ArticlePDF AvailableLiterature Review

Mitochondrial Autoantibodies and the Role of Apoptosis in Pemphigus Vulgaris

Authors:
  • Hackensack Meridian School of Medicine

Abstract and Figures

Pemphigus vulgaris (PV) is an IgG autoantibody-mediated, potentially fatal mucocutaneous disease manifested by progressive non-healing erosions and blisters. Beyond acting to inhibit adhesion molecules, PVIgGs elicit a unique process of programmed cell death and detachment of epidermal keratinocytes termed apoptolysis. Mitochondrial damage by antimitochondrial antibodies (AMA) has proven to be a critical link in this process. AMA act synergistically with other autoantibodies in the pathogenesis of PV. Importantly, absorption of AMA inhibits the ability of PVIgGs to induce blisters. Pharmacologic agents that protect mitochondrial function offer a new targeted approach to treating this severe immunoblistering disease.
Content may be subject to copyright.
Citation: Hutchison, D.M.; Hosking,
A.-M.; Hong, E.M.; Grando, S.A.
Mitochondrial Autoantibodies and
the Role of Apoptosis in Pemphigus
Vulgaris. Antibodies 2022,11, 55.
https://doi.org/10.3390/
antib11030055
Academic Editor: Kyle T. Amber
Received: 3 August 2022
Accepted: 23 August 2022
Published: 25 August 2022
Publisher’s Note: MDPI stays neutral
with regard to jurisdictional claims in
published maps and institutional affil-
iations.
Copyright: © 2022 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/).
antibodies
Review
Mitochondrial Autoantibodies and the Role of Apoptosis in
Pemphigus Vulgaris
Dana M. Hutchison 1,2,3 , Anna-Marie Hosking 1, Ellen M. Hong 2,4 and Sergei A. Grando 1,5,6,*
1Department of Dermatology, University of California Irvine, Irvine, CA 92697, USA
2Beckman Laser Institute, University of California Irvine, Irvine, CA 92612, USA
3Department of Internal Medicine, Riverside Community Hospital, Riverside, CA 92501, USA
4Hackensack Meridian School of Medicine, Nutley, NJ 07110, USA
5Department of Biochemistry, University of California Irvine, Irvine, CA 92697, USA
6Institute for Immunology, University of California Irvine, Irvine, CA 92697, USA
*Correspondence: sgrando@hs.uci.edu; Tel.: +1-949-824-2713; Fax: +1-949-824-2993
Abstract:
Pemphigus vulgaris (PV) is an IgG autoantibody-mediated, potentially fatal mucocuta-
neous disease manifested by progressive non-healing erosions and blisters. Beyond acting to inhibit
adhesion molecules, PVIgGs elicit a unique process of programmed cell death and detachment of
epidermal keratinocytes termed apoptolysis. Mitochondrial damage by antimitochondrial antibodies
(AMA) has proven to be a critical link in this process. AMA act synergistically with other autoan-
tibodies in the pathogenesis of PV. Importantly, absorption of AMA inhibits the ability of PVIgGs
to induce blisters. Pharmacologic agents that protect mitochondrial function offer a new targeted
approach to treating this severe immunoblistering disease.
Keywords: pemphigus vulgaris; apoptosis; apoptosis; antimitochondrial autoantibodies
1. Introduction
Pemphigus encompasses a family of rare, potentially lethal autoimmune blistering
dermatoses involving the skin and mucosal surfaces. The word ‘pemphigus’ derives from
the Greek “pemphix”, which means blister. Its earliest use dates back to Hippocrates in
460–370 B.C. [
1
]. In modern history, the disease was first described by an Irish physician
in 1788 [
2
], however, our current understanding of pemphigus pathophysiology began
in 1964 with the discovery of autoantibodies in the sera of pemphigus vulgaris (PV) pa-
tients directed against the cell surface of keratinocytes [
3
]. The disease is associated with
both circulating and tissue-bound IgG autoantibodies, and manifested by the loss of cell–
cell adhesion of keratinocytes (acantholysis), and formation of non-healing suprabasal
intraepidermal blisters.
IgG antibodies against desmoglein-1 (Dsg1) and desmoglein-3 (Dsg3), calcium-dependent
cell adhesion molecules of the cadherin family, have been considered to play a primary role in
the development of PV. However, explanation of the pathogenesis remains controversial [
4
].
Clinically, the detection of anti-Dsg3 reactivity, with or without anti-Dsg1 reactivity, is helpful
in diagnosing PV. However, there have been a number of reports of patients in whom no
reactivity was found, challenging the notion of an exclusive role these proteins have in the
biologic mechanism of keratinocyte cohesion and their autoantibodies in blister formation in
patients (reviewed in Ref. [5]).
Proteomic studies have led to the discovery of additional major defined types of non-
Dsg proteins targeted by pemphigus autoantibodies, including: mitochondrial proteins,
desmocollin 1 and 3 (Dsc1 and Dsc3), various nicotinic and muscarinic acetylcholine recep-
tor subtypes, thyroid peroxidase, human leukocyte antigen (HLA) molecules, and secretory
pathway Ca
2+
/Mn
2+
-ATPase isoform 1 (SPCA1) encoded by the ATP2C1 gene, which is
mutated in Hailey-Hailey disease [
6
]. A “multiple hit” hypothesis has been proposed [
7
],
Antibodies 2022,11, 55. https://doi.org/10.3390/antib11030055 https://www.mdpi.com/journal/antibodies
Antibodies 2022,11, 55 2 of 8
wherein various non-Dsg autoantibodies against keratinocytes act synergistically with
anti-Dsg autoantibodies to cause blistering. These non-Dsg autoantibodies can induce
changes seen in PV, including keratinocyte shrinkage, cell–cell detachment, and triggering
of apoptotic signaling events (reviewed in Ref. [
4
]). For example, non-Dsg autoantibodies
against Dsc3, M3 muscarinic acetylcholine receptor (M3AR), and SPCA1 isolated from the
sera of patients with anti-Dsg1/3 autoantibody-negative PV were found to be pathogenic,
working synergistically with each other to cause acantholysis [
8
]. Thus, recent discoveries
of numerous non-Dsg autoantibody species further develop our understanding of PV and
implicate additional cell metabolism and signaling pathways involved in acantholysis [
6
,
9
].
In this article, we focus on targets of PV autoimmunity within the mitochondrion, as an-
timitochondrial antibodies (AMA) have proven to be a critical link in the
pathogenesis of PV.
2. Apoptolysis
Beyond acting at the keratinocyte cell membrane to block the function of adhesion
molecules, PVIgGs elicit the signaling events that trigger the keratinocyte cell death pro-
gram. The term “apoptolysis” has been coined to describe the distinct autoantibody-
induced process of keratinocyte structural damage and detachment (acantholysis) followed
by death (apoptosis), which is unique to PV. Acantholysis and apoptosis are inseparable in
PV and are mediated by the same cell death enzymes [
10
]. Apoptosis refers to programmed
cell death—a pathway not activated by inflammation, but rather by cysteine aspartate
proteases, or caspases. This cell death pathway can be activated by cellular damage (intrin-
sic pathway) or by signaling molecules (extrinsic, death receptor-initiated pathway), and
ultimately results in the formation of apoptotic bodies which are then cleared by phago-
cytic cells [
11
]. The best-known cell death pathways are apoptosis, oncosis, and necrosis,
however others have recently been described by the Nomenclature Committee on Cell
Death (NCCD) [
12
]. Additional, newly described apoptolytic pathways that play a role in
the pathophysiology of skin blistering characteristic of PV should be further investigated.
Mitochondria play a critical role in programmed cell death (reviewed in Ref. [
13
]).
Initiation of apoptotic pathways ultimately disrupt the inner mitochondrial membrane,
resulting in loss of the mitochondrial transmembrane potential and leakage of pro-apoptotic
proteins into the cell cytosol, including the release of cytochrome c (CytC), a marker
for mitochondrial outer membrane permealization and early apoptosis, and subsequent
activation of caspases [
14
]. The disruption of mitochondrial energy production, combined
with cleavage of adhesion and structural molecules, causes cytoskeleton collapse and the
keratinocyte to shrink [
10
]. The fundamental feature of apoptolysis is that anti-keratinocyte
antibodies in PV cause basal keratinocytes only to shrink, but not to die, giving rise to
their “tombstone” appearance on histopathology. This is distinct from the classic apoptotic
processes in the epidermis of patients with Stevens-Johnson Syndrome/Toxic Epidermal
Necrolysis, in which apoptosis leads to sloughing of the entire epidermis, including its
basal layer [15].
