Next Article in Journal
Differential Regulation of the EGFR/PI3K/AKT/PTEN Pathway between Low- and High-Grade Gliomas
Next Article in Special Issue
Secondary Impact of COVID-19 Pandemic on People with Parkinson’s Disease—Results of a Polish Online Survey
Previous Article in Journal
Virtual Reality for Aggression Assessment: The Development and Preliminary Results of Two Virtual Reality Tasks to Assess Reactive and Proactive Aggression in Males
Previous Article in Special Issue
Accelerated Early Progression of Amyotrophic Lateral Sclerosis over the COVID-19 Pandemic
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Can SARS-CoV-2 Infection Lead to Neurodegeneration and Parkinson’s Disease?

Department of Neurology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
*
Author to whom correspondence should be addressed.
Brain Sci. 2021, 11(12), 1654; https://doi.org/10.3390/brainsci11121654
Submission received: 26 November 2021 / Revised: 8 December 2021 / Accepted: 16 December 2021 / Published: 18 December 2021
(This article belongs to the Special Issue COVID-19, Parkinson’s Disease and Other Neurodegenerative Disorders)

Abstract

:
The SARS-CoV-2 pandemic has affected the daily life of the worldwide population since 2020. Links between the newly discovered viral infection and the pathogenesis of neurodegenerative diseases have been investigated in different studies. This review aims to summarize the literature concerning COVID-19 and Parkinson’s disease (PD) to give an overview on the interface between viral infection and neurodegeneration with regard to this current topic. We will highlight SARS-CoV-2 neurotropism, neuropathology and the suspected pathophysiological links between the infection and neurodegeneration as well as the psychosocial impact of the pandemic on patients with PD. Some evidence discussed in this review suggests that the SARS-CoV-2 pandemic might be followed by a higher incidence of neurodegenerative diseases in the future. However, the data generated so far are not sufficient to confirm that COVID-19 can trigger or accelerate neurodegenerative diseases.

1. Introduction

The aim of this review is to summarize the data on the link between COVID-19, other viral infections and Parkinson’s disease. Therefore, different mechanisms of the SARS-CoV-2 virus affecting cerebral functions and inducing neurodegeneration have to be considered. First, a direct neurotoxic effect of the virus resulting from neuroinvasion is possible as well as secondary effects due to systemic inflammatory alterations. In the first section, we present the knowledge on SARS-CoV-2 neurotropism, neuropathology, neuroinflammation and changes in levels of biomarkers that have been observed during infection. This is followed by a brief overview on the connection between other viral infections and neurodegenerative diseases. In the third chapter, we highlight the link between COVID-19 and neurodegeneration focusing on Parkinson’s disease (PD) and cognitive dysfunction/Alzheimer’s disease (AD). At last, we discuss how the pandemic has influenced symptoms and psychosocial aspects in PD patients underlining the tremendous impact that the infection has had especially on people with preexisting neurological conditions and disabilities.

2. Methods

Literature research for this review was done in PubMed using the search terms “COVID-19”, “SARS-CoV-2”, “Parkinson’s disease”, “Alzheimer’s disease”, “neurodegeneration”, “viral infection” and “infection” in different combinations. Only articles published in English in international peer-reviewed journals were included in the selection process. Articles were selected by screening the abstracts for eligibility; publications contributing relevant data to the core content of this review were included.

