ArticlePDF AvailableLiterature Review

Potential of Sulforaphane as a Natural Immune System Enhancer: A Review

Authors:

Abstract and Figures

Brassicaceae are an outstanding source of bioactive compounds such as ascorbic acid, polyphenols, essential minerals, isothiocyanates and their precursors, glucosinolates (GSL). Recently, GSL gained great attention because of the health promoting properties of their hydrolysis products: isothiocyanates. Among them, sulforaphane (SFN) became the most attractive one owing to its remarkable health-promoting properties. SFN may prevent different types of cancer and has the ability to improve hypertensive states, to prevent type 2 diabetes–induced cardiomyopathy, and to protect against gastric ulcer. SFN may also help in schizophrenia treatment, and recently it was proposed that SFN has potential to help those who struggle with obesity. The mechanism underlying the health-promoting effect of SFN relates to its indirect action at cellular level by inducing antioxidant and Phase II detoxifying enzymes through the activation of transcription nuclear factor (erythroid-derived 2)-like (Nrf2). The effect of SFN on immune response is generating scientific interest, because of its bioavailability, which is much higher than other phytochemicals, and its capacity to induce Nrf2 target genes. Clinical trials suggest that sulforaphane produces favorable results in cases where pharmaceutical products fail. This article provides a revision about the relationship between sulforaphane and immune response in different diseases. Special attention is given to clinical trials related with immune system disorders.
Content may be subject to copyright.
molecules
Review
Potential of Sulforaphane as a Natural Immune System
Enhancer: A Review
Andrea Mahn 1, * and Antonio Castillo 2


Citation: Mahn, A.; Castillo, A.
Potential of Sulforaphane as a
Natural Immune System Enhancer:
A Review. Molecules 2021,26, 752.
https://doi.org/10.3390/
molecules26030752
Academic Editors: Maria
JoséRodríguez-Lagunas and
Malen Massot-Cladera
Received: 30 December 2020
Accepted: 28 January 2021
Published: 1 February 2021
Publisher’s Note: MDPI stays neutral
with regard to jurisdictional claims in
published maps and institutional affil-
iations.
Copyright: © 2021 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
1Departamento de Ingeniería Química, Facultad de Ingeniería, Universidad de Santiago de Chile (USACH),
Santiago 8330111, Chile
2Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile (USACH),
Santiago 8330111, Chile; antonio.castillo@usach.cl
*Correspondence: andrea.mahn@usach.cl; Tel.: +56-227-181-833
Abstract:
Brassicaceae are an outstanding source of bioactive compounds such as ascorbic acid,
polyphenols, essential minerals, isothiocyanates and their precursors, glucosinolates (GSL). Recently,
GSL gained great attention because of the health promoting properties of their hydrolysis products:
isothiocyanates. Among them, sulforaphane (SFN) became the most attractive one owing to its
remarkable health-promoting properties. SFN may prevent different types of cancer and has the
ability to improve hypertensive states, to prevent type 2 diabetes–induced cardiomyopathy, and to
protect against gastric ulcer. SFN may also help in schizophrenia treatment, and recently it was
proposed that SFN has potential to help those who struggle with obesity. The mechanism underlying
the health-promoting effect of SFN relates to its indirect action at cellular level by inducing antioxidant
and Phase II detoxifying enzymes through the activation of transcription nuclear factor (erythroid-
derived 2)-like (Nrf2). The effect of SFN on immune response is generating scientific interest, because
of its bioavailability, which is much higher than other phytochemicals, and its capacity to induce
Nrf2 target genes. Clinical trials suggest that sulforaphane produces favorable results in cases
where pharmaceutical products fail. This article provides a revision about the relationship between
sulforaphane and immune response in different diseases. Special attention is given to clinical trials
related with immune system disorders.
Keywords: sulforaphane; immunological response; cellular mechanism
1. Introduction
Many of the current synthetic drugs come from natural products of plant origin. Even
some plant-derived bioactive compounds have been proposed as possible therapeutic
solutions to fight highly prevalent diseases such as cancer [1].
Sulforaphane (SFN), an isothiocyanate (ITC) widely distributed in Brassicaceae
plants, has generated great interest in the last 15 years, with an exponentially growing
number of scientific articles reaching around 250 in 2020 and a total of 2315 since 1948
(PubMed, https://pubmed.ncbi.nlm.nih.gov/, last access 21 December 2020). This is due
to the outstanding health promoting properties of SFN which are related with its high
capacity to induce Phase II detoxifying enzymes, being 14-fold higher than other potent
phytochemicals such as quercetin. Additionally, SFN exhibits the highest bioavailability
among well-known antioxidant phytochemicals, such as quercetin (20-fold higher) [
2
] and
curcumin (80-fold higher) [
3
]. This confers SFN a high potential to be used as nutraceutical
to improve health status or as pharmaceutical to treat some disease states.
SFN comes from the enzymatic hydrolysis of glucoraphanin, a glucosinolate stored
as inactive precursor in the plant cells. The hydrolysis of glucoraphanin occurs through
myrosinase (E.C. 3.2.1.147), which is compartmentalized in the vegetable inside myrosin
cells. The reaction proceeds after tissue disruption that can be produced by insects and
herbivores attack or by processing and chewing the vegetable [
4
]. The products of the
Molecules 2021,26, 752. https://doi.org/10.3390/molecules26030752 https://www.mdpi.com/journal/molecules
Molecules 2021,26, 752 2 of 14
hydrolysis reaction vary depending on the chemical conditions where the reaction occurs.
The myrosinase—glucosinolate system belongs to the defense system of the plant and there-
fore some of the products that come from glucosinolates hydrolysis are toxic [
5
].
Figure 1
depicts the myrosinase—glucosinolate (glucoraphanin) system in plants. In mammals,
SFN can be administered directly in its active form or as glucoraphanin which undergoes
the hydrolysis during digestion by the action of vegetable and gut microflora myrosinases.
After intake, SFN follows the mercapturic acid pathway until its conversion in dithiocarba-
mates and is finally excreted [
6
]. Figure 2shows the formation and metabolic degradation
of SFN.
Molecules 2021, 26, x 2 of 14
hydrolysis reaction vary depending on the chemical conditions where the reaction occurs.
The myrosinase—glucosinolate system belongs to the defense system of the plant and
therefore some of the products that come from glucosinolates hydrolysis are toxic [5]. Fig-
ure 1 depicts the myrosinase—glucosinolate (glucoraphanin) system in plants. In mam-
mals, SFN can be administered directly in its active form or as glucoraphanin which un-
dergoes the hydrolysis during digestion by the action of vegetable and gut microflora my-
rosinases. After intake, SFN follows the mercapturic acid pathway until its conversion in
dithiocarbamates and is finally excreted [6]. Figure 2 shows the formation and metabolic
degradation of SFN.
Figure 1. Myrosinase—glucoraphanin system in Brassicaceae plants. Glucosinolates are located in
specialized glucosinolate-containing cells, while myrosinase is stored in the vacuoles of the my-
rosin cells. After mechanical disruption of plant tissue, the substrate and enzyme come in contact
and the hydrolysis occurs, resulting in different products, among which it is found sulforaphane
[7].
Figure 1.
Myrosinase—glucoraphanin system in Brassicaceae plants. Glucosinolates are located in
specialized glucosinolate-containing cells, while myrosinase is stored in the vacuoles of the myrosin
cells. After mechanical disruption of plant tissue, the substrate and enzyme come in contact and the
hydrolysis occurs, resulting in different products, among which it is found sulforaphane [7].
Several efforts have been conducted in order to exploit the health-promoting effects
of SFN on humans. Its direct administration has been limited because of the instability
of SFN. Some research about SFN stabilization is being conducted [
9
]. Another way to
administer SFN to humans is through broccoli sprout extracts or minimally processed
broccoli. Some processing conditions that maximize SFN content in processed broccoli
have been reported [
10
14
], resulting in an important amount of information about the
processing conditions that achieve this goal. Given the instability of SFN and the possibility
to maximize SFN in processed vegetable, most clinical studies about the effect of SFN use
broccoli extracts or powder, and focused on validating the efficacy of SFN-rich food, not in
SFN as a drug.
The first report about the effect of sulforaphane on health dates from 1992, when
Zhang et al. [
15
] suggested that SFN was a potent activator of cellular defense systems.
Later Bonnesen et al. [
16
] informed that SFN and other isothiocyanates showed a preventive
effect on colon tumorigenesis since these compounds stimulate apoptosis and enhance
Molecules 2021,26, 752 3 of 14
cell defense against molecules that produce gene toxicity. Since then, several
in vitro
and
in vivo
studies about the effect of SFN have been conducted, including clinical trials,
resulting in relevant information regarding prevention and treatment of diseases such as
pancreatic cancer [
17
], breast cancer [
18
], diffuse axonal injury [
19
], lymphomas [
20
], liver
cancer [
21
], leukemia [
22
], and prostate cancer [
23
]. Moreover, SFN has cardio protective
properties [
24
], the ability to prevent aging and neurodegeneration [
25
], and to protect
from gastric ulcer [
26
]. Additionally, several clinical trials are currently in progress or
already finished.
Molecules 2021, 26, x 3 of 14
Figure 2. Formation and metabolization of sulforaphane. Sulforaphane (SFN) is formed by the hydrolysis of glucoraphanin
catalyzed by either plant or bacterial myrosinase. After intake, SFN is metabolized through the mercapturic acid pathway.
Initially, isothiocyanates are conjugated with glutathione (GSH) in a glutathione transferase (GST)-catalyzed reaction.
Then, successive cleavage reactions catalyzed by γ-glutamyltranspeptidase, cysteinylglycinase, and N-acetyltransferase
occur to generate sulforaphane-N-acetylcysteine (SFR-NAC) [8].
Several efforts have been conducted in order to exploit the health-promoting effects
of SFN on humans. Its direct administration has been limited because of the instability of
SFN. Some research about SFN stabilization is being conducted [9]. Another way to ad-
minister SFN to humans is through broccoli sprout extracts or minimally processed broc-
coli. Some processing conditions that maximize SFN content in processed broccoli have
been reported [10–14], resulting in an important amount of information about the pro-
cessing conditions that achieve this goal. Given the instability of SFN and the possibility
to maximize SFN in processed vegetable, most clinical studies about the effect of SFN use
broccoli extracts or powder, and focused on validating the efficacy of SFN-rich food, not
in SFN as a drug.
The first report about the effect of sulforaphane on health dates from 1992, when
Zhang et al. [15] suggested that SFN was a potent activator of cellular defense systems.
Later Bonnesen et al. [16] informed that SFN and other isothiocyanates showed a preven-
tive effect on colon tumorigenesis since these compounds stimulate apoptosis and en-
hance cell defense against molecules that produce gene toxicity. Since then, several in vitro
and in vivo studies about the effect of SFN have been conducted, including clinical trials,
resulting in relevant information regarding prevention and treatment of diseases such as
pancreatic cancer [17], breast cancer [18], diffuse axonal injury [19], lymphomas [20], liver
cancer [21], leukemia [22], and prostate cancer [23]. Moreover, SFN has cardio protective
properties [24], the ability to prevent aging and neurodegeneration [25], and to protect
from gastric ulcer [26]. Additionally, several clinical trials are currently in progress or al-
ready finished.
This review aims at presenting the most recent advances of research about the effects
of SFN on the immune system, considering in vitro studies, which were performed using
animal or human cells in culture, and in vivo studies, which were animal or human clini-
cal intervention trials.
2. Mechanisms of Action of SFN on Immune System
Sulforaphane exerts a pleiotropic effect on immunological response. The mechanism
is based on activation of nuclear factor (erythroid-derived 2)-like (Nrf2) which triggers
cellular defense mechanisms. There is induction of Phase II detoxifying enzymes as well
as antioxidant enzymes, and down regulation of Phase I enzymes by inactivation of NFκβ.
Figure 2.
Formation and metabolization of sulforaphane. Sulforaphane (SFN) is formed by the hydrolysis of glucoraphanin
catalyzed by either plant or bacterial myrosinase. After intake, SFN is metabolized through the mercapturic acid pathway.