In PV, the apoptotic pathway is activated long before morphological evidence of
acantholysis [
16
,
17
]. The hypothetical sequence of apoptolysis development in PV has
five consecutive steps: (i) Pathogenic autoantibodies bind PV antigens on the surface of
keratinocytes and pro-apoptolytic signals are transduced. (ii) Activation of EGFR, mTOR,
Src, p38 MAPK and other signaling pathways increase intracellular calcium and initiate
programmed cell death enzymatic cascades predominately in basal keratinocytes. (iii)
Executioner caspases cleave tonofilaments, leading to their collapse and retraction, while
inter-desmosomal adhesion complexes are phosphorylated and dissociate. This results in
basal cell contraction, a crossing step of both the apoptotic and early acantholytic pathways.
The majority of desmosomes remain intact and bridge collapsing keratinocytes. (iv) The
continued degradation of structural proteins by the programmed cell death enzymes lead
to cytoskeleton collapse and complete separation of shrinking keratinocytes (visible acan-
tholysis). The sloughed cell membrane pieces trigger production of scavenging (secondary)
autoantibodies to Dsg, Dsc, E-cadherin and other adhesion molecules attached to the cell
Antibodies 2022,11, 55 3 of 8
membrane. (v) The end result is rounding up and apoptotic death of acantholytic cells
resulting from irreversible damage to mitochondrial and nuclear proteins by the same
cell death enzymes giving rise to a “tombstone” appearance of the surviving basal ker-
atinocytes [
10
]. The more recent observations of the pathogenic role of AMA, however,
indicate that damage of mitochondria occurs at an early stage of apoptolysis.
3. Mitochondrial Damage by AMA in PV
Patients with PV produce PVIgG antibodies targeting a variety of proteins, including
those at the inner and outer mitochondrial membrane, as well as the mitochondrial ma-
trix [
18
20
]. Utilizing protein microarray, the most common antigen targets recognized by
AMA in PV have been identified from a large cohort of patients (Table 1) [
18
]. Based on
the known functions of these proteins, AMA likely lead to mitochondrial dysfunction by
altering cellular ability to produce or inactivate reactive oxygen species, perform oxidative
phosphorylation, and participate in oxygen respiration. The exact mechanism of mitochon-
drial damage likely varies greatly among PV patients, consistent with the strikingly wide
spectrum of disease severity and treatment response [
20
]. Importantly, absorption of AMA
inhibits the ability of PVIgGs to induce keratinocyte detachment and blistering [19].
Table 1. Mitochondrial autoantibodies in patients with PV (adapted from Kalantari et al. [18]) *.
Symbol Antigen Localization on
Mitochondria
Frequency
PV (%) Control (%)
ABAT-V1 4-Aminobutyrate aminotransferase, mitochondrial; 50 kDa Matrix 19 4
ALDH4A1 Aldehyde dehydrogenase 4 family, member A1 Matrix 23 5
CPT1B Carnitine O-palmitoyltransferase 1B Outer membrane 18 5
CRAT Carnitine O-acetyltransferase Inner membrane 28 7
CYB5B Cytochrome b5type B; 21 kDa Outer membrane 19 1
ETFA Electron transfer flavoprotein, αprotein Matrix 19 4
ETFB Electron transfer flavoprotein, βprotein Matrix 21 3
FDXR-V2 NADPH:adrenodoxin oxidoreductase Matrix 25 6
FH Fumarate hydratase (fumarase) Mitochondrion 29 3
MAOB Amine oxidase (flavin-containing) B Outer membrane 27 5
ME2 NAD-dependent malic enzyme Matrix 18 6
ME3 NADP-dependent malic enzyme, mitochondrial Matrix 23 8
MLYCD Malonyl-CoA decarboxylase Mitochondrion 29 4
NDUFA9 NADH dehydrogenase [ubiquinone] 1αsubcomplex subunit 9; 39 kDa Matrix 20 3
NDUFA13
NADH dehydrogenase [ubiquinone] 1
α
subcomplex subunit 13; 16 kDa
Inner membrane 24 6
NDUFB10 NADH dehydrogenase [ubiquinone] 1βsubcomplex subunit 10 Matrix 17 2
NDUFV3 NADH dehydrogenase [ubiquinone] flavoprotein 3; 9 kDa Inner membrane 19 4
NDUFS6 NADH dehydrogenase [ubiquinone] iron-sulfur protein 6; 13 kDa Inner membrane 24 6
PC Pyruvate carboxylase Matrix 32 5
PDK4 Pyruvate dehydrogenase kinase, isozyme 4 Matrix 24 4
PDHA1 Pyruvate dehydrogenase E1 component αsubunit, somatic form Glycolysis 30 3
PMPCB Mitochondrial processing peptidase βsubunit Mitochondrial
organization 31 4
PRODH Proline oxidase Matrix 25 6
SOD2 Superoxide dismutase [Mn] Matrix 23 2
TIMM44 Mitochondrial import inner membrane translocase subunit Inner membrane 20 4
* Every PV serum analyzed in the referenced study contained an autoantibody to at least one mitochondrial
protein (data not shown).
Antibodies to mitochondrial proteins, among anti-keratinocyte antibodies and sev-
eral other soluble pathogenic factors, act synergistically to activate keratinocyte cell death
pathways in PV (Figure 1). In an organ culture of neonatal mouse skin—an
in vitro
model
of PV—AMA and anti-Dsg1/3 autoantibodies acted synergistically to induce acantholy-
sis [
21
]. In that study, treatment with AMA alone did not result in acantholysis, however
the combination of AMA and a mixture of anti-Dsg antibodies induced acantholysis. The
AMA/anti-Dsg3 combination induced epidermal splitting suprabasally and the AMA/anti-
Dsg1 combination induced epidermal splitting subcorneally, which is characteristic of
pemphigus foliaceus. Moreover, while acantholysis was observed following treatment with
high concentrations of the human anti-Dsg single-chain variable fragment (scFv), at low,
Antibodies 2022,11, 55 4 of 8
physiologic doses the scFv was not able to induce keratinocyte detachment. Acantholysis
was only observed when it was combined with AMA [
21
]. The synergy of AMA with other
autoantibodies in PV implies that a simultaneous hit is required to alter the keratinocyte
ability to maintain epidermal integrity. It is theorized that the binding of a single type of au-
toantibody only induces reversible changes in the keratinocyte, such that the cell maintains
its ability to recover via self-repair mechanisms. Irreversible keratinocyte damage only
occurs following presumed synchronized inactivation of salvage pathways by partnering
autoantibodies, leading to loss of epidermal integrity [21].
Antibodies 2022, 11, x FOR PEER REVIEW 4 of 9
Table 1. Mitochondrial autoantibodies in patients with PV (adapted from Kalantari et al. [18]) *.
