The Immunomodulatory Effect of Alpha-Lipoic Acid

Review Article
The Immunomodulatory Effect of Alpha-Lipoic Acid in
Autoimmune Diseases

Wei Liu ,

1 Lian-jie Shi,2 and Sheng-guang Li 2

1
Department of Respiratory and Critical Care Medicine, e 900th Hospital of the Joint Logistic Support Force, PLA,
Fujian Medical University, Fuzhou 350025, China
2
Department of Rheumatology and immunology, Peking University International Hospital, Beijing 102206, China
Correspondence should be addressed to Sheng-guang Li; lishengguang@vip.sina.com
Received 10 September 2018; Revised 11 February 2019; Accepted 21 February 2019; Published 20 March 2019
Academic Editor: Hai-Feng Pan
Copyright © 2019 Wei Liu et al. Tis is an open access article distributed under the Creative Commons Attribution License, which
permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Αlpha-lipoic acid is a naturally occurring antioxidant in human body and has been widely used as an antioxidant clinically.
Accumulating evidences suggested that �-lipoic acid might have immunomodulatory efects on both adaptive and innate immune
systems. Tis review focuses on the evidences and potential targets involved in the immunomodulatory efects of �-lipoic acid. It
highlights the fact that �-lipoic acid may have benefcial efects in autoimmune diseases once the immunomodulatory efects can
be confrmed by further investigation.

1. Background
Αlpha-lipoic acid (ALA) is a naturally occurring dithiol
compound that is wildly synthesized in the mitochondrion by
plants and animals. Physiologically, ALA is a cofactor for �-

ketoglutarate dehydrogenase complex to protect mitochon-
dria from oxidative attack.

ALA and dihydrolipoic acid (DHLA) are the oxidized
form and reduced form of LA, respectively. Tey are a pair of
powerful redox couple which can directly scavenge reactive
oxygen species (ROS), chelate metals, and regenerate other
antioxidants to show antioxidant biochemical properties.
With both liposoluble and water-soluble dual properties,

ALA and DHLA can fully function intracellularly and extra-
cellularly [1, 2].

Based on its cogent antioxidant properties and proven
safety, ALA has been widely used to treat oxidative stress
associated diseases, such as diabetes, neurological diseases,
and cardiovascular diseases. More and more studies on LA
had been performed; better understanding had been achieved
towards the mechanisms of molecules, including the fact that
ALA stimulated glucose uptake in insulin-sensitive cells and
enhanced both the antioxidant defenses and the function
of endothelial vascular cells [3]. Several lines of evidence
suggested that ALA might have immunomodulatory efect.

With the progress of investigation of basic and clinical
immunology, the role of oxidative stress on the pathogenesis
of some autoimmune diseases has been generally recognized
and interaction of ROS with the immune system well proven.
On one hand, ROS may have a physiological role in signal
transduction of all kinds of immune cells. For example,
macrophage produced ROS to kill bacteria and regulatory T
cells (Treg) released ROS to suppress activation of other T
cells [4]. On the other hand, in pathological status, immune
cells produce excessive ROS which exacerbated infammation
and broke balance in the immune system. For example,
oxidative stress was one of the contributors to immune system
dysregulation and dysfunction [5], which in turn led to
deteriorate oxidative stress in systemic lupus erythematosus
(SLE) [6, 7]. Both oxidative stress and immune dysfunction
participated in the development and progression of SLE.

Recently, redox-controlled activation of the mechanistic tar-
get of rapamycin (mTOR) has been recognized to play a

critical regulatory role in the immune system [8], which
highly implies that mTOR is a key bridge of metabolic stress
and autoimmunity. It can be speculated that the antioxidant
may be used for the treatment of certain autoimmune diseases
based on evidence that ROS clearance played a role in
immune regulation. Here in, we summarize the evidence

Hindawi
BioMed Research International
Volume 2019, Article ID 8086257, 11 pages
https://doi.org/10.1155/2019/8086257

2 BioMed Research International
of the immunomodulatory efects of ALA and possible
mechanisms involved by literature review.
1.1. e Involved Signaling Pathways of ALA

1.1.1. IKK�, Ras/Erk1/2, and PI3K/Akt/mTOR Signaling Path-
ways. Te mTOR, which could drive expansion of plas-
mablast [9] and T follicular helper (Tf) cells [10], induce

the diferentiation of T1 and T17 [11], and restrict the
diferentiation of Treg [12] and CD8 memory T cells [13], is
an essential mediator of immunity.
ALA had been shown to regulate upstream kinases of
mTOR in multiple pathological conditions [1].

ALA blocked TNF-� induced IKK/NF-�B signaling cas-
cades in RA-FLS and human umbilical vein endothelial cells

(HUVECs) [14, 15]. TNF-� activated mTORC1 pathway via
IKK� activation in tumor angiogenesis and insulin resistance
[16, 17].Terefore, it can be speculated that ALA may suppress
IKK�-mediated activation of mTORC1.
ALA inhibited Erk signaling to improve atherosclerotic

lesions and inhibit vascular smooth muscle cells prolifer-
ation [18]. ALA also inhibited activation of Erk mediated

by 5-hydroxytryptamine (5-HT) [19] and epidermal growth
factor (EGF), basic fbroblast growth factor (bFGF), and
platelet-derived growth factor (PDGF) [20]. Te activation
of Akt/S6K1 and Erk suppressed by ALA attenuated hepatic
stellate cell activation and ROS generation stimulated by
TGF-�/PDGF [21].
However, Erk signaling activated by ALA has been

reported to protect cardiovascular system and nervous sys-
tem. ALA increased heme oxygenase-1 (HO-1) to protect vas-
cular smooth muscle cells [22], ameliorated glucose/glucose

oxidase- (G/GO-) induced injury of rat cardiomyoblast

[23], inhibited adipocyte diferentiation [24], protected cor-
tical neurons from 4-hydroxy-2-nonenal- (HNE-) mediated

oxidative damage and neurotoxicity [25], and promoted
neurite outgrowth via activation of Erk [26].
Te efect of bidirectional regulation of Erk1/2 kinases on
mouse fbroblasts mediated by ALA was dependent on the
cell culture medium containing serum or not [27, 28], which,
to some extent, can interpret the fact that ALA regulated the
same kinase in diferent directions in diferent pathological
states.
ALA activated Akt kinase to protect pancreatic beta cells
from hydrogen peroxide-mediated oxidative stress [29]. In
rat L6 muscle cells, ALA mitigated insulin resistance via Akt
activation and Erk inhibition [30].

ALA enhanced apoptosis and suppressed cell prolifera-
tion of human breast cancer cell line [31]. ALA also induced

hepatoma cells apoptosis [32] to exert antitumor efects by
suppression of PI3K/Akt pathway. ALA also inhibited leptin
production of adipocytes and ameliorated insulin resistance
of Goto-Kakizaki (GK) rat to improve disorders of glucose
and lipid metabolism [33, 34].
Cytoprotective efect of ALA also could be mediated

through phosphorylation of Akt kinase to ameliorate endo-
plasmic reticulum stress-induced FRTL5 thyroid cell death

[35], protect neurons from injury induced by bupivacaine,

amyloid and hydrogen peroxide [36, 37], reduce ischemia-
reperfusion injury [38, 39] and oxidative stress injury of rat L6

muscle cells induced by TNF� and palmitate [30], decrease
hydrogen peroxide-induced apoptosis of pancreatic beta
cells [29], attenuate LPS-induced cardiac dysfunction [40],
monocyte activation, and acute infammatory responses [41],
and ameliorate vascular endothelial dysfunction [42, 43].
1.1.2. AMPK Signaling Pathway. ALA has been reported
to activate AMPK to upregulate adipose triglyceride lipase
(ATGL) to reduce body weight and visceral fat content of
diabetic mice [44].
Trough AMPK/mTORC1/S6K1 signaling pathway,
leucine and glucose induced insulin resistance which

could be attenuated by ALA via TSC2-mTOR inhibitors-
phosphorylation [45, 46] and AMPK activation [47] in

skeletal muscle. ALA also activated AMPK to downregulate
expression of S6K1 [48] leading to inhibition of insulin
secretion in pancreatic beta cells, which implies involvement
of mTOR.