Initially, isothiocyanates are conjugated with glutathione (GSH) in a glutathione transferase (GST)-catalyzed reaction. Then,
successive cleavage reactions catalyzed by
γ
-glutamyltranspeptidase, cysteinylglycinase, and N-acetyltransferase occur to
generate sulforaphane-N-acetylcysteine (SFR-NAC) [8].
This review aims at presenting the most recent advances of research about the effects
of SFN on the immune system, considering
in vitro
studies, which were performed using
animal or human cells in culture, and
in vivo
studies, which were animal or human clinical
intervention trials.
2. Mechanisms of Action of SFN on Immune System
Sulforaphane exerts a pleiotropic effect on immunological response. The mechanism
is based on activation of nuclear factor (erythroid-derived 2)-like (Nrf2) which triggers
cellular defense mechanisms. There is induction of Phase II detoxifying enzymes as well
as antioxidant enzymes, and down regulation of Phase I enzymes by inactivation of NF
κβ
.
The final effect of SFN varies with cell type. In T-cells, the response to SFN exposure is the
generation of a pro-oxidant environment, with an increase of intracellular reactive oxygen
species (ROS) and a decrease in intracellular glutathione (GSH) levels, that produces a block
of the T-cell-mediated immune response. SFN is able to create a pro-oxidative ROS enriched
milieu in primary human
T-cells
. It inhibits co-stimulation initiated
T-cell
activation and
proliferation by depletion of GSH and oxidation of proteins at redox active cysteine residues.
Importantly, SFN also enhances the ROS levels in lymphocytes within whole blood of RA
(rheumatoid arthritis) patients and inhibited the production of pro-inflammatory TH17
related cytokines. This immunosuppressive effect of SFN on T-cells can be desirable in
autoimmune or inflammatory diseases, but it would be detrimental in other chronic diseases
such as cancer since the T-cell-mediated immune response is important for immune surveil-
lance of tumors. Therefore, caution should be exercised, as SFN could interfere with the
successful application of immunotherapy by immune checkpoint inhibitors (e.g., CTLA-4
Molecules 2021,26, 752 4 of 14
antibodies and PD-1/PD-L1 antibodies) or CAR (chimeric antigen receptors) T-cells in
cancer patients, and a combination of both treatments could not be advisable [27].
Although there is a good amount of evidence that indicates that SFN is a potent
anticancer compound and that its main mechanism of action would occur through the
activation of Nrf2, recent publications present controversial results that indicate that the
activation of Nrf2 contributes to the whole process of pathogenesis, promotes cancer
progression and metastasis while conferring resistance to chemo- and radiotherapy, and
has a poor prognosis, a phenomenon that has been described as the “dark side” of Nrf2 [
28
].
Therefore, in accordance with the above, Nrf2 could be a promising target in cancer therapy.
However, research related to Nrf2 inhibitors is still incipient [29].
In monocytes and macrophages, SFN inhibits pro-inflammatory cytokines and acti-
vates antioxidant enzymes through Nrf2 modulation, resulting in an anti-inflammatory
effect (Figure 3) useful for the treatment of bacterial and viral related diseases. SFN is
widely recognized as among the most powerful natural anti-cancer agents, but its mecha-
nism of action is not fully understood so far. This owes to the multi-factorial nature of this
disease and to the pleiotropic effect of SFN. However there is evidence that supports SFN
to exert an antioxidant effect in tumor cells [
30
]. The mechanisms that underlay SFN effect
on immunological system in different diseases are presented below.
Molecules 2021, 26, x 7 of 14
Freeman [56] suggested that clinical trials including administration of Nrf2-activating
molecules, such as SFN, are imperative to support a possible three-party strategy to fight
the COVID-19 pandemic, which includes prevention, diagnostic, and treatment.
2.4. Bacterial Diseases
Research about the effect of SFN on immunological system during bacterial infec-
tions is incipient. Currently there are reports that consider H. pylori, S. aureus, E. coli and
M. pneumoniae. Although SFN exhibits direct bactericidal activity, it triggers an immuno-
logical response to H. pylori infection in the stomach mucosa. SFN acts by activating Nrf2
and downregulating NF-κB, whose joint action modulates antioxidant and anti-inflam-
matory response in the host [57,58]. As a consequence, SFN exerts a protective effect from
gastritis and gastric ulcer. Yanaka [59] conducted in vitro and in vivo studies about the
effect of SFN on H. pylori infection. The outcomes demonstrated that SFN significantly
reduced the bacterium viability and alleviated gastritis in animal models and in humans.
Haodang et al. [60] studied the response of monocytes stimulated with Mycoplasma
pneumoniae lipopeptide to SFN exposure. Pathological injury of M. pneumoniae in lungs
relates with inflammation that stimulates immune response of the host triggered by lipid
polysaccharide (LPS) excretion by the bacteria. The authors found that SFN inhibited the
expression of pro-inflammatory cytokines and activated the expression of HO-1 after the
induction of Nrf2. As a result, SFN reduced lung inflammation in an animal model. The
mechanism proposed by [60] is depicted in Figure 3.
Ali et al. [61] investigated the effect of four Nrf2 activators on bacteria-infected mac-
rophages, among them, SFN. Macrophages were infected either with E. coli or S. aureus
and the intracellular viability of bacteria was evaluated. SFN significantly reduced intra-
cellular bacteria survival in PBMC-derived macrophages. Even though the authors do not
present any mechanism, they propose that the intra and extra cellular bactericidal effect
of SFN relies on the anti-inflammatory and antioxidant milieu produced inside the mac-
rophages. SFN, as a potent Nrf2 activator, seems a promising therapeutic option for Gram
(+) and Gram () bacterial infections since it modulates antioxidant and anti-inflammatory
responses. Deramaudt et al. [62] studied the intracellular survival of S. aureus in human
and mice macrophages treated with SFN. They proposed a mechanism consisting in mod-
ulation of p38/JNK signal pathway induced by SFN in macrophages, thus reducing in-
flammatory response. Additionally, the authors reported that SFN affected S. aureus in-
tracellular survival by inducing apoptosis in the bacterium. Then, the combination of both
mechanisms supports SFN as a possible treatment for S. aureus infection.
Finally, Belchamber and Donnelly [63] suggested that SFN stimulates phagocytic
pathways and improves macrophage phagocytosis of S. pneumoniae, P. aeruginosa and H.
influenzae by upregulating Nrf2 in alveolar cells from COPD9 (chronic obstructive pulmo-
nary disease).
Figure 3. Mechanism of inflammatory response suppression induced by sulforaphane in bacteria-infected monocytes.
Figure 3. Mechanism of inflammatory response suppression induced by sulforaphane in bacteria-infected monocytes.
2.1. Autoimmune/Inflamatory Diseases
SFN exerts its effect on immune system through different biochemical and cellular
mechanisms, among them the downregulation of pro-inflammatory cytokines, T-cells
suppressing, and activation of adenosine monophosphate activated protein kinase (AMPK)
signaling pathway. Even though these processes have a suppressive effect, this is desirable
in cases of autoimmune/inflammatory diseases.
Townsend and Johnson [
31
] studied the effect of sulforaphane on pro-inflammatory
markers and target genes of nuclear factor erythroid 2 (NFE2)—related factor 2 (Nrf2) in
mice subjected to lipopolysaccharide (LPS) challenge. They found that SFN decreased pro-
inflammatory markers such as interleukin 1-
β
(IL-1
β
) and interleukin 6 (IL6) as response to
LPS-treatment. The authors propose that the anti-inflammatory effect of SFN was regulated
by the Nrf2 pathway.
Deng et al. [
32
] demonstrated that SFN delivered as broccoli nanoparticles to mice
is involved in prevention of colitis, an autoimmune disease that can lead to ulcers. The
mechanism consists in the induction of tolerogenic dendritic cells by adenosine monophos-
phate activated protein kinase (AMPK), thus regulating the intestinal immune homeostasis.
Accordingly, SFN could have preventive or therapeutic application on some intestinal
inflammatory diseases due to its activating effect of AMPK signaling pathway.
Liang et al. [
33
] studied the effect of sulforaphane on the redox regulation in human
T-cells, in order to uncover the mechanism that underlays the immunosuppressive effect of
Molecules 2021,26, 752 5 of 14
SFN in chronic Th17-related diseases, such as rheumatoid arthritis. They reported that SFN
exerts a redox-related immunosuppressive effect on untransformed human T-cells, downreg-
ulation of the pro-inflammatory Th17 cytokines associated with autoimmune/inflammatory
diseases (IL-17A, IL-17F and IL-22), and inhibition of cartilage-disruptive proteins. These
processes produce a significant reduction in the clinical symptoms. Since this study was
conducted ex vivo, the results cannot be extrapolated to the effect in humans.
Some authors investigated the effect of sulforaphane on immune-associated inflam-
matory diseases of the central nervous system (CNS), such as Alzheimer and Parkinson,
concluding that SFN has anti-inflammatory and anti-oxidant effect [
34
,
35
]. The mecha-
nism that underlies this kind of disease relies on promotion of leukocyte traffic across the
blood-brain barrier by the action of reactive oxygen species (ROS) [
36
]. ROS induce myelin
breakdown and neuronal injury, among other effects. Additionally, the infiltrated cells
increase the production of ROS, thus contributing to the advance of the CNS diseases [
37
].
Yoo et al. [
38
] administered SFN (orally, 50 mg/kg/day over 14 days) to an autoimmune
encephalomyelitis model (mouse). The clinical symptoms of SFN–treated animals were
diminished significantly in comparison with those observed in control animals. This was
attributed to the anti-inflammatory and anti-oxidative effects of SFN, resulting in neuro-
protection. Accordingly, SFN seems a promising alternative to traditional drugs, which are
expensive and most importantly have undesirable side-effects.
2.2. Pulmonary Diseases
Information about the effect of SFN on immune system in lung diseases is poorly
documented so far. Recently, Patel et al. [
39
] presented evidence that SFN can act as
prophylactic in hyperoxia-induced lung injury or hyperoxia-compromised macrophage
function in phagocytosis. The results presented in this study suggest that SFN can alleviate
hyperoxia-induced inflammatory acute lung injury by increasing macrophage phagocytosis
via inhibiting the accumulation of extracellular HMGB1 (high-mobility group box 1 pro-
tein). Thus, by reducing the toxic effects of extracellular HMGB1, it is possible to maintain
the functions of pulmonary macrophages and the integrity of lung tissues under oxidative
stress. This is the first report in which is shown that SFN attenuate hyperoxia-induced
macrophage dysfunction through an HMGB1-mediated pathway. The authors concluded
that the supplementation of SFN during oxygen therapy may prevent lung damage and pre-
serve lung cell functions and lung tissue integrity, thus providing a promising therapeutic
approach for patients receiving mechanical ventilation.
2.3. Viral Diseases
Literature about the effect of SFN on immunological system during viral infection is
scarce. There are studies showing that SFN may help an organism to fight against some
types of virus, mainly HIV, influenza, hepatitis C, and most recently COVID-19. These
studies suggest that SFN acts by restoring the immune system and downregulating free
radicals production, mediated through modulation of antioxidant genes expression by the
transcription factor Nrf2.
Jin-Nyoung et al. [
40
] studied the effect of administering isothiocyanates (benzyl
isothiocyanate, indolo[3,2-b]carbazole, indole-3-carbinol, phenethyl isothiocyanate, and
sulforaphane) on the life span of leukemia retrovirus infected-mice. The authors reported
that mice treated with benzyl isothiocyanate, phenethyl isothiocyanate, or sulforaphane
significantly extended their life span in comparison with the control retrovirus-infected
group. Accordingly, those three ITC retarded the evolution of the infection with LP-BM5
retrovirus to murine AIDS. Furuya et al. [
41
] investigated the effect of SFN on human
macrophages and T-cells after infection with HIV. The authors demonstrated that, unlike
other viruses like Dengue virus (DENV) or Marburg virus (MARV) that benefit from
Nrf2, HIV infection is blocked with the activation of Nrf2 in primary macrophages. This
effect was not detected in T-cells. SFN modulates Nrf2 and results in reprogramming
Molecules 2021,26, 752 6 of 14
gene expression in macrophages. Finally, it was proposed that SFN is capable to induce
an antiviral response in human macrophages against HIV, arising as a promising therapy.