Symbol Antigen Localization on Mitochondria
Frequency
PV (%) Control (%)
ABAT-V1 4-Aminobutyrate aminotransferase, mitochondrial; 50 kDa Matrix 19 4
ALDH4A1 Aldehyde dehydrogenase 4 family, member A1 Matrix 23 5
CPT1B Carnitine O-palmitoyltransferase 1B Outer membrane 18 5
CRAT Carnitine O-acetyltransferase Inner membrane 28 7
CYB5B Cytochrome b5 type B; 21 kDa Outer membrane 19 1
ETFA Electron transfer flavoprotein, α protein Matrix 19 4
ETFB Electron transfer flavoprotein, β protein Matrix 21 3
FDXR-V2 NADPH:adrenodoxin oxidoreductase Matrix 25 6
FH Fumarate hydratase (fumarase) Mitochondrion 29 3
MAOB Amine oxidase (flavin-containing) B Outer membrane 27 5
ME2 NAD-dependent malic enzyme Matrix 18 6
ME3 NADP-dependent malic enzyme, mitochondrial Matrix 23 8
MLYCD Malonyl-CoA decarboxylase Mitochondrion 29 4
NDUFA9 NADH dehydrogenase [ubiquinone] 1α subcomplex subunit 9; 39 kDa Matrix 20 3
NDUFA13 NADH dehydrogenase [ubiquinone] 1α subcomplex subunit 13; 16 kDa Inner membrane 24 6
NDUFB10 NADH dehydrogenase [ubiquinone] 1β subcomplex subunit 10 Matrix 17 2
NDUFV3 NADH dehydrogenase [ubiquinone] flavoprotein 3; 9 kDa Inner membrane 19 4
NDUFS6 NADH dehydrogenase [ubiquinone] iron-sulfur protein 6; 13 kDa Inner membrane 24 6
PC Pyruvate carboxylase Matrix 32 5
PDK4 Pyruvate dehydrogenase kinase, isozyme 4 Matrix 24 4
PDHA1 Pyruvate dehydrogenase E1 component α subunit, somatic form Glycolysis 30 3
PMPCB Mitochondrial processing peptidase β subunit Mitochondrial organization 31 4
PRODH Proline oxidase Matrix 25 6
SOD2 Superoxide dismutase [Mn] Matrix 23 2
TIMM44 Mitochondrial import inner membrane translocase subunit Inner membrane 20 4
* Every PV serum analyzed in the referenced study contained an autoantibody to at least one mito-
chondrial protein (data not shown).
Figure 1. Hypothetical multipathogenic mechanism of interconnected signaling cascades leading to
keratinocyte apoptolysis in pemphigus (modified from Marchenko et al. [19]). AMA: antimitochon-
drial antibodies; Anti-Dsg3 PVAb: anti-desmoglein 3 PV antibody; Cs: caspase; Dsg3: desmoglein-
3; EGFR: epidermal growth factor receptor; FasL: Fas ligand; FasR: Fas receptor; JNK: c-Jun N-ter-
minal kinase; mTOR: mammalian target of rapamycin; NO: nitric oxide; nDPVAb: non-Dsg PV an-
tibodies; OPVAg: other PV antigens; PKC: protein kinase C; PVAb: PV antibody; Src: SRC proto-
oncogene, nonreceptor tyrosine kinase; TNF-α: tumor necrosis factor-α.
Figure 1.
Hypothetical multipathogenic mechanism of interconnected signaling cascades leading to
keratinocyte apoptolysis in pemphigus (modified from Marchenko et al. [
19
]). AMA: antimitochon-
drial antibodies; Anti-Dsg3 PVAb: anti-desmoglein 3 PV antibody; Cs: caspase; Dsg3: desmoglein-3;
EGFR: epidermal growth factor receptor; FasL: Fas ligand; FasR: Fas receptor; JNK: c-Jun N-terminal
kinase; mTOR: mammalian target of rapamycin; NO: nitric oxide; nDPVAb: non-Dsg PV antibodies;
OPVAg: other PV antigens; PKC: protein kinase C; PVAb: PV antibody; Src: SRC proto-oncogene,
nonreceptor tyrosine kinase; TNF-α: tumor necrosis factor-α.
Human skin contains a complex non-neuronal cholinergic network composed of
the cytotransmitter acetylcholine and its nicotinic and muscarinic receptors [
22
]. These
receptors are involved in keratinocyte cell–cell and cell-matrix adhesion and some are
targeted by PV autoantibodies (reviewed in Ref. [
6
]). In a neonatal mouse model of PV,
preabsorption of PVIgGs with recombinant pemphaxin, a low-affinity dual muscarinic and
nicotinic receptor for acetylcholine, eliminated the acantholytic activity of PVIgGs. In turn,
the acantholytic activity could be restored by the addition of anti-pemphaxin antibody
back to the preabsorbed PVIgG fraction [
23
]. In addition to being present on the surface
of keratinocytes, nicotinic acetylcholine receptors have been found on the mitochondrial
outer membrane (mt-nAChRs) and are one of the targets of AMA. Stimulation of mt-
nAChR prevents apoptosis by inhibiting mitochondrial permeability transition pore (mPTP)
opening, thus preventing CytC release from the organelle [
20
]. Interestingly, nicotinergic
stimulation has been shown to protect keratinocytes from apoptolysis [20].
Among non-Dsg autoantibodies in PV patient sera, that increase activity of pathways
involved in apoptolysis, a combination of anti-M3AR, anti-SPCA1 and Dsc3 has been
identified [
8
]. When each of the above autoantibodies was tested alone in a neonatal mouse
skin explant model, none were able to solely induce acantholysis. However, a mixture of
all three produced an acantholytic effect similar to that of PVIgGs. When the combination
Antibodies 2022,11, 55 5 of 8
was further tested in a model of PV in BABL/c mice, it was also found to be sufficient to
disrupt epidermal integrity
in vivo
[
8
]. Thus, antibodies altering vital cell functions (i.e.,
anti-M3AR), cell adhesion (i.e., anti-Dsc3) and Ca
2+
metabolism (i.e., anti-SPCA1) appear
to work synergistically to produce an acantholytic effect similar to that of total PVIgGs in a
clinically relevant manner.
It has been shown that the binding of anti-M3AR or anti-SPCA1 autoantibodies to
keratinocytes leads to mitochondrial damage and the release of CytC and activation of the
caspase 9 pathway [
24
]. Anti-SPCA1 produced a 10-fold, and anti-M3AR autoantibody a
4–5 fold, increase in levels of CytC and Cs-9, respectively [
24
]. Further, anti-M3AR and
anti-SPCA1 autoantibodies worked synergistically in a 3D culture of human epidermis.
A mixture of anti-SPCA1 and anti-M3AR autoantibodies resulted in changes in the mor-
phology of human epidermis consistent with acantholysis, including “bubbling” at the
epidermis while basal cells remained intact at the dermal-epidermal junction [
24
]. However,
when given alone, anti-M3AR did not cause morphological changes in that
in vitro
model,
while anti-SPCA1 given alone resulted in shrinkage of epidermal keratinocytes [24].
SPCA1 is located on the Golgi apparatus, and it is thought that a defect in SPCA1 Ca
2+
sequestration contributes to Golgi stress leading to apoptosis [
25
]. The Golgi complex is
capable of transducing pro-apoptotic signals which are partially mediated through caspase
2 (Cs-2) that localizes to the Golgi apparatus [
26
,
27
]. A recent study demonstrated that the
effects of PV anti-SPCA1 autoantibody on mitochondrial CytC release were abolished in
the presence of a Cs-2 inhibitor [
24
]. These findings suggest that anti-SPCA1 autoantibodies
alter mitochondrial function through Cs-2, triggering early pro-apoptotic events. Notably,
SPCA1 is encoded by the ATP2C1 gene, which is mutated in benign chronic pemphigus
(also known as Hailey-Hailey disease) [28].
The neonatal Fc receptor (FcRn) may, in part, mediate the pathogenic effects of PV
autoantibodies to intracellular self-antigens, including SPCA1 and those present in mito-
chondria. Following binding of PVIgG to FcRn on the cell membrane of keratinocytes,
complexes of PVIgG-FcRn are internalized and trafficked to the mitochondria, where they
are released from endosomes [
21
]. The complexes dissociate and AMA reach mitochon-
dria, triggering early apoptotic events and cell shrinkage. However, this AMA-induced
damage is reversible. Interestingly, cells lacking FcRn do not internalize PVIgGs and AMA
is therefore unable to reach the mitochondria [
21
]. Further, pretreatment of mouse ker-
atinocytes with anti-FcRn antibody, which functionally inactivates FcRn, prevents shrinkage
of keratinocytes as well as other AMA-dependent changes in mitochondrial integrity and
metabolism [
21
]. Since FcRn is predominantly expressed in the basal epidermal layer [
29
],
basal and suprabasal keratinocytes should respond differently to the PVIgGs entering
keratinocytes via FcRn-mediated mechanism. This may explain the suprabasal location of
epidermal split in PV, as only basal keratinocytes shrink, thereby separating themselves
from suprabasal keratinocytes. However, the exact mechanism by which PVIgGs enter ker-
atinocytes to reach the mitochondrial target antigens, and why other cell types in the body
that contain the same mitochondrial antigens are not affected by PVIgGs,
is still unknown.
New research has implicated that the thioredoxin-2 (Trx2)/apoptosis signal-regulating
kinase 1 (ASK1) pathway may play a key role in mediating mitochondrial injury in PV [
30
].