3. Chapter 1

3.1. SARS-CoV-2 Neurotropism

SARS-CoV-2, the cause of the current pandemic, belongs to the well-known family of Coronaviridae. Previously defined Coronaviridae (HCov-OC43, HCov-229E, SARS-CoV, MERS-Cov) were detected in human brain samples, which proves their neurotropism and their ability to cause persistent infections of the central nervous system (CNS) [1,2,3]. As early as 1999 it was shown in vitro that neuroblastoma, neuroglioma and glial cells were susceptible to infection with human Coronaviridae and that the infection could persist for at least 130 days of culture time [2]. In animal models, this persistent infection led to neuronal loss and long-term sequelae such as reduced activity and hippocampal neuronal volumes as signs of a neurodegenerative phenotype [4,5,6].
There is an ongoing debate about whether SARS-CoV-2 can enter and persist in cerebral structures. Indeed, there are some data supporting the theory of SARS-CoV-2 neurotropism that will be outlined in the following paragraphs.
The ACE-2-receptor was identified as one of SARS-CoV-2′s most common entry receptors [7]. However, it is not highly expressed in the brain compared to other tissues [7]. Its expression in the CNS was shown on glial cells (astrocytes), capillary endothelium, monocytes/macrophages and neurons [8,9]. A comparably high expression of ACE-2-receptor was detected in brainstem areas leading to the hypothesis that SARS-CoV-2 invasion might impair brainstem structures involved in regulating cardiovascular functions [10]. An infection of ACE-2-receptor transgenic mice also led to the expression of viral antigens in neurons especially in the thalamus, cerebrum and brainstem, while the cerebellum remained uninfected [5]. The affected brain areas showed neuronal loss and microglial activation in the absence of other inflammatory signs [5].
In contrast, there is increasing evidence that the ACE-2-receptor may not be the primary way of invasion into the CNS. Instead, other receptors such as neuropilin 1 (NRP-1) could contribute substantially to the invasion of SARS-CoV-2 into cerebral structures [11,12,13]. NRP-1 was found to be highly expressed in neurons and astrocytes [12].
There are three main routes that are speculated to lead to SARS-CoV-2 invasion into the CNS that are presented below [10,11,14,15,16,17,18].
The first possible way of viral invasion is the transneural way starting in the nasal epithelium and the olfactory nerves progressing into the brain via axonal transport [5,14,19]. This route of neurotropism was shown for SARS-CoV and HCov-OC43 after intranasal infection [4,20]. In transgenic ACE-2-receptor expressing mice, intranasal infection with SARS-CoV resulted in neuronal loss [3,20]. Additionally, the mouse equivalent of the human coronavirus, the mouse hepatitis virus, entered the brain via the olfactory nerve after intranasal inoculation [21].
Supporting this idea of neurotropism is the fact that COVID-19 frequently causes olfactory dysfunction (OD). A wide variation in the incidence of OD associated with COVID-19 (5–98%) was observed mostly due to missing objective testing [22]. An Iranian study using objective smell testing in 60 COVID-19 patients revealed that 98% demonstrated smell loss, but only 35% were subjectively aware of their OD, which underlines the importance of objective testing for this symptom [23,24]. Gustation dysfunctions are also common in COVID-19 and can be confused with olfactory problems [25]. OD appears to be a very early symptom in the course of COVID-19 [26]. In general, two mechanisms can lead to OD: First, an obstruction of the olfactory cleft by swelling or rhinorrhea, which could not be detected in COVID-19 patients [24,25,27]. Secondly, defects of sensorineural transmission can impair the sense of smell [25]. A detailed imaging study using CT and MRI on COVID-19 patients with prolonged OD (minimum 1 month) revealed decreased volumes of the olfactory bulb (43.5%) and shallow olfactory sulci (60.9%) as evidence for this underlying pathology of OD in COVID-19 [27]. However, ACE-2-receptors are absent in olfactory sensory neurons and can only be found in supporting cells such as sustentacular cells and horizontal basal cells (reserve stem cells) in the olfactory and respiratory epithelium [22,28]. It has to be acknowledged that OD is generally a common symptom in the elderly, as it occurs in 10% of people over 65 years and in 62–80% over 80 years [24]. OD is also known to be a common symptom in early PD and AD [24]. Interestingly, OD in COVID-19 occurs more often in younger patients and is inversely correlated with death [29]. This supports the contrasting hypothesis that OD is a sign of defense against the virus to prevent it from reaching cerebral structures rather than allowing entry into the CNS [30].
Alternative transneural ways of CNS invasion by SARS-CoV-2 via the trigeminal or vagal nerve have also been discussed [10,11].
The second proposed route of viral invasion into the CNS is the hematogenous pathway with subsequent crossing of the blood–brain barrier or infection of the choroid plexus [10,11,14,15]. This was described for various other viruses, e.g., HIV, HSV, HCMV and enteroviruses [6].
The endothelial cells in blood vessels and choroid plexus could be the target of invasion in this route of infection, as they were shown to express the ACE-2-receptor [8,11]. Additionally, the SARS-CoV-2 spike protein can cross and impair the blood–brain barrier itself by inducing an inflammatory response within the microvascular endothelium [31,32].
Another mechanism supporting this route of invasion could be an increase of the blood–brain barrier permeability due to elevated IL-6 levels that are present in acute COVID-19 disease [14,33].
The third possible pathway of neurotropism for SARS-CoV-2 is the so-called the “Trojan horse mechanism” which describes the viral infection of immune cells (neutrophils, macrophages, monocytes, CD4+-lymphocytes) that reach the CNS via bloodstream and then migrate into cerebral structures by diapedesis [10,11,12,15,16,17,18]. Once they are in the cerebral tissue, the virus or viral particles could be released by those immune cells [12].