In contrast to the effect of Nrf2 on the HIV infection, the oxidative stress generation
during DENV infection stimulates the transcription factor Nrf2, which tightly regulates
ROS levels as well as innate immune and apoptotic responses to DENV infection, limiting
both antiviral and cell death responses to the virus by feedback modulation of oxidative
stress. Confirming the above, silencing of Nrf2 by RNA interference increased DENV-
associated immune and apoptotic responses [
42
]. On the other hand, MARV directly
increases Nrf2 levels through a protein called VP24. This protein, like SFN, interacts with
Keap1 (Kelch-like ECH-associated protein 1), a negative regulator of Nrf2. Binding of
VP24 to Keap1 Kelch domain releases Nrf2 from Keap1-mediated inhibition, promoting
persistent activation of diverse cytoprotective genes implicated in cellular responses to
oxidative stress and regulation of inflammatory responses. The authors demonstrated
that there is increased expression of Nrf2-dependent genes both during MARV infection
and upon transient expression of MARV VP24. Finally, Nrf2-deficient (Nrf2
-/-
) mice
can control MARV infection when compared to lethal infection in wild-type animals,
indicating that Nrf2 is critical for MARV infection [43].
Hepatitis C Virus (HCV) is susceptible to heme-oxigenase-1 (HO-1) which interferes
with the replication of viruses like HIV and Hepatitis B [
44
,
45
]. Since SFN is a potent
activator of phase II antioxidant enzymes, like HO-1, Yu et al. [
46
] studied the effect of
SFN on Huh-7 cells infected with HCV. The authors demonstrated that SFN suppresses
replication of HCV by inducing HO-1 expression through activation of Nrf2 pathway.
Efforts have been made to elucidate the role of SFN in immunological response to
influenza. Some phytochemicals have shown to enhance immunological response against
influenza, such as glucans [47] and sulforaphane [48], the latter associated to Nrf2 expres-
sion that blocks influenza A entry and replication in human nasal epithelial cells. Vaclav
and Jana [
49
] investigated the effect of a glucan–SFN combination on influenza in a mouse
model. They evaluated immunological response by assessing some immune reactions,
virus concentration, and animal survival. The results suggested that both phytochemicals
had a synergistic effect on stimulation of immunological system. Müller et al. [
50
] con-
ducted a clinical trial to evaluate the effect of SFN-rich broccoli sprouts homogenate on
peripheral blood mononuclear cells (PBMC) after administering a nasal vaccine dose of live
attenuated influenza virus (LAIV) to healthy subjects. They found significant differences
between the response to BSH (broccoli sprout homogenates) and placebo, observing that
LAIV significantly reduced NKT (natural killer T) and T-cell populations. The authors con-
clude that nasal influenza infection may induce complex changes in peripheral blood NK
cell activation, and that BSH (rich in SFN) effect may be important for enhanced antiviral
defense responses. Li et al. [
51
] studied the effect of SFN on influenza A virus replication in
Madin-Darby canine kidney cells. They detected an increased accumulation of Nrf2 factor
triggered by SFN, resulting in a decrease of virus replication.
During the last year, the world has been shocked by the abrupt irruption of COVID-19
and the scientific community has been devoted to find insights that help fight against this
disease. A way to reduce the severity and mortality generated by acute respiratory distress
syndrome (ARDS) produced by SARS-COV 2 is to strengthen the immune system. ARDS
produces a dysregulation of the immunological system, and in the most severe cases, the
release of pro-inflammatory cytokines and loss of T-cells in the infected organism [
52
].
There is evidence of the antiviral effect of Nrf2 on respiratory syncytial virus infection [
53
]
and on SARS-COV 1 [
54
]. Based on information about viruses that belong to the same
family, it has been proposed that compounds that activate Nrf2 could probably help to
diminish these effects. Cuadrado et al. [
55
] suggested that due to its ability to activate
Nrf2, induce antioxidant enzymes, reduce pro-inflammatory cytokines, and its efficacy and
safety, SFN is a promising candidate to counteract inflammatory reaction and protect lungs
from severe damage during SARS COV 2 infection. Finally, Horowitz and Freeman [
56
]
suggested that clinical trials including administration of Nrf2-activating molecules, such as
Molecules 2021,26, 752 7 of 14
SFN, are imperative to support a possible three-party strategy to fight the COVID-19
pandemic, which includes prevention, diagnostic, and treatment.
2.4. Bacterial Diseases
Research about the effect of SFN on immunological system during bacterial infections
is incipient. Currently there are reports that consider H. pylori,S. aureus,E. coli and M. pneu-
moniae. Although SFN exhibits direct bactericidal activity, it triggers an immunological
response to H. pylori infection in the stomach mucosa. SFN acts by activating Nrf2 and
downregulating NF-
κ
B, whose joint action modulates antioxidant and anti-inflammatory
response in the host [
57
,
58
]. As a consequence, SFN exerts a protective effect from gastritis
and gastric ulcer. Yanaka [
59
] conducted
in vitro
and
in vivo
studies about the effect of
SFN on H. pylori infection. The outcomes demonstrated that SFN significantly reduced the
bacterium viability and alleviated gastritis in animal models and in humans.
Haodang et al. [
60
] studied the response of monocytes stimulated with Mycoplasma
pneumoniae lipopeptide to SFN exposure. Pathological injury of M. pneumoniae in lungs
relates with inflammation that stimulates immune response of the host triggered by lipid
polysaccharide (LPS) excretion by the bacteria. The authors found that SFN inhibited the
expression of pro-inflammatory cytokines and activated the expression of HO-1 after the
induction of Nrf2. As a result, SFN reduced lung inflammation in an animal model. The
mechanism proposed by [60] is depicted in Figure 3.
Ali et al. [
61
] investigated the effect of four Nrf2 activators on bacteria-infected
macrophages, among them, SFN. Macrophages were infected either with E. coli or S. au-
reus and the intracellular viability of bacteria was evaluated. SFN significantly reduced
intracellular bacteria survival in PBMC-derived macrophages. Even though the authors
do not present any mechanism, they propose that the intra and extra cellular bacterici-
dal effect of SFN relies on the anti-inflammatory and antioxidant milieu produced inside
the macrophages. SFN, as a potent Nrf2 activator, seems a promising therapeutic option
for Gram (+) and Gram (
) bacterial infections since it modulates antioxidant and anti-
inflammatory responses. Deramaudt et al. [
62
] studied the intracellular survival of S. aureus
in human and mice macrophages treated with SFN. They proposed a mechanism consisting
in modulation of p38/JNK signal pathway induced by SFN in macrophages, thus reducing
inflammatory response. Additionally, the authors reported that SFN affected S. aureus
intracellular survival by inducing apoptosis in the bacterium. Then, the combination of
both mechanisms supports SFN as a possible treatment for S. aureus infection.
Finally, Belchamber and Donnelly [
63
] suggested that SFN stimulates phagocytic
pathways and improves macrophage phagocytosis of S. pneumoniae,P. aeruginosa and
H. influenzae by upregulating Nrf2 in alveolar cells from COPD9 (chronic obstructive
pulmonary disease).
2.5. Cancer
Cancer is a multi-factorial disease responsible for around 10 million deaths worldwide
per year. The WHO estimates that 30–50% of cancer cases could be prevented. Accordingly,
several efforts are made to discover new strategies to fight and most importantly, prevent
this disease. SFN is widely recognized as the most potent natural anti-cancer compound.
This phytochemical acts at different cancer stages, from development to progression,
by exerting a pleiotropic effect. SFN can trigger apoptosis, reducing angiogenesis and
metastasis in cancerous cells. At the molecular level, it activates Nrf2, consequently
modulating cellular redox homeostasis and stimulating the immune system [
64
]. Figure 4
shows the mechanism action of SFN as chemoprotective and chemotherapeutic agent.
Singh et al. [
65
] studied the effect of administering a SFN analogue on prostate car-
cinogenesis and pulmonary metastasis in an animal model. The results showed that SFN
stimulates NK cells cytotoxicity, thus enhancing immunological function. Also, SFN increased
the infiltration of lymphocyte T-cells in prostate tumors resulting in a reduction of metastasis.
Molecules 2021,26, 752 8 of 14
Molecules 2021, 26, x 8 of 14
2.5. Cancer
Cancer is a multi-factorial disease responsible for around 10 million deaths world-
wide per year. The WHO estimates that 30–50% of cancer cases could be prevented. Ac-
cordingly, several efforts are made to discover new strategies to fight and most im-
portantly, prevent this disease. SFN is widely recognized as the most potent natural anti-
cancer compound. This phytochemical acts at different cancer stages, from development
to progression, by exerting a pleiotropic effect. SFN can trigger apoptosis, reducing angi-
ogenesis and metastasis in cancerous cells. At the molecular level, it activates Nrf2, con-
sequently modulating cellular redox homeostasis and stimulating the immune system
[64]. Figure 4 shows the mechanism action of SFN as chemoprotective and chemothera-
peutic agent.
Singh et al. [65] studied the effect of administering a SFN analogue on prostate car-
cinogenesis and pulmonary metastasis in an animal model. The results showed that SFN
stimulates NK cells cytotoxicity, thus enhancing immunological function. Also, SFN in-
creased the infiltration of lymphocyte T-cells in prostate tumors resulting in a reduction
of metastasis.
The efficacy of SFN as a possible therapeutic compound has been assayed in different
types of cancer cells and tissues. Bessler and Djaldetti [66] investigated the effect of SFN
on immunological interaction between PBMC and human colon cancer cell lines. The au-
thors detected a concentration-dependent effect of sulforaphane that inhibited production
of pro-inflammatory cytokines in PBMC. SFN acts against colon cancer by different mech-
anisms: (1) induction of DNA damage in cancerous cells by acetylating the DNA repair
protein; (2) activation of pro-apoptotic proteins resulting in induction of apoptosis; (3)
activation of Phase II detoxifying proteins through Nrf2; (4) cell cycle arrest by suppress-
ing histone deacetylase inhibitor and telomerase reverse transcriptase [67,68]. Suzuki et
al. [69] studied the effect of daily intake of SFN delivered as fresh broccoli sprouts on colon
cancer animal model and in humans. Their results indicate that SFN treatment suppressed
the formation of aberrant crypt foci and macroscopic tumors in mice and in colon cancer
patients.
Palliyaguru et al. [70] investigated the effect of SFN on breast cancer development in
a mouse model exposed to estradiol. The authors found that SFN enhanced cytoprotection
by mitigating DNA damage and suppressing lipogenesis. These effects were attributed to
activation of Nrf2 by SFN.
Figure 4. Chemoprotective and chemotherapeutic mechanisms of SFN in cancer cells. SFN exerts
chemoprevention by inducing HO-1 and Phase II enzymes, and increasing GSH concentration
Figure 4.
Chemoprotective and chemotherapeutic mechanisms of SFN in cancer cells. SFN exerts
chemoprevention by inducing HO-1 and Phase II enzymes, and increasing GSH concentration
expression and activating NK cells, as well as downregulating pro-inflammatory cytokines. In both
cases, the effect on immune system is mediated by transcription factors Nrf2 and Nfκβ.
The efficacy of SFN as a possible therapeutic compound has been assayed in different
types of cancer cells and tissues. Bessler and Djaldetti [
66
] investigated the effect of SFN on
immunological interaction between PBMC and human colon cancer cell lines. The authors
detected a concentration-dependent effect of sulforaphane that inhibited production of pro-
inflammatory cytokines in PBMC. SFN acts against colon cancer by different mechanisms:
(1) induction of DNA damage in cancerous cells by acetylating the DNA repair protein;
(2) activation of pro-apoptotic proteins resulting in induction of apoptosis; (3) activation
of Phase II detoxifying proteins through Nrf2; (4) cell cycle arrest by suppressing histone
deacetylase inhibitor and telomerase reverse transcriptase [
67
,
68
]. Suzuki et al. [
69
] studied
the effect of daily intake of SFN delivered as fresh broccoli sprouts on colon cancer animal
model and in humans. Their results indicate that SFN treatment suppressed the formation
of aberrant crypt foci and macroscopic tumors in mice and in colon cancer patients.