However, ALA was also reported to inhibit the phospho-
rylation of AMPK, which suppressed appetite and prevented

obesity in the hypothalamus [49–51] consistent with the efect
of ALA on peripheral tissue to improve insulin resistance and
decrease lipid accumulation and lipogenesis.
Overall, ALA regulated some upstream kinase of mTOR
in inconsistent directions in diverse cell types of diferent
diseases. It has been proven that mTOR can modulate T cell
diferentiation and inhibit Treg cells which are defcient in
SLE patients [52, 53]. N-acetylcysteine (NAC), a well-known
antioxidant, has been reported to inhibit mTOR in vitro [54]
and improve the outcome of murine lupus [55] and even SLE
patients [56]. It has also been observed that disease activity
could be reduced and Treg populations could be reversed by
mTOR blockade in treating SLE patients [57]. Although these
existing indirect evidence was tempting to conclude that ALA
had efect on regulation of mTOR signaling, regulation of the
mTOR pathway by ALA in immune cells is worthy of further
investigation for patients with autoimmune diseases of high
relapse rate and poor responsiveness to traditional treatment.
2. Effects of ALA on Immune System
Te proven efects of ALA on adaptive immune cells,
including T and B cells, are briefy summarized in Table 1
and Figure 1, and the proven efects on innate immune
cells including NK cells, macrophages, and monocytes are
summarized in Table 1, all of which will be discussed in detail
below.
2.1. Effects on Adaptive Immune Cells
2.1.1. Effects on T Cells. Multiple sclerosis (MS) is an
autoimmune disease in central nervous system, which is
characterized by the migration and the long-term survival
of myelin-specifc T lymphocytes into the central nervous
system (CNS). A common model of MS is experimental
autoimmune encephalomyelitis (EAE). Studies demonstrated
the benefcial efects of ALA on treating EAE by suppressing

BioMed Research International 3

Table 1: Evidence of ALA on adaptive immune cells.

T cell B cell

Animal model

EAE

Decrease the number of T17 and T1 in

CNS;
Increase Treg numbers in spleen;
Reduce migration.

High fat diet mice

Recover transcriptional levels of the
diferentiation-related genes of jejunal T

cells.

Restore transcriptional levels of BCR signaling
pathway relating genes;
Decrease the apoptotic percentage of splenic B

lymphocytes.

Atopic dermatitis Suppress production of IFN-� and IL-4

by CD4+T. Reduce total serum IgE levels.

Models of established
atherosclerosis

Reduce T cell migration in response to
chemokines.

Endotoxemia mice Increase the number of splenic B cells.

Patients

AIDS

Increase the number of T cells;
Improve the lymphocyte proliferation

response;

Ameliorate the impaired mitochondrial
function of CD4+T cells.
Advanced cancer Induce lymphocyte progression from
G0/G1 to S phase.
Jurkat T cells Inhibit NF-�B activation induced by TNF
Reduce migration.

Normal human

Increase cAMP which afects
proliferation and activation of T cells;
Down-regulate the expression of CD4
molecules;
Reduce migration.

ALA: �-lipoic acid.
EAE: experimental autoimmune encephalomyelitis.
T17: T helper cell 17.
T1: T helper cell 1.
CNS: central nervous system.
Treg: regulatory T cells.
BCR: B-cell receptor.
IFN-�: interferon-�.
AIDS: acquired immunodefciency syndrome.
NF-�B: nuclear factor kappa B.
TNF: tumor necrosis factor.
cAMP: cyclic adenosine monophosphate.

the infltration of infammatory cells [58–61]. Recently, Wang
and colleagues [60] reported that ALA reduced the number
of T17 and T1 cells in CNS and increased the number
of splenic Treg cells in EAE mice. It highly implied that
ALA showed immunomodulatory efects on diferentiation
and proliferation of T cells. ALA has also been reported to
increase cAMP synthesis through activation of prostaglandin
receptors (EP2 and EP4) in peripheral blood T cells [62]
(Figure 1, A). Te elevated levels of intracellular cAMP
decreased the expression of IL-2 and IL-2R� (CD25) [63]
(Figure 1, B), which in turn afected proliferation and
activation of T cells [64].
More studies indicated that ALA could regulate function
of T cells in many ways. ALA could ameliorate the impaired

mitochondrial function of CD4+T cells of acquired immun-
odefciency syndrome (AIDS) patients [65], downregulate

the expression of CD4 molecules of human peripheral blood

T cells [66], inhibit nuclear factor kappa B (NF-�B) activation
induced by tumor necrosis factor-alpha (TNF) in Jurkat T
cells [67] (Figure 1, C), suppress production of interferon-
� (IFN-�) and interleukin-4 (IL-4) (Figure 1, D) by CD4+T
cells to reduce the severity of atopic dermatitis lesions in mice
model [68], and induce lymphocyte progression from G0/G1
to S phase (Figure 1, E), which might be related to restore the
function of immune system in advanced cancer patients [69].
Besides efects on T cell proliferation, diferentiation,
and the cytokines produced by them, ALA could also
inhibit migration of T cells. Ying and colleagues found that
ALA could directly reduce T cell migration in response
to chemokines to reduce T cell numbers in atherosclerotic
plaque in models of established atherosclerosis [70]. ALA
was also reported to reduce migration of T cell [58, 71],
lymphocyte and monocyte of models of MS [61], and Jurkat
T cells [72], which were associated with downregulated

4 BioMed Research International

ΔΨG

A
A

2
2
H

0

C

G

1 1 1 1

B B

D

E F

ATP
cAMP

CREBAP-1
CD25
IL-2

ALA
mPTP opening ROS
MHP

Nrf2 sMaf
ARE

mTOR

S6K1
4E-BP1

Ribosomal biosynthesis
mRNA biosynthesis
G1 to S phase traverse
CD4
IL-4

EP2
EP4
ALA

AC

VLA-4

ATP depletion

Necrosis
Spontaneous
apoptosis

GSH

AMPK AKT ERK

Inhibits
Activates
Indirectly activates
Indirectly inhibits
Unknown relationship

‘M
‘M

CD86

CD28
CD2

INF-

NF-B

IKK
TNFR

?

Figure 1: Effect of ALA on T cell. A ALA increase cAMP synthesis through activation of prostaglandin receptors (EP2 and EP4) in peripheral
blood T cells. B Te expression of IL-2 and IL-2R� (CD25) could be inhibited when the level of cAMP was increased by ALA. C ALA could

inhibit NF-�B activation induced by TNF in Jurkat T cells. D ALA could suppress production of interferon-� (IFN-�) and interleukin -4 (IL-
4). E ALA could induce lymphocyte progression from G0/G1 to S phase. F ALA was also reported to reduce migration of T cell, lymphocyte

and monocyte of models of MS, and Jurkat T cells, which were associated with down-regulated expression of very late activation-4 antigen
(VLA-4). G Uncontrolled mPTP opening leads to decrease Ψm irreversibly until dissipation which results in apoptosis and necrosis of
cells. H Persistent MHP in SLE T cells could enhance ROS production. 0 Studies showed that ALA and DHLA promoted mPTP opening
in mitochondria of rat liver. 1 mTORC1 was a central common regulator of a complex signaling network In cytoplasm, in which Ras/Erk,
PI3K/Akt, and IKK� activated mTORC1 while Dsh/GSK3 and LKBl/AMPK inactivated mTORC1. 2 Activated mTORC promoted protein
synthesis by phosphorylating the eukaryotic initiation factor 4E-binding protein 1 (4E-BP1) and p70 ribosomal S6 kinase 1 (S6K1).

expression of very late activation-4 antigen (VLA-4) (Figure 1,
F) and inhibition of MMP-9 activity by ALA [72].