ASK1, a serine/threonine kinase, is activated by oxidative stress and triggers apoptosis.
One of the functions of Trx2 is to inactivate ASK1 by forming a complex with the molecule,
thereby preventing its phosphorylation and activation. Elevated levels of reactive oxygen
species, which are found following mitochondrial injury by AMA, oxidize the cysteine
residues of Trx2, promoting the dissociation of the Trx2-ASK1 complex and allowing
activated ASK1 to trigger apoptotic events. In an
in vitro
study of keratinocytes cultured
with PV sera, the Trx2/ASK1 cascade was abnormally activated, with decreased local
expression of Trx2, an increased amount of phosphorylated ASK1, and an increased rate
of apoptosis compared to control cells [
29
]. In a mouse model, the overexpression of
Trx2 decreased ASK1 phosphorylation, the apoptotic rate, and relieved acantholysis and
blister formation. Thus, Trx2 appears to have a protective role in mitochondrial injury
Antibodies 2022,11, 55 6 of 8
and compounds targeting the Trx2/ASK1 pathway may help prevent progression of PV
in the future.
4. Efficacy of Mitochondrion Protective Agents in Pemphigus Patients
While the antigen specificities of AMA produced by individual PV patients is highly
variable (Table 1), uniform protection from mitochondrial damage can be achieved with non-
steroidal mitochondrion-protective pharmacologic agents [
18
]. Growing evidence suggests
that the use of mitochondrion-protecting drugs, such as cyclosporine A (CsA), tetracyclines
and nicotinamide (also called niacinamide), are justified in the treatment of PV [
18
]. In
addition to inhibiting the production of cytokines involved in T-cell activation, CsA can
protect mitochondria by binding cyclophilin D and inhibiting opening of mPTP, allowing
the mitochondrion to retain a high transmembrane potential (
ψ
m) [
31
33
]. Tetracyclines,
such as minocycline and doxycyline, inhibit mPTP opening by reducing mitochondrial
uptake of Ca
2+
, inhibiting loss of
ψ
m and preventing CytC release [
34
]. Niacinamide, one
of the two principle forms of vitamin B
3
, is a precursor of the coenzyme NAD
+
consumed
during ATP generation in the mitochondrial electron transport chain. Vitamin B
3
is thought
to help cells retain high-quality mitochondria by activating autophagy of mitochondria
with low
ψ
m, which indicates a damaged (depolarized) cell [
33
]. Animal studies pro-
vided evidence that pharmacologic protection of mitochondria with CsA, minocycline,
and nicotinamide prevents PVIgGs-mediated induction of skin blisters in mouse skin [
18
],
which is in keeping with clinical reports that PV lesions can be partially controlled by these
agents in the absence of systemic steroids [3537].
One interesting observation is that nicotine competes with PVIgGs for binding to
mt-nAChRs, thereby inhibiting mitochondrial CytC release in a dose-dependent fashion,
and prevents intrinsic apoptosis in keratinocytes [
20
]. The potential therapeutic effect
of nicotinergic stimulation in PV has been reported in a case study [
38
] as well as in
epidemiological data showing a beneficial effect of smoking on PV [
39
41
]. This may be in
part due to nicotinic agonism at mt-nAChRs protecting mitochondria.
Additionally, sirolimus (also known as rapamycin) has been proposed to protect
keratinocytes from PVIgG aggression through a poorly understood mechanism. In an
experimental setting, pretreatment with sirolimus prevented acantholysis in a mouse
model of PV [
42
]. In a clinical setting, within two weeks of initiating therapy with sirolimus,
PV lesions on a man with severe side effects to prednisone completely healed, allowing him
to rapidly taper off prednisone and remain lesion free on a maintenance dose of 2 mg/day
of sirolimus. Studies in other mitochondrial disorders have suggested that sirolimus
improves cellular function by reducing the number of dysfunctional mitochondria within
an organelle, thereby preserving mitochondrial integrity [43].
At the UC Irvine Immunobullous Clinic, PV patients are successfully treated with
a multidrug therapeutic approach including mitochondrion-protecting agents (minocy-
cline or doxycycline 200 mg/day + niacinamide 1.5 g/day), in addition to intravenous
immunoglobulin, or IVIg, systemic corticosteroids, and an immunosuppressive cytotoxic
drug (mycophenolate mofetil, azathioprine or cyclophosphamide) [
44
]. The synergy of the
drugs utilized in this protocol allows for rapid achievement and maintenance of clinical re-
mission in approximately 88% of pemphigus patients with a smaller than usual cumulative
dose of systemic corticosteroids. Indeed, while these mitochondrion protective agents are
already utilized in the treatment of PV, novel pharmacologic prospects which protect and
or compensate for disrupted mitochondrial function may offer an even safer, nonsteroidal
approach to treating PV in the future.
5. Conclusions and Future Directions
The acantholytic process in PV is complex and involves autoantibodies directed
against various keratinocyte proteins that maintain adhesion and other vital cell functions.
While different pathogenic autoantibodies act synergistically in the pathogenesis of PV,
pharmacological protection or the elimination of a single antibody may suffice to protect
Antibodies 2022,11, 55 7 of 8
epidermal integrity and halt development of the disease. Further characterization of the
role of individual AMA causing mitochondrial injury in the pathogenesis of PV may lead
to development of personalized pharmacologic therapies to correct mitochondrial abnor-
malities unique to individual PV patients. Future studies to improve our understanding
of the immunopathogenesis of PV should, therefore, aid in the development of novel and
more efficient therapeutic modalities.
Author Contributions:
Conceptualization, D.M.H., S.A.G.; Writing—Original Draft Preparation,
D.M.H.; Writing—Review and Editing, E.M.H., A.-M.H., S.A.G.; Figures, D.M.H., E.M.H. All authors
have read and agreed to the published version of the manuscript.
Funding: This research received no external funding.
Institutional Review Board Statement: Not applicable.
Informed Consent Statement: Not applicable.
Data Availability Statement:
No new data were created or analyzed in this study. Data sharing is
not applicable to this article.
Conflicts of Interest: The authors declare no conflict of interest.
Disclaimer:
The views expressed in this publication represent those of the author(s) and do not
necessarily represent the official views of HCA Healthcare or any of its affiliated entities.
References
1. Lever, W.F.; Talbott, J.H. Pemphigus: A historical study. Arch. Dermatol. Syphilol. 1942,46, 800–823. [CrossRef]
2. Dickson, S. Observations on Pemphigus. Lond. Med. J. 1788,9, 309–324. [PubMed]
3.
Beutner, E.H.; Jordon, R.E. Demonstration of skin antibodies in sera of pemphigus vulgaris patients by indirect immunofluorescent
staining. Proc. Soc. Exp. Biol. Med. 1964,117, 505–510. [CrossRef]
4.
Ahmed, A.R.; Carrozzo, M.; Caux, F.; Cirillo, N.; Dmochowski, M.; Alonso, A.E.; Gniadecki, R.; Hertl, M.; López-Zabalza, M.J.;
Lotti, R.; et al. Monopathogenic vs. multipathogenic explanations of pemphigus pathophysiology. Exp. Dermatol.
2016
,25,
839–846. [CrossRef] [PubMed]
5. Grando, S.A. Pemphigus autoimmunity: Hypotheses and realities. Autoimmunity 2012,45, 7–35. [CrossRef] [PubMed]
6.
Amber, K.T.; Valdebran, M.; Grando, S.A. Non-Desmoglein Antibodies in Patients with Pemphigus Vulgaris. Front. Immunol.
2018,9, 1190. [CrossRef]
7.
Grando, S.A. Autoimmunity to keratinocyte acetylcholine receptors in pemphigus. Dermatology
2000
,201, 290–295. [CrossRef] [PubMed]
8.
Chernyavsky, A.; Amber, K.T.; Agnoletti, A.F.; Wang, C.; Grando, S.A. Synergy among non-desmoglein antibodies contributes to
the immunopathology of desmoglein antibody-negative pemphigus vulgaris. J. Biol. Chem. 2019,294, 4520–4528. [CrossRef]
9.
Sharma, P.; Mao, X.; Payne, A.S. Beyond steric hindrance: The role of adhesion signaling pathways in the pathogenesis of
pemphigus. J. Dermatol. Sci. 2007,48, 1–14. [CrossRef]
10.