Palliyaguru et al. [
70
] investigated the effect of SFN on breast cancer development in
a mouse model exposed to estradiol. The authors found that SFN enhanced cytoprotection
by mitigating DNA damage and suppressing lipogenesis. These effects were attributed to
activation of Nrf2 by SFN.
3. Clinical Trials Regarding the Effect of Sulforaphane on Immune System
Over the last decade, there are 74 clinical studies that have aimed at evaluating the
effect of sulforaphane on different diseases; four of them focused on immune system
disorders (www.clinicaltrials.gov). Table 1shows details about the clinical trials.
Trial NCT01357070 was designed to test whether consuming a “broccoli smoothie”
containing sulforaphane could protect white blood cells from activation in the presence
of experimental stress and how long this protective effect would last. To do this, the
researchers analyzed inflammatory changes in blood samples taken at different times
during the study. The investigators suggest that inducing anti-oxidant enzymes indirectly
may be an effective means of providing vascular protection. To date, no results are available.
Molecules 2021,26, 752 9 of 14
Table 1. Clinical trials regarding the effect of sulforaphane on immune system (www.clinicaltrials.gov).
Clinical Trial
Identifier Title Study Population Duration Sulforaphane (Dose) Results Phase Status
NCT01357070
Effect of Broccoli Sprout on
Blood Levels of
Sulforaphane to Reduce
Responsiveness of
Immune System
6 healthy volunteers,
London, United
Kingdom
34 months
(May 2011–January 2014)
broccoli sprout
homogenate (70 g dry
weight, orally by three
consecutive days).
N.I. 1N.A. 2Completed
NCT01183923
Dietary Interventions in
Asthma Treatment:
Sprouts Study
1 adult, asthmatic,
male, white, United
States of America
14 months
(November 2010–
February 2012)
N.I. 1(broccoli sprouts,
one serving per day, 7
days, in a sandwich)
N.I. 1N.A. 2Halted
NCT01269723
Effects of Sulforaphane
(SFN) on Immune
Response to Live
Attenuated Influenza Virus
in Smokers and
Nonsmokers
51 adults, healthy,
smokers or
nonsmokers, United
States of America
28 months
(December 2010–
March 2013)
N.I.1(broccoli sprouts
homogenate) N.I. 1N.A. 2Completed
NCT01845493
Sulforaphane
Supplementation in Atopic
Asthmatics (brasma)
16
adults, asthmatics,
United States
of America
17 months
(April 2013–
October 2014)
N.I. 1(broccoli sprouts
homogenate orally daily,
three days)
N.I. 11 Completed
NCT01845220 Prevention of Alcohol
Intolerance
30 adults, older adults,
sensitive to alcohol on
the skin, Japanese,
United States
of America
27 months
(May 2013–July 2015)
150 nmol of
sulforaphane/cm2 of
skin in 80% acetone
SFN increased
erythema (affected skin
area) as response to
alcohol exposure
2 Completed
NCT02885025 Effects of Broccoli Sprout
Extract on Allergy Rhinitis
47 adults, older adults,
allergic rhinitis
or healthy
18 months
(October 2016–
March 2019)
N.I. 1(broccoli
sprouts extract)
SFN reduced
pro-inflammatory
cytokines with and
without combination
with fluticasone
2 Completed
1Not Informed. 2Not Applicable.
Molecules 2021,26, 752 10 of 14
Trial NCT01183923 hypothesized that since SFN is an inducer of Phase II antioxidant
enzymes and broccoli sprouts (BS) are rich in SFN, administration of BS would improve
lung and airways function in asthmatic subjects. As a consequence, oxidative stress and
inflammation markers would decrease after exposure to allergens. Recruited subjects
(n = 1)
ingested BS and were exposed to environmental mouse allergen challenge. After,
seven daily BS intake markers (nasal epithelial gene expression, urinary oxidative stress
biomarkers, serum inflammatory and oxidative stress biomarkers, and basophil activation)
were assessed. Unfortunately, this trial was halted because of an adverse event. While
no results are available at www.clinicaltrialls.gov, the outcomes of this trial can be found
elsewhere [
71
]. Asthmatic subjects (n = 40) ingested 100 g of BS daily for three days. Effect
of SFN was assessed by measuring antioxidant genes expression in nasal epithelial and
PBMC, inflammation, and oxidative stress biomarkers, among others. Determinations were
conducted before and after BS intake. Since no change in biomarkers and cytoprotective
genes expression could be detected, the authors concluded that despite the increase in
blood concentration of SFN, BS intake did not improve lung inflammatory response nor
antioxidant biomarkers in asthmatic subjects.
Trial NCT01269723 aimed at evaluating the short-term immunological response to live
attenuated influenza virus, and to compare the reaction between smokers and nonsmokers
treated with BS (or placebo) homogenate. As response to the treatment, they evaluated
the virus charge and inflammation biomarkers (IL6, cytokines, NK cells activation) in
nasal mucosa. There are no results available at www.clinicaltrials.gov, however the out-
comes of this trial were published in [
50
]. The main conclusions of this trial was that BS
homogenate enhanced immune response against influenza virus, demonstrated by an
increase in granzyme B production in peripheral NK cells.
Trial NCT01845493 consisted of a pilot study about the effect of SFN administration
(in the form of BS homogenate) on Nrf2 and Phase II enzymes induction. A total of
16 asthmatic subjects ingested BS homogenate for three days and the outcomes were
compared with SFN and placebo controls. Even though the trial ended on 2014, the results
are not available so far.
Trial NCT01845220 aimed at evaluating the effect of SFN (as BS extract) exposure in
alcohol-intolerant subjects. After SFN application, subjects (n = 30) were topically exposed
to alcohol, and reddened skin area was measured as indicator of SFN protection against
irritation. The outcomes indicate that topical application of SFN increased erythema after
exposure to alcohol in alcohol-sensitive subjects.
Trial NCT02885025 studied the effect of administering SFN (as BS extract) to subjects
(n = 47) suffering allergic rhinitis to grass. The randomized trial considered a three-
week treatment aiming at evaluating the effect of BS extract intake in comparison with
administration of corticosteroid (fluticasone) and the combination of both. Before treatment,
subjects were exposed (nasal way) to different varieties of grass. The results showed
that SFN alone or in combination with fluticasone reduced pro-inflammatory cytokines
expression. However SFN exhibited a more limited effect than fluticasone alone.
4. Conclusions
Despite the huge amount of information about the effect of SFN on several diseases,
especially cancer, research about the effect of SFN on immune system response at molecular
and cellular levels is scarce, as well as clinical trials focused on immune system diseases.
Sulforaphane exerts a pleiotropic effect on immunological response, and the final effect
depends on the cell type. In lymphocyte T-cells, SFN induces ROS production, GSH de-
pletion, and repression of inflammatory cytokines, resulting in suppression of immune
and inflammatory responses. This may help in treatment on autoimmune/inflammatory
diseases symptoms. In monocytes and macrophages, SFN stimulates immune response by
inducing Nrf2, thus triggering antioxidant and anti-inflammatory responses. As a conse-
quence, bacteria survival decreases in infected cells, and virus-infected cells are neutralized
by induction of antioxidant enzymes such as HO-1. Additionally, SFN improves immune
Molecules 2021,26, 752 11 of 14
system, thus helping in prevention and reducing severity of viral pulmonary diseases.
In cancer cells, SFN induces apoptosis and cell cycle arrest, as well as antioxidant enzymes
that stimulate cellular immune response. Finally, the few clinical trials about the effect of
SFN on immune system are not conclusive; this kind of study should be encouraged.
Author Contributions:
Conceptualization, A.M.; methodology, A.M. and A.C.; formal analysis, A.M.
and A.C.; investigation, A.M.; resources, A.M. and A.C.; data curation, A.M. and A.C.; writing—
original draft preparation, A.M.; writing—review and editing, A.C.; project administration, A.M.;
funding acquisition, A.M. and A.C. All authors have read and agreed to the published version of
the manuscript.
Funding:
This research was funded by Agencia Nacional de Investigación y Desarrollo (ANID),
Fondecyt program, grant number 1201418. The APC was funded by Fondecyt 1201418. Ethical review
and approval were not applicable.
Conflicts of Interest: The authors declare no conflict of interest.
References
1.
Raskin, I.; Ribnicky, D.M.; Komarnytsky, S.; Ilic, N.; Poulev, A.; Borisjuk, N.; Brinker, A.; Moreno, D.A.; Ripoll, C.; Yakoby, N.;
et al. Plants and human health in the twenty-first century. Trends Biotechnol. 2002,20, 522–531. [CrossRef]
2.
Warwick, E.; Cassidy, A.; Hanley, B.; Jouni, Z.E.; Bao, Y. Effect of phytochemicals on phase II enzyme expression in infant human
primary skin fibroblast cells. Br. J. Nutr. 2012,108, 2158–2165. [CrossRef] [PubMed]
3.
Houghton, C.A. Sulforaphane: Its “Coming of Age” as a Clinically Relevant Nutraceutical in the Prevention and Treatment of
Chronic Disease. Oxid. Med. Cell. Longev. 2019,2019, 2716870. [CrossRef] [PubMed]
4.
Dinkova-Kostova, A.T.; Kostov, R.V. Glucosinolates and isothiocyanates in health and disease. Trends Mol. Med.
2012
,18, 337–347.
[CrossRef] [PubMed]
5.
Jeschke, V.; Kearney, E.E.; Schramm, K.; Kunert, G.; Shekhov, A.; Jonathan Gershenzon, J.; Vassão, D.G. How Glucosinolates
Affect Generalist Lepidopteran Larvae: Growth, Development and Glucosinolate Metabolism. Front. Plant Sci.
2017
,8, 1995.
[CrossRef]
6.
Yoko Yagishita, Y.; Fahey, J.W.; Dinkova-Kostova, A.T.; Kensler, T.W. Broccoli or Sulforaphane: Is it the Source or Dose That
Matters? Molecules 2019,24, 3593. [CrossRef]
7.
Mahn, A.; Quintero, J.; Castillo, N.; Comett, R. Effect of ultrasound-assisted blanching on myrosinase activity and sulforaphane
content in broccoli florets. Catalysts 2020,10, 616. [CrossRef]
8.
Vanduchova, A.; Anzenbacher, P.; Anzenbacherova, E. Isothiocyanate from Broccoli, Sulforaphane, and Its Properties. J. Med.
Food 2019,22, 121–126. [CrossRef]
9.
Zambrano, V.; Bustos, R.; Mahn, A. Insights about stabilization of sulforaphane through microencapsulation. Heliyon
2019
,5,
e02951. [CrossRef]
10.
Pérez, C.; Barrientos, H.; Román, J.; Mahn, A. Optimization of a blanching step to maximize sulforaphane synthesis in broccoli
florets. Food Chem. 2014,145, 264–271. [CrossRef]
11.
Quintero, J.; Román, D.; Salazar, J.; Mahn, A. Economic assessment of a small-scale plant for production of sulforaphane-rich
broccoli flour in Chile. Biofuel. Bioprod. Bior. 2020,14, 544–552. [CrossRef]
12.
Mahn, A.; Reyes, A. An overview of health-promoting compounds of broccoli (Brassica oleracea var italica) and the effect of
processing. Food Sci. Technol. Int. 2012,18, 503–514. [CrossRef] [PubMed]
13.
Cai, Y.X.; Augustin, M.A.; Jegasothy, H.; Wang, J.H.; Terefe, N.S. Mild heat combined with lactic acid fermentation: A novel
approach for enhancing sulforaphane yield in broccoli puree. Food Funct. 2020,11, 779–786. [CrossRef] [PubMed]
14.
