You are on page 1of 28

1

Abstract

2
Table of Contents
Abstract ........................................................................................................................................................ 2
1. Introduction ............................................................................................................................................. 5
1.1 Etiology and Pathogenesis ............................................................................................................ 5
1.2 Symptoms and treatment ............................................................................................................... 7
1.3 Parkinson’s disease stages ........................................................................................................... 8
1.3.1 Clinical stages ........................................................................................................................... 8
1.3.2 Pathological stages .................................................................................................................. 8
1.4 Mechanisms of neuronal death ..................................................................................................... 9
1.5. Role of the adaptive immune system in the onset of Parkinson’s disease ....................... 11
1.5.1 Genetic and environmental risk factors.............................................................................. 11
1.5.2 Clinical and postmortem evidence ...................................................................................... 11
1.6 Submandibular gland as an accessible organ to study the pathogenesis of Parkinson’s
disease.................................................................................................................................................... 13
1.7 Digital pathology and artificial intelligence............................................................................... 14
2. Objectives .............................................................................................................................................. 14
3. Materials and methods ........................................................................................................................ 15
3.1 Human post-mortem brain and submandibular gland tissue ................................................ 15
3.2 Immunohistochemistry ................................................................................................................. 15
3.3 Algorithm training (deep learning artificial intelligence) ........................................................ 16
3.4 Quantitative analysis ..................................................................................................................... 17
3.4.1 Neuromelanin-containing dopaminergic neurons density in the substantia Nigra
pars compacta ................................................................................................................................... 17
3.4.2 CD8+ T cells density in the substantia nigra pars compacta ......................................... 17
3.4.3 Adipose tissue density in the submandibular gland ........................................................ 17
3.4.4 Tyrosine Hydroxylase-positive fibers density in the submandibular gland ................ 17
3.4.5 CD8+ T cell density in the submandibular gland .............................................................. 17
3.5 Statistical analysis ......................................................................................................................... 18
4. Results ................................................................................................................................................... 18
4.1 Algorithm development................................................................................................................. 18
4.2 Quantification of the neuromelanin-containing dopaminergic neurons density in the
SNpc using OlyVIA algorithm............................................................................................................. 19
5. Discussion ............................................................................................................................................. 20
6. Conclusions .......................................................................................................................................... 20
References ................................................................................................................................................. 21

3
4
Parkinson’s Disease

1. Introduction

1.1 Etiology and Pathogenesis


Parkinson’s disease (PD) is a progressive neuronal disorder that primarily affects the elderly
population and is known as the second most frequent form of neurodegenerative disease
after Alzheimer’s disease (1). Although it is widely accepted that PD is mainly caused by the
loss of dopaminergic (DA) neurons of the substantia nigra pars compacta (SNpc) in the
midbrain, its etiology remains unknown(2). DA neurons accumulate a brownish pigment
called neuromelanin (NM) that allows seeing the loss of these neurons macroscopically in
postmortem brain tissue of individuals with PD (3). NM accumulation increases with age and
subsequently makes the individual more susceptible to neuronal loss ((4,5). Other vulnerable
brain regions in PD include locus coeruleus (LC), nucleus basalis of Meynert (NBM), dorsal
motor nucleus of the vagus (DMV), raphe nuclei (RN), pedunculopontine nucleus (PPN),
ventral tegmental area (VTA) and the olfactory bulb (Ob) (6,7).

One of the neuropathological hallmarks of PD is the presence of intraneuronal α-synuclein


aggregates in the form of Lewy bodies (LB) and Lewy neurites (LN) in the SNpc (8). α-
synuclein is a 140 amino-acidic protein encoded by a gene called SNCA, located on
chromosome 4 (4q21) which is mainly expressed the presynaptic nerve terminals in the CNS,
close to the synaptic vesicles (9–12). It seems to be involved in neurotransmitter release and
vesicle trafficking (9). Normally in neurons, there is a balance between the cytosolic (unfolded
monomers) and the membrane-bound (helical multimeric structure) states (13,14). On the
contrary, the α-synuclein monomers conformationally can turn into β-sheet and crossed-b
structures, forming amyloid-like fibrils that finally appear as pathologic Lewy inclusions and
neurites (13,14). While spindle-shaped Lewy neurites appear in the neuronal branches, the
eosinophilic Lewy bodies are mostly detected in the cytoplasm. These amyloid-like
characteristics prevent their degradation via autophagy or other endogenous cellular removal
mechanisms (14,15). Regarding this, LNs seem to be the first detectable marks of
synucleinopathy (14).

5
Up to now, the exact role of these α-synuclein aggregates, whether they appear as a
neuroprotective response to a pathological threat or they actually contribute to
neurodegeneration by disturbing the normal cell functioning, is unclear (15). Moreover, there
are many different posttranslational modifications (PTMs) that might be associated with α-
synuclein toxicity such as α-synuclein phosphorylation, glycation, glycosylation,
glutathionylation acetylation, ubiquitination, nitration, SUMOylation and truncation (16,17).

Over the course of PD progression, in addition to brain regions like SNpc, LC, hypothalamus
and subnuclei of the thalamus, amygdala and Ob, α-synucleinopathy can affect the peripheral
nervous system (PNS), enteric nervous system (ENS), and the autonomic nervous system
(ANS) including the vagus nerve, colon, paraspinal sympathetic ganglia, skin, submandibular
gland (SG) and lower esophagus. It can also be found in biological fluids of PD patients
including their serum, saliva and cerebrospinal fluid (CSF) (18–23)The accumulation of α-
synuclein has been shown to somehow be associated with DA neurodegeneration in PD,
which seems to precede the appearance of the cardinal motor symptoms by 7-10 years (20).
Moreover, the SG and lower esophagus reportedly contain higher densities of α-synuclein
aggregation among other PNS sites. A rostro-caudal gradient of α -synucleinopathy in the
gastrointestinal tract (GI) has also been detected (24,25). However, there is no consensus
on whether this synucleinopathy first appears in the brain or within the organs innervated by
the PNS.

Even though the cause of DA cell loss is still unexplained, it seems that having a genetic
background increases the chances of PD manifestation later in life (26).The idiopathic form
of PD occurs as a result of both genetic and environmental risk factors and comprises the
majority of PD cases (1). However, low percentage of PD cases are familial, which has been
found to be related to monogenic mutations such as SNCA, LRRK2, VPS35, PINK1, Parkin
and DJ-1 (27–29). Reportedly, GBA genetic mutations are one of the genetic risk factors for
PD development (Do et al., 2019). It is well known that mutations in SNCA were the first ones
to be linked to familial PD (12). It seems that in addition to point mutations in the SNCA gene
that encodes -synuclein, there are duplications and triplications of this gene that might be
associated with the age of onset and severity of the disease in familial PD (30). Point
mutations in α-synuclein including A53T, A30P, and E46K can somehow lead to an enhanced
expression of α-synuclein which is related to a higher risk for sporadic PD (31).

6
Among all the environmental risk factors, age seems to be the most important one in
idiopathic PD. For about one fourth of PD patients the age of onset is younger than 65 years
old and for 5%-10% of them, it is before 50 years old (32). Moreover, the frequency seems to
be higher in men with a ratio from 1.3 to 2 (9). Among the environmental risk factors, exposure
to pesticides, head injury, cancer, methamphetamine and consumption of dairy products are
known to be associated with an increased risk of PD development whereas tobacco and
nicotine consumption, coffee intake, antioxidants like urate, physical activity, and non-
steroidal anti-inflammatory drugs (NSAIDS) are suggested as protective factors (27,33). In
addition to autoimmune diseases, a link between PD and various infections including
measles, hepatitis C, influenza, Helicobacter pylori and infections of the CNS have been
reported (33).