Grando, S.A.; Bystryn, J.C.; Chernyavsky, A.I.; Fruši´c-Zlotkin, M.; Gniadecki, R.; Lotti, R.; Milner, Y.; Pittelkow, M.R.; Pincelli, C.
Apoptolysis: A novel mechanism of skin blistering in pemphigus vulgaris linking the apoptotic pathways to basal cell shrinkage
and suprabasal acantholysis. Exp. Dermatol. 2009,18, 764–770. [CrossRef]
11.
Galluzzi, L.; Vitale, I.; Aaronson, S.A.; Abrams, J.M.; Adam, D.; Agostinis, P.; Alnemri, E.S.; Altucci, L.; Amelio, I.; Andrews, D.W.;
et al. Molecular mechanisms of cell death: Recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death
Differ. 2018,25, 486–541. [CrossRef] [PubMed]
12.
Yan, G.; Elbadawi, M.; Efferth, T. Multiple cell death modalities and their key features (Review). World Acad. Sci. J.
2020
,2, 39–48. [CrossRef]
13.
Gupta, S.; Kass, G.E.; Szegezdi, E.; Joseph, B. The mitochondrial death pathway: A promising therapeutic target in diseases. J.
Cell. Mol. Med. 2009,13, 1004–1033. [CrossRef] [PubMed]
14. Elmore, S. Apoptosis: A Review of Programmed Cell Death. Toxicol. Pathol. 2007,35, 495–516. [CrossRef]
15.
Hasegawa, A.; Abe, R. Recent advances in managing and understanding Stevens-Johnson syndrome and toxic epidermal
necrolysis [version 1; peer review: 2 approved]. F1000Research 2020,9, 612. [CrossRef]
16.
Arredondo, J.; Chernyavsky, A.I.; Karaouni, A.; Grando, S.A. Novel mechanisms of target cell death and survival and of
therapeutic action of IVIg in Pemphigus. Am. J. Pathol. 2005,167, 1531–1544. [CrossRef]
17.
Lotti, R.; Shu, E.; Petrachi, T.; Marconi, A.; Palazzo, E.; Quadri, M.; Lin, A.; O’Reilly, L.A.; Pincelli, C. Soluble Fas Ligand Is
Essential for Blister Formation in Pemphigus. Front. Immunol. 2018,9, 370. [CrossRef] [PubMed]
18.
Kalantari-Dehaghi, M.; Chen, Y.; Deng, W.; Chernyavsky, A.; Marchenko, S.; Wang, P.H.; Grando, S.A. Mechanisms of mitochon-
drial damage in keratinocytes by pemphigus vulgaris antibodies. J. Biol. Chem. 2013,288, 16916–16925. [CrossRef]
19.
Marchenko, S.; Chernyavsky, A.I.; Arredondo, J.; Gindi, V.; Grando, S.A. Antimitochondrial autoantibodies in pemphigus
vulgaris: A missing link in disease pathophysiology. J. Biol. Chem. 2010,285, 3695–3704. [CrossRef]
Antibodies 2022,11, 55 8 of 8
20.
Chernyavsky, A.; Chen, Y.; Wang, P.H.; Grando, S.A. Pemphigus vulgaris antibodies target the mitochondrial nicotinic acetyl-
choline receptors that protect keratinocytes from apoptolysis. Int. Immunopharmacol. 2015,29, 76–80. [CrossRef]
21.
Chen, Y.; Chernyavsky, A.; Webber, R.J.; Grando, S.A.; Wang, P.H. Critical Role of the Neonatal Fc Receptor (FcRn) in the
Pathogenic Action of Antimitochondrial Autoantibodies Synergizing with Anti-desmoglein Autoantibodies in Pemphigus
Vulgaris. J. Biol. Chem. 2015,290, 23826–23837. [CrossRef] [PubMed]
22. Grando, S.A. Cholinergic control of epidermal cohesion. Exp. Dermatol. 2006,15, 265–282. [CrossRef]
23.
Nguyen, V.T.; Ndoye, A.; Grando, S.A. Pemphigus vulgaris antibody identifies pemphaxin. A novel keratinocyte annexin-like
molecule binding acetylcholine. J. Biol. Chem. 2000,275, 29466–29476. [CrossRef] [PubMed]
24.
Chernyavsky, A.; Patel, K.G.; Grando, S.A. Mechanisms of synergy of autoantibodies to M3 muscarinic acetylcholine receptor and
secretory pathway Ca(2+)/Mn(2+)-ATPase isoform 1 in patients with non-desmoglein pemphigus vulgaris. Int. Immunopharmacol.
2020,80, 106149. [CrossRef]
25.
Okunade, G.W.; Miller, M.L.; Azhar, M.; Andringa, A.; Sanford, L.P.; Doetschman, T.; Prasad, V.; Shull, G.E. Loss of the Atp2c1
secretory pathway Ca(2+)-ATPase (SPCA1) in mice causes Golgi stress, apoptosis, and midgestational death in homozygous
embryos and squamous cell tumors in adult heterozygotes. J. Biol. Chem. 2007,282, 26517–26527. [CrossRef]
26.
Mancini, M.; Machamer, C.E.; Roy, S.; Nicholson, D.W.; Thornberry, N.A.; Casciola-Rosen, L.A.; Rosen, A. Caspase-2 is localized
at the Golgi complex and cleaves golgin-160 during apoptosis. J. Cell Biol. 2000,149, 603–612. [CrossRef] [PubMed]
27.
Qin, Y.; Vanden Hoek, T.L.; Wojcik, K.; Anderson, T.; Li, C.-Q.; Shao, Z.-H.; Becker, L.B.; Hamann, K.J. Caspase-dependent
cytochrome c release and cell death in chick cardiomyocytes after simulated ischemia-reperfusion. Am. J. Physiol. Heart Circ.
Physiol. 2004,286, H2280–H2286. [CrossRef]
28.
Micaroni, M.; Giacchetti, G.; Plebani, R.; Xiao, G.G.; Federici, L. ATP2C1 gene mutations in Hailey-Hailey disease and possible
roles of SPCA1 isoforms in membrane trafficking. Cell Death Dis. 2016,7, e2259. [CrossRef]
29.
Cauza, K.; Hinterhuber, G.; Dingelmaier-Hovorka, R.; Brugger, K.; Klosner, G.; Horvat, R.; Wolff, K.; Foedinger, D. Expression of
FcRn, the MHC Class I-Related Receptor for IgG, in Human Keratinocytes. J. Investig. Dermatol. 2005,124, 132–139. [CrossRef]
30.
Wei, B.; Li, F. Mechanisms of Trx2/ASK1-Mediated Mitochondrial Injury in Pemphigus Vulgaris. BioMed Res. Int.
2021
,2021,
2471518. [CrossRef]
31.
Halestrap, A.P.; Connern, C.P.; Griffiths, E.J.; Kerr, P. Cyclosporin A binding to mitochondrial cyclophilin inhibits the permeability
transition pore and protects hearts from ischaemia/reperfusion injury. Mol. Cell. Biochem. 1997,174, 167–172. [CrossRef] [PubMed]
32.
Russell, G.; Graveley, R.; Seid, J.; Al-Humidan, A.-K.; Skjodt, H. Mechanisms of action of cyclosporine and effects on connective
tissues. Semin. Arthritis Rheum. 1992,21, 16–22. [CrossRef]
33.
Kang, H.T.; Hwang, E.S. Nicotinamide enhances mitochondria quality through autophagy activation in human cells. Aging Cell
2009,8, 426–438. [CrossRef] [PubMed]
34.
Wang, X.; Zhu, S.; Drozda, M.; Zhang, W.; Stavrovskaya, I.G.; Cattaneo, E.; Ferrante, R.J.; Kristal, B.S.; Friedlander, R.M.
Minocycline inhibits caspase-independent and -dependent mitochondrial cell death pathways in models of Huntington’s disease.
Proc. Natl. Acad. Sci. USA 2003,100, 10483–10487. [CrossRef]
35.
Barthelemy, H.; Frappaz, A.; Cambazard, F.; Mauduit, G.; Rouchouse, B.; Kanitakis, J.; Souteyrand, P.; Claudy, A.; Thivolet, J.