Aguiló-Aguayo, I.; Suarez, M.; Plaza, L.; Hossain, M.B.; Brunton, N.; Lyng, J.G.; Rai, D.K. Optimization of pulsed electric field
pre-treatments to enhance health-promoting glucosinolates in broccoli flowers and stalk. J. Sci. Food Agric.
2015
,95, 1868–1875.
[CrossRef] [PubMed]
15.
Zhang, Z.; Talalay, P.; Cho, C.G.; Posner, G.H. A major inducer of anticarcinogenic protective enzymes from broccoli: Isolation
and elucidation of structure. Proc. Natl. Acad. Sci. USA 1992,89, 2399–2403. [CrossRef]
16.
Bonnesen, C.; Eggleston, I.M.; Hayes, J.D. Dietary Indoles and Isothiocyanates That Are Generated from Cruciferous Vegetables
Can Both Stimulate Apoptosis and Confer Protection against DNA Damage in Human Colon Cell Lines. Cancer Res.
2001
,
61, 6120–6130.
17.
Georgikou, C.; Yin, L.; Gladkich, J.; Xiao, X.; Sticht, C.; De la Torre, C.; Gretz, N.; Gross, W.; Schäfer, M.; Karakhanova, S.; et al.
Inhibition of miR30a-3p by sulforaphane enhances gap junction intercellular communication in pancreatic cancer. Cancer Lett.
2020,469, 238–245. [CrossRef]
18.
Hossain, S.; Liu, Z.; Wood, R.J. Histone deacetylase activity and vitamin D-dependent gene expressions in relation to sulforaphane
in human breast cancer cells. J. Food Biochem. 2019,44, e13114. [CrossRef]
19.
Wu, D.; Zheng, Z.; Fan, S.; Zhang, Z.; Chen, G.; Lu, J. Sulforaphane administration alleviates diffuse axonal injury (DAI) via
regulation signaling pathway of NRF2 and HO-1. J. Cell. Biochem. 2020,121, 430–442. [CrossRef]
Molecules 2021,26, 752 12 of 14
20.
Ishiura, Y.; Ishimaru, H.; Watanabe, T.; Fujimuro, M. Sulforaphane Exhibits Cytotoxic Effects against Primary Effusion Lymphoma
Cells by Suppressing p38MAPK and AKT Phosphorylation. Biol. Pharm. Bull. 2019,42, 2109–2112. [CrossRef]
21.
Xu, Y.; Han, X.; Li, Y.; Min, H.; Zhao, X.; Zhang, Y.; Qi, Y.; Shi, J.; Qi, S.; Bao, Y.; et al. Sulforaphane Mediates Glutathione Depletion
via Polymeric Nanoparticles to Restore Cisplatin Chemosensitivity. ACS Nano 2019,13, 13445–13455. [CrossRef] [PubMed]
22.
Lin, L.; Yeh, C.; Kuo, C.; Lee, C.; Yen, G.; Wang, L.; Wu, C.; Yang, W.; Wu, A.T.H. Sulforaphane Potentiates the Efficacy of Imatinib
against Chronic Leukemia Cancer Stem Cells through Enhanced Abrogation of Wnt/
β
-Catenin Function. J. Agric. Food Chem.
2012,60, 7031–7039. [CrossRef] [PubMed]
23.
Carrasco-Pozo, C.; Tan, K.N.; Rodriguez, T.; Avery, V.M. The Molecular Effects of Sulforaphane and Capsaicin on Metabolism
upon Androgen and Tip60 Activation of Androgen Receptor. Int. J. Mol. Sci. 2019,20, 5384. [CrossRef]
24.
Silva-Palacios, A.; Ostolga-Chavarría, M.; Sánchez-Garibay, C.; Rojas-Morales, P.; Galván-Arzate, S.; Buelna-Chontal, M.; Pavón,
N.; Pedraza-Chaverrí, J.; Königsberg, M.; Zazueta, C. Sulforaphane protects from myocardial ischemia-reperfusion damage
through the balanced activation of Nrf2/AhR. Free Radic. Biol. Med. 2019,143, 331–340. [CrossRef] [PubMed]
25.
Santín-Márquez, R.; Alarcón-Aguilar, A.; López-Diazguerrero, N.E.; Chondrogianni, N.; Königsberg, M. Sulforaphane-role in
aging and neurodegeneration. GeroScience 2019,41, 655–670. [CrossRef]
26.
Zeren, S.; Bayhan, Z.; Kocak, F.E.; Kocak, C.; Akcılar, R.; Bayat, Z.; Simsek, H.; Duzgun, S.A. Gastroprotective effects of
sulforaphane and thymoquinone against acetylsalicylic acideinduced gastric ulcer in rats. J. Surg. Res.
2016
,203, 348–359.
[CrossRef]
27.
Liang, J.; Hänsch, G.M.; Hübner, K.; Samstag, Y. Sulforaphane as anticancer agent: A double-edged sword? Tricky balance
between effects on tumor cells and immune cells. Adv. Biol. Regul. 2019,71, 79–87. [CrossRef]
28. De la Vega, M.R.; Chapman, E.; Zhang, D.D. NRF2 and the hallmarks of cancer. Cancer Cell 2018,34, 21–43. [CrossRef]
29.
Singh, A.; Daemen, A.; Nickles, D.; Jeon, S.M.; Foreman, O.; Sudini, K.; Gnad, F.; Lajoie, S.; Gour, N.; Mitzner, W.; et al. NRF2
Activation Promotes Aggressive Lung Cancer and Associates with Poor Clinical Outcomes. Clin. Cancer Res. 2020. [CrossRef]
30. Bailly, C. Regulation of PD-L1 expression on cancer cells with ROS-modulating drugs. Life Sci. 2020,246, 117403. [CrossRef]
31.
Townsend, B.E.; Johnson, R.W. Sulforaphane reduces lipopolysaccharide-induced proinflammatory markers in hippocampus and
liver but does not improve sickness behaviour. Nutr. Neurosci. 2017,20, 195–202. [CrossRef] [PubMed]
32.
Deng, Z.; Rong, Y.; Teng, Y.; Mu, J.; Zhuang, X.; Tseng, M.; Samykutty, A.; Zhang, L.; Yan, J.; Miller, D.; et al. Broccoli-Derived
Nanoparticle Inhibits Mouse Colitis by Activating Dendritic Cell AMP-Activated Protein Kinase. Mol. Ther.
2017
,25, 1641–1654.
[CrossRef] [PubMed]
33.
Liang, J.; Jahraus, B.; Balta, E.; Ziegler, J.D.; Hübner, K.; Blank, N.; Niesler, B.; Wabnitz, G.H.; Samstag, Y. Sulforaphane Inhibits
Inflammatory Responses of Primary Human T-Cells by Increasing ROS and Depleting Glutathione. Front. Immunol.
2018
,9, 2584.
[CrossRef] [PubMed]
34.
Tarozzi, A.; Angeloni, C.; Malaguti, M.; Morroni, F.; Hrelia, S.; Hrelia, P. Sulforaphane as a potential protective phytochemical
against neurodegenerative diseases. Oxid. Med. Cell. Longev. 2013,2013, 415078. [CrossRef] [PubMed]
35.
Holloway, P.M.; Gillespie, S.; Becker, F.; Vital, S.A.; Nguyen, V.; Alexander, J.S.; Evans, P.C.; Gavins, F.N.E. Sulforaphane induces
neurovascular protection against a systemic inflammatory challenge via both Nrf2-dependent and independent pathways. Vascul.
Pharmacol. 2016,85, 29–38. [CrossRef] [PubMed]
36.
Yong, H.; Chartier, G.; Quandt, J. Modulating inflammation and neuroprotection in multiple sclerosis. J. Neurosci. Res.
2018
,96,
927–950. [CrossRef]
37.
Adamczyk, B.; Adamczyk-Sowa, M. New insights into the role of oxidative stress mechanisms in pathophysiology and treatment
of multiple sclerosis. Oxid. Med. Cell. Longev. 2016,2016, 1973834. [CrossRef]
38.
Yoo, I.H.; Kim, M.J.; Kim, J.; Sung, J.J.; Park, S.T.; Ahn, S.W. The Anti-Inflammatory Effect of Sulforaphane in Mice with
Experimental Autoimmune Encephalomyelitis. J. Korean Med. Sci. 2019,34, e197. [CrossRef]
39.
Patel, V.; Dial, K.; Wu, J.; Gauthier, A.G.; Wu, W.; Lin, M.; Espey, M.G.; Thomas, D.D.; Ashby, C.R., Jr.; Mantell, L.L. Dietary
Antioxidants Significantly Attenuate Hyperoxia-Induced Acute Inflammatory Lung Injury by Enhancing Macrophage Function
via Reducing the Accumulation of Airway HMGB1. Int. J. Mol. Sci. 2020,21, 977. [CrossRef]
40.
Jin-Nyoung, H.O.; Kang, E.R.; Yoon, H.G.; Jeon, H.; Jun, W.; Watson, R.R.; Lee, J. Inhibition of Premature Death by
Isothiocyanates through Immune Restoration in LP-BM5 Leukemia Retrovirus-Infected C57BL/6 Mice. Biosci. Biotechnol.
Biochem. 2011,75, 1234–1239.
41.
Furuya, A.K.M.; Sharifi, H.J.; Jellinger, R.M.; Cristofano, P.; Shi, B.; de Noronha, C.M.C. Sulforaphane Inhibits HIV Infection of
Macrophages through Nrf2. PLoS Pathog. 2016,12, e1005581. [CrossRef] [PubMed]
42.
Olagnier, D.; Peri, S.; Steel, C.; van Montfoort, N.; Chiang, C.; Beljanski, V.; Slifker, M.; He, Z.; Nichols, C.N.; Lin, R.; et al. Cellular
Oxidative Stress Response Controls the Antiviral and Apoptotic Programs in Dengue Virus-Infected Dendritic Cells. PLoS Pathog.
2014,10, e1004566. [CrossRef] [PubMed]
43.
Page, A.; Volchkova, V.A.; Reid, S.P.; Mateo, M.; Bagnaud-Baule, A.; Nemirov, K.; Shurtleff, A.C.; Lawrence, P.; Reynard, O.;
Ottmann, M.; et al. Marburgvirus Hijacks Nrf2-Dependent Pathway by Targeting Nrf2-Negative Regulator Keap1. Cell. Rep.
2014,6, 1026–1036. [CrossRef] [PubMed]
44.
Qiu, L.; Fan, H.; Jin, W.; Zhao, B.; Wang, Y.; Ju, Y.; Chen, L.Z.; Chen, Y.; Duan, Z.P.; Meng, S.D. MiR-122-induced down-
regulation of HO-1 negatively affects miR-122-mediated suppression of HBV. Biochem. Biophys. Res. Commun.
2010
,398, 771–777.
[CrossRef] [PubMed]
Molecules 2021,26, 752 13 of 14
45.
Davinelli, S.; Scapagnini, G.; Denaro, F.; Calabrese, V.; Benedetti, F.; Krishnan, S.; Curreli, S.; Bryant, J.; Zella, D. Altered expression
pattern of Nrf2/HO-1 axis during accelerated-senescence in HIV-1 transgenic rat. Biogerontology
2014
,15, 449–461. [CrossRef]
[PubMed]
46.
Yu, J.S.; Chen, W.C.; Tseng, C.K.; Lin, C.K.; Hsu, Y.C.; Chen, Y.H.; Lee, J.C. Sulforaphane Suppresses Hepatitis C Virus Replication
by Up-Regulating Heme Oxygenase-1 Expression through PI3K/Nrf2 Pathway. PLoS ONE 2016,11, e0152236. [CrossRef]
47.
Vetvicka, V.; Vetvickova, J. Glucan supplementation enhances the immune response against an influenza challenge in mice.
Ann. Transl. Med. 2015,3, 22. [CrossRef]
48.
Kesic, M.J.; Simmons, S.O.; Bauer, R. Nrf2 expression modifies influenza A entry and replication in nasal epithelial cells. Free
Radic. Biol. Med. 2011,51, 444–453. [CrossRef]
49.