2.1.2. Effects on B Cells. Te studies showed that ALA supple-
ment might play a role in high fat diet mice to prevent the

development of oxidative stress and to attenuate B-cell injury
by increasing the gene expression of the B-cell receptor (BCR)

signaling pathway and decrease the apoptotic percentage of
splenic B lymphocytes [73], which was also relevant to the
improvement of gene expression level of BCR [74]. It has been
proven that ALA increased the number of splenic B cells in
endotoxemia mice [75] and reduced total serum IgE levels of
atopic dermatitis mice model [68].Tese experiments suggest
that ALA plays a regulating role on proliferation, apoptosis
and function of B cells (Table 1).

BioMed Research International 5

Table 2: Evidence of ALA on innate immune cells.
NK cell Macrophage Monocyte

Animal
model

EAE Inhibit the phagocytosis of
myelin.

Decrease monocytes infltration
into the CNS.

High fat diet
mice

Suppress infltration and
activation of macrophage to
attenuate visceral adipose
infammation.

BMDM or
RAW 264.7

Decrease the production of
MCP-1 and TNF-� induced by
LPS.

Normal
human

Increase cAMP
production to
suppress cytotoxicity
and IFN-�
production.

Induce the expression of HO-1 by

Nrf2.

ALA: �-lipoic acid.
NK cell: natural killer cell.
EAE: experimental autoimmune encephalomyelitis.
CNS: central nervous system.
BMDM: bone marrow-derived macrophages.
MCP-1: monocyte chemotactic protein 1.
TNF-�: tumor necrosis factor-alpha.
LPS: lipopolysaccharide.
cAMP: cyclic adenosine monophosphate.
IFN-�: interferon-�.
HO-1: heme oxygenase-1.
Nrf2: nuclear factor-erythroid 2-related factor.

2.1.3. Effects on Innate Immune Cells
Natural Killer Cell (NK Cell), Macrophage, and Monocyte
(See Table 2). Cytotoxicity and cytokines secretion are two
main functions of NK cells. Te former is associated with
the release of granzymes (perforin and proteases) from their
cytoplasm. INF-� secretion is a representative of the latter.

INF-� is a potent macrophage activator for both phagocy-
tosis and lysis. IFN-� secretion induced by IL-12/IL-18 and

cellular cytotoxicity in NK cells could be inhibited by ALA,
which increased cAMP production via G protein-coupled
receptors- (GPCRs-) dependent and GPCRs-independent

mechanisms [62, 76, 77]. In addition, it has also been demon-
strated that both cAMP and cAMP-inducing agents (PGE1,

theophylline, and histamine) suppressed cytolytic function
of NK cells [78]. PGE2, another cAMP elevating agent, also

suppressed cytotoxicity and IFN-� production induced by IL-
15 [79]. Terefore, ALA could suppress NK function in a few

ways.
It has also been found that ALA regulated activation,
phagocytosis, and migration of macrophage by either direct
or indirect means. ALA inhibited the phagocytosis of myelin
by macrophages [80], which was the main autoantigen in EAE
mice, and decreased the production of monocyte chemotactic

protein 1 (MCP-1) and TNF� induced by lipopolysaccha-
ride (LPS) of macrophages [81, 82]. Also, ALA decreased

monocytes infltration into the CNS and stabilized brain
endothelial cells in EAE rat [61], which might be associated
with the downregulated intracellular adhesion molecule-1
(ICAM-1) expression of monocytes [83] and the upregulated

vascular cell adhesion molecule-1 (VCAM-1) expression of
endothelial cells [84]. In addition, ALA could induce the

expression of heme oxygenase-1(HO-1) by nuclear factor-
erythroid 2-related factor 2 (Nrf2) in human monocytic cells

[85].
3. Other Potential Targets of ALA
Immunomodulatory Effects
ALA has been widely used for decades in clinic and studied
in various experimental models. Terefore, we found the
potential targets for immunomodulatory efects of ALA in
these researches.

3.1. Mitochondrial Membrane Potential (Ψm). Mitochon-
dria provide place for the citric acid cycle and oxidative

phosphorylation, which is the energy station of cells and
is involved in cell diferentiation, cell cycle regulation, and

cell death. Te stability of Ψm is essential for the mainte-
nance of normal physiological function of cells. Te electron

transport chain and the F0F1-ATPase complex maintain an
electrochemical gradient namely “Ψm”, and vice versa,
Ψm tightly regulates the production of ROS and ATP
synthesis [86]. Mitochondrial permeability transition pore
(mPTP) is a series of protein channels which are located
in the inner and outer mitochondrial membrane. mPTP
closes completely to stabilize Ψm whereas mPTP opens
transiently to a low conductance state to result in lowering
Ψm. Uncontrollable mPTP opening leads to decrease Ψm

6 BioMed Research International
irreversibly until dissipation which results in apoptosis and
necrosis of cells [87, 88] (Figure 1, G). In the process of
both activation and apoptosis of T lymphocytes, Ψm was
transiently reversibly elevated, that is what mitochondrial
hyperpolarization (MHP) in physiological status should be
[89]. However, persistent MHP in T cells of SLE patients
would enhance ROS production (Figure 1 H), which resulted
in activation of macrophages [90] and dendritic cells [91]
to exacerbate infammation [92, 93] and resulted in ATP
depletion which increased IL-10 production and spontaneous
apoptosis of T cells [94]. T cell apoptosis not only provided
a source of nuclear antigens but also was correlated with

SLE disease activity [95, 96]. Increased production of IL-
10 could promote T cell apoptosis [97] and contribute to

the production of autoantibodies by hyperactive SLE B
lymphocytes [98, 99]. Tere were proofs showing that ALA
and DHLA promoted mPTP opening of mitochondria in rat
liver [3, 93, 100] (Figure 1, 0). Tus, the authors speculate
that ALA may attenuate mitochondrial dysfunction in SLE
from several aspects. ALA opens mPTP to reduce ΔΨm,
which improves pathological MHP of SLE T cells and directly
quenches ROS to correct dysfunction of T cells and B
cells.
3.2. Neutrophil Extracellular Traps (NETs). Neutrophils play
a very important role in innate immune system and are the
frst leukocytes to be recruited to the site of infection to
eliminate pathogens by multiple mechanisms which include
phagocytosis, infammatory mediators secretion, and NETs
release which is also known as NETosis. NETs are composed
of nucleic acids, histone proteins, and granule proteins with
or without death of neutrophils [101]. NETs only fght against
pathogens but also have been implicated in pathogenesis of
the autoimmune diseases (e.g., SLE [102, 103], RA [104, 105],
psoriasis [106], and autoimmune small-vessel vasculitis [107])
and thrombosis [108]. Nuclear material of NETs components
became autoantigens afer it was extruded from the cell
to induce autoantibodies production [109]. NETs formation
was dependent on autophagy and ROS generation [110, 111]
and regulated by mTOR signaling pathway [112, 113]. It

has been speculated that ALA may be capable of quench-
ing ROS and regulating mTOR signaling, which suggests