Treatment of nine cases of pemphigus vulgaris with cyclosporine. J. Am. Acad. Dermatol. 1988,18, 1262–1266. [CrossRef]
36.
Chaffins, M.L.; Collison, D.; Fivenson, D.P. Treatment of pemphigus and linear IgA dermatosis with nicotinamide and tetracycline:
A review of 13 cases. J. Am. Acad. Dermatol. 1993,28, 998–1000. [CrossRef]
37.
Sawai, T.; Kitazawa, K.; Danno, K.; Sugie, N.; Mochizuki, T.; Sugiura, H.; Uehara, M. Pemphigus vegetans with oesophageal
involvement: Successful treatment with minocycline and nicotinamide. Br. J. Dermatol. 1995,132, 668–670. [CrossRef]
38.
Mehta, J.N.; Martin, A.G. A Case of Pemphigus Vulgaris Improved by Cigarette Smoking. Arch. Dermatol.
2000
,136, 15–17. [CrossRef]
39.
Brenner, S.; Tur, E.; Shapiro, J.; Ruocco, V.; D’Avino, M.; Ruocco, E.; Tsankov, N.; Vassileva, S.; Drenovska, K.; Brezoev, P.; et al.
Pemphigus vulgaris: Environmental factors. Occupational, behavioral, medical, and qualitative food frequency questionnaire.
Int. J. Dermatol. 2001,40, 562–569. [CrossRef]
40. Sullivan, T.P.; Elgart, G.W.; Kirsner, R.S. Pemphigus and smoking. Int. J. Dermatol. 2002,41, 528–530. [CrossRef]
41. Valikhani, M.; Kavusi, S.; Chams-Davatchi, C.; Hallaji, Z.; Esmaili, N.; Ghandi, N.; Farahani, F.; Lajevardi, V. Impact of smoking
on pemphigus. Int. J. Dermatol. 2008,47, 567–570. [CrossRef] [PubMed]
42.
Pretel, M.; España, A.; Marquina, M.; Pelacho, B.; López-Picazo, J.M.; López-Zabalza, M.J. An imbalance in Akt/mTOR is
involved in the apoptotic and acantholytic processes in a mouse model of pemphigus vulgaris. Exp. Dermatol.
2009
,18, 771–780.
[CrossRef] [PubMed]
43.
Cheema, N.; Cameron, J.M.; Hood, D.A. Effect of rapamycin on mitochondria and lysosomes in fibroblasts from patients with
mtDNA mutations. Am. J. Physiol.-Cell Physiol. 2021,321, C176–C186. [CrossRef] [PubMed]
44.
Grando, S.A. Retrospective analysis of a single-center clinical experience toward development of curative treatment of 123
pemphigus patients with a long-term follow-up: Efficacy and safety of the multidrug protocol combining intravenous im-
munoglobulin with the cytotoxic immunosuppressor and mitochondrion-protecting drugs. Int. J. Dermatol.
2019
,58, 114–125.
[CrossRef] [PubMed]
... Neuronal-type nAChRs expressed by non-excitable cells are involved in the pathogenesis of palmoplantar pustulosis, psoriasis, pemphigus [97][98][99][100], rheumatic diseases [101], inflammatory bowel diseases, asthma and some types of cancers [102]. The neuronal cholinergic system is abundant in keratinocytes of the stratified epithelium and plays an important role in controlling the viability, the proliferation, and the migration of these cells [103][104][105]. ...
... ACh is produced in the skin tissue by keratinocytes, endothelial cells and immune cells entering the skin at inflammation sites [106]. A few studies suggest the possible presence of antibodies to neuronal-type nAChRs in the above diseases [97][98][99][100]107,108]. However, even if such antibodies do occur in some patients with these diseases, their pathogenic function is yet unknown while the available data do not significantly support such a role. ...
Article
Full-text available
Nicotinic acetylcholine receptors (nAChRs) are widely expressed in many and diverse cell types, participating in various functions of cells, tissues and systems. In this review, we focus on the autoimmunity against neuronal nAChRs, the specific autoantibodies and their mechanisms of pathological action in selected autoimmune diseases. We summarize the current relevant knowledge from human diseases as well as from experimental models of autoimmune neurological disorders related to antibodies against neuronal nAChR subunits. Despite the well-studied high immunogenicity of the muscle nAChRs where autoantibodies are the main pathogen of myasthenia gravis, autoimmunity to neuronal nAChRs seems infrequent, except for the autoantibodies to the ganglionic receptor, the α3 subunit containing nAChR (α3-nAChR), which are detected and are likely pathogenic in Autoimmune Autonomic Ganglionopathy (AAG). We describe the detection, presence and function of these antibodies and especially the recent development of a cell-based assay (CBA) which, contrary to until recently available assays, is highly specific for AAG. Rare reports of autoantibodies to the other neuronal nAChR subtypes include a few cases of antibodies to α7 and/or α4β2 nAChRs in Rasmussen encephalitis, schizophrenia, autoimmune meningoencephalomyelitis, and in some myasthenia gravis patients with concurrent CNS symptoms. Neuronal-type nAChRs are also present in several non-excitable tissues, however the presence and possible role of antibodies against them needs further verification. It is likely that the future development of more sensitive and disease-specific assays would reveal that neuronal nAChR autoantibodies are much more frequent and may explain the mechanisms of some seronegative autoimmune diseases.
... Mitochondrial nAChRs control cytochrome c (CytC) release by blocking mitochondrial permeability transition pore (mPTP) opening, thus preventing mitochondrial apoptosis. Therefore, nAChRs play a key role in maintaining keratinocyte viability [31]. ...
... Antimitochondrial antibodies (AMA) and anti-Dsg1/3 autoantibodies are thought to act synergistically to induce acantholysis in mouse skin. For example, the removal of AMA abolishes the acantholytic effect of PV serum, but the treatment of cells with AMA alone does not result in acantholysis; however, the treatment of these cells with a mixture of AMA and anti-Dsg antibodies induced acantholysis [31]. Different acantholytic factors are amenable to specific treatment strategies; therefore, it is not surprising that mitochondrion-protecting drugs, such as tetracyclines and niacinamide, are used in the potentially curative treatment of PV [42]. ...
Article
Full-text available
The importance of acetylcholine (ACh) in keratinocyte adhesion and acantholysis has been investigated over the last three decades, particularly in the pathophysiology of autoimmune blistering dermatoses. Pemphigus vulgaris (PV) is an autoimmune blistering skin disease where autoantibody-mediated suprabasilar intraepidermal splitting causes flaccid blisters and non-healing erosions of the oral mucosa and sometimes also of the skin. Historically, acantholysis in PV was thought to be driven by anti-desmoglein (Dsg) antibodies. Herein, we describe the role of autoantibodies against keratinocyte muscarinic and nicotinic acetylcholine receptors, as well as the annexin-like molecule pemphaxin that also binds ACh, in the immunopathogenesis of PV. The identification of targets in this disease is important, as they may lead to novel diagnostic and therapeutic options in the future for this potentially deadly disease.
... It is an autoimmune disease in which the body's immune system incorrectly recognizes skin proteins as foreign and thus produces antibodies to attack the foreign protein [15,16]. Trigger factors include medications [17] such as antibiotics, calcium channel blockers, ACE inhibitors, NSAIDs, salicylates, and interleukins, vaccines such as influenza [18,19], swine flu [20], tetanus toxoid [21], and COVID-19 vaccines [22-24], viral infections such as herpes simplex [25], hepatitis B and C [26-28], bacterial infections such as Helicobacter pylori [29], parasitic infections such as Toxoplasma gondii [30,31], and following organ transplantation [32,33]. ...
Article
Full-text available
Acute skin failure is rarely the primary diagnosis that necessitates admission to an intensive care unit. Dermatological manifestations in critically ill patients, on the other hand, are relatively common and can be used to make a key diagnosis of an adverse drug reaction or an underlying systemic illness, or they may be caused by factors related to a prolonged stay or invasive procedures. In intensive care units, their classification is based on the aetiopathogenesis of the cutaneous lesion and, in the meantime, distinguishes critical patients. When evaluating dermatological manifestations, several factors must be considered: onset, morphology, distribution, and associated symptoms and signs. This review depicts dermatological signs in critical patients in order to lay out better recognition.