Vaclav, V.; Jana, V.A. Novel Glucan-Sulforaphane Combination Stimulates Immune Response to Influenza in Mouse Model. Am. J.
Immunol. 2016,12, 20–28. [CrossRef]
50.
Müller, L.; Meyer, M.; Bauer, R.N.; Zhou, H.; Zhang, H.; Jones, S.; Robinette, C.; Noah, T.L.; Jaspers, I. Effect of Broccoli Sprouts
and Live Attenuated Influenza Virus on Peripheral Blood Natural Killer Cells: A Randomized, Double-Blind Study. PLoS ONE
2016,11, e0147742. [CrossRef]
51.
Li, Z.; Liu, Y.; Fang, Z.; Yang, L.; Zhuang, M.; Zhang, Y.; Lv, H. Natural Sulforaphane From Broccoli Seeds Against Influenza A
Virus Replication in MDCK Cells. Nat. Prod. Commun. 2019,14, 1–8. [CrossRef]
52.
Tahaghoghi-Hajghorbani, S.; Zafari, P.; Masoumi, E.; Rajabinejad, M.; Jafari-Shakib, R.; Hasani, B.; Rafiei, A. The role of
dysregulated immune responses in COVID-19 pathogenesis. Virus Res. 2020,290, 198197. [CrossRef] [PubMed]
53.
Cho, H.Y.; Imani, F.; Miller-DeGraff, L.; Walters, D.; Melendi, G.A.; Yamamoto, M.; Polack, F.P.; Kleeberger, S.R. Antiviral activity
of Nrf2 in a murine model of respiratory syncytial virus disease. Am. J. Respir. Crit. Care Med.
2009
,179, 138–150. [CrossRef]
[PubMed]
54.
Lin, C.H.; Yao, C.A. Potential Role of Nrf2 Activators with Dual Antiviral and Anti-Inflammatory Properties in the Management
of Viral Pneumonia. Infect. Drug Resist. 2020,13, 1735–1741. [CrossRef] [PubMed]
55.
Cuadrado, A.; Pajares, M.; Benito, C.; Jiménez-Villegas, J.; Escoll, M.; Fernández-Ginés, R.; Garcia Yagüe, A.J.; Lastra, D.; Manda,
G.; Rojo, A.I.; et al. Can Activation of NRF2 Be a Strategy against COVID-19? Trends Pharmacol. Sci.
2020
,41, 598–610. [CrossRef]
56.
Horowitz, R.I.; Freeman, P.R. Three novel prevention, diagnostic, and treatment options for COVID-19 urgently necessitating
controlled randomized trials. Med. Hypotheses 2020,143, 109851. [CrossRef]
57.
Yanaka, A. Sulforaphane enhances protection and repair of gastric mucosa against oxidative stress
in vitro
, and demonstrates
anti-inflammatory effects on helicobacter pylori infected gastric mucosae in mice and human subjects. Curr. Pharm. Des.
2011
,17,
1532–1540. [CrossRef]
58.
Yanaka, A. Role of NRF2 in protection of the gastrointestinal tract against oxidative stress. J. Clin. Biochem. Nutr.
2018
,63, 18–25.
[CrossRef]
59.
Yanaka, A. Role of Sulforaphane in Protection of Gastrointestinal Tract Against H. pylori and NSAID-Induced Oxidative Stress.
Curr. Phar. Des. 2017,23, 4067–4075.
60.
Haodang, L.; Lianmei, Q.; Ranhui, L.; Liesong, C.; Jun, H.; Yihua, Z.; Cuiming, Z.; Yimou, W.; Xiaoxing, Y. HO-1 mediates the
anti-inflammatory actions of Sulforaphane in monocytes stimulated with a mycoplasmal lipopeptide. Chem. Biol. Interact.
2019
,
306, 10–18. [CrossRef]
61.
Ali, M.; Bonay, M.; Vanhee, V.; Vinit, S.; Deramaudt, T.B. Comparative effectiveness of 4 natural and chemical activators of Nrf2
on inflammation, oxidative stress, macrophage polarization, and bactericidal activity in an
in vitro
macrophage infection model.
PLoS ONE 2020,15, e0234484. [CrossRef]
62.
Dermaudt, T.B.; Ali, M.; Vinit, S.; Bonay, M. 1-3Sulforaphane reduces intracellular survival of Staphylococcus aureus in macrophages
through inhibition of JNK and p38 MAPK-induced inflammation. Int. J. Mol. Med. 2020,45, 1927–1941.
63.
Belchamber, K.B.R.; Donnelly, L.E. Targeting defective pulmonary innate immunity–A new therapeutic option? Pharmacol. Ther.
2020,209, 107500. [CrossRef] [PubMed]
64.
Russo, M.; Spagnuolo, C.; Russo, G.L.; Skalicka-Wo´zniak, K.; Daglia, M.; Sobarzo-Sánchez, E.; Nabavi, S.F.; Nabavi, S.M. Nrf2
targeting by sulforaphane: A potential therapy for cancer treatment. Crit. Rev. Food Sci. Nutr.
2017
,58, 1391–1405. [CrossRef]
[PubMed]
65.
Singh, S.V.; Warin, R.; Xiao, D.; Powolny, A.A.; Stan, S.D.; Arlotti, J.A.; Zeng, Y.; Hahm, E.R.; Marynowski, S.W.; Bommareddy, A.;
et al. Sulforaphane Inhibits Prostate Carcinogenesis and Pulmonary Metastasis in TRAMP Mice in Association with Increased
Cytotoxicity of Natural Killer Cells. Cancer Res. 2009,69, 2117–2125. [CrossRef]
66.
Bessler, H.; Djaldetti, M. Broccoli and human health: Immunomodulatory effect of sulforaphane in a model of colon cancer. Int. J.
Food. Sci. Nutr. 2018,69, 946–953. [CrossRef]
67.
Soundararajan, P.; Kim, J.S. Anti-Carcinogenic Glucosinolates in Cruciferous Vegetables and Their Antagonistic Effects on
Prevention of Cancers. Molecules 2018,23, 2983. [CrossRef]
68.
AL-Ishaq, R.K.; Overy, A.J.; Büsselberg, D. Phytochemicals and Gastrointestinal Cancer: Cellular Mechanisms and Effects to
Change Cancer Progression. Biomolecules 2020,10, 105. [CrossRef]
69.
Suzuki, H.; Mutoh, M.; Kamoshida, Y.; Kakinoki, N.; Yoshida, S.; Ebihara, T.; Hirose, M.; Hyodo, I.; Yanaka, A. Chemoprevention
against colon cancer by dietary intake of sulforaphane. Funct. Food Health. Dis. 2019,9, 392–411.
Molecules 2021,26, 752 14 of 14
70.
Palliyaguru, D.L.; Yang, L.; Chartoumpekis, D.V.; Wendell, S.G.; Fazzari, M.; Skoko, J.J.; Liao, Y.; Oesterreich, S.; Michalopoulos,
J.K.; Kensler, T.W. Sulforaphane Diminishes the Formation of Mammary Tumors in Rats Exposed to 17
β
-Estradiol. Nutrients
2020
,
12, 2282. [CrossRef]
71.
Sudini, K.; Diette, G.B.; Breysse, P.N.; McCormack, M.C.; Bull, D.; Biswal, S.; Zhai, S.; Brereton, N.; Peng, R.D.; Matsui, E.C.A.
Randomized Controlled Trial of the Effect of Broccoli Sprouts on Anti-oxidant Gene Expression and Airway Inflammation in
Asthmatics. J. Allergy Clin. Immunol. Pract. 2016,4, 932–940. [CrossRef]
... Cruciferae or Brassicaceae are an outstanding source of bioactive compounds such as polyphenols, essential minerals, ascorbic acid, and isothiocyanates [85]. This family consist of edible green plants species such as Brassica oleracea var. ...
... These were accompanied by amelioration of other abnormalities like brains' oxidative stress, inflammation, acetylcholinesterase (AChE)activity, and cerebral cortex histopathological changes myrosinase, a ß-thioglucosidase. Notably, this enzyme hydrolysis glucoraphanin to liberate glucose and form an unstable aglucone that spontaneously rearranges to a range of reactive products, such as isothiocyanate sulforaphane [85,86]. Sulforaphane, 1-isothiocyanato-4-(methylsulfinyl)butane), is a sulfur-rich compound, known of its role as an antioxidant and potent stimulator of endogenous detoxifying enzymes [87]. ...
... No doubt there has been a need to exploit the health-promoting effects of sulforaphane in humans. Briefly, in humans, sulforaphane can be administered directly in its active form or as glucoraphanin which undergoes the hydrolysis during digestion by the action of vegetable and gut microflora myrosinase [8,85,102]. Humans or mammalian do not produce the enzyme, myrosinase; however, the conversion of glucoraphanin to sulforaphane still occurs. ...
... The health-promoting effects of SFN are largely documented, and they include the prevention and treatment of several types of cancer and protection against cardiac diseases, hypertension, and neurodegenerative diseases, among others [1,2]. This is associated with its capacity to induce detoxifying Phase 2 enzymes [3]. ...
Article
Full-text available
Sulforaphane (SFN) is a bioactive compound widely studied for its potential applications in pharmaceutical, nutraceutical, and food industries since it offers health benefits due to its nature as a Phase 2 enzyme inducer. Its application in the food industry has been limited because SFN is unstable at high temperatures in an aqueous milieu. An option to increase SFN stability and protect it from thermal degradation is microencapsulation. The aim of this work was to optimize a microencapsulation process using oil-in-water emulsion to increase the thermal stability of SFN. The operation conditions that gave the highest entrapment efficiency were determined via experimental design and response surface methodology. Thermal degradation of microencapsulated SFN was studied at 37, 50, 60, and 70 °C. The optimum microencapsulation conditions were 8 min stirring, SFN/Gum Arabic ratio of 0.82, and surfactant/oil ratio of 1.0, resulting in an entrapment efficiency of 65%, which is the highest reported so far. The thermal stability of microencapsulated SFN was greatly enhanced compared with free SFN, with a 6-fold decrease in the degradation kinetic constant and a 41% increase in the activation energy. These results will contribute to a more efficient incorporation of SFN in various food matrices and explore new microencapsulation technologies to maximize the efficiency and stability of SFN.
... Sulforaphane (SFN), a compound found naturally in cruciferous vegetables, has potential therapeutic properties, including detoxi cation, antimicrobial, anti-in ammatory, and redox balancing [12]. SFN's therapeutic and protective properties may be attributed to the induction of the nuclear factor-erythroid-2-related factor 2 (Nrf2) transcription factor, which regulates antioxidant response elements, in ammation, non-enzyme antioxidants, and phase II detoxi cation enzymes [13]. SFN has been shown to induce the expression of quinone reductase and glutathione transferases, phase II anticarcinogenic enzymes, in murine hepatoma cells [14][15][16]. ...
Preprint
Full-text available
Background: In cancer therapy, combining treatments can result in additive and synergistic outcomes, reducing the development of drug resistance compared with monotherapy. We propose that combining Paclitaxel (Taxol, PTX) with Sulforaphane (SFN) may result in better treatment outcomes in prostate cancer. Understanding the mechanism of drug synergy, as opposed to simply knowing which drugs to combine, enables further optimization of advantageous drug interactions and can provide efficient therapeutic strategies in preclinical research. Methods: We measured apoptosis, cell cycle, and expression of Bax and Bcl2 in response to individual and combined treatments of PTX and SFN in PC-3 and LNCaP cells. Different concentrations of PTX, SFN, and their combination were used. We conducted Annexin V/PI positivity and data analysis using a flow cytometer and guava data acquisition and analysis software. Statistical analyses and graph generation were performed using Graph-Pad Prism 6 and Microsoft Excel software. Student’s t-tests or one-way analysis of variance with Tukey’s correction were used to determine the significant difference between mono- and combination treatments. Results: The effect of PTX or SFN treatments on reducing cell viability increased in a dose-dependent manner. Combined treatment enhanced PTX’s effects and reduced the EC50 values of both drugs compared to individual treatments. Flow cytometry analysis revealed that PTX or SFN treatments redistributed cell-cycle phases by inducing S-phase arrest and increasing the apoptotic cell population in PC-3 cells. These effects were further enhanced in the PTX+SFN combination group. Interestingly, the combination treatments did not affect the necrotic cells. Caspase-3 cleavage and morphological deformations of the cell nuclei were examined by western blot and fluorescent microscopy in response to mono- and combination treatments, indicating apoptotic cell death. Conclusion: The PTX or SFN treatments differentially modulated the expression of Bax and Bcl2 in PC-3 and LNCaP cell lines, and the combined treatment enhanced these effects in favor of cell apoptosis versus survival. Our data indicated that combination therapy of PTX and SFN significantly increased Bax protein expression and Bax: Bcl2 ratio compared to PTX or SFN individual treatments. These findings could help develop new biomarkers and guide therapy choices. Understanding the mechanism of drug synergy is essential to optimize drug interactions and provide efficient therapeutic strategies in preclinical research.