that it may have benefcial efects on NETs formation to
reduce the autoantibodies production and protect vascular
endothelium.
3.3. Nrf2 Signaling Pathway. ALA is well recognized to
be an activator of Nrf2 signaling [1, 114]. Te nuclear
factor-erythroid 2-related factor 2 (Nrf2), a central regulator
of cellular resistance to oxidant stress, binds antioxidant
response element (ARE) to regulate expression of a lot
of ARE-containing genes to play a pivotal role in control
of oxidant homeostasis [115]. Tere is a little evidence to
show association between Nrf2 signaling and pathogenesis
of autoimmune disease. It has been demonstrated that Nrf2-
defcient female mice developed severe nephritis similar to
lupus [116] and Nrf2 gene variant was relevant with nephritis

in childhood-onset SLE patients [117]. Nrf2 (-/-) mice devel-
oped regenerative immune-mediated hemolytic anemia [118]

and disruption of Nrf2 aggravated [119] while activation of
Nrf2 attenuated [120] neuroinfammatory disorders in EAE.
Hence, ALA may have regulatory efects in immune system
via Nrf2 pathway.
4. Safety of ALA
ALA, a naturally occurring antioxidant in human body and
available from common dietary sources, has been used to
treat diabetic neuropathy and retinopathy for over 50 years
in Germany. A number of clinical trials have reported that
oral LA supplementation up to 2400 mg/d and intravenous
LA supplementation up to 600 mg/d for three weeks showing
no adverse efects versus placebo [1, 2]. Moreover, fexible
regulatory efects of ALA have been shown by Sen and
colleagues that ALA promoted apoptosis induced by Fas

in Jurkat cells but not healthy peripheral blood lympho-
cytes [121]. Te data of these studies supports the safety of

ALA.
5. Summary
In conclusion, ALA, a natural ingredient of human body,
not only acts as a powerful antioxidant but also is able to
regulate the immune system in either direct or indirect ways.
Studies reviewed above might suggest that ALA is used to
treat autoimmune diseases including SLE, RA, and primary
vasculitis as well as MS. Te current therapies for systemic
rheumatic diseases are efective. However, there was still a
high percent of patients with not enough or no response to
the therapies. Terefore, if the immunomodulatory efects
of ALA could be confrmed by further investigation, it
might have benefcial efects in conjunction with the current
treatment of rheumatic diseases.
Conflicts of Interest
Te authors declare that they have no conficts of interest.
Authors’ Contributions
Wei Liu and Lian-jie Shi contributed equally to the paper.
Acknowledgments
Te authors acknowledge Guo-xiang Lai, M.D., from the
900th Hospital of the Joint Logistic Support Force, PLA,
for his contribution to the fgure drawing and medical
writing assistance while drafing this manuscript.Tis work is
supported by grants from Fuzhou General Hospital (2018 J05)
and Clinical Key Specialty Construction Project in Fujian
Province (Min Wei Medical Letter [2015] no. 593).

BioMed Research International 7
References
[1] K. P. Shay, R. F. Moreau, E. J. Smith, A. R. Smith, and T. M.
Hagen, “Alpha-lipoic acid as a dietary supplement: molecular

mechanisms and therapeutic potential,” Biochimica et Biophys-
ica Acta, vol. 1790, no. 10, pp. 1149–1160, 2009.

[2] A. Gora ̧ca, H. Huk-Kolega, A. Piechota, P. Kleniewska, E.
Ciejka, and B. Skibska, “Lipoic acid—biological activity and
therapeutic potential,” Pharmacological Reports, vol. 63, no. 4,
pp. 849–858, 2011.
[3] L. Rochette, S. Ghibu, C. Richard, M. Zeller, Y. Cottin, and C.
Vergely, “Direct and indirect antioxidant properties of �-lipoic
acid and therapeutic potential,” Molecular Nutrition & Food
Research, vol. 57, no. 1, pp. 114–125, 2013.
[4] C. Nathan and A. Cunningham-Bussel, “Beyond oxidative
stress: an immunologist’s guide to reactive oxygen species,”
Nature Reviews Immunology, vol. 13, no. 5, pp. 349–361, 2013.
[5] A. Perl, “Oxidative stress in the pathology and treatment of
systemic lupus erythematosus,” Nature Reviews Rheumatology,
vol. 9, no. 11, pp. 674–686, 2013.
[6] Y. L. Lightfoot, L. P. Blanco, and M. J. Kaplan, “Metabolic
abnormalities and oxidative stress in lupus,” Current Opinion in
Rheumatology, vol. 29, no. 5, pp. 442–449, 2017.
[7] X. Huang, J. Li, S. Dorta-Estremera et al., “Neutrophils regulate
humoral autoimmunity by restricting interferon-� production
via the generation of reactive oxygen species,” Cell Reports, vol.
12, no. 7, pp. 1120–1132, 2015.
[8] N. Huang and A. Perl, “Metabolism as a target for modulation
in autoimmune diseases,” Trends in Immunology, vol. 39, no. 7,
pp. 562–576, 2018.

[9] M. Torigoe, S. Iwata, S. Nakayamada et al., “Metabolic repro-
gramming commits diferentiation of human CD27(+)IgD(+)

B cells to plasmablasts or CD27(-)IgD(-) cells,” e Journal of
Immunology, vol. 199, no. 2, pp. 425–434, 2017.
[10] W. Yi, S. Gupta, E. Ricker et al., “Te mTORC1-4E-BP-eIF4E
axis controls de novo Bcl6 protein synthesis in T cells and
systemic autoimmunity,” Nature Communications, vol. 8, no. 1,
2017.
[11] G. M. Delgofe, K. N. Pollizzi, A. T. Waickman et al., “Te kinase
mTOR regulates the diferentiation of helper T cells through
the selective activation of signaling by mTORC1 and mTORC2,”
Nature Immunology, vol. 12, no. 4, pp. 295–304, 2011.
[12] J. Wei, L. Long, K. Yang et al., “Autophagy enforces functional
integrity of regulatory T cells by coupling environmental cues
and metabolic homeostasis,” Nature Immunology, vol. 17, no. 3,
pp. 277–285, 2016.
[13] X. Xu, K. Araki, S. Li et al., “Autophagy is essential for
efector CD8(+) T cell survival and memory formation,” Nature
Immunology, vol. 15, no. 12, pp. 1152–1161, 2014.
[14] C.-K. Lee, E. Y. Lee, Y. G. Kim, S. H. Mun, H.-B. Moon,
and B. Yoo, “Alpha-lipoic acid inhibits TNF-� induced NF-�B
activation through blocking of MEKK1-MKK4-IKK signaling
cascades,” International Immunopharmacology, vol. 8, no. 2, pp.
362–370, 2008.
[15] Z. Ying, T. Kampfrath, Q. Sun, S. Parthasarathy, and S.

Rajagopalan, “Evidence that �-lipoic acid inhibits NF-�B acti-
vation independent of its antioxidant function,” Inflammation

Research, vol. 60, no. 3, pp. 219–225, 2011.
[16] D. F. Lee, H. P. Kuo, C. T. Chen et al., “IKKbeta suppression of
TSC1 function links the mTOR pathway with insulin resistance,”
International Journal of Molecular Medicine, vol. 22, no. 5, pp.
633–638, 2008.

[17] D. F. Lee, H. P. Kuo, C. T. Chen et al., “IKK beta suppression of
TSC1 links infammation and tumor angiogenesis via the mTOR
pathway,” Cell, vol. 130, no. 3, pp. 440–455, 2007.
[18] W.-R. Lee, A. Kim, K.-S. Kim et al., “Alpha-lipoic acid attenuates
atherosclerotic lesions and inhibits proliferation of vascular
smooth muscle cells through targeting of the Ras/MEK/ERK
signaling pathway,” Molecular Biology Reports, vol. 39, no. 6, pp.
6857–6866, 2012.
[19] J. S. Grewal, Y. V. Mukhin, M. N. Garnovskaya, J. R. Raymond,
and E. L. Greene, “Serotonin 5-HT(2A) receptor induces TGF-
�1 expression in mesangial cells via ERK: Proliferative and

fbrotic signals,” American Journal of Physiology-Renal Physiol-
ogy, vol. 276, no. 6, pp. F922–F930, 1999.