3.2. SARS-CoV-2 Neuropathology

In general, neuropathological hallmarks of COVID-19 autopsy cases are diffuse edema, gliosis with activation of microglia and astrocytes, ischemic lesions, intracranial bleeds, arteriosclerosis, hypoxic–ischemic injury, encephalitis/meningitis and diffuse inflammation [34,35]. Patients suffering from severe COVID-19 showed a reduction in the numbers of neurons and an elevation in the number of activated microglial cells and astrocytes, as well as higher levels of proinflammatory cytokines measured by qPCR [36].
Matching the hypothesis of hematogenous invasion into the brain, Paniz-Mondolfi et al. detected the virus in capillary endothelium and in neurons in frontal lobe tissue from a patient with COVID-19 [11,37]. The virus was not observed in glial cells in vivo [11]. Another group similarly found SARS-CoV-2 to favor CNS endothelial cells with the ACE-2-receptor expressed in smooth muscle cells of blood vessels [38]. Small vessel disease was identified in five out of nine COVID-19 autopsy cases; however, SARS-CoV-2 was only detected in one case using immunohistochemistry [39]. Detection of SARS-CoV-2 in the brain using PCR was equally difficult; the highest viral load was documented in the olfactory bulb, while SARS-CoV-2 PCR was repeatedly negative in the substantia nigra [30,34,40]. Viral presence is, however, rarely detected in viral encephalitis in general (e.g., in herpesvirus-, arbovirus- or enterovirus-induced encephalitis) [6].
The brains of COVID-19 autopsy cases showed microglial activation in the olfactory bulb, frontal cortex, hippocampus and most prominently in the brainstem, whereas lymphocytes did not appear to be activated [39]. Interestingly, patients with a history of delirium during COVID-19 demonstrated more microglial activation in the hippocampus [39]. Patients with and without sepsis could not be distinguished neuropathologically, contradicting the common hypothesis that neuropathology develops secondary to a cytokine storm during septic disease [39].