... Although SFN was isolated and identified in 1959, it did not gain people's attention until 1992 when Prochaska et al. developed a method for screening fruits and vegetable extracts that can induce phase 2 enzymes (Prochaska et al., 1992). Current research has confirmed that sulforaphane not only has detoxifying (Alkharashi et al., 2019), antioxidant (Akbari and Namazian, 2020), anti-inflammatory (Al-Bakheit and Abu-Qatouseh, 2020), antibacterial (Deramaudt et al., 2020), immune-regulating (Mahn and Castillo, 2021), obesityreducing (Çakır et al., 2022), cardiovascular disease-improving (Zhang et al., 2022), and diabetes-improving (Bose et al., 2020) effects but also has significant anti-cancer effects in many cancers, such as lung cancer (Wang et al., 2004), breast cancer (Fowke et al., 2003), colon cancer (Lin et al., 1998), and prostate cancer (Joseph et al., 2004). Due to the potent pharmacological effects of SFN, more and more studies are focusing on its impact on BC cells. ...
Article
Full-text available
Bladder cancer (BC) is a common form of urinary tract tumor, and its incidence is increasing annually. Unfortunately, an increasing number of newly diagnosed BC patients are found to have advanced or metastatic BC. Although current treatment options for BC are diverse and standardized, it is still challenging to achieve ideal curative results. However, Sulforaphane, an isothiocyanate present in cruciferous plants, has emerged as a promising anticancer agent that has shown significant efficacy against various cancers, including bladder cancer. Recent studies have demonstrated that Sulforaphane not only induces apoptosis and cell cycle arrest in BC cells, but also inhibits the growth, invasion, and metastasis of BC cells. Additionally, it can inhibit BC gluconeogenesis and demonstrate definite effects when combined with chemotherapeutic drugs/carcinogens. Sulforaphane has also been found to exert anticancer activity and inhibit bladder cancer stem cells by mediating multiple pathways in BC, including phosphatidylinositol-3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR), mitogen-activated protein kinase (MAPK), nuclear factor kappa-B (NF-κB), nuclear factor (erythroid-derived 2)-like 2 (Nrf2), zonula occludens-1 (ZO-1)/beta-catenin (β-Catenin), miR-124/cytokines interleukin-6 receptor (IL-6R)/transcription 3 (STAT3). This article provides a comprehensive review of the current evidence and molecular mechanisms of Sulforaphane against BC. Furthermore, we explore the effects of Sulforaphane on potential risk factors for BC, such as bladder outlet obstruction, and investigate the possible targets of Sulforaphane against BC using network pharmacological analysis. This review is expected to provide a new theoretical basis for future research and the development of new drugs to treat BC.
... Sulforaphane, present in broccoli sprout extracts, has been studied in over 70 completed or recruiting clinical trials since the late 1990s including malignancy, diabetes, gastroesophageal reflux, schizophrenia, asthma, and influenza [12]. Sulforaphane has anti-inflammatory, antioxidant, and antiviral activity through its activity on the master gene regulator nuclear factor erythroid 2-related factor 2 (Nrf2) [15][16][17]. Nrf2 is a basic region leucine-zipper transcription factor predominantly known for its ability to bind and activate the antioxidant response element (ARE) of various genes to promote the expression of numerous phase II detoxification enzymes including glutathione S-transferase (GST), NADPH quinine oxidoreductase 1 (NQO-1), and heme-oxygenase 1 (HO-1) [18,19]. The evidence for an antioxidant effect from sulforaphane via its upregulation of Nrf2 has been summarised in a number of recent reviews [20][21][22]. ...
Article
Full-text available
Since its discovery in late 2019, the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been estimated to be responsible for at least 769.3 million infections and over 6.95 million deaths. Despite significant global vaccination efforts, there are limited therapies that are considered safe and effective for use in the management of COVID-19 during pregnancy despite the common knowledge that pregnant patients have a much higher risk of adverse outcomes. A bioactive compound found in broccoli sprout—sulforaphane—is a potent inducer of phase-II detoxification enzymes promoting a series of potentially beneficial effects notably as an antioxidant, anti-inflammatory, and anti-viral. A pilot, double-blinded, placebo-controlled randomised trial is to be conducted in Melbourne, Australia, across both public and private hospital sectors. We will assess a commercially available broccoli sprout extract in pregnant women between 20+0 and 36+0 weeks gestation with SARS-CoV-2 infection to investigate (i) the duration of COVID-19 associated symptoms, (ii) maternal and neonatal outcomes, and (iii) biomarkers of infection and inflammation. We plan to enrol 60 outpatient women with COVID-19 irrespective of vaccination status diagnosed by PCR swab or RAT (rapid antigen test) within five days and randomised to 14 days of oral broccoli sprout extract (42 mg of sulforaphane daily) or identical microcrystalline cellulose placebo. The primary outcome of this pilot trial will be to assess the feasibility of conducting a larger trial investigating the duration (days) of COVID-19-associated symptoms using a broccoli sprout supplement for COVID-19-affected pregnancies. Pregnant patients remain an at-risk group for severe disease following infection with SARS-CoV-2 and currently unclear consequences for the offspring. Therefore, this study will assess feasibility of using a broccoli sprout supplement, whilst providing important safety data for the use of sulforaphane in pregnancy.
Article
Open heart surgery is often an unavoidable procedure for treatment of coronary artery disease. The procedure associated reperfusion injury affects postoperative cardiac performance and long-term outcome. We addressed here whether cardioplegia essential for cardiopulmonary bypass surgery activates Nrf2, a transcription factor regulating the expression of antioxidant and detoxification genes. With commonly used cardioplegic solutions, High K ⁺ , Low K ⁺ , Del Nido (DN), histidine-tryptophan-ketoglutarate (HTK) and Celsior (CS), we found that DN caused a significant increase of Nrf2 protein in AC16 human cardiomyocytes. Tracing the ingredients in DN led to the discovery of KCl at the concentration of 26 to 60 mM capable of significant Nrf2 protein induction. The Antioxidant Response Element (ARE) luciferase reporter assay confirmed Nrf2 activation by DN or KCl. Transcriptomic profiling using RNA-seq revealed that oxidation-reduction as a main gene ontology group affected by KCl. KCl indeed elevated the expression of classical Nrf2 downstream targets, including TXNRD1, AKR1C, AKR1B1, SRXN1 and G6PD. DN or KCl induced Nrf2 elevation is Ca ²⁺ concentration dependent. We found that KCl decreased Nrf2 protein ubiquitination and extended the half-life of Nrf2 from 17.8 to 25.1 mins. Knocking out Keap1 blocked Nrf2 induction by K ⁺ . Nrf2 induction by DN or KCl correlates with the protection against ROS generation or loss of viability by H 2 O 2 treatment. Our data support that high K ⁺ concentration in DN cardioplegic solution can induce Nrf2 protein and protect cardiomyocytes against oxidative damage.
Article
Full-text available
Elevated levels of estrogen are a risk factor for breast cancer. In addition to inducing DNA damage, estrogens can enhance cell proliferation as well as modulate fatty acid metabolism that collectively contributes to mammary tumorigenesis. Sulforaphane (SFN) is an isothiocyanate derived from broccoli that is currently under evaluation in multiple clinical trials for prevention of several diseases, including cancer. Previous studies showed that SFN suppressed DNA damage and lipogenesis pathways. Therefore, we hypothesized that administering SFN to animals that are co-exposed to 17β-estradiol (E2) would prevent mammary tumor formation. In our study, 4–6 week old female August Copenhagen Irish rats were implanted with slow-release E2 pellets (3 mg x 3 times) and gavaged 3x/week with either vehicle or 100 μmol/kg SFN for 56 weeks. SFN-treated rats were protected significantly against mammary tumor formation compared to vehicle controls. Mammary glands of SFN-treated rats showed decreased DNA damage while serum free fatty acids and triglyceride species were 1.5 to 2-fold lower in SFN-treated rats. Further characterization also showed that SFN diminished expression of enzymes involved in mammary gland lipogenesis. This study indicated that SFN protects against breast cancer development through multiple potential mechanisms in a clinically relevant hormonal carcinogenesis model.
Article
Full-text available
Acute respiratory distress syndrome (ARDS) caused by SARS-CoV-2 is largely the result of a dysregulated host response, followed by damage to alveolar cells and lung fibrosis. Exacerbated pro-inflammatory cytokines release (cytokine storm) and loss of T-lymphocytes (leucopenia) characterize the most aggressive presentation. Here we propose that a multi-faceted anti-inflammatory strategy based on pharmacological activation of nuclear factor erythroid 2 p45-related factor 2 (NRF2), can be deployed against the virus. The strategy provides robust cytoprotection by restoring the redox and protein homeostasis, promoting resolution of inflammation, and facilitating repair. NRF2 activators such as sulforaphane and bardoxolone methyl are already in clinical trials. The safety and efficacy information of these modulators in humans, together with their well-documented cytoprotective and anti-inflammatory effects in preclinical models, highlight the potential of this armamentarium for deployment to the battlefield against COVID-19.
Article
Full-text available
Chih-Yin Lin,1 Chun-An Yao2 1Department of Neurology, Chang Gung Memorial Hospital, Linkou 333, Taiwan; 2Department of Dermatology, Cathay General Hospital, Taipei, TaiwanCorrespondence: Chun-An YaoDepartment of Dermatology, Cathay General Hospital, 280 Renai Road Sec. 4, Taipei, TaiwanTel +886-2-27082121Fax +886-2-2709-6521Email chunany115@gmail.comAbstract: The outbreak of coronavirus disease 2019 (COVID-19) pandemic has already caused a huge burden to the global healthcare system, with the death toll reached tens of thousands. Although some antiviral agents were identified and used to inhibit viral replication, the management of cytokine storm is also a critical issue. In this article, we reviewed the literature on drug candidates for severe acute respiratory syndrome (SARS-CoV-1) and provided a brief overview of a class of drugs that exert antiviral and anti-inflammatory effects. These molecules mitigated inflammatory cytokine cascades induced by viral infections via Nrf2 activating capacity and might have additional anti-fibrotic and anti-remodeling properties. Besides, their effects on the regulation of scavenger receptors expression by macrophages may offer some benefits to the pulmonary antibacterial defense system after viral infection. The potential roles of these agents assessed on the basis of the pathophysiology of viral pneumonia and acute respiratory distress syndrome were also discussed. Further research is needed to ascertain whether Nrf2 activators are useful in the management of viral pneumonia.Keywords: COVID-19, viral pneumonia, Nrf2 activators, curcumin, sulforaphane, macrolide
Article
Full-text available
Inflammation plays a crucial role in the defense response of the innate immune system against pathogen infection. In this study, we selected 4 compounds for their potential or proven anti-inflammatory and/or anti-microbial properties to test on our in vitro model of bacteria-infected THP-1-derived macrophages. We first compared the capacity of sulforaphane (SFN), wogonin (WG), oltipraz (OTZ), and dimethyl fumarate (DMF) to induce the nuclear factor erythroid 2-related factor 2 (Nrf2), a key regulator of the antioxidant, anti-inflammatory response pathways. Next, we performed a comparative evaluation of the antioxidant and anti-inflammatory efficacies of the 4 selected compounds. THP-1-derived macrophages and LPS-stimulated macrophages were treated with each compound and expression levels of genes coding for inflammatory cytokines IL-1β, IL-6, and TNF-α were quantified by RT-qPCR. Moreover, expression levels of genes coding for M1 (IL-23, CCR7, IL-1β, IL-6, and TNF-α) and M2 (PPARγ, MRC1, CCL22, and IL-10) markers were determined in classically-activated M1 macrophages treated with each compound. Finally, the effects of each compound on the intracellular bacterial survival of gram-negative E. coli and gram-positive S. aureus in THP-1-derived macrophages and PBMC-derived macrophages were examined. Our data confirmed the anti-inflammatory and antioxidant effects of SFN, WG, and DMF on LPS-stimulated THP-1-derived macrophages. In addition, SFN or WG treatment of classically-activated THP-1-derived macrophages reduced expression levels of M1 marker genes, while SFN or DMF treatment upregulated the M2 marker gene MRC1. This decrease in expression of M1 marker genes may be correlated with the decrease in intracellular S. aureus load in SFN- or DMF-treated macrophages. Interestingly, an increase in intracellular survival of E. coli in SFN-treated THP-1-derived macrophages that was not observed in PBMC-derived macrophages. Conversely, OTZ exhibited pro-oxidant and proinflammatory properties, and affected intracellular survival of E. coli in THP-1-derived macrophages. Altogether, we provide new potential therapeutic alternatives in treating inflammation and bacterial infection.