[20] M. N. Budisavljevic, L. Hodge, K. Barber et al., “Oxidative

stress in the pathogenesis of experimental mesangial prolifera-
tive glomerulonephritis,” American Journal of Physiology-Renal

Physiology, vol. 285, no. 6, pp. F1138–F1148, 2003.
[21] N.-P. Foo, S.-H. Lin, Y.-H. Lee, M.-J. Wu, and Y.-J. Wang, “�-
Lipoic acid inhibits liver fbrosis through the attenuation of
ROS-triggered signaling in hepatic stellate cells activated by
PDGF and TGF-�,” Toxicology, vol. 282, no. 1-2, pp. 39–46, 2011.
[22] P.-Y. Cheng, Y.-M. Lee, N.-L. Shih, Y.-C. Chen, and M.-H. Yen,

“Heme oxygenase-1 contributes to the cytoprotection of alpha-
lipoic acid via activation of p44/42 mitogen-activated protein

kinase in vascular smooth muscle cells,” Free Radical Biology &
Medicine, vol. 40, no. 8, pp. 1313–1322, 2006.
[23] Y. Yao, R. Li, Y. Ma et al., “�-Lipoic acid increases tolerance of
cardiomyoblasts to glucose/glucose oxidase-induced injury via
ROS-dependent ERK1/2 activation,” Biochimica et Biophysica
Acta (BBA) – Molecular Cell Research, vol. 1823, no. 4, pp. 920–
929, 2012.
[24] K. Cho, H. Moon, H. Moini, L. Packer, D. Yoon, and A.
Chung, “�-Lipoic acid inhibits adipocyte diferentiation by

regulating pro-adipogenic transcription factors via mitogen-
activated protein kinase pathways,” e Journal of Biological

Chemistry, vol. 278, no. 37, pp. 34823–34833, 2003.
[25] H. Mohmmad Abdul and D. A. Butterfeld, “Involvement of
PI3K/PKG/ERK1/2 signaling pathways in cortical neurons to
trigger protection by cotreatment of acetyl-L-carnitine and �-

lipoic acid against HNE-mediated oxidative stress and neu-
rotoxicity: Implications for Alzheimer’s disease,” Free Radical

Biology & Medicine, vol. 42, no. 3, pp. 371–384, 2007.
[26] X. Wang, Z. Wang, Y. Yao et al., “Essential role of ERK activation
in neurite outgrowth induced by alpha-lipoic acid,” Biochimica
et Biophysica Acta, vol. 1813, no. 5, pp. 827–838, 2011.
[27] S. S. Shi, R. M. Day, A. D. Halpner, J. B. Blumberg, and Y.
J. Suzuki, “Homocysteine and �-lipoic acid regulate p44/42
MAP kinase phosphorylation in NIH/3T3 cells,” Antioxidants
& Redox Signaling, vol. 1, no. 1, pp. 123–128, 1999.
[28] Y. J. Suzuki, S. S. Shi, R. M. Day, and J. B. Blumberg, “Diferential
regulation of MAP kinase signaling by pro- and antioxidant
biothiols,” Annals of the New York Academy of Sciences, vol. 899,
pp. 159–167, 2000.

[29] B. W. Lee, S. J. Kwon, H. Y. Chae et al., “Dose-related cytopro-
tective efect of alpha-lipoic acid on hydrogen peroxide-induced

oxidative stress to pancreatic beta cells,” Free Radical Research,
vol. 43, no. 1, pp. 68–77, 2009.

[30] M. Ishiki, Y. Nishida, H. Ishibashi et al., “Impact of diver-
gent efects of astaxanthin on insulin signaling in L6 cells,”

Endocrinology, vol. 154, no. 8, pp. 2600–2612, 2013.

[31] E. Dozio, M. Ruscica, L. Passafaro et al., “Te natural antiox-
idant alpha-lipoic acid induces p27Kip1-dependent cell cycle

8 BioMed Research International
arrest and apoptosis in MCF-7 human breast cancer cells,”
European Journal of Pharmacology, vol. 641, no. 1, pp. 29–34,
2010.
[32] D.-Y. Shi, H.-L. Liu, J. S. Stern, P.-Z. Yu, and S.-L. Liu,
“Alpha-lipoic acid induces apoptosis in hepatoma cells via the
PTEN/Akt pathway,” FEBS Letters, vol. 582, no. 12, pp. 1667–
1671, 2008.
[33] P. L. Prieto-Hontoria, P. Perez-Matute, M. Fern ́ andez-Galilea, ́
J. A. Mart ́ınez, and M. J. Moreno-Aliaga, “Lipoic acid inhibits
leptin secretion and Sp1 activity in adipocytes,” Molecular
Nutrition & Food Research, vol. 55, no. 7, pp. 1059–1069, 2011.
[34] M. S. Bitar, S. Wahid, C. W. Pilcher, E. Al-Saleh, and F. Al-Mulla,
“�-lipoic Acid Mitigates Insulin Resistance in Goto-Kakizaki
Rats,” Hormone and Metabolic Research, vol. 36, no. 8, pp. 542–
549, 2004.
[35] S. J. Lee, S. H. Kim, J. G. Kang et al., “Alpha-lipoic acid inhibits
endoplasmic reticulum stress-induced cell death through
PI3K/Akt signaling pathway in FRTL5 thyroid cells,” Hormone
and Metabolic Research, vol. 43, no. 7, pp. 445–451, 2011.

[36] X. Wang, X. Zhang, Y. Cheng et al., “�-Lipoic acid pre-
vents bupivacaine-induced neuron injury in vitro through a

PI3K/Akt-dependent mechanism,” NeuroToxicology, vol. 31, no.
1, pp. 101–112, 2010.
[37] L. Zhang, G. q. Xing, J. L. Barker et al., “�-lipoic acid protects
rat cortical neurons against cell death induced by amyloid
and hydrogen peroxide through the Akt signalling pathway,”
Neuroscience Letters, vol. 312, no. 3, pp. 125–128, 2001.
[38] C. Deng, Z. Sun, G. Tong et al., “Alpha-lipoic acid reduces
infarct size and preserves cardiac function in rat myocardial
ischemia/reperfusion injury through activation of pi3k/akt/nrf2
pathway,” PLoS ONE, vol. 8, no. 3, Article ID e58371, 2013.
[39] C. Muller, F. D ̈ unschede, E. Koch, A. M. Vollmar, and A. ̈

K. Kiemer, “�-Lipoic acid preconditioning reduces ischemia-
reperfusion injury of the rat liver via the PI3-kinase/Akt

pathway,” American Journal of Physiology-Gastrointestinal and
Liver Physiology, vol. 285, no. 4, pp. G769–G778, 2003.

[40] S. Jiang, W. Zhu, C. Li et al., “�-Lipoic acid attenuates LPS-
induced cardiac dysfunction through a PI3K/Akt-dependent

mechanism,” International Immunopharmacology, vol. 16, no. 1,
pp. 100–107, 2013.
[41] W. J. Zhang, H. Wei, T. Hagen, and B. Frei, “�-Lipoic acid
attenuates LPS-induced infammatory responses by activating

the phosphoinositide 3-kinase/Akt signaling pathway,” Proceed-
ings of the National Acadamy of Sciences of the United States of

America, vol. 104, no. 10, pp. 4077–4082, 2007.
[42] M. S. Bitar, A. K. Ayed, S. M. Abdel-Halim, E. R. Isenovic, and F.
Al-Mulla, “Infammation and apoptosis in aortic tissues of aged

type II diabetes: amelioration with �-lipoic acid through phos-
phatidylinositol 3-kinase/Akt- dependent mechanism,” Life Sci-
ences, vol. 86, no. 23-24, pp. 844–853, 2010.