3.3. SARS-CoV-2 Neuroinflammation/Biomarkers

Apart from a direct influence of SARS-CoV-2 on the brain by invasion into the CNS, secondary effects on cerebral functions due to systemic alterations in the course of the disease are widely discussed. Investigations of brain tissue, biofluids and the systemic reaction showed a (neuro-)inflammatory response triggered by COVID-19.
Multiple cytokines were found to be elevated in the blood during acute COVID-19, while increased levels of proinflammatory markers were not detected in the cerebrospinal fluid (CSF) [41]. Serum levels of IL-4, IL-10, IL-6 and IL1β were elevated in COVID-19 patients [33,42,43]. IL-1- and IL-6 are known to trigger neuroinflammation [9].
SARS-CoV-2 antibodies were frequently detected in the CSF of COVID-19 patients, although this does not prove intrathecal antibody production [41,44,45]. The detection of the virus via PCR from CSF was impossible in most cases [41,44,45]. Only few authors described sporadic positive results in SARS-CoV-2 PCR from CSF in patients with severe cerebral symptoms [46,47,48].
Analysis of markers indicating CNS lesions revealed elevated levels of neurofilament light chain (NfL) and glial fibrillary acidic protein (GFAP) in plasma of patients with moderate to severe COVID-19 [17,49]. Additionally, three of eight patients with severe COVID-19 had signs of a disrupted blood–brain barrier, one had a specific intrathecal antibody synthesis and four were positive for 14-3-3 in the CSF [44]. The data on CSF pleocytosis are controversial so far. A case series of 15 patients and a review summarizing CSF white blood cell counts of 409 COVID-19 patients with neurological symptoms observed frequent pleocytosis (defined as >5 cells/µL) in 36% of 15 and 17% of 409 cases [30,50]. On the other hand, a case series of 13 patients with COVID-19 and encephalopathy or seizures reported CSF pleocytosis in only one case, similar to a study with 18 patients with COVID-19 and neurological complications that discovered pleocytosis in four cases and reported all four to be likely due to blood contamination [51,52].
Sun et al. investigated the cargo of neuronal-enriched extracellular vesicles and interestingly found elevated NfL, amyloid-β, neurogranin, tau and phosphorylated tau in COVID-19 patients implicating possible neurodegenerative processes [42].
Take home messages of Chapter 1 (Section 3):
  • It is likely that SARS-CoV-2 can be neurotropic, since this was shown for other human coronaviruses (HCov-OC43, Hcov-229E, SARS-CoV, MERS-Cov) in the past.
  • There are three possible routes of SARS-CoV-2 neuroinvasion: The transneural route via the olfactory nerve, the hematogenous route via vascular endothelium or a permeable blood–brain barrier and the “Trojan-horse-mechanism” by infiltration of immune cells and subsequent invasion into the CNS via diapedesis.
  • Neuropathologically, SARS-CoV-2 leads to microglial activation in distinct CNS areas.
  • SARS-CoV-2 triggers a neuroinflammatory response with increased serum levels of several cytokines (e.g., IL-1, IL-6) and elevated markers such as NfL and GFAP in the CSF indicating CNS lesions.