Article
Aim To quantify the presence of apoptotic bodies in the blood samples of pemphigus vulgaris cases and to correlate levels of the inflammatory cytokines TNF-α and IL-2 with the disease severity. Materials and Methods Sera were collected from eight subjects including four patients with pemphigus vulgaris and four from healthy volunteers taken as controls. Patients are divided into two groups, one on regular steroid therapy (1–2 mg per kg per day) for at least 2 weeks, other from untreated patients. The levels of apoptotic bodies were evaluated using flow cytometry and levels of IL-2 and TNF-α were evaluated using enzyme-linked immunosorbent assay. Results The mean value of annexin V, propidium iodide, and a combination of both stained cells showed 3.962%, 7.375%, and 8.988%. The mean value of IL-2 and TNF-α are 4.568 and 56.175 pg/mL, respectively. Conclusion The levels of apoptotic cells increased with the disease progression and also differed between the patients with and without steroidal therapy. The study also shows the increase in levels of TNF-α with increased severity of the disease and also among patients without treatment which in turn correlated with the levels of apoptotic cells. Levels of IL-2 did not show much difference in the levels among the healthy controls and the diseased.
Article
Full-text available
Mitochondria are intracellular organelles that play a critical role in numerous cellular processes including the regulation of metabolism, cellular stress response, and cell fate. Mitochondria themselves are subject to well-orchestrated regulation in order to maintain organelle and cellular homeostasis. Wound healing is a multifactorial process that involves the stringent regulation of several cell types and cellular processes. In the event of dysregulated wound healing, hard-to-heal chronic wounds form and can place a significant burden on healthcare systems. Importantly, treatment options remain limited owing to the multifactorial nature of chronic wound pathogenesis. One area that has received more attention in recent years is the role of mitochondria in wound healing. With regards to this, current literature has demonstrated an important role for mitochondria in several areas of wound healing and chronic wound pathogenesis including metabolism, apoptosis, and redox signalling. Additionally, the influence of mitochondrial dynamics and mitophagy has also been investigated. However, few studies have utilised patient tissue when studying mitochondria in wound healing, instead using various animal models. In this review we dissect the current knowledge of the role of mitochondria in wound healing and discuss how future research can potentially aid in the progression of wound healing research.
Article
Full-text available
Objective: Apoptotic events mediated by mitochondrial injury play an important role on the onset of Pemphigus vulgaris (PV). The thioredoxin-2 (Trx2)/apoptosis signal-regulating kinase 1 (ASK1) signaling pathway is considered a key cascade involved on the regulation of mitochondrial injury. Hence, we have investigated the regulatory mechanism of the Trx2/ASK1 signaling in PV-induced mitochondrial injury. Methods: Serum and tissue samples were collected from clinical PV patients to detect the oxidative stress factors, cell apoptosis, and expression of members from Trx2/ASK1 signaling. HaCaT cells were cultured with the serum of PV patients and transfected with Trx2 overexpression or silencing vector. Changes in the levels of reactive oxygen species (ROS), mitochondrial membrane potential (△ψm), and apoptosis were further evaluated. A PV mouse model was established and administered with Trx2-overexpressing plasmid. The effect of ectopic Trx2 expression towards acantholysis in PV mice was observed. Results: A series of cellular and molecular effects, including (i) increased levels of oxidative stress products, (ii) destruction of epithelial cells in the skin tissues, (iii) induction of apoptosis in keratinocytes, (iv) reduction of Trx2 protein levels, and (v) enhanced phosphorylation of ASK1, were detected in PV patients. In vitro experiments confirmed that Trx2 can inhibit ASK1 phosphorylation, alleviate ROS release, decrease △ψm, and lower the apoptotic rate. Injection of Trx2-overexpressing vectors in vivo could also relieve acantholysis and blister formation in PV mice. Conclusion: The Trx2/ASK1 signaling pathway regulates the incidence of PV mediated by mitochondrial injury.
Article
Full-text available
Stevens-Johnson syndrome (SJS) and toxic epidermal necrolysis (TEN) are life-threatening diseases characterized by detachment of the epidermis and mucous membrane. SJS/TEN are considered to be on the same spectrum of diseases with different severities. They are classified by the percentage of skin detachment area. SJS/TEN can also cause several complications in the liver, kidneys, and respiratory tract. The pathogenesis of SJS/TEN is still unclear. Although it is difficult to diagnose early stage SJS/TEN, biomarkers for diagnosis or severity prediction have not been well established. Furthermore, optimal therapeutic options for SJS/TEN are still controversial. Several drugs, such as carbamazepine and allopurinol, are reported to have a strong relationship with a specific human leukocyte antigen (HLA) type. This relationship differs between different ethnicities. Recently, the usefulness of HLA screening before administering specific drugs to decrease the incidence of SJS/TEN has been investigated. Skin detachment in SJS/TEN skin lesions is caused by extensive epidermal cell death, which has been considered to be apoptosis via the Fas-FasL pathway or perforin/granzyme pathway. We reported that necroptosis, i.e. programmed necrosis, also contributes to epidermal cell death. Annexin A1, released from monocytes, and its interaction with the formyl peptide receptor 1 induce necroptosis. Several diagnostic or prognostic biomarkers for SJS/TEN have been reported, such as CCL-27, IL-15, galectin-7, and RIP3. Supportive care is recommended for the treatment of SJS/TEN. However, optimal therapeutic options such as systemic corticosteroids, intravenous immunoglobulin, cyclosporine, and TNF-α antagonists are still controversial. Recently, the beneficial effects of cyclosporine and TNF-α antagonists have been explored. In this review, we discuss recent advances in the pathophysiology and management of SJS/TEN.
Article
Full-text available
Pemphigus vulgaris (PV) is a potentially lethal mucocutaneous blistering disease characterized by IgG autoantibodies (AuAbs) binding to epidermal keratinocytes and inducing this devastating disease. Here, we observed that non-desmoglein (Dsg) AuAbs in serum of individuals with Dsg1/3 AuAb-negative acute PV are pathogenic, since IgGs from these patients induced skin blistering in neonatal mice due to suprabasal acantholysis. Serum levels of AuAbs to desmocollin 3 (Dsc3), M3 muscarinic acetylcholine receptor (M3AR), and secretory pathway Ca²⁺/Mn²⁺-ATPase isoform 1 (SPCA1) correlated with the disease stage of PV. Moreover, AuAb absorption on recombinant Dsc3, M3AR, or SPCA1 prevented both skin blistering in the passive transfer of AuAbs model of PV in BALB/c mice and significantly decreased the extent of acantholysis in a neonatal mouse skin explant model. Although acantholytic activities of each of these immunoaffinity-purified AuAbs could not induce a PV-like phenotype in vivo, their mixture produced a synergistic effect manifested by a positive Nikolsky sign in the skin of neonatal mice. The downstream signaling of all pathogenic non-Dsg AuAbs involved p38 mitogen-activated protein kinase (MAPK)-mediated phosphorylation and elevation of cytochrome c release and caspase 9 activity. Anti-Dsc3 and anti-SPCA1 AuAbs also activated SRC proto-oncogene, non-receptor tyrosine kinase (SRC). Of note, although a constellation of non-Dsg AuAbs apparently disrupted epidermal integrity, elimination of a single pathogenic AuAb could prevent keratinocyte detachment and blistering. Therefore, anti-Dsg1/3 AuAb–free PV can be a model for elucidating the roles of non-Dsg antigen–specific AuAbs in the physiological regulation of keratinocyte cell–cell adhesion and blister development.
Article
Full-text available
Pemphigus vulgaris (PV) is a potentially life-threatening mucocutaneous autoimmune blistering disease. Patients develop non-healing erosions and blisters due to cell–cell detachment of keratinocytes (acantholysis), with subsequent suprabasal intraepidermal splitting. Identified almost 30 years ago, desmoglein-3 (Dsg3), a Ca²⁺-dependent cell adhesion molecule belonging to the cadherin family, has been considered the “primary” autoantigen in PV. Proteomic studies have identified numerous autoantibodies in patients with PV that have known roles in the physiology and cell adhesion of keratinocytes. Antibodies to these autoantibodies include desmocollins 1 and 3, several muscarinic and nicotinic acetylcholine receptor subtypes, mitochondrial proteins, human leukocyte antigen molecules, thyroid peroxidase, and hSPCA1—the Ca²⁺/Mn²⁺-ATPase encoded by ATP2C1, which is mutated in Hailey–Hailey disease. Several studies have identified direct pathogenic roles of these proteins, or synergistic roles when combined with Dsg3. We review the role of these direct and indirect mechanisms of non-desmoglein autoantibodies in the pathogenesis of PV.