1.2 Symptoms and treatment


PD patients suffer from an excessive reduced quality of life. DA depletion is the underlying
cause of the cardinal motor symptoms observed in PD patients including resting tremors,
postural and gait instability, rigidity and bradykinesia (9,19,34). The degeneration of DA
neurons of the SNpc whose axonal projections are part of the nigrostriatal pathway from the
SNpc to the putamen and caudate nuclei (known as striatum) of the basal ganglia, an
important area for muscle control and movement, leads to motor dysfunction (3,35–37).

The prodromal phase which occurs up to 15-20 years before the onset of motor dysfunction,
is accompanied by manifestations such as olfactory dysfunction (hyposmia and anosmia),
rapid eye movement sleep disorder (RBD), depression, gastrointestinal problems especially
constipation and autonomic dysfunction (9,19). Several other non-motor symptoms including
psychosis, hypersialorrhea or drooling, pain, cognitive impairment and dementia might
appear along with the motor symptoms (9,19,38). Furthermore, about 80% of PD patients
experience dysphagia (i.e. having difficulty while swallowing food) and hypersialorrhea as a
result of hyposalivation (39–41).

Up to date, the best drug treatment for PD patients is Levodopa (L-3,4-


dihydroxyphenylalanine; L-DOPA), a dopamine precursor that unlike dopamine itself, can
pass through the blood-brain barrier (BBB) (37,42). However, there are patients who develop
resistance to L-DOPA after a period of time (42). Several other complications such as
dyskinesia, psychosis and fluctuations can occur as a result of chronic dopaminergic

7
treatment (9,19). Administration of anticholinergics, monoamine oxidase inhibitors (MAOIs),
dopamine agonists or less commonly, amantadine has also been suggested (9,19). Other
types of surgical and medical interventions include deep brain stimulation, gene therapy and
cell transplantation (19,43–46). A PD diagnosis is mainly based on the appearance of the
cardinal motor symptoms (47), and it is further confirmed by a positive symptomatic response
to the administration of L-DOPA (24,26).

1.3 Parkinson’s disease stages


1.3.1 Clinical stages
The motor and non-motor features of PD led to the introduction of clinical staging systems
that help clinicians have a classification according to the severity and symptoms manifested
in each patient. The standard staging reference for PD is the Hoehn and Yahr (HY) scale,
even though it has limitations of mixing impairment and disability, nonlinear character
between the stages and its tendency towards postural instability. HY correlates with
neuroimaging studies of DA cell loss (48). Unified Parkinson’s disease Rating Scale (UPDRS)
is another staging system that exclusively evaluates the main clinical symptoms of PD and
monitors the severity of the disease (49). Later on, the modified UPDRS by Movement
Disorder Society Task Force on Rating Scales (MDS-UPDRS) was introduced which includes
the non-motor symptoms in addition to the previous ones and shows a higher sensitivity in
PD diagnosis (48,50).

1.3.2 Pathological stages


The prodromal phase of PD can be explained according to the neuropathological events
involved. Braak was the first person who suggested a staging system for PD depending on
when the synucleinopathy first emerges in different regions of the brain (51). He defined this
staging system by including cases with synucleinopathy that displayed symptoms and cases
who lacked them. The following classification was the result including 6 stages: stage 1,
medulla oblongata; stage 2, medulla oblongata and pontine tegmentum; stage 3, midbrain
particularly SNpc and the basal nucleus of Meynert; stage 4, basal prosencephalon and
mesocortex; stage 5, neocortex and prefrontal neocortex; stage 6, premotor and sensory
association areas of the neocortex. Based on Braak’s staging system, synucleinopathy in
brainstem was a representation of the early stages of PD (51,52). Braak was also a pioneer
in including the Ob as one of the earliest areas affected by synucleinopathy in PD (51). He

8
suggested the vagus nerve as another initially affected area by synucleinopathy which
remains dubious because reportedly, patients who lack -synuclein aggregation in the brain
do not seem to show synucleinopathy in the vagus nerve (53).

Subsequently, the Unified Staging System for Lewy Body Disorders (USSLBD) was proposed
by Beach and his colleagues to improve the Braak’s staging system which can be applied to
all the Lewy body disorders (PD, DLB iLBD and AD) . USSLBD suggests the next model
which is determined by the α-synucleinopathy density score in the examined brain regions: I.
Olfactory bulb only; IIa. Brainstem Predominant; IIb. Brainstem Predominant; III. Brainstem
and limbic; IV. Neocortical (23). According to USSLBD, the presence of -synuclein
aggregates in the Ob increases the chances of developing Lewy body disorders later in life
(23). Regarding that, clinical observations and postmortem evidence coincide in the olfactory
problems being an initial sign of the disease (23). These classifications help us better
understand the prodromal stages of PD, known as incidental Lewy body disease (iLBD) cases
who probably were in the initial phase and were not clinically diagnosed before passing away
(23,54). Tyrosine Hydroxylase (TH) levels, an essential enzyme in dopamine biosynthesis
(55), have been found to be lower in the iLBD cases than in PD patients, which is a proof of
dissimilarity between them and healthy controls (56,57). Moreover, it has been reported that
during the first two stages of Braak’s staging system when no trace of -synuclein
aggregations is apparent yet, there are lower numbers of TH-containing neurons as well as
more neuronal loss in the SNpc (58), therefore suggesting that neuronal loss probably
predates synucleinopathy.

1.4 Mechanisms of neuronal death


The main reason why DA neurons are pushed toward death in PD is not known yet. Different
cell death mechanisms including NM accumulation, oxidative stress and mitochondrial
dysfunction, -synuclein aggregation, lysosomal dysfunction and innate and adaptive
immune response have been suggested to be associated with this neuronal loss, which either
triggers the initiation of apoptosis or promotes necrosis (5,9). Understanding these
pathophysiological mechanisms will help us both in disease prevention and treatment.
However, there is no universal consensus on whether these mechanisms independently or
together are provoking the detrimental events in the brain (9).

9
By using an animal model overexpressing NM, an association between neuronal loss and
age-related NM production in the DA neurons of the SNpc has been found. This has led to
the definition of a threshold for NM accumulation inside these cells. Interestingly, while it
seems that in PD patients and iLBD cases the amount of intracellular NM might cross this
threshold, in healthy controls it does not reach that. This points out to the possible role of the
age-dependent accumulating NM inside the DA neurons of the SNpc as an element that
makes them more vulnerable to degeneration (5).

Oxidative stress, as a result of an unbalanced production of Reactive oxygen species (ROS)


whose accumulation can cause oxidative damage to proteins, DNA and membrane lipids, can
be a cause of deregulated activity of complex I (3,9,59). Mitochondrial dysfunction causes a
chronic cycle of ROS production (60). It has been shown that the impairment of the complex
I might increase the chances of neurodegeneration by making the DA neurons more
susceptible to neurotoxin exposure (60–62). The presence of a defective complex I as a result
of mitochondrial dysfunction in the SNpc of PD patients has been reported (63). Inhibition of
complex I can reversely increase the production of ROS, especially in the DA neurons of the
SNpc that seems to be more sensitive to this mechanism due to auto-oxidation of NM and
dopamine catabolism (3,7).

Abnormal -synuclein misfolding has been reported to be toxic for DA neurons which can be
one of the underlying causes of neuronal loss in PD (9). Oxidative stress and gene mutations
in PD seem to affect these conformational changes of -synuclein (7). Moreover,
mitochondrial damage has been found to be a cause of reduced glucocerebrosidase and
increased accumulation of oxidized dopamine products (9,64). Since recycling the -
synuclein aggregates is primariliy dependent on the function of lysosomes, lysosomal
dysfunction is another factor that can contribute to neuronal death by decreasing the turnover
of these aggregates and therefore, increasing their intracellular accumulation (65). On the
other hand, the adaptive immune response, either to endogenous antigens (aforementioned
mechanisms) hence leading to autoimmunity or to exogenous antigens such as viral particles
(9,53).