[43] A. R. Smith and T. M. Hagen, “Vascular endothelial dysfunction

in aging: Loss of Akt-dependent endothelial nitric oxide syn-
thase phosphorylation and partial restoration by (R)-�-lipoic

acid,” Biochemical Society Transactions, vol. 31, no. 6, pp. 1447–
1449, 2003.
[44] W. Chen, C. Kang, S. Wang et al., “Alpha-Lipoic acid regulates
lipid metabolism through induction of sirtuin 1 (SIRT1) and
activation of AMP-activated protein kinase,” Diabetologia, vol.
55, no. 6, pp. 1824–1835, 2012.
[45] A. K. Saha, X. J. Xu, E. Lawson et al., “Downregulation of AMPK
accompanies leucine- and glucose-induced increases in protein

synthesis and insulin resistance in rat skeletal muscle,” Diabetes,
vol. 59, no. 10, pp. 2426–2434, 2010.
[46] A. K. Saha, X. J. Xu, T. W. Balon, A. Brandon, E. W. Kraegen,
and N. B. Ruderman, “Insulin resistance due to nutrient excess:
is it a consequence of AMPK downregulation?” Cell Cycle, vol.
10, no. 20, pp. 3447–3451, 2011.
[47] W. J. Lee, K. H. Song, and E. H. Koh, “Alpha-lipoic acid increases
insulin sensitivity by activating AMPK in skeletal muscle,”
Biochemical and Biophysical Research Communications, vol. 332,
no. 3, pp. 885–891, 2005.
[48] E. D. Targonsky, F. Dai, V. Koshkin et al., “alpha-lipoic acid
regulates AMP-activated protein kinase and inhibits insulin
secretion from beta cells,” Diabetologia, vol. 49, no. 7, pp. 1587–
1598, 2006.
[49] M.-S. Kim, J.-Y. Park, C. Namkoong et al., “Anti-obesity efects

of �-lipoic acid mediated by suppression of hypothalamic AMP-
activated protein kinase,” Nature Medicine, vol. 10, no. 7, pp. 727–

733, 2004.
[50] W. J. Lee, H. K. Eun, C. W. Jong, M.-S. Kim, J.-Y. Park, and K.-U.
Lee, “Obesity: Te role of hypothalamic AMP-activated protein
kinase in body weight regulation,” e International Journal of
Biochemistry & Cell Biology, vol. 37, no. 11, pp. 2254–2259, 2005.
[51] M. S. Kim and K. U. Lee, “Role of hypothalamic 5�

-AMP-
activated protein kinase in the regulation of food intake and

energy homeostasis,” Journal of Molecular Medicine, vol. 83, no.
7, pp. 514–520, 2005.
[52] D. Fernandez and A. Perl, “mTOR signaling: a central pathway
to pathogenesis in systemic lupus erythematosus?” Discovery
Medicine, vol. 9, no. 46, pp. 173–178, 2010.
[53] D. Fernandez and A. Perl, “Metabolic control of T cell activation
and death in SLE,” Autoimmunity Reviews, vol. 8, no. 3, pp. 184–
189, 2009.
[54] A. O’Loghlen, M. I. Perez-Morgado, M. Salinas, and M. ́

E. Mart ́ın, “N-acetyl-cysteine abolishes hydrogen peroxide-
induced modifcation of eukaryotic initiation factor 4F activity

via distinct signalling pathways,” Cellular Signalling, vol. 18, no.
1, pp. 21–31, 2006.
[55] S. Suwannaroj, A. Lagoo, D. Keisler, and R. W. McMurray,
“Antioxidants suppress mortality in the female NZB × NZW F1
mouse model of systemic lupus erythematosus (SLE),” Lupus,
vol. 10, no. 4, pp. 258–265, 2001.
[56] Z.-W. Lai, R. Hanczko, E. Bonilla et al., “N-acetylcysteine
reduces disease activity by blocking mammalian target of
rapamycin in T cells from systemic lupus erythematosus
patients: a randomized, double-blind, placebo-controlled trial,”
Arthritis & Rheumatism, vol. 64, no. 9, pp. 2937–2946, 2012.
[57] Z.-W. Lai, R. Kelly, T. Winans et al., “Sirolimus in patients with
clinically active systemic lupus erythematosus resistant to, or

intolerant of, conventional medications: a single-arm, open-
label, phase 1/2 trial,” e Lancet, vol. 391, no. 10126, pp. 1186–

1196, 2018.
[58] G. H. Marracci, R. E. Jones, G. P. McKeon, and D. N. Bourdette,
“Alpha lipoic acid inhibits T cell migration into the spinal
cord and suppresses and treats experimental autoimmune
encephalomyelitis,” Journal of Neuroimmunology, vol. 131, no. 1-
2, pp. 104–114, 2002.
[59] M. Morini, L. Roccatagliata, R. Dell’Eva et al., “Alpha-lipoic
acid is efective in prevention and treatment of experimental
autoimmune encephalomyelitis,” Journal of Neuroimmunology,
vol. 148, no. 1-2, pp. 146–153, 2004.
[60] K.-C. Wang, C.-P. Tsai, C.-L. Lee et al., “�-lipoic acid enhances
endogenous peroxisome-proliferator-activated receptor-� to

BioMed Research International 9
ameliorate experimental autoimmune encephalomyelitis in
mice,” Clinical Science, vol. 125, no. 7, pp. 329–340, 2013.
[61] G. Schreibelt, R. J. P. Musters, A. Reijerkerk et al., “Lipoic
acid afects cellular migration into the central nervous system
and stabilizes blood-brain barrier integrity,” e Journal of
Immunology, vol. 177, no. 4, pp. 2630–2637, 2006.
[62] R. V. Schillace, N. Pisenti, N. Pattamanuch et al., “Lipoic acid
stimulates cAMP production in T lymphocytes and NK cells,”
Biochemical and Biophysical Research Communications, vol. 354,
no. 1, pp. 259–264, 2007.

[63] K. A. Anderson, T. J. Ribar, M. Illario, and A. R. Means, “Defec-
tive survival and activation of thymocytes in transgenic mice

expressing a catalytically inactive form of Ca2+/calmodulin-
dependent protein kinase IV,” Molecular Endocrinology, vol. 11,

no. 6, pp. 725–737, 1997.
[64] E. M. Kuklina and S. V. Shirshev, “Role of cAMP-dependent
signal transduction in the control of T lymphocyte activation,”
Biochemistry (Moscow), vol. 65, no. 6, pp. 629–639, 2000.

[65] L. Milazzo, B. Menzaghi, I. Caramma et al., “Efect of antioxi-
dants on mitochondrial function in HIV-1-related lipoatrophy:

A pilot study,” AIDS Research and Human Retroviruses, vol. 26,
no. 11, pp. 1207–1214, 2010.
[66] G. H. Marracci, W. E. Marquardt, A. Strehlow et al., “Lipoic
acid downmodulates CD4 from human T lymphocytes by
dissociation of p56Lck,” Biochemical and Biophysical Research
Communications, vol. 344, no. 3, pp. 963–971, 2006.
[67] Y. J. Suzuki, B. Aggarwal, and L. Packer, “Alpha-lipoic acid is
a potent inhibitor of NF-kappa B activation in human T cells,”
Biochemical and Biophysical Research Communications, vol. 189,
no. 3, pp. 1709–1715, 1992.
[68] G. Kim, T. Kim, A. Jang, H. Ahn, Y. S. Park, and C. Park,
“�-Lipoic acid suppresses the development of DNFB-induced

atopic dermatitis-like symptoms in NC/Nga mice,” Experimen-
tal Dermatology, vol. 20, no. 2, pp. 97–101, 2011.