4. Chapter 2

Viral Infection and Neurodegeneration

Besides the COVID-19 pandemic, there is broad (epidemiological) evidence linking other viral infections to neurodegenerative diseases, especially PD and AD, which will be reviewed in the following chapters.
The idea, that viral infections can promote neurodegeneration first developed with encephalitis lethargica after the Spanish flu epidemic at the beginning of the 20th century [53]. Since then, a connection between infections and neurodegenerative diseases has been assumed repeatedly.
A meta-analysis of 287,773 PD patients and 7,102,901 controls revealed that individuals with reported infections in the past had an elevated risk for PD (odds ratio, 1.20) [54]. This effect was foremost attributable to bacterial infections [54]. In line with this, a more recent study found a “higher infectious burden” defined by the existence of more antibodies against different viruses and bacteria in the blood of PD patients [55]. More specifically, PD risk was shown to be elevated after VZV infection (adjusted hazard ratio, 1.17) and PD patients were more often seropositive for EBV [56,57]. HCV is a well-established risk factor for PD as is HSV-1 infection for the development of AD [58,59,60,61].
Influenza viruses were brought into context with PD, since encephalitis lethargica had a parkinsonian phenotype and an influenza virus was proposed as the infectious agent of the Spanish flu [53]. Furthermore, H1N1 infection led to persistent microglial activation as a sign of chronic neuroinflammation in wildtype mice [62]. H5N1 accordingly led to microglial activation and α-synuclein aggregation in mice resulting in the loss of dopaminergic neurons in the substantia nigra, which is recognized as the pathological hallmark of PD [63]. Furthermore, the influenza A virus was detected postmortem in the substantia nigra of PD patients [64]. A recent case–control study using data from the Danish National Patient Registry revealed that an influenza diagnosis was associated with the development of PD up to ten years later (odds ratio 1.73) [65]. Thus, a strong association between influenza viruses and PD is suspected but needs to be further elucidated.
Japanese encephalitis virus causes a parkinsonian phenotype during acute disease, but even persistent parkinsonism with MRI lesions in the substantia nigra was observed three to five years after viral infection [66].
West Nile virus can also induce parkinsonism during acute infection. In postmortem studies, elevated α-synuclein levels were detected in patients infected with West Nile virus [57,67,68]. An interesting hypothesis about the function of α-synuclein was developed in an α-synuclein-knockout mouse model after West Nile infection [67]. The absence of α-synuclein in this model led to disastrous disease progression, suggesting a protective role of α-synuclein against viral infection [57,67]. It was postulated that α-synuclein entraps viral particles as a cellular defense mechanism, which persists after the infection leading to its pathological aggregation and subsequent neurotoxic effects. The same mechanism was proposed for β-amyloid, which can entrap HSV-1 and inhibit its viral replication and entry in vitro and in vivo [69,70]. HSV-1 infection was implicated as a disease risk factor foremost in AD but also in PD in different in vitro and in vivo investigations [71,72]. A 2.56-fold increased risk of developing dementia was reported in a retrospective cohort study with 8362 patients with acute HSV-1 or HSV-2 infections [60]. A phase 2 study investigating whether valaciclovir can slow the progression of AD in patients with HSV-1 is currently ongoing (clinicaltrials.gov NCT03282916) [70].
There are different studies suggesting an involvement of the adaptive immune system in the development of neurodegeneration. Genome-wide association studies have found an association of specific major histocompatibility complex II gene alleles with PD and T-cells of PD patients were shown to react to α-synuclein epitopes [73]. Another group showed that Th17-T-cells contribute to PD pathogenesis in a cell culture model of PD using induced pluripotent stem cells (iPSCs) [74]. Recently, T-cells were found to be adjacent to Lewy bodies and dopaminergic neurons in brains of Lewy-body-dementia patients and stimulation of CD4+ T-cells with a phosphorylated α-synuclein epitope resulted in increased IL-17 production as a sign of a Th17-response [75].
Take home messages of Chapter 2 (Section 4):
  • Multiple epidemiological studies link different (viral) infections to PD, as individuals with certain infections have an elevated risk for PD.
  • The protein α-synuclein might physiologically act as an infection defense mechanism, entrapping viral particles, which could lead to its pathological aggregation and subsequent neurotoxic PD effects.
  • The involvement of the adaptive immune system in the development of neurodegenerative diseases has been increasingly implicated supporting the hypothesis that infections, and thus activation of the immune system can trigger neurodegenerative cascades.

5. Chapter 3

5.1. General Implications of SARS-CoV-2 in Neurodegeneration

The previously discussed mechanisms of viral neurotropism and neuroinflammation raise the question of whether long-term neurodegeneration has to be expected after COVID-19 disease.
SARS-CoV-2 and potentially pathogenic proteins involved in neurodegeneration have been linked by different studies. It was observed that the spike protein receptor binding domain binds to heparin and heparin binding proteins including amyloid-β, α-synuclein, tau, prion and TDP-43, which may initiate the pathological aggregation of these proteins resulting in neurodegeneration [76,77]. The same mechanism is described for HSV-1, which catalyzes the aggregation of amyloid-β in vitro and in vivo and is a well-established risk factor for AD [76,78]. Recently, it was demonstrated that viral particles (including SARS-CoV-2 spike protein) facilitate the spreading of proteopathic seeds by altering intercellular cargo transfer [79].
Viruses use different strategies to take control over host cellular functions, such as interfering with autophagy and mitochondrial or lysosomal functions, which are implicated in the development of neurodegenerative disease as well [80]. SARS-CoV-2 alters autophagy and mitochondrial and lysosomal functions in infected lung cells [81].
Furthermore, viral changes of proteostasis of the host cell can lead to accelerated “aging” of the infected tissue, which may then boost neurodegenerative processes that are common in senescent cells [80].
Ferrosenescence is an iron-mediated premature aging process of cells that results in an the iron-induced disruption of DNA repair and, thus, in neurodegeneration [82]. An interesting aspect of viral capabilities is the induction of ferrosenescence in host cells to facilitate viral replication [82].
These data support the notion that SARS-CoV-2 infections can induce alterations promoting neurodegenerative cascades.