10
1.5. Role of the adaptive immune system in the onset of Parkinson’s
disease
1.5.1 Genetic and environmental risk factors
There are various genetic and environmental risk factors involved in PD pathogenesis that
might also contribute to this altered immune response (32). In familial PD, mutations in genes
such as SNCA, LRRK2, DJ1 and PINK1, which seem to play an important role in regulation
and homeostasis of the immune response (especially T cells), can also contribute to PD
development (66). Environmental risk factors including exposure to chemicals, infectious
organisms (e.g. bacteria or viruses) and more importantly, age-related physiological changes
of the immune system, immunosenescence and inflammaging can provide us with an
explanation for the underlying cause of this altered immune response (32,67,68).

Accumulating evidence supports the involvement of the adaptive immune system in the
etiopathogenesis of PD (69). Genome-wide association studies (GWAS) analyses have
shown a clear association between PD and other autoimmune disorders due to common
genetic risk factors, which sheds light on the possible role of autoimmunity in PD
pathogenesis (70,71). Moreover, various single nucleotide polymorphisms (SNPs) such as
SNPs of some genes related to the lymphocyte function, T cell receptor (TCR) signaling and
regulatory variants in the HLA region were identified as the possible genetic risk factors (72–
75). Additionally, various genetic modifications in MHCII and LRRK2 have been shown to
mutually be associated with an increased immune response and a higher risk of PD
development (71,76).

1.5.2 Clinical and postmortem evidence


There is increasing evidence supporting the crucial role of the adaptive immune system,
specifically CD8+ T lymphocytes, in PD pathogenesis (2). Increased infiltration of T cells into
the brain parenchyma of PD patients, especially during the early phases of the disease in the
absence of nigral synucleinopathy has been reported (2,77). This sheds light on the role of
the immune system not only in the disease initiation but also in its progression.

Brochard et al. (2009) described the CD4+ T cells as the responsible components of the
adaptive immune system in MPTP animal models and PD patients, even when the CD8+ T
cell infiltration preponderated (78). The results of a recent study done by our team clearly
demonstrate the importance of the nigral cytotoxic attack of the CD8+ T cells after infiltration,

11
which precedes the α-synuclein aggregation and neurodegeneration during the premotor
phases (2). We assessed both CD4+ and CD8+ T cell densities in the parenchyma and the
perivascular space of PD and iLBD cases. We could only find the infiltration of CD8+ T cells
in the parenchyma of PD patients and perivascular CD4+ T cells. A positive correlation
between neurodegeneration and CD8+ T cell densities was observed whereas no such thing
was seen with CD4+ T cells (2). Our findings demonstrate that the CD8+ T cells were in close
proximity with the NM-containing neurons (2). Accordingly, this cytotoxic attack could be
triggering the initiation of this deadly cascade of neurodegeneration as well as its progression
(2). These results can explain the role of the adaptive immune system in the PD
pathogenesis. During the first stage, which happens almost concomitantly with the first stage
of Braak’s staging system, there is a large number of CD8+ T cells infiltrating the SNpc. Then,
following a sudden reduction in the infiltrating cells, another wave of infiltration occurs which
also is accompanied with the appearance of Lewy bodies, Lewy neurites and DA
neurodegeneration (2). This somehow implies that CD8+ T cells are not cytotoxic at the
beginning (2). Nonetheless, whether this α-synuclein accumulation acts as a neuroprotective
shield or has a detrimental role in PD etiopathogenesis remains enigmatic (2).

Moreover, microglial hyperactivation in the SNpc is considered as one of the pathological


characteristics of PD (31), in relation to which, animal model studies have reported an
abundance of reactive human leukocyte antigen-DR (HLA-DR)–positive microglial cells in
that region (79). Although the process of neuroinflammation in PD is not clear yet, there is an
increased expression of proinflammatory cytokine levels such as TNF-α, IL-1β, and IFN-γ by
the microglia in the striatum of the postmortem brain and CSF compared with age-matched
controls (31,80).

The clinical symptoms present in PD patients together with the presence of synucleinopathy
in the periphery brought up the hypothesis that a neurotropic pathogen, probably a viral agent,
might enter the brain either through the nasal cavity or after ingesting the nasal secretions in
saliva (81). Having in mind both exposure to a neurotropic pathogen and the self-antigens of
the individual that might be triggering an autoimmune response, these results support the
hypothesis that there might have be an infection or an autoimmune response that leads to
the following deleterious events.

In PD and other neurodegenerative diseases in general, having samples from the brain tissue
would be the ideal condition in order to study the etiology of the disease. However, since that

12
is not possible until we have access to the postmortem tissue, finding adequate substitutes
would be really helpful. This is why we are interested in studying the possible association
between the immunological responses, mainly CD8+ T cell infiltration, and α-synucleinopathy
in the SG which may be mirroring the immunologic events that occur in the brain. Moreover,
this might help us in the future to design inidividualized therapies for each patient knowing
the little differences that exist during the early stages of PD.

1.6 Submandibular gland as an accessible organ to study the


pathogenesis of Parkinson’s disease
The submandibular gland (SG) is a seromucous salivary gland that is located subcutaneously
in the posterior part of the submandibular triangle, under the mandible which is known as the
second largest salivary gland after the parotid gland (82,83). The paired SGs are responsible
for 70% of saliva production in unstimulated conditions (82).

Histologically, the SG is made up of parenchymal (secretory acini and ducts) and stromal
components (84). Moreover, the SG is divided into superficial and deep lobes by the
mylohyoid muscle, with the superficial part lying beneath the deep cervical fascia that forms
a capsule of connective tissue wrapping around the gland (82). This division further arranges
a series of lobules inside the gland that contain the ducts and provide the blood supply and
innervation of the tissue (84). There are serous and mucous acini with serous cells being
predominantly distributed throughout the SG tissue (83). The produced saliva is excreted
through the intercalated, striated and excretory ducts towards the Wharton duct which opens
into the oral cavity (82,84).

The parasympathetic (cholinergic) post ganglionic fibers innervate the SG coming from the
submandibular ganglion, while the sympathetic (adrenergic) innervation is extended through
the branches of the external carotid artery, originating from the superior cervical ganglion
(82,85). The parasympathetic nerves mainly stimulate the serous end pieces that are
distributed predominantly in the parenchyma, whereas the sympathetic ones are mostly
involved in protein secretion (82,83).

Beach et al. (2016) confirmed the possible detection of Lewy pathology in the SG by showing
the presence of α-synuclein aggregations in 91% of PD subjects and 11% of iLBD subjects
(18). This suggests that in a high percentage of PD cases show α-synucleinopathy in this
gland. It seems that Lewy pathology initially affects the peripheral tissues of the ANS, even

13
before the involvement of the SNpc (86). Moreover, it has been reported that α-
synucleinopathy in the SG exists concomitantly with its presence in the brain (18). During the
advanced stages of disease, the SG showed higher sensitivity and specificity in α-synuclein
detection compared with colon or skin (21). Regarding this, the SG has been suggested as a
good biopsy site to look for synucleinopathy. While it is an accessible and safe-to-be-biopsied
organ, some authors believe that it increases the possibility of having a more accurate
diagnosis while the individual is still alive (18).

1.7 Digital pathology and artificial intelligence


Digital pathology is an almost recent phenomenon in pathology laboratory, which aids the
pathologists and pathology researchers to not only obtain full-coverage scans of the sections
but also have access to this more detailed information anytime they need it (87). One of the
important characteristics of this digital tool is that in addition to giving us high-quality digitized
images, it encompasses both machine learning and deep learning, both of which help us
create algorithms that help us analyze the data we have on the sections (88,89).

