studies pertaining to the bioactive potential of fisetin and quercetin, especially in the modulation of a range of cancer signaling pathways. Further emphasis has also been made to highlight the molecular action of quercetin and fisetin so that one could explore cancer initiation pathways and progression, which could be helpful in designing effective treatment strategies.

148 citations

References
More filters
Journal ArticleDOI
19 Mar 2010-Cell
TL;DR: The principal mechanisms that govern the effects of inflammation and immunity on tumor development are outlined and attractive new targets for cancer therapy and prevention are discussed.

8,664 citations


"Roles of NF-κB in Cancer and Inflam..." refers background in this paper

  • ...In inflammatory cells, continuous NF-κB activity could promote the production of reactive oxygen species, thereby damaging DNA of surrounding epithelial cells [53]....

    [...]

Journal ArticleDOI
TL;DR: Recent progress has been made in understanding the details of the signaling pathways that regulate NF-kappaB activity, particularly those responding to the proinflammatory cytokines tumor necrosis factor-alpha and interleukin-1.
Abstract: NF-κB (nuclear factor-κB) is a collective name for inducible dimeric transcription factors composed of members of the Rel family of DNA-binding proteins that recognize a common sequence motif. NF-κ...

4,724 citations


"Roles of NF-κB in Cancer and Inflam..." refers background or methods in this paper

  • ...After processing of p105 and p100 by the ubiquitin/proteasome pathway that involves selective degradation of their C-terminal region containing ankyrin repeats, mature NF-κB subunits such as p50 and p52 are generated [16,17]....

    [...]

  • ...The IκB proteins mask the nuclear localization signals (NLS) of NF-κB proteins and keep them sequestered in an inactive state in the cytoplasm by virtue of their ankyrin repeat domains [11,16]....

    [...]

  • ...A subfamily of NF-κB proteins, including RelA (p65), RelB and c-Rel has a transactivation domain in their C-termini [16,17]....

    [...]

Journal ArticleDOI
TL;DR: The inhibition of NF-kappa B activation by antioxidants and specific protease inhibitors may provide a pharmacological basis for interfering with these acute processes in suppressing toxic/septic shock, graft-vs-host reactions, acute inflammatory reactions, severe phase response, and radiation damage.
Abstract: NF-kappa B is a ubiquitous transcription factor. Nevertheless, its properties seem to be most extensively exploited in cells of the immune system. Among these properties are NF-kappa B's rapid posttranslational activation in response to many pathogenic signals, its direct participation in cytoplasmic/nuclear signaling, and its potency to activate transcription of a great variety of genes encoding immunologically relevant proteins. In vertebrates, five distinct DNA binding subunits are currently known which might extensively heterodimerize, thereby forming complexes with distinct transcriptional activity, DNA sequence specificity, and cell type- and cell stage-specific distribution. The activity of DNA binding NF-kappa B dimers is tightly controlled by accessory proteins called I kappa B subunits of which there are also five different species currently known in vertebrates. I kappa B proteins inhibit DNA binding and prevent nuclear uptake of NF-kappa B complexes. An exception is the Bcl-3 protein which in addition can function as a transcription activating subunit in th nucleus. Other I kappa B proteins are rather involved in terminating NF-kappa B's activity in the nucleus. The intracellular events that lead to the inactivation of I kappa B, i.e. the activation of NF-kappa B, are complex. They involve phosphorylation and proteolytic reactions and seem to be controlled by the cells' redox status. Interference with the activation or activity of NF-kappa B may be beneficial in suppressing toxic/septic shock, graft-vs-host reactions, acute inflammatory reactions, acute phase response, and radiation damage. The inhibition of NF-kappa B activation by antioxidants and specific protease inhibitors may provide a pharmacological basis for interfering with these acute processes.

4,708 citations


"Roles of NF-κB in Cancer and Inflam..." refers background in this paper

  • ...NF-κB controls expression of a number of genes that regulate immune responses, cell growth and proliferation, survival and apoptosis, stress responses and embryogenesis and development of a variety of stimuli [7,9]....

    [...]

Journal ArticleDOI
08 Feb 2008-Cell
TL;DR: The authors synthesize some of the basic principles that have emerged from studies of NF-kappaB, and aim to generate a more unified view of the regulation of the transcription factor.

3,996 citations

Journal ArticleDOI
TL;DR: An overview of established NF-kappaB signaling pathways is provided with focus on the current state of research into the mechanisms that regulate IKK activation and NF- kappaB transcriptional activity.
Abstract: The transcription factor NF-kappaB has been the focus of intense investigation for nearly two decades. Over this period, considerable progress has been made in determining the function and regulation of NF-kappaB, although there are nuances in this important signaling pathway that still remain to be understood. The challenge now is to reconcile the regulatory complexity in this pathway with the complexity of responses in which NF-kappaB family members play important roles. In this review, we provide an overview of established NF-kappaB signaling pathways with focus on the current state of research into the mechanisms that regulate IKK activation and NF-kappaB transcriptional activity.

3,829 citations


"Roles of NF-κB in Cancer and Inflam..." refers background in this paper

  • ...Among them, the p50/65 heterodimer clearly represents the most abundant of Rel dimers, being found in almost all cell types [15]....

    [...]