5.2. COVID-19 and Possible Mechanisms Connected to Parkinson’s Disease

There are several links between COVID-19 and the development of PD that are elaborated in this section.
In 1985, it was observed that infection of mice with the mouse hepatitis virus (that has been identified as a murine analogue of the human Coronaviridae) resulted in mild encephalitis and the deposition of viral antigens mostly in the nucleus subthalamicus and the substantia nigra [83]. This led to subsequent gliosis in those regions, suggesting a link between the virus and PD/postencephalitic parkinsonism [83]. Antibodies against Coronaviridae were found to be elevated in the CSF of PD patients compared to controls as early as 1992 [84].
Thus far, three case reports of PD onset in timely correlation to COVID-19 disease have been reported; however, a clear causal link could not be established [85].
Two cases of patients developing COVID-19-associated encephalitis with progressive atypical parkinsonism and FDG-PET alterations reminiscent of postencephalitic parkinsonism were published recently [86].
Several mechanisms by which COVID-19 might contribute to the development of PD were previously reviewed and discussed: Vascular insults in the nigrostriatal system could lead to subsequent parkinsonism [87]. Furthermore, the cytokine storm associated with severe COVID-19 triggers neuroinflammation and, subsequently, neurodegeneration [33,87]. Systemic levels of IL-6 are elevated in COVID-19, and a small prospective observational study revealed that a higher level of IL-6 was associated with an increased risk of developing PD [88].
Another possible mechanism of inducing PD would be viral neurotropism resulting in direct neuronal damage in strategic areas. IPSC-derived midbrain dopaminergic neurons were shown to be submissive to SARS-CoV-2-infection, which triggered an inflammatory response and subsequently cellular senescence in vitro [89]. RNA-sequencing analysis of the ventral midbrain tissue of COVID-19 patients revealed a comparable phenotype of inflamed neurons and identified low levels of SARS-CoV-2 transcripts [89]. These data underline that there may be a special susceptibility to SARS-CoV-2 of particularly vulnerable midbrain regions involved in the development of PD.
The general susceptibility of central nervous structures to SARS-CoV-2 was shown by Ramani et al., who infected brain organoids and observed viral entry especially in neurons. The infection induced an altered distribution and hyperphosphorylation of tau and subsequent neuronal death [90].
A link between NF-κB and PD was previously established because NF-κB was increased in the substantia nigra of MPTP-treated mice [91]. MPTP treatment is a common animal model of PD as the neurotoxin leads to nigrostriatal degeneration and loss of dopaminergic neurons [91,92]. Suppressing NF-κB in this model led to prevention of the degeneration of dopaminergic neurons [91]. In an in vitro model of dopaminergic neurons, treatment with 6-OHDA led to NF-κB activation, caspase activation and apoptotic death that was prevented by inhibition of NF-κB [93]. NF-κB is activated by SARS-CoV-2 via pattern recognition receptors, which might be a neurodegenerative trigger [93].
Other interesting aspects are the shared implications for the angiotensin–aldosterone system in COVID-19 and PD. Angiotensinogen is produced by astrocytes as part of a local independent renin–angiotensin system (RAS) [94,95]. Its pathological overactivation (that also results from the degeneration of dopaminergic neurons) led to oxidative stress and inflammation, whereas its inhibition was considered as a treatment option in several neurodegenerative diseases including PD and AD [96,97]. SARS-CoV-2 uses the ACE2-receptor as an entryway into host cells and, therefore, intervenes with the RAS as well [10].