An open access software such as QuPath and a closed-access one like OlyVIA are great
examples that incorporate useful tools for annotating and viewing in both bright-field and
fluorescence images. They make it possible to train algorithms that are beneficial for a great
variety of tasks such as quantitative analyses and cell counting. More features include cell
segmentation, tissue microarray dearraying, batch processing and stain estimation and cell
detection tools (68,89). This highly potential technology that we use in this study, accelerates
the process of sample evaluation and also increases the quantitative accuracy by decreasing
the risk of bias (89,90).

2. Objectives
We hypothesize that, there is a CD8+ T cell infiltration in PNS regions affected by
synucleinopathy such as the submandibular gland in PD patients, which supposedly is
mirroring the pathogenic events observed in the brain. We hypothesize that this infiltration
precedes the α-synuclein aggregation just as it does in the SNpc. Moreover, we advance the
hypothesis that there is a denervation of TH+ fibers in the SG. Therefore, we propose the SG
as an organ for studying the etiopathogenesis of PD.

14
The main goal of this study is to determine whether there are similar pathological events
occurring in the SG and the SNpc regarding the CD8+ T cell infiltration in FFPE postmortem
tissue sections. To accomplish this main goal, we define the following specific objectives:

1. To train two deep learning algorithms in OlyVIA software to automatically count the
CD8+ T cells and to assess the TH+ fibers in human postmortem SG tissue sections.
2. To train an algorithm in OlyVIA to quantify the NM-containing neurons in human
postmortem SNpc tissue sections and thus, assess the DA neuronal density in Healthy
controls, iLBD1, iLBD2 and PD groups.
3. To ascertain whether there is an increase of CD8+ T cell infiltration in the SG of PD
patients and if so, whether it precedes the synucleinopathy.
4. To determine if there is neuronal loss in the SG of PD patients.
5. To determine whether there is a positive correlation between the CD8+ T cell densities
in the SG and SNpc.

3. Materials and methods

3.1 Human post-mortem brain and submandibular gland tissue


Formalin-Fixed Paraffin-Embedded (FFPE) SNpc and SG sections (thickness 5 µm) from
deceased PD patients with or without synucleinopathy in the SNpc (n= 34) and SG (n= 24)
and age-matched healthy control individuals were provided by Banner Sun Health Research
Institute (BSHRI), Sun City, Arizona. The Brain and Body Donation Program operates with
the approval of Western Institutional Review Board (Puyallup, WA). Cases were selected
from individuals of both sexes, who had volunteered for the BSHRI Brain and Body Donation
Program (BBDP), and who had full autopsy reports, clinical history and neuropathological
information, including the SG. Two cases were obtained from the Neurological Tissue
BioBank at IDIBAPS-Hospital Clinic (Barcelona). For each case, three sections were used.

3.2 Immunohistochemistry
Briefly, tissues were deparaffinized at 60°C for 30 min. Sections were then rehydrated in two
solutions of xylene (5 min each) and then transferred to 100%, 95% ethanol (10 min each),
70% ethanol and distilled water (5 min each), respectively. Then sections were rinsed in 0.1
M Tris buffered saline (1x TBS) 100 mM pH 7.4 (Ref. S3014; Trizma base, Sigma-Aldrich

15
Ref. T6066; NaCl, Sigma-Aldrich) between each incubation period. An antigen retrieval
protocol was performed using sodium citrate (10 mM; pH 6) in a water bath at 95°C for 20
min to unmask the epitopes. Afterwards, the slides were immersed in 10% methanol (vol/vol)
and 3% hydrogen peroxide (vol/vol) for 10 min to inhibit the endogenous peroxidase activity.
A permeabilization step was performed by washing the sections in 1x TBS + 0.3% Triton TM X-
100 two times (5 min each) and Normal Goat serum (Ref. S-1000-20; NGS, Vector) was used
to block for 1h at room temperature. An additional incubation with Avidin/Biotin blocking kit
was also performed on the SG sections (Ref. SP-2001; Vector). Sections were then incubated
overnight with primary antibodies at 4°: rat anti-human CD8α (Supernatant; Ref. NOR132;
CNIO) and rabbit anti-human tyrosine hydroxylase (1:1000; Ref. 657012; Sigma-aldrich). The
following day, appropriate biotinylated secondary antibodies were applied to the sections for
a 1-hour incubation at room temperature. ABC kit (Ref. 32050; Thermo Fischer Scientific),
Vector SG (Ref. SK-4700; Vector), DAB (Ref. D3939-1SET; Sigma) were then applied to
sections to detect the antigen-antibody reaction. The SG tissue sections were counterstained
with Hematoxylin (Ref. GHS316-500ML; Sigma-Aldrich) & Eosin (Ref. HT110116-500ML;
Sigma-Aldrich). Sections were finally dehydrated through 70% and 95% (1 min each), 100%
ethanol (2 min) and finally in two changes of xylene (5 min each). Tissue sections were then
coverslipped using DPX non-aqueous mounting medium (Ref. 1.005.790.500; Sigma-
Aldrich). All antibodies were previously tested in positive control tissue to optimize
immunohistochemistry conditions.

In order to train the NM algorithm, previously immunostained SNpc tissue sections were used
including rabbit anti-human CD4 (1:100; Ref. HPA004252; Sigma-Aldrich), rat anti-human
CD8 α (Supernatant; Ref. NOR132; CNIO), rabbit anti-human GFAP (2μg/mL; Ref.
HPA056030, Sigma-Aldrich) and mouse anti-human pS129 a-syn (1:100; Ref. 015-25191,
Wako).

3.3 Algorithm training (deep learning artificial intelligence)


Part of this project was dedicated to the development of different algorithms for the
automatized quantification of different antigens in both substantia nigra pars compacta
(SNpc) and submandibular gland (SG). In this regard, all sections were scanned with the
Slide scanner Olympus SV200 using a multi-layer image with a 20x objective scan mode with
EFI 1.5. All images were focused semi-automatically and manually corrected before saving.

16
Digitized images were then processed with Olympus SV200 desktop software for algorithm
training and further analysis.

3.4 Quantitative analysis


3.4.1 Neuromelanin-containing dopaminergic neurons density in the substantia
Nigra pars compacta
In order to evaluate the neuronal degeneration in the SNpc, the area was drawn manually in
HC (n=6), iLBD1 (n=8), iLBD2 (n=5) and PD (n=15) by using Olympus SV200 desktop
software. The NM algorithm was run in the regions of interest and the NM-containing neurons
were quantified using the count and measure tool (FIGURE). Finally, the density values were
assessed by dividing the total number of neurons by the delimitated area of SNpc (cells/mm2).

3.4.2 CD8+ T cells density in the substantia nigra pars compacta


CD8+ T cell densities were quantified using X sections per case, on the same tissue sections
used in section 3.4.1. These sections were immunohistochemically stained for CD8+ T cells
detection. Quantification was performed manually using QuPath software, differentiating
between parenchymal and perivascular CD8+ T cells (FIGURE). Afterwards, the number of
CD8+ T cells was divided by the area of the SNpc that was calculated for the DA neuron
quantification (CD8+ T cells/mm2).

3.4.3 Adipose tissue density in the submandibular gland


QuPath software was used to manually determine SG and adipose tissue areas, as fatty
infiltration is an age-associated phenomenon (91). The SG tissue sections used here were
counterstained with Hematoxylin & Eosin (H&E). In order to compare the cases, the sections
were normalized.

3.4.4 Tyrosine Hydroxylase-positive fibers density in the submandibular gland


Quantification of TH+ fibers in the SG was calculated by dividing the area occupied by the
TH+ fibers obtained with the TH algorithm by the total area of the SG (TH+ area/mm2).

3.4.5 CD8+ T cell density in the submandibular gland


The SG total areas were drawn manually using QuPath software. The density was further
measured by dividing the number of CD8+ T cells detected by the algorithm by the total area
of the SG (cell counts/mm2).