Article
Full-text available
Pemphigus is a blistering disease characterized by pemphigus autoantibodies (PVIgG) directed mostly against desmogleins (Dsgs), resulting in the loss of keratinocyte adhesion (acantholysis). Yet, the mechanisms underlying blister formation remain to be clarified. We have shown previously that anti-Fas ligand (FasL) antibody (Ab) prevents PVIgG-induced caspase-8 activation and Dsg cleavage in human keratinocytes, and that sera from pemphigus patients contain abnormally increased levels of FasL. Here, we demonstrate that recombinant FasL induces the activation of caspases prior to Dsg degradation, and anti-FasL Ab prevents acantholysis in cultured keratinocytes. Moreover, the silencing of FasL reduces PVIgG-induced caspase-8 activation and Dsg3 cleavage. Following injection of PVIgG into mice, FasL is upregulated at 1–3 h and is followed by caspase-8-mediated keratinocyte apoptosis, before blister formation. The administration of anti-FasL Ab after PVIgG injection blocks blister formation in mice. Furthermore, we injected PVIgG into two different gene-targeted mutant mice that selectively lack either secreted soluble FasL (sFasL), FasLΔs/Δs mice, or the membrane-bound form of FasL (mFasL), FasLΔm/Δm mice. After PVIgG treatment, blisters are only visible in FasLΔm/Δm animals, lacking mFasL, but still producing sFasL, similar to wild-type (C57BL/6) animals. By contrast, a significant decrease in the relative acantholytic area is observed in the FasLΔs/Δs animals. These results demonstrate that soluble FasL plays a crucial role in the mechanisms of blister formation, and blockade of FasL could be an effective therapeutic approach for pemphigus.
Article
Full-text available
Over the past decade, the Nomenclature Committee on Cell Death (NCCD) has formulated guidelines for the definition and interpretation of cell death from morphological, biochemical, and functional perspectives. Since the field continues to expand and novel mechanisms that orchestrate multiple cell death pathways are unveiled, we propose an updated classification of cell death subroutines focusing on mechanistic and essential (as opposed to correlative and dispensable) aspects of the process. As we provide molecularly oriented definitions of terms including intrinsic apoptosis, extrinsic apoptosis, mitochondrial permeability transition (MPT)-driven necrosis, necroptosis, ferroptosis, pyroptosis, parthanatos, entotic cell death, NETotic cell death, lysosome-dependent cell death, autophagy-dependent cell death, immunogenic cell death, cellular senescence, and mitotic catastrophe, we discuss the utility of neologisms that refer to highly specialized instances of these processes. The mission of the NCCD is to provide a widely accepted nomenclature on cell death in support of the continued development of the field.
Article
Maintaining mitochondrial function and dynamics is crucial for cellular health. In muscle, defects in mitochondria result in severe myopathies where accumulation of damaged mitochondria causes deterioration and dysfunction. Importantly, understanding the role of mitochondria in disease is a necessity to determine future therapeutics. One of the most common myopathies is mitochondrial encephalopathy lactic acidosis stroke-like episodes (MELAS), which has no current treatment. Recently, MELAS patients treated with rapamycin exhibited improved clinical outcomes. However, the cellular mechanisms of rapamycin effects in MELAS patients are currently unknown. In this study, we used cultured skin fibroblasts as a window into the mitochondrial dysfunction evident in MELAS cells, as well as to study the mechanisms of rapamycin action, compared to control, healthy individuals. We observed that mitochondria from patients were fragmented, had a 3-fold decline in the average speed of motility, a 2-fold reduced mitochondrial membrane potential and a 1.5-2-fold decline in basal respiration. Despite the reduction in mitochondrial function, mitochondrial import protein Tim23 was elevated in patient cell lines. MELAS fibroblasts exhibited increased MnSOD levels and lysosomal function when compared to healthy controls. Treatment of MELAS fibroblasts with rapamycin for 24 hrs resulted in increased mitochondrial respiration compared to control cells, a higher lysosome content, and a greater localization of mitochondria to lysosomes. Our studies suggest that rapamycin has the potential to improve cellular health even in the presence of mtDNA defects, primarily via an increase in lysosomal content.
Article
Pemphigus vulgaris (PV) is a potentially lethal mucocutaneous blistering disease characterized by IgG autoantibodies (AuAbs) binding to epidermal keratinocytes and inducing a devastating blistering disease affecting oral and/or esophageal surfaces and, sometimes, also the skin. Anti-keratinocyte AuAbs developed by the desmoglein (Dsg) 1/3 AuAb-negative acute PV patients are pathogenic, as they induced acantholysis and epidermal split in the experimental models of PV in vitro and in vivo. These PV patients have various combinations of AuAbs to keratinocyte muscarinic acetylcholine receptor subtype M3 (M3AR), the secretory pathway Ca2+/Mn2+-ATPase isoform 1 (SPCA1), and desmocollin 3 whose relative concentrations correlate with the disease activity. In this study, we identified new molecular mechanisms of the synergistic cooperation of AuAbs to M3AR and SPCA1 in inducing acantholysis in the anti-Dsg 1/3 AuAb-negative PV patients. Anti-M3AR AuAb was found to play an important role in determining the level of intraepidermal split just above the basal cells, caspase to mediate early pro-apoptotic events triggered by anti-SPCA1 AuAb, and the neonatal Fc receptor (FcRn) to contribute to the pathobiological actions of both anti-M3AR and anti-SPCA1 AuAbs. Altogether, these novel results support our original hypothesis that pemphigus acantholysis is a complex disease process (also known as apoptolysis) initiated by AuAbs directed against different keratinocyte proteins that play important roles in supporting cell viability and regulating vital cell functions.
Article
Background Pemphigus vulgaris (PV) is a life‐long IgG autoantibody‐mediated blistering disease affecting the mucosal surfaces lined by the stratified epithelium (oral, nasal, genital) and sometimes also the skin. While corticosteroid treatment is life saving, the high dose and prolonged courses required for disease control are associated with significant adverse effects, including death. Although introduction of rituximab (RTX) provided for a favorable outcome, the high relapse rate, that is, up to 80%, precludes successful use of RTX as a monotherapy. Intravenous immunoglobulin (IVIg) is being increasingly utilized as off‐label therapy for a variety of autoimmune and inflammatory diseases, including PV and pemphigus foliaceus (PF). Aims The goal of pemphigus research is to develop an effective treatment modality that would allow patients to achieve and maintain a stable clinical remission without the need for additional treatments, or cure. Materials and Methods This article summarizes clinical outcome of 123 pemphigus patients treated with a combination of IVIg, an immunosuppressive cytotoxic drug (ICD) and mitochondrion‐protecting drugs in the Blistering Disease Clinic at the University of California, Irvine from 2007 to 2017. Results The mean time to disease control was 0.2 months and time to complete remission – 1.7 months. Duration of complete remission on drugs until relapse or end of treatment was 19.3 months. The mean duration of complete remission off drugs until relapse was 15.8 months. That until end of follow up was 48.4 months, with a minimum of 14 and a maximum of 91 months. The overall complete remission rate off all drugs was 100%, with 12% overall relapse rate. Most relapses, 8.1 vs. 3.3%, occurred during the time of treatment, compared to posttreatment. No patients had more than a single relapse. The duration of the posttreatment follow‐up ranged from 9 to 97 months with a mean of 64.8 months, or 5.4 years. The total number of IVIg cycles ranged from 26 in patients without a relapse to 37 in patients with a relapse. The clinical outcome in patients that received IVIg with RTX or another ICD were found to be very similar. Discussion Thus, the multidrug IVIg regimen allowed to achieve three principal treatment objectives: (i) rapid control of pemphigus symptoms; (ii) stable disease remission; and (iii) overall safety of treatment. Conclusions While the individualized therapeutic approaches to eradicate the autoreactive B cell clones causing disease in each particular PV or PF patient are being developed, all pemphigus patients can benefit from the treatment protocol described in this study.