A previously observed connection between H1N1 influenza virus and α-synuclein aggregation could potentially be relevant for SARS-CoV-2, too. H1N1 led to the aggregation of endogenous α-synuclein in LUHMES cells [98]. As a reason for the pathological α-synuclein aggregation following H1N1 infection, an impairment of the autophagosome of infected LUHMES-cells was proposed [98]. Interestingly, α-synuclein aggregates were also seen in the olfactory bulb after intranasal instillation of H1N1 [98].
Early symptoms of PD are olfactory and vegetative dysfunction including obstipation as well as the prodromal syndrome REM sleep behavior disorder (RBD). Olfactory dysfunction is a very common early symptom of COVID-19, and the olfactory route is discussed as one way of viral entry into the CNS [21,26]. Therefore, it seems plausible that COVID-19 might influence the pathogenesis of PD as SARS-CoV-2 can take a route of spreading that was described for the developing neuropathology in PD [99,100].
Polysomnographic investigations in 11 patients four months after initial infection with SARS-CoV-2 revealed episodes of REM sleep without atonia in 4 patients, which is a characteristic (prodromal) sign of RBD [101].
Another interesting aspect is that the development of PD is linked to the gut microbiome and its dysbiosis [102]. SARS-CoV-2 causes an imbalance of the gut microbiome (dysbiosis) and intestinal inflammation indicated by elevated fecal calprotectin in COVID-19-associated diarrhea, which proposes a possible link to PD [103,104]. SARS-CoV-2 RNA was detected in the feces of about 50% of patients with COVID-19, supporting the hypothesis of intestinal infection [105].
Molecular investigations have established links between COVID-19 and PD focusing on protein interactions. In all, 44 proteins in the CNS implicated in PD were found to interact with 24 host proteins from the lung that interact with SARS-CoV-2 viral proteins [106]. The two most promising interaction candidates were Rab7a and NUP62 [106]. Rab7a is a lysosomal protein reducing the proportion of cells with α-synuclein particles as well as the toxicity of α-synuclein, whereas NUP62 is involved in autophagosome formation [106]. The comparison of transcriptomic modulations induced by SARS-CoV-2 and PD also revealed significant overlap in several pathways [107].
On the other hand, a protective role of α-synuclein against COVID-19 was proposed since α-synuclein, like β-amyloid, is upregulated in the face of viral infections and can restrict viral replication acting as a defense mechanism in the brain [108]. This leads to the speculation that PD patients with higher α-synuclein levels in the brain might have some protection against SARS-CoV-2 infection [109]. Before the COVID-19 pandemic, a Japanese retrospective cohort study showed that hospitalized PD patients were less likely than other patients to die from pneumonia [109].
If the viral infection leads to an upregulation of α-synuclein as a defense mechanism, it may induce prolonged inflammation and neuronal death triggering the development of PD on the long run as it was shown earlier for West Nile virus infections [87].
Interestingly, a hypothetical connection between COVID-19 and atypical parkinsonism can be established as well, although data on this topic are rare so far. It was demonstrated that atypical Parkinson syndromes such as multisystem atrophy and progressive supranuclear palsy are associated with microglial activation as a sign of neuroinflammation and that the microglial activation contributes to the progression of neurodegeneration [110,111,112]. Recently, it was shown that microglial activation can be visualized by PET imaging, which might function as a biomarker for tauopathies [113,114]. Microglial activation and neuroinflammation are seen in COVID-19 as described in chapter 1, creating a link between atypical parkinsonism and COVID-19 [39].