17
3.5 Statistical analysis
All statistical analyses were performed using GraphPad Prism 6 software (version 6,
GraphPad Software, USA). We analyzed the obtained data for normal distribution using the
Shapiro-Wilks test. For parametric analyses, One-way ANOVA with Tukey's post-hoc multiple
comparisons analysis was performed. For our non-parametric analysis, Kruskal-Wallis with
Dunnet post-hoc multiple comparisons analysis was performed. P < 0.05 is considered
significant. Error bars in all graphs are represented as ± standard deviation (SD). Correlations
between two numerical variables were done using the Pearson correlation test, where results
are considered significant for P value < 0.05.

4. Results

4.1 Algorithm development


One of the main goals of our study was to train an algorithm that could count our target cells,
in our case the NM-containing neurons in the SNpc, the CD8+ T cells in the SG and the TH+
fibers of the SG. Before that, this quantification was performed manually and from now on,
the possibility of using machine learning algorithms to automatically count the cells exists.

The NM algorithm was designed and trained in order to count dopaminergic neurons and
evaluate neuronal cell death. To train the algorithm, a total number of 24 SNpc sections that
were previously undergone immunohistochemical staining for CD8, CD4, GFAP and
phosphorylated-synuclein were selected (6 sections per group). DA-neurons are identified by
the presence of unstained neuromelanin brown pigment in SNpc, which are easy to
distinguish from the background without any counterstaining. Therefore, two label classes
(NM and background) were added in these sections. About 100 NM object annotations and
50 annotations marking the background were drawn manually in the regions of interest
(FIGURE).

The selection of four groups for the training helped us improve the accuracy of the algorithm
by introducing different combinations of NM-containing neurons and the background. For SG,
two algorithms were designed independently. On one hand, CD8+ T cell SG algorithm was
designed using X images with X object annotations named as CD8+ T cells and background
(FIGURE). On the other hand, for TH+ fiber quantification the TH+ algorithm was made, using
a total of X images with X object annotations drawn named as TH+ fiber and background

18
(FIGURE).. All algorithms were trained using a non-iteration limit (accepting as good when
the algorithm reached a checkpoint value of at least 0.75).

Thus, the algorithms were created at checkpoints, X (NM algorithm), X (CD8+ T cell SG
algorithm) and X (TH+ algorithm). Once the algorithms were created, they were manually
checked and if necessary adjusted. CD8+ T cell algorithm as it had a huge amount of CD8+
T cells made some mistakes when counting CD8+ T cells that were close to one another
identifying them as just one. This error was corrected by applying the “automatically split
objects” tool. Finally, each algorithm was validated by being compared with manually
counts of the same sections, to make sure that the algorithm is counting correctly.

4.2 Quantification of the neuromelanin-containing dopaminergic


neurons density in the SNpc using OlyVIA algorithm
Based on the presence of -synuclein aggregations in the brain, four groups were defined as
HC, iLBD1, iLBD2 and PD. Individuals in the iLBD1 group represent the earliest stage of PD
that lack synucleinopathy in the SNpc, whereas the iLBD2 group shows synucleinopathy in
the SNpc. After the data analysis, a significantly less density of DA neurons was observed
in the PD group (number of neurons/mm2=?) compared with that of the HC (number of
neurons/mm2=?) and the iLBD groups (number of neurons/mm2=?) (p=?). Our results
demonstrate that the loss of NM-containing neurons severely affects the SNpc during the
advanced stages of the disease. Moreover, an increased tendency towards
neurodegeneration is observed as the disease proceeds (FIGURE X).

19
5. Discussion

6. Conclusions

20
References

1. Bartels AL, Leenders KL. Parkinson’s disease: The syndrome, the pathogenesis and pathophysiology.
Cortex. 2009 Sep 1;45(8):915–21.
2. Galiano-Landeira J, Torra A, Vila M, Bové J. CD8 T cell nigral infiltration precedes synucleinopathy in
early stages of Parkinson’s disease. Brain [Internet]. 2020 Dec 1 [cited 2023 Jun 16];143(12):3717–33.
Available from: https://dx.doi.org/10.1093/brain/awaa269
3. Blandini F, Nappi G, Tassorelli C, Martignoni E. Functional changes of the basal ganglia circuitry in
Parkinson’s disease. Prog Neurobiol. 2000 Sep 1;62(1):63–88.
4. Carballo-Carbajal I, Laguna A, Romero-Giménez J, Cuadros T, Bové J, Martinez-Vicente M, et al. Brain
tyrosinase overexpression implicates age-dependent neuromelanin production in Parkinson’s disease
pathogenesis. Nature Communications 2019 10:1 [Internet]. 2019 Mar 7 [cited 2023 Jun 17];10(1):1–
19. Available from: https://www.nature.com/articles/s41467-019-08858-y
5. Vila M. Neuromelanin, aging, and neuronal vulnerability in Parkinson’s disease. Movement Disorders
[Internet]. 2019 Oct 1 [cited 2023 Jun 17];34(10):1440–51. Available from:
https://onlinelibrary.wiley.com/doi/full/10.1002/mds.27776
6. Sulzer D, Surmeier DJ. Neuronal vulnerability, pathogenesis and Parkinson’s disease. Mov Disord
[Internet]. 2013 Jan [cited 2023 Jun 16];28(1):41. Available from: /pmc/articles/PMC3578396/
7. Giguère N, Nanni SB, Trudeau LE. On Cell Loss and Selective Vulnerability of Neuronal Populations in
Parkinson’s Disease. Front Neurol [Internet]. 2018 Jun 19 [cited 2023 Jun 16];9(JUN):455. Available
from: /pmc/articles/PMC6018545/
8. Tansey MG, Romero-Ramos M. Immune system responses in Parkinson’s Disease: early and dynamic.
Eur J Neurosci [Internet]. 2019 Feb 1 [cited 2023 Jun 16];49(3):364. Available from:
/pmc/articles/PMC6391192/
9. Jankovic J, Tan EK. Parkinson’s disease: etiopathogenesis and treatment. J Neurol Neurosurg
Psychiatry [Internet]. 2020 [cited 2023 Jun 16];91:795–808. Available from: http://jnnp.bmj.com/
10. Wakabayashi K, Mori F, Takahashi H. Progression patterns of neuronal loss and Lewy body pathology
in the substantia nigra in Parkinson’s disease. Parkinsonism Relat Disord [Internet]. 2006 Oct 1 [cited
2023 Jun 16];12(SUPPL. 2):S92–8. Available from: http://www.prd-
journal.com/article/S1353802006001088/fulltext
11. Goedert M. Alpha-synuclein and neurodegenerative diseases. Nature Reviews Neuroscience 2001 2:7
[Internet]. 2001 Jul [cited 2023 Jun 17];2(7):492–501. Available from:
https://www.nature.com/articles/35081564
12. Mochizuki H, Choong CJ, Masliah E. A refined concept: α-synuclein dysregulation disease. Neurochem
Int. 2018 Oct 1;119:84–96.
13. Campo F, Carletti R, Fusconi M, Pellicano C, Pontieri FE, Di Gioia CR, et al. Alpha-synuclein in salivary
gland as biomarker for Parkinson’s disease. Rev Neurosci [Internet]. 2019 Jul 1 [cited 2023 Jun
17];30(5):455–62. Available from: https://www.degruyter.com/document/doi/10.1515/revneuro-
2018-0064/html