Article
Full-text available
Sulforaphane (SFN) is a health-promoting compound occurring in broccoli. It is formed by action of myrosinase in a two-step reaction that also yields undesirable compounds such as nitriles and isothionitriles. Different techniques affecting enzyme activity and tissue integrity were proposed to increase SFN content in the edible parts and discards of broccoli. Ultrasound processing is an emerging technology that produces these effects in foods, but has been poorly explored in broccoli so far. The aim of this work was to study the effect of ultrasound-assisted blanching on myrosinase activity and SFN content in broccoli florets. Myrosinase showed first-order inactivation kinetics in blanching at different temperatures with and without ultrasound processing. The inactivation rate was faster using ultrasound, with kinetic constants two orders of magnitude higher than without ultrasound. The activation energy (Ea) in traditional blanching (57.3 kJ mol−1) was higher than in ultrasound-assisted blanching (15.8 kJ mol−1). Accordingly, ultrasound accelerates myrosinase inactivation. The blanching time and temperature significantly affected myrosinase activity and SFN content. At 60 °C and 4 min of ultrasound-assisted blanching, myrosinase activity was minimum and SFN content was the highest. These findings may help to design SFN enrichment processes and will contribute to the valorization of agro-industrial wastes.
Article
Full-text available
Purpose Asymptomatic or minimally symptomatic infection with COVID-19 can result in silent transmission to large numbers of individuals, resulting in expansion of the pandemic with a global increase in morbidity and mortality. New ways of screening the general population for COVID-19 are urgently needed along with novel effective prevention and treatment strategies. Hypothesis A hypothetical three-part prevention, diagnostic, and treatment approach based on an up-to-date scientific literature review for COVID- 19 is proposed. Regarding diagnosis, a validated screening questionnaire and digital app for COVID-19 could help identify individuals who are at risk of transmitting the disease, as well as those at highest risk for poor clinical outcomes. Global implementation and online tracking of vital signs and scored questionnaires that are statistically validated would help health authorities properly allocate essential health care resources to test and isolate those at highest risk for transmission and poor outcomes. Second, regarding prevention, no validated protocols except for physical distancing, hand washing, and isolation exist, and recently ivermectin and the antimalarial drugs chloroquine and hydroxychloroquine has been published to have anti-viral properties against COVID-19. A randomized trial of ivermectin or hydroxychloroquine alone, and/or nutraceuticals that have been published to support immune function including Vitamin C, zinc, and immunomodulatory supplements (3,6 Beta glucan) could be beneficial in preventing transmission or lessening symptomatology but requires statistical validation. Third, concerning treatment, COVID-19 induced inflammation and “cytokine storm syndrome” with hemophagocytic lymphohistiocytosis (HLH)/Macrophage Activation Syndrome (MAS) have resulted in extreme morbidity and mortality in those with certain comorbidities, secondary to “acute respiratory distress syndrome” (ARDS) and multiorgan dysfunction with disseminated intravascular coagulation (DIC). Deficiency in red blood cell, serum and alveolar glutathione has been published in the medical literature for ARDS, as well as viral and bacterial pneumonias, resulting from increased levels of free radical/oxidative stress. A randomized controlled trial of blocking NFKappa B and cytokine formation using glutathione precursors (N-acetyl-cysteine [NAC] and alpha lipoic acid) and PO/IV glutathione should be performed, along with an evaluation of Nrf2 activators (curcumin, sulforaphane glucosinolate) which have been scientifically proven to lower inflammation. Since high mortality rates from sepsis induced DIC due to COVID 19 infection has also been associated with thrombotic events and elevated levels of D-dimer, randomized controlled trials of using anticoagulant therapy with heparin is urgently required. This is especially important in patients on ventilators who have met certain sepsis induced coagulopathy (SIC) criteria. The use of acetazolamide with or without sildenafil also needs to be explored with or without heparin, since increased oxygen delivery to vital organs through prevention of thrombosis/pulmonary emboli along with carbonic anhydrase inhibition may help increase oxygenation and prevent adverse clinical outcomes. Conclusion and Implications a three-part prevention, diagnostic, and treatment plan is proposed for addressing the severe complications of COVID-19. Digital monitoring of symptoms to clinically diagnose early exposure and response to treatment; prevention with hydroxychloroquine and/or ivermetin as well as nutritional therapies that support a healthy immune response; treatment with anti-inflammatory therapies that block NFKappaB and activate Nrf2 pathways, as well as novel therapies that address COVID-19 pneumonia and ARDS with DIC including anticoagulation and/ or novel respiratory therapies with or without acetazolamide and sildenafil. These three broad-based interventions urgently need to be subjected to randomized, controlled trials.
Article
Full-text available
Macrophages are active contributors to the innate immune defense system. As macrophage activation is clearly affected by the surrounding microenvironment, the present study investigated the effect of sulforaphane (SFN) on the bactericidal activity of macrophages and the underlying molecular mechanisms involved in this process. Human THP‑1‑derived macrophages, primary human peripheral blood mononuclear cell‑derived macrophages, and primary mouse bone marrow derived‑macrophages (BMDMs) pretreated with SFN or DMSO were utilized in a model of Staphylococcus aureus infection. The results suggested that SFN pretreatment of macrophages effectively repressed the intracellular survival of S. aureus through modulation of p38/JNK signaling and decreased S. aureus‑induced caspases‑3/7‑dependent cell apoptosis, potentially through downregulation of microRNA (miR)‑142‑5p and miR‑146a‑5p. As SFN is a well‑known activator of nuclear factor erythroid 2‑related factor 2 (Nrf2), Nrf2‑/‑ BMDMs were used to demonstrate that the SFN‑mediated inhibitory effect was independent of Nrf2. Nevertheless, an increase in intracellular bacterial survival in Nrf2‑deficient macrophages was observed. In addition, SFN pretreatment suppressed S. aureus‑induced transcriptional expression of genes coding for the proinflammatory cytokines interleukin (IL)‑1β, IL‑6, and tumor necrosis factor‑α (TNF‑α), as well as for the M1 markers C‑C motif chemokine receptor 7, IL‑23 and inducible nitric oxide synthase (iNOS). Western blot analysis indicated that S. aureus challenge activated p38 mitogen‑activated protein kinase (MAPK) (p38) and c‑Jun N‑terminal kinase (JNK) MAPK signaling pathways, while SFN pretreatment prevented p38 and JNK phosphorylation. Pretreatment with 2 specific inhibitors of p38 and JNK, SB203580 and SP600125, respectively, resulted in a decrease in S. aureus‑induced proinflammatory gene expression levels compared with those observed in the SFN‑pretreated macrophages. Furthermore, THP‑1‑derived macrophages pretreated with SB203580 or SP600125 prior to bacterial infection exhibited a significant inhibition in intracellular S. aureus survival. In conclusion, we hypothesize that concomitant targeting of the p38/JNK‑inflammatory response and the S. aureus‑induced apoptosis with SFN may be a promising therapeutic approach in S. aureus infection.
Article
Purpose: Stabilization of the transcription factor NRF2 through genomic alterations in KEAP1 and NFE2L2 occurs in a quarter of lung adenocarcinoma (LUAD) and a third of lung squamous (LUSC) patients. In LUAD, KEAP1 loss often co-occurs with STK11 loss and KRAS activating alterations. Despite its prevalence, the impact of NRF activation on tumor progression and patient outcomes is not fully defined. Experimental design: We model NRF2 activation, STK11 loss and KRAS activation in vivo using novel genetically engineered mouse models. Further, we derive a NRF2 activation signature from human non-small cell lung tumors that we use to dissect how these genomic events impact outcomes and immune contexture of participants in the OAK and IMpower131 immunotherapy trials. Results: Our in vivo data reveal roles for NRF2 activation in (i) promoting rapid-onset, multi-focal intra-bronchiolar carcinomas, leading to lethal pulmonary dysfunction, and (ii) decreasing elevated redox stress in KRAS-mutant, STK11-null tumors. In patients with non-squamous tumors, the NRF2 signature is negatively prognostic independently of STK11 loss. LUSC patients with low NRF2 signature survive longer when receiving anti-PD-L1 treatment. Conclusions: Our in vivo modeling establishes NRF2 activation as a critical oncogenic driver, cooperating with STK11 loss and KRAS activation to promote aggressive LUAD. In patients, oncogenic events alter the tumor immune contexture, possibly impacting treatment responses. Importantly, patients with NRF2 activated non-squamous or squamous tumors have poor prognosis and show limited response to anti-PD-L1 treatment.
Article
The coronavirus disease-2019 (COVID-19) which caused by severe acute respiratory syndrome-related coronavirus (SARS-CoV-2), is a pandemic threat to global public health. It has a wide spectrum of clinical manifestations from mild to critical illness, the most serious of which is the complications of acute respiratory distress syndrome (ARDS). SARS-CoV-2 infection appears mild in infants and children, however, in adults, it can lead to serious consequences. In this review, we highlighted the differences between the immune responses of the lung in children and adults, immune dysregulation and their possible role in clinical manifestations in COVID-19. There is a reduction in population of immunocompetent cells during aging and subsequently induced ineffective inflammation in the faces of some infections. Dysregulation in the immune system can lead to an unappropriated local and systemic immune responses and subsequently the rapid spread of the virus, leading to severe COVID-19 disease. Therefore, recognizing the differences in the immune responses of various hosts as well as to improve the immune system disorder should always be part of research and treatment protocols.
Article
Broccoli offers several health benefits due to its high content of bioactive compounds, especially sulforaphane – a promising anticancer compound. In Chile, the broccoli crop is expanding rapidly, creating opportunities to industrialize this vegetable and exploit its functional properties. Various processes have been studied with the aim of maximizing the sulforaphane content in processed broccoli using different dehydration methods. These have shown large differences in terms of recovery and cost. This work shows an economic feasibility study of a small‐scale plant for the production of sulforaphane‐rich broccoli flour considering three drying alternatives: tray drying (TD), pulsed fluidized‐bed drying, and freeze‐drying (FD). Freeze drying resulted in the highest sulforaphane content in the broccoli flour; however, this process entails the highest production cost. Tray drying showed the lowest production cost and a comparatively high sulforaphane content in the final product. Pulsed fluidized bed drying (PFBD) resulted in the highest sulforaphane loss. Although the three drying technologies resulted in positive net present values (NPV), the internal rate of return (IRR) was the lowest for freeze drying as a result of the high investment – over 20% higher than the investment required by the other alternatives. The NPV was practically independent of the drying technology when the sulforaphane content was considered in the estimation of the selling price. © 2020 Society of Chemical Industry and John Wiley & Sons, Ltd