5.3. Alzheimer’s Disease, Cognitive Deficits and COVID-19

There is cumulating evidence demonstrating a close connection between cognitive disturbances and COVID-19. A prospective longitudinal study revealed that cognitive decline measured by Montreal Cognitive Assessment (MOCA) was apparent in 21% of mild COVID-19 patients vs. 2% of seronegative individuals [115]. Another study found pathological MOCA results in 18 of 26 COVID-19 patients and also FDG-PET abnormalities (frontoparietal hypometabolism) in 10 patients matching the clinical deficit [45].
Cognitive decline was not only observed during acute infection, but there are also reports of persistent cognitive impairment after recovering from COVID-19, as MOCA abnormalities were detected in a group of post-COVID-19 patients [116]. Another study confirmed that cognitive deficits persisted in 70% of COVID-19 patients for at least 1 month after hospital discharge [116,117]. Similarly, 46 of 57 recovering COVID-19 patients (81%) had signs of cognitive impairment [13,118]. Interestingly, persisting memory and concentration deficits were found after SARS-Cov-1 and MERS infections in 15–20% of cases [119].
Another interesting study used transcranial magnetic stimulation to investigate recovered COVID-19 patients who suffered from severe disease with ICU stay and neurological complications reporting fatigue and showing abnormal scores in the frontal assessment battery during the subacute phase [120]. The transcranial magnetic stimulation in these patients revealed severe impairment of GABAergic intracortical circuits while glutamatergic transmission was intact [120]. GABAergic impairments are usually common in frontotemporal dementia and executive dysfunction [120,121]. However, it has to be noted that cognitive impairment is a common problem after suffering from acute respiratory distress syndrome (ARDS), which can have multiple reasons other than COVID-19 [116,122,123,124]. After ARDS, cognitive disturbances persisted in long-term follow-ups in about 10% of cases [116,122]. Other studies found cognitive deficits and psychiatric disorders (mainly depression and anxiety) in up to 60% of ARDS survivors after 12 months [125].
Dementia was found to be one of the strongest risk factors for COVID-19 and associated with a higher mortality [126,127,128,129,130]. Apparently, patients with dementia have difficulties in following hygiene rules, mask requirements, behavioral instructions and distancing rules due to cognitive deficits [124,131]. Dementia patients frequently live in nursing homes where a higher risk for infection with the virus was present in many areas [124]. COVID-19 disease in dementia patients often appeared atypical presenting foremost with delirium/confusion and few infectious symptoms [129,132]. Confusion and mood and behavioral disturbances persisted in 19.2% of survivors [129].
An analysis of the network-based relationship for the gene/protein sets between virus and host factors as well as different neurological diseases in an interactome network model showed close proximity between COVID-19 and cognitive decline as well as AD and PD [13].
Postmortem studies demonstrated that ACE2-expression was increased in the brains of AD patients. Especially in severe dementia, ACE2-expression was elevated, which could lead to a higher susceptibility for COVID-19 [123,124,133].
Ischemic white matter damage occurs early in AD contributing to the progression of dementia. COVID-19 can induce vascular lesions due to hypercoagulability and can be expected to accelerate disease progression in AD patients [123,134].
It was hypothesized that amyloid-β, the protein implicated in AD development, is an antimicrobial peptide involved in fighting cerebral SARS-CoV-2 infection as previously described for α-synuclein in PD [123,135]. It could be speculated that amyloid-β is upregulated as a defense mechanism during infection leading to an overactivation with pathological deposition of amyloid-β in the long run [123,135].
Apoε4, an established risk factor for AD, was also recognized as a prominent risk factor for COVID-19, potentially linking the two pathophysiologies [136]. COVID-19 severity could be statistically predicted by the Apoε4 genotype [136]. In human iPSCs models with Apoε4 genotype, neurons and astrocytes were more susceptible to SARS-CoV-2 infection than non-Apoε4 cells and brain organoids [12].
Another point of overlap is IL-6, which was shown to be elevated in COVID-19 and was also considered a biomarker with prognostic value in AD [49,123,137].
SARS-CoV-2 is likely to disturb autonomic functions in vagal regulation centers in the brainstem [127]. In AD, autonomic functions are impaired as well, since higher cardiac sympathetic function and lower parasympathetic function are reported in patients [127]. Therefore, noninvasive (auricular) vagal nerve stimulation is discussed as a therapeutic strategy for AD as well as severe COVID-19, since a downregulation of inflammatory pathways (reduction of IL-6 levels) is expected as a result [127]. Supporting this theory, transauricular vagal nerve stimulation was able to reduce cognitive dysfunction in a preclinical murine model of AD [138].
AD leads to alterations in calcium homeostasis in the brain; RNA-viruses use the same mechanism to facilitate viral replication [124]. Therefore, viral replication might be easier in the brain of AD patients where calcium homeostasis is already abnormal [124].
There is an association between AD and diabetes type II, which elevates the risk to develop AD [139]. AD and diabetes type II were both found to be strong risk factors for COVID-19 endangering those patient groups particularly and proposing a mechanistic link between these diseases that could explain an overlapping pathophysiology [139].
Take home messages of Chapter 3 (Section 5):
  • Viruses have different strategies to take control over host cellular functions, for example, impairing autophagy and mitochondrial or