21
14. Del Tredici K, Braak H. Review: Sporadic Parkinson’s disease: development and distribution of α-
synuclein pathology. Neuropathol Appl Neurobiol [Internet]. 2016 Feb 1 [cited 2023 Jun 17];42(1):33–
50. Available from: https://onlinelibrary.wiley.com/doi/full/10.1111/nan.12298
15. Jellinger KA. Formation and development of Lewy pathology: A critical update. J Neurol [Internet].
2009 Aug 27 [cited 2023 Jun 16];256(SUPPL. 3):270–9. Available from:
https://link.springer.com/article/10.1007/s00415-009-5243-y
16. Outeiro TF, Koss DJ, Erskine D, Walker L, Kurzawa-Akanbi M, Burn D, et al. Dementia with Lewy
bodies: an update and outlook. Molecular Neurodegeneration 2019 14:1 [Internet]. 2019 Jan 21 [cited
2023 Jun 17];14(1):1–18. Available from:
https://molecularneurodegeneration.biomedcentral.com/articles/10.1186/s13024-019-0306-8
17. Manzanza N de O, Sedlackova L, Kalaria RN. Alpha-Synuclein Post-translational Modifications:
Implications for Pathogenesis of Lewy Body Disorders. Front Aging Neurosci. 2021 Jun 25;13:690293.
18. Beach TG, Adler CH, Serrano G, Sue LI, Walker DG, Dugger BN, et al. Prevalence of Submandibular
Gland Synucleinopathy in Parkinson’s Disease, Dementia with Lewy Bodies and other Lewy Body
Disorders. J Parkinsons Dis [Internet]. 2016 Mar 30 [cited 2023 Jun 16];6(1):153. Available from:
/pmc/articles/PMC5498170/
19. Kalia L V., Lang AE. Parkinson’s disease. The Lancet [Internet]. 2015 Aug 29 [cited 2023 Jun
17];386(9996):896–912. Available from:
http://www.thelancet.com/article/S0140673614613933/fulltext
20. Du T, Wang L, Liu W, Zhu G, Chen Y, Zhang J. Biomarkers and the Role of α-Synuclein in Parkinson’s
Disease. Front Aging Neurosci [Internet]. 2021 Mar 23 [cited 2023 Jun 17];13. Available from:
/pmc/articles/PMC8021696/
21. Chahine LM, Beach TG, Brumm MC, Adler CH, Coffey CS, Mosovsky S, et al. In vivo distribution of α-
synuclein in multiple tissues and biofluids in Parkinson disease. Neurology [Internet]. 2020 Sep 1
[cited 2023 Jun 17];95(9):e1267–84. Available from: https://n.neurology.org/content/95/9/e1267
22. Braak H, Rüb U, Gai WP, Del Tredici K. Idiopathic Parkinson’s disease: Possible routes by which
vulnerable neuronal types may be subject to neuroinvasion by an unknown pathogen. J Neural
Transm [Internet]. 2003 May 1 [cited 2023 Jun 17];110(5):517–36. Available from:
https://link.springer.com/article/10.1007/s00702-002-0808-2
23. Beach TG, Adler CH, Lue LF, Sue LI, Bachalakuri J, Henry-Watson J, et al. Unified Staging System for
Lewy Body Disorders: Correlation with Nigrostriatal Degeneration, Cognitive Impairment and Motor
Dysfunction. Acta Neuropathol [Internet]. 2009 [cited 2023 Jun 17];117(6):613. Available from:
/pmc/articles/PMC2757320/
24. Beach TG, Adler CH, Dugger BN, Serrano G, Hidalgo J, Henry-Watson J, et al. Submandibular Gland
Biopsy for the Diagnosis of Parkinson Disease. J Neuropathol Exp Neurol [Internet]. 2013 Feb [cited
2023 Jun 17];72(2):130. Available from: /pmc/articles/PMC3571631/
25. Breen DP, Halliday GM, Lang AE. Gut–brain axis and the spread of α-synuclein pathology: Vagal
highway or dead end? Movement Disorders [Internet]. 2019 Mar 1 [cited 2023 Jun 17];34(3):307–16.
Available from: https://onlinelibrary.wiley.com/doi/full/10.1002/mds.27556

22
26. de Lau LM, Breteler MM. Epidemiology of Parkinson’s disease. Lancet Neurol [Internet]. 2006 Jun 1
[cited 2023 Jun 17];5(6):525–35. Available from:
http://www.thelancet.com/article/S1474442206704719/fulltext
27. Bloem BR, Okun MS, Klein C. Parkinson’s disease. The Lancet [Internet]. 2021 Jun 12 [cited 2023 Jun
16];397(10291):2284–303. Available from:
http://www.thelancet.com/article/S014067362100218X/fulltext
28. Kim CY, Alcalay RN. Genetic Forms of Parkinson’s Disease. Semin Neurol [Internet]. 2017 Apr 1 [cited
2023 Jun 16];37(2):135–46. Available from: http://www.thieme-
connect.com/products/ejournals/html/10.1055/s-0037-1601567
29. Day JO, Mullin S. The Genetics of Parkinson’s Disease and Implications for Clinical Practice. Genes
(Basel) [Internet]. 2021 Jul 1 [cited 2023 Jun 17];12(7). Available from: /pmc/articles/PMC8304082/
30. Nussbaum RL. Genetics of Synucleinopathies. Cold Spring Harb Perspect Med. 2018 Jun
1;8(6):a024109.
31. Hartmann A. Postmortem studies in Parkinson’s disease. Dialogues Clin Neurosci [Internet]. 2004
[cited 2023 Jun 17];6(3):281. Available from: /pmc/articles/PMC3181805/
32. Kline EM, Houser MC, Herrick MK, Seibler P, Klein C, West A, et al. Genetic and environmental factors
in Parkinson’s converge on immune function and inflammation. Mov Disord [Internet]. 2021 Jan 1
[cited 2023 Jun 16];36(1):25. Available from: /pmc/articles/PMC8285924/
33. Ascherio A, Schwarzschild MA. The epidemiology of Parkinson’s disease: risk factors and prevention.
Lancet Neurol [Internet]. 2016 Nov 1 [cited 2023 Jun 17];15(12):1257–72. Available from:
http://www.thelancet.com/article/S1474442216302307/fulltext
34. Davie CA. A review of Parkinson’s disease. Br Med Bull [Internet]. 2008 Jun 1 [cited 2023 Jun
16];86(1):109–27. Available from: https://dx.doi.org/10.1093/bmb/ldn013
35. Grealish S, Jönsson ME, Li M, Kirik D, Björklund A, Thompson LH. The A9 dopamine neuron
component in grafts of ventral mesencephalon is an important determinant for recovery of motor
function in a rat model of Parkinson’s disease. Brain [Internet]. 2010 [cited 2023 Jun 16];133(2):482.
Available from: /pmc/articles/PMC2822634/
36. Gordián-Vélez WJ, Chouhan D, España RA, Chen HI, Burdick JA, Duda JE, et al. Restoring lost
nigrostriatal fibers in Parkinson’s disease based on clinically-inspired design criteria. Brain Res Bull.
2021 Oct 1;175:168–85.
37. Beckers M, Bloem BR, Verbeek MM. Mechanisms of peripheral levodopa resistance in Parkinson’s
disease. npj Parkinson’s Disease 2022 8:1 [Internet]. 2022 May 11 [cited 2023 Jun 17];8(1):1–9.
Available from: https://www.nature.com/articles/s41531-022-00321-y
38. Poewe W. Non-motor symptoms in Parkinson’s disease. Eur J Neurol [Internet]. 2008 Apr [cited 2023
Jun 17];15(SUPPL. 1):14–20. Available from: https://onlinelibrary.wiley.com/doi/full/10.1111/j.1468-
1331.2008.02056.x
39. Proulx M, de Courval FP, Wiseman MA, Panisset M. Salivary production in Parkinson’s disease.
Movement Disorders [Internet]. 2005 Feb 1 [cited 2023 Jun 17];20(2):204–7. Available from:
https://onlinelibrary.wiley.com/doi/full/10.1002/mds.20189