[69] G. Mantovani, A. Maccio, C. Madeddu et al., “Antioxi- `
dant agents are efective in inducing lymphocyte progression
through cell cycle in advanced cancer patients: assessment of the
most important laboratory indexes of cachexia and oxidative
stress,” Journal of Molecular Medicine, vol. 81, no. 10, pp. 664–
673, 2003.
[70] Z. Ying, N. Kherada, B. Farrar et al., “Lipoic acid efects on
established atherosclerosis,” Life Sciences, vol. 86, no. 3-4, pp.
95–102, 2010.
[71] P. Chaudhary, G. H. Marracci, and D. N. Bourdette, “Lipoic acid
inhibits expression of ICAM-1 and VCAM-1 by CNS endothelial
cells and T cell migration into the spinal cord in experimental
autoimmune encephalomyelitis,” Journal of Neuroimmunology,
vol. 175, no. 1-2, pp. 87–96, 2006.
[72] G. H. Marracci, G. P. McKeon, W. E. Marquardt, R. W. Winter,
M. K. Riscoe, and D. N. Bourdette, “� lipoic acid inhibits human
T-cell migration: Implications for multiple sclerosis,” Journal of
Neuroscience Research, vol. 78, no. 3, pp. 362–370, 2004.
[73] J. Cui, Y. Xiao, Y. Shi, G. Le, and X. Miao, “Comparative

proteome analysis of splenic lymphocytes in long-term high-
fat diet and dietary supplement with lipoic acid mice,” Cellular

Immunology, vol. 264, no. 2, pp. 156–162, 2010.
[74] J. Cui, Y. Xiao, Y.-H. Shi, B. Wang, and G.-W. Le, “Lipoic acid

attenuates high-fat-diet-induced oxidative stress and B-cell-
related immune depression,” Nutrition Journal , vol. 28, no. 3,

pp. 275–280, 2012.
[75] B. Wessner, E.-M. Strasser, N. Manhart, and E. Roth, “Supply of
R-�-lipoic acid and glutamine to casein-fed mice infuences the

number of B lymphocytes and tissue glutathione levels during
endotoxemia,” Wiener Klinische Wochenschri

, vol. 118, no. 3-4,

pp. 100–107, 2006.
[76] S. Salinthone, R. V. Schillace, G. H. Marracci, D. N. Bourdette,
and D. W. Carr, “Lipoic acid stimulates cAMP production
via the EP2 and EP4 prostanoid receptors and inhibits IFN
gamma synthesis and cellular cytotoxicity in NK cells,” Journal
of Neuroimmunology, vol. 199, no. 1-2, pp. 46–55, 2008.

[77] S. Salinthone, R. V. Schillace, C. Tsang, J. W. Regan, D. N. Bour-
dette, and D. W. Carr, “Lipoic acid stimulates cAMP production

via G protein-coupled receptor-dependent and -independent
mechanisms,” e Journal of Nutritional Biochemistry, vol. 22,
no. 7, pp. 681–690, 2011.
[78] J. C. Roder and M. Klein, “Target-efector interaction in the
natural killer cell system. IV. Modulation by cyclic nucleotides,”
e Journal of Immunology, vol. 123, no. 6, pp. 2785–2790, 1979.
[79] P. C. Joshi, X. Zhou, M. Cuchens, and Q. Jones, “Prostaglandin
E2 suppressed IL-15-mediated human NK cell function through

down-regulation of common �-chain,” e Journal of Immunol-
ogy, vol. 166, no. 2, pp. 885–891, 2001.

[80] A. Van Der Goes, J. Brouwer, K. Hoekstra, D. Roos, T. K.
Van Den Berg, and C. D. Dijkstra, “Reactive oxygen species
are required for the phagocytosis of myelin by macrophages,”
Journal of Neuroimmunology, vol. 92, no. 1-2, pp. 67–75, 1998.
[81] J. A. Deiuliis, T. Kampfrath, Z. Ying, A. Maiseyeu, and S.
Rajagopalan, “Lipoic acid attenuates innate immune infltration
and activation in the visceral adipose tissue of obese insulin
resistant mice,” Lipids, vol. 46, no. 11, pp. 1021–1032, 2011.
[82] A. K. Kiemer, C. Muller, and A. M. Vollmar, “Inhibition of LPS- ̈
induced nitric oxide and TNF-� production by �-lipoic acid
in rat Kupfer cells and in RAW 264.7 murine macrophages,”
Immunology & Cell Biology, vol. 80, no. 6, pp. 550–557, 2002.
[83] H. A. Lee and D. A. Hughes, “Alpha-lipoic acid modulates NF-
�B activity in human monocytic cells by direct interaction with
DNA,” Experimental Gerontology, vol. 37, no. 2-3, pp. 401–410,
2002.
[84] T. Kunt, T. Forst, A. Wilhelm et al., “�-Lipoic acid reduces
expression of vascular cell adhesion molecule-1 and endothelial
adhesion of human monocytes afer stimulation with advanced
glycation end products,” Clinical Science, vol. 96, no. 1, pp. 75–
82, 1999.
[85] R. M. Ogborne, S. A. Rushworth, and M. A. O’Connell, “�-
lipoic acid-induced heme oxygenase-1 expression is mediated

by nuclear factor erythroid 2-related factor 2 and p38 mitogen-
activated protein kinase in human monocytic cells,” Arterioscle-
rosis, rombosis, and Vascular Biology, vol. 25, no. 10, pp. 2100–

2105, 2005.
[86] A. Perl, P. Gergely Jr., G. Nagy, A. Koncz, and K. Banki,
“Mitochondrial hyperpolarization: A checkpoint of T-cell life,
death and autoimmunity,” Trends in Immunology, vol. 25, no. 7,
pp. 360–367, 2004.

[87] A. P. Halestrap, “What is the mitochondrial permeability tran-
sition pore?” Journal of Molecular and Cellular Cardiology, vol.

46, no. 6, pp. 821–831, 2009.

[88] D. Siemen and M. Ziemer, “What is the nature of the mitochon-
drial permeability transition pore and What is it Not?” IUBMB

Life, vol. 65, no. 3, pp. 255–262, 2013.
[89] G. Nagy, A. Koncz, D. Fernandez, and A. Perl, “Nitric oxide,
mitochondrial hyperpolarization, and T cell activation,” Free
Radical Biology & Medicine, vol. 42, no. 11, pp. 1625–1631, 2007.

10 BioMed Research International
[90] J. J. Cohen, R. C. Duke, V. A. Fadok, and K. S. Sellins, “Apoptosis
and programmed cell death in immunity,” Annual Review of
Immunology, vol. 10, pp. 267–293, 1992.
[91] B. Sauter, M. L. Albert, L. Francisco, M. Larsson, S. Somersan,
and N. Bhardwaj, “Consequences of cell death: exposure to
necrotic tumor cells, but not primary tissue cells or apoptotic
cells, induces the maturation of immunostimulatory dendritic
cells,” e Journal of Experimental Medicine, vol. 191, no. 3, pp.
423–433, 2000.
[92] P. Gergely Jr., C. Grossman, B. Niland et al., “Mitochondrial
hyperpolarization and ATP depletion in patients with systemic
lupus erythematosus,”Arthritis & Rheumatism, vol. 46, no. 1, pp.
175–190, 2002.
[93] N. Saris, A. Karjalainen, V. Teplova, and K. Lindros, “Te
stimulation of the mitochondrial permeability transition by

dihydrolipoate and �-lipoate,” Biochemistry and Molecular Biol-
ogy International, vol. 44, no. 1, pp. 127–134, 1998.

[94] P. Gergely Jr., B. Niland, N. Gonchorof, R. Pullmann Jr., P. E.

Phillips, and A. Perl, “Persistent mitochondrial hyperpolariza-
tion, increased reactive oxygen intermediate production, and

cytoplasmic alkalinization characterize altered IL-10 signaling
in patients with systemic lupus erythematosus,” e Journal of
Immunology, vol. 169, no. 2, pp. 1092–1101, 2002.
[95] W. Emlen, J. Niebur, and R. Kadera, “Accelerated in vitro
apoptosis of lymphocytes from patients with systemic lupus
erythematosus,” e Journal of Immunology, vol. 152, no. 7, pp.
3685–3692, 1994.
[96] L. A. Casciola-Rosen, G. Anhalt, and A. Rosen, “Autoantigens
targeted in systemic lupus erythematosus are clustered in two
populations of surface structures on apoptotic keratinocytes,”
e Journal of Experimental Medicine, vol. 179, no. 4, pp. 1317–
1330, 1994.
[97] L. Georgescu, R. K. Vakkalanka, K. B. Elkon, and M. K. Crow,
“Interleukin-10 promotes activation-induced cell death of SLE
lymphocytes mediated by Fas ligand,” e Journal of Clinical
Investigation, vol. 100, no. 10, pp. 2622–2633, 1997.
[98] L. Llorente, W. Zou, Y. Levy et al., “Role of interleukin 10 in
the B lymphocyte hyperactivity and autoantibody production

of human systemic lupus erythematosus,” e Journal of Exper-
imental Medicine, vol. 181, no. 3, pp. 839–844, 1995.

[99] H. Peng, W. Wang, M. Zhou, R. Li, H.-F. Pan, and D.-Q. Ye,
“Role of interleukin-10 and interleukin-10 receptor in systemic
lupus erythematosus,” Clinical Rheumatology, vol. 32, no. 9, pp.
1255–1266, 2013.
[100] H. Moini, L. Packer, and N.-E. L. Saris, “Antioxidant and
prooxidant activities of �-lipoic acid and dihydrolipoic acid,”
Toxicology and Applied Pharmacology, vol. 182, no. 1, pp. 84–90,
2002.
[101] B. G. Yipp and P. Kubes, “NETosis: how vital is it?” Blood, vol.
122, no. 16, pp. 2784–2794, 2013.
[102] E. Villanueva, S. Yalavarthi, C. C. Berthier et al., “Netting
neutrophils induce endothelial damage, infltrate tissues, and

expose immunostimulatory molecules in systemic lupus ery-
thematosus,” e Journal of Immunology, vol. 187, no. 1, pp. 538–

552, 2011.
[103] A. Hakkim, B. G. Furnrohr, K. Amann et al., “Impairment ̈
of neutrophil extracellular trap degradation is associated with
lupus nephritis,” Proceedings of the National Acadamy of Sciences
of the United States of America, vol. 107, no. 21, pp. 9813–9818,
2010.

[104] N. Dwivedi and M. Radic, “Citrullination of autoantigens
implicates NETosis in the induction of autoimmunity,” Annals
of the Rheumatic Diseases, vol. 73, no. 3, pp. 483–491, 2014.
[105] R. Khandpur, C. Carmona-Rivera, A. Vivekanandan-Giri et al.,
“NETs are a source of citrullinated autoantigens and stimulate

infammatory responses in rheumatoid arthritis,” Science Trans-
lational Medicine, vol. 5, no. 178, Article ID 178ra40, 2013.

[106] A. M. Lin, C. J. Rubin, R. Khandpur et al., “Mast cells and
neutrophils release IL-17 through extracellular trap formation
in psoriasis,” e Journal of Immunology, vol. 187, no. 1, pp. 490–
500, 2011.
[107] K. Kessenbrock, M. Krumbholz, U. Schonermarck et al., “Net- ̈
ting neutrophils in autoimmune small-vessel vasculitis,” Nature
Medicine, vol. 15, no. 6, pp. 623–625, 2009.
[108] T. A. Fuchs, A. Brill, D. Duerschmied et al., “Extracellular
DNA traps promote thrombosis,” Proceedings of the National
Acadamy of Sciences of the United States of America, vol. 107, no.
36, pp. 15880–15885, 2010.
[109] E. Darrah and F. Andrade, “NETs: the missing link between
cell death and systemic autoimmune diseases?” Frontiers in
Immunology, vol. 3, p. 428, 2013.
[110] S. Hahn, S. Giaglis, C. S. Chowdury, I. Hosli, and P. Hasler, ̈

“Modulation of neutrophil NETosis: interplay between infec-
tious agents and underlying host physiology,” Seminars in

Immunopathology, vol. 35, no. 4, pp. 439–453, 2013.

[111] Q. Remijsen, T. V. Berghe, E. Wirawan et al., “Neutrophil extra-
cellular trap cell death requires both autophagy and superoxide

generation,” Cell Research, vol. 21, no. 2, pp. 290–304, 2011.
[112] A. Itakura and O. J. T. McCarty, “Pivotal role for the mTOR
pathway in the formation of neutrophil extracellular traps via
regulation of autophagy,” American Journal of Physiology-Cell
Physiology, vol. 305, no. 3, pp. C348–C354, 2013.
[113] A. M. McInturf, M. J. Cody, E. A. Elliott et al., “Mammalian
target of rapamycin regulates neutrophil extracellular trap
formation via induction of hypoxia-inducible factor 1 ,” Blood,
vol. 120, no. 15, pp. 3118–3125, 2012.
[114] D. Fratantonio, A. Speciale, M. S. Molonia et al., “Alpha-lipoic
acid, but not di-hydrolipoic acid, activates Nrf2 response in
primary human umbilical-vein endothelial cells and protects
against TNF-� induced endothelium dysfunction,” Archives of
Biochemistry and Biophysics, vol. 655, pp. 18–25, 2018.
[115] Q. Ma, “Role of Nrf2 in oxidative stress and toxicity,” Annual
Review of Pharmacology and Toxicology, vol. 53, pp. 401–426,
2013.
[116] K. Yoh, K. Itoh, A. Enomoto et al., “Nrf2-defcient female

mice develop lupus-like autoimmune nephritis,” Kidney Inter-
national, vol. 60, no. 4, pp. 1343–1353, 2001.

[117] E. J. Cordova, R. Vel ́ azquez-Cruz, F. Centeno, V. Baca, and L. ́
Orozco, “Te NRF2 gene variant, -653G/A, is associated with
nephritis in childhood-onset systemic lupus erythematosus,”
Lupus, vol. 19, no. 10, pp. 1237–1242, 2010.

[118] J.-M. Lee, K. Chan, Y. W. Kan, and J. A. Johnson, “Tar-
geted disruption of Nrf2 causes regenerative immune-mediated

hemolytic anemia,” Proceedings of the National Acadamy of
Sciences of the United States of America, vol. 101, no. 26, pp. 9751–
9756, 2004.
[119] D. A. Johnson, S. Amirahmadi, C. Ward, Z. Fabry, and

J. A. Johnson, “Te absence of the pro-antioxidant tran-
scription factor Nrf2 exacerbates experimental autoimmune

encephalomyelitis,” Toxicological Sciences, vol. 114, no. 2, pp.
237–246, 2009.

BioMed Research International 11
[120] T. K. Pareek, A. Belkadi, S. Kesavapany et al., “Triterpenoid

modulation of IL-17 and Nrf-2 expression ameliorates neu-
roinfammation and promotes remyelination in autoimmune

encephalomyelitis,” Scientific Reports, vol. 1, no. 1, 2011.
[121] C. K. Sen, S. Roy, and L. Packer, “Fas mediated apoptosis of
human Jurkat T-cells: Intracellular events and potentiation by
redox-active
�-lipoic acid,” Cell Death & Differentiation, vol. 6,
no. 5, pp. 481–491, 1999.

Leave a reply