23
40. Suttrup I, Warnecke T. Dysphagia in Parkinson’s Disease. Dysphagia [Internet]. 2016 Feb 1 [cited 2023
Jun 17];31(1):24–32. Available from: https://link.springer.com/article/10.1007/s00455-015-9671-9
41. Barbe AG, Heinzler A, Derman SHM, Hellmich M, Timmermann L, Noack MJ. Hyposalivation and
xerostomia among Parkinson’s disease patients and its impact on quality of life. Oral Dis [Internet].
2017 May 1 [cited 2023 Jun 17];23(4):464–70. Available from:
https://onlinelibrary.wiley.com/doi/full/10.1111/odi.12622
42. Lopalco A, Cutrignelli A, Denora N, Lopedota A, Franco M, Laquintana V. Transferrin Functionalized
Liposomes Loading Dopamine HCl: Development and Permeability Studies across an In Vitro Model of
Human Blood–Brain Barrier. Nanomaterials [Internet]. 2018 [cited 2023 Jun 17];8(3). Available from:
/pmc/articles/PMC5869669/
43. Kordower JH, Bjorklund A. Trophic Factor Gene Therapy for Parkinson’s Disease. Movement Disorders
[Internet]. 2013 Jan 1 [cited 2023 Jun 17];28(1):96–109. Available from:
https://onlinelibrary.wiley.com/doi/full/10.1002/mds.25344
44. Bjorklund A, Kordower JH. Cell Therapy for Parkinson’s Disease: What Next? Movement Disorders
[Internet]. 2013 Jan 1 [cited 2023 Jun 17];28(1):110–5. Available from:
https://onlinelibrary.wiley.com/doi/full/10.1002/mds.25343
45. Lindvall O. Developing dopaminergic cell therapy for Parkinson’s disease—give up or move forward?
Movement Disorders [Internet]. 2013 Mar 1 [cited 2023 Jun 17];28(3):268–73. Available from:
https://onlinelibrary.wiley.com/doi/full/10.1002/mds.25378
46. Charles D, Konrad PE, Neimat JS, Molinari AL, Tramontana MG, Finder SG, et al. Subthalamic Nucleus
Deep Brain Stimulation in Early Stage Parkinson’s Disease. Parkinsonism Relat Disord [Internet]. 2014
[cited 2023 Jun 17];20(7):731. Available from: /pmc/articles/PMC4103427/
47. Mei J, Desrosiers C, Frasnelli J. Machine Learning for the Diagnosis of Parkinson’s Disease: A Review of
Literature. Front Aging Neurosci. 2021 May 6;13:633752.
48. Skorvanek M, Martinez-Martin P, Kovacs N, Rodriguez-Violante M, Corvol JC, Taba P, et al. Differences
in MDS‐UPDRS Scores Based on Hoehn and Yahr Stage and Disease Duration. Mov Disord Clin Pract
[Internet]. 2017 Jul 1 [cited 2023 Jun 17];4(4):536. Available from: /pmc/articles/PMC6174385/
49. Fahn S. Unified Parkinson’s disease rating scale. Recent developments in Parkinson’s disease.
1987;153–63.
50. Goetz CC. The Unified Parkinson’s Disease Rating Scale (UPDRS): Status and recommendations.
Movement Disorders [Internet]. 2003 Jul 1 [cited 2023 Jun 17];18(7):738–50. Available from:
https://onlinelibrary.wiley.com/doi/full/10.1002/mds.10473
51. Braak H, Del Tredici K, Rüb U, De Vos RAI, Jansen Steur ENH, Braak E. Staging of brain pathology
related to sporadic Parkinson’s disease. Neurobiol Aging. 2003 Mar 1;24(2):197–211.
52. Braak H, Del Tredici K. Neuropathological Staging of Brain Pathology in Sporadic Parkinson’s disease:
Separating the Wheat from the Chaff. J Parkinsons Dis [Internet]. 2017 [cited 2023 Jun 17];7(Suppl
1):S71. Available from: /pmc/articles/PMC5345633/
53. Beach TG, Adler CH, Sue LI, Shill HA, Driver-Dunckley E, Mehta SH, et al. Vagus Nerve and Stomach
Synucleinopathy in Parkinson’s Disease, Incidental Lewy Body Disease, and Normal Elderly Subjects:
Evidence Against the “Body-First” Hypothesis. J Parkinsons Dis. 2021 Oct 12;11(4):1833–43.

24
54. Braak H, Del Tredici K, Rüb U, De Vos RAI, Jansen Steur ENH, Braak E. Staging of brain pathology
related to sporadic Parkinson’s disease. Neurobiol Aging. 2003 Mar 1;24(2):197–211.
55. Daubner SC, Le T, Wang S. Tyrosine Hydroxylase and Regulation of Dopamine Synthesis. Arch Biochem
Biophys [Internet]. 2011 Apr 4 [cited 2023 Jun 17];508(1):1. Available from:
/pmc/articles/PMC3065393/
56. Dickson DW, Fujishiro H, DelleDonne A, Menke J, Ahmed Z, Klos KJ, et al. Evidence that incidental
Lewy body disease is pre-symptomatic Parkinson’s disease. Acta Neuropathol. 2008 Apr 9;115(4):437–
44.
57. Beach TG, Adler CH, Sue LI, Peirce JB, Bachalakuri J, Dalsing-Hernandez JE, et al. Reduced striatal
tyrosine hydroxylase in incidental Lewy body disease. Acta Neuropathol. 2008 Apr 6;115(4):445–51.
58. Milber JM, Noorigian J V., Morley JF, Petrovitch H, White L, Ross GW, et al. Lewy pathology is not the
first sign of degeneration in vulnerable neurons in Parkinson disease. Neurology [Internet]. 2012 Dec
11 [cited 2023 Jun 18];79(24):2307–14. Available from: https://n.neurology.org/content/79/24/2307
59. Trist BG, Hare DJ, Double KL. Oxidative stress in the aging substantia nigra and the etiology of
Parkinson’s disease. Aging Cell [Internet]. 2019 Dec 1 [cited 2023 Jun 17];18(6). Available from:
/pmc/articles/PMC6826160/
60. Bose A, Beal MF. Mitochondrial dysfunction in Parkinson’s disease. J Neurochem [Internet]. 2016 Oct
1 [cited 2023 Jun 17];139:216–31. Available from:
https://onlinelibrary.wiley.com/doi/full/10.1111/jnc.13731
61. Perier C, Bové J, Vila M. Mitochondria and Programmed Cell Death in Parkinson’s Disease: Apoptosis
and Beyond. https://home.liebertpub.com/ars [Internet]. 2012 Mar 5 [cited 2023 Jun 18];16(9):883–
95. Available from: https://www.liebertpub.com/doi/10.1089/ars.2011.4074
62. Bové J, Perier C. Neurotoxin-based models of Parkinson’s disease. Neuroscience. 2012 Jun 1;211:51–
76.
63. Schapira AHV. Mitochondrial Pathology in Parkinson’s Disease. Mount Sinai Journal of Medicine: A
Journal of Translational and Personalized Medicine [Internet]. 2011 Nov 1 [cited 2023 Jun
17];78(6):872–81. Available from: https://onlinelibrary.wiley.com/doi/full/10.1002/msj.20303
64. Wong YC, Luk K, Purtell K, Burke Nanni S, Stoessl AJ, Trudeau LE, et al. Neuronal Vulnerability in
Parkinson Disease and Putative Therapeutics: Should the focus be on axonal and synaptic terminals?
Mov Disord [Internet]. 2019 Oct 1 [cited 2023 Jun 17];34(10):1406. Available from:
/pmc/articles/PMC6879792/
65. Navarro-Romero A, Montpeyó M, Martinez-Vicente M. The Emerging Role of the Lysosome in
Parkinson’s Disease. Cells [Internet]. 2020 Nov 2 [cited 2023 Jun 17];9(11). Available from:
/pmc/articles/PMC7692401/
66. Magistrelli L, Contaldi E, Vignaroli F, Gallo S, Colombatto F, Cantello R, et al. Immune Response
Modifications in the Genetic Forms of Parkinson’s Disease: What Do We Know? Int J Mol Sci
[Internet]. 2022 Apr 1 [cited 2023 Jun 18];23(7). Available from: /pmc/articles/PMC8998358/
67. Costantini E, D’Angelo C, Reale M. The role of immunosenescence in neurodegenerative diseases.
Mediators Inflamm. 2018;2018.

25
68. Bankhead P, Loughrey MB, Fernández JA, Dombrowski Y, McArt DG, Dunne PD, et al. QuPath: Open
source software for digital pathology image analysis. Sci Rep [Internet]. 2017 Dec 1 [cited 2023 Jun
17];7(1). Available from: /pmc/articles/PMC5715110/
69. Mosley RL, Hutter-Saunders JA, Stone DK, Gendelman HE. Inflammation and Adaptive Immunity in
Parkinson’s Disease. Cold Spring Harb Perspect Med [Internet]. 2012 [cited 2023 Jun 17];2(1).
Available from: /pmc/articles/PMC3253034/
70. Witoelar A, Jansen IE, Wang Y, Desikan RS, Gibbs JR, Blauwendraat C, et al. Genome-wide Pleiotropy
Between Parkinson Disease and Autoimmune Diseases. JAMA Neurol [Internet]. 2017 Jul 1 [cited 2023
Jun 17];74(7):780. Available from: /pmc/articles/PMC5710535/
71. Tan EK, Chao YX, West A, Chan LL, Poewe W, Jankovic J. Parkinson disease and the immune system -
associations, mechanisms and therapeutics. Nat Rev Neurol [Internet]. 2020 Apr 24 [cited 2023 Jun
17];16(6):303–18. Available from: https://europepmc.org/article/med/32332985
72. Singleton A, Hardy J. Progress in the genetic analysis of Parkinson’s disease. Hum Mol Genet
[Internet]. 2019 Nov 11 [cited 2023 Jun 18];28(R2):R215. Available from: /pmc/articles/PMC6872433/
73. Srinivasan BS, Doostzadeh J, Absalan F, Mohandessi S, Jalili R, Bigdeli S, et al. Whole genome survey of
coding SNPs reveals a reproducible pathway determinant of Parkinson disease. Hum Mutat [Internet].
2009 Feb [cited 2023 Jun 18];30(2):228. Available from: /pmc/articles/PMC2793088/
74. Ahmed I, Tamouza R, Delord M, Krishnamoorthy R, Tzourio C, Mulot C, et al. Association between
Parkinson’s disease and the HLA-DRB1 locus. Movement Disorders [Internet]. 2012 Aug 1 [cited 2023
Jun 18];27(9):1104–10. Available from: https://onlinelibrary.wiley.com/doi/full/10.1002/mds.25035
75. Wissemann WT, Hill-Burns EM, Zabetian CP, Factor SA, Patsopoulos N, Hoglund B, et al. Association of
Parkinson Disease with Structural and Regulatory Variants in the HLA Region. Am J Hum Genet
[Internet]. 2013 Nov 11 [cited 2023 Jun 18];93(5):984. Available from: /pmc/articles/PMC3824116/
76. Garretti F, Agalliu D, Arlehamn CSL, Sette A, Sulzer D. Autoimmmunity in parkinson’s disease: The role
of α:-synuclein-specific T cells. Front Immunol. 2019 Feb 25;10(FEB):442306.
77. Williams GP, Schonhoff AM, Jurkuvenaite A, Gallups NJ, Standaert DG, Harms AS. CD4 T cells mediate
brain inflammation and neurodegeneration in a mouse model of Parkinson’s disease. Brain [Internet].
2021 Jul 1 [cited 2023 Jun 17];144(7):2047. Available from: /pmc/articles/PMC8370411/
78. Brochard V, Combadière B, Prigent A, Laouar Y, Perrin A, Beray-Berthat V, et al. Infiltration of CD4+
lymphocytes into the brain contributes to neurodegeneration in a mouse model of Parkinson disease.
J Clin Invest [Internet]. 2009 Jan 1 [cited 2023 Jun 17];119(1):182. Available from:
/pmc/articles/PMC2613467/
79. McGeer PL, Schwab C, Parent A, Doudet D. Presence of reactive microglia in monkey substantia nigra
years after 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine administration. Ann Neurol [Internet]. 2003
Nov 1 [cited 2023 Jun 17];54(5):599–604. Available from:
https://onlinelibrary.wiley.com/doi/full/10.1002/ana.10728
80. Nagatsu T, Mogi M, Ichinose H, Togari A. Cytokines in Parkinson’s disease. Journal of Neural
Transmission, Supplement. 2000;(58):143–51.

26
81. Hawkes CH, Del Tredici K, Braak H. Parkinson’s disease: a dual‐hit hypothesis. Neuropathol Appl
Neurobiol [Internet]. 2007 Dec [cited 2023 Jun 17];33(6):599. Available from:
/pmc/articles/PMC7194308/
82. Grewal JS, Jamal Z, Ryan J. Anatomy, Head and Neck, Submandibular Gland. StatPearls [Internet].
2022 Dec 11 [cited 2023 Jun 17]; Available from: https://www.ncbi.nlm.nih.gov/books/NBK542272/
83. Holmberg K V., Hoffman MP. Anatomy, biogenesis, and regeneration of salivary glands. Monogr Oral
Sci [Internet]. 2014 [cited 2023 Jun 17];24:1. Available from: /pmc/articles/PMC4048853/
84. Pedersen AML, Sørensen CE, Proctor GB, Carpenter GH, Ekström J. Salivary secretion in health and
disease. J Oral Rehabil [Internet]. 2018 Sep 1 [cited 2023 Jun 17];45(9):730–46. Available from:
https://onlinelibrary.wiley.com/doi/full/10.1111/joor.12664
85. Ryumon S, Hage D, Ibaragi S, Okui T, Tubbs RS, Iwanaga J. Dual innervation of the submandibular
gland by nerve to mylohyoid and chorda tympani. Morphologie. 2021 Dec 1;105(351):316–8.
86. Tredici K Del, Hawkes CH, Ghebremedhin E, Braak H. Lewy pathology in the submandibular gland of
individuals with incidental Lewy body disease and sporadic parkinson’s disease. Acta Neuropathol
[Internet]. 2010 Jun 14 [cited 2023 Jun 17];119(6):703–13. Available from:
https://link.springer.com/article/10.1007/s00401-010-0665-2
87. Niazi MKK, Parwani A V., Gurcan MN. Digital Pathology and Artificial Intelligence. Lancet Oncol
[Internet]. 2019 May 1 [cited 2023 Jun 17];20(5):e253. Available from: /pmc/articles/PMC8711251/
88. van der Laak J, Litjens G, Ciompi F. Deep learning in histopathology: the path to the clinic. Nature
Medicine 2021 27:5 [Internet]. 2021 May 14 [cited 2023 Jun 17];27(5):775–84. Available from:
https://www.nature.com/articles/s41591-021-01343-4
89. Dawson H. Digital pathology – Rising to the challenge. Front Med (Lausanne) [Internet]. 2022 Jul 22
[cited 2023 Jun 17];9. Available from: /pmc/articles/PMC9354827/
90. Baxi V, Edwards R, Montalto M, Saha S. Digital pathology and artificial intelligence in translational
medicine and clinical practice. Modern Pathology 2021 35:1 [Internet]. 2021 Oct 5 [cited 2023 Jun
17];35(1):23–32. Available from: https://www.nature.com/articles/s41379-021-00919-2
91. Klein A, Klein J, Chacham M, Kleinman S, Shuster A, Peleg O, et al. Acinar Atrophy, Fibrosis and Fatty
Changes Are Significantly More Common than Sjogren’s Syndrome in Minor Salivary Gland Biopsies.
Medicina (B Aires) [Internet]. 2022 Feb 1 [cited 2023 Jun 17];58(2). Available from:
/pmc/articles/PMC8879368/

27
28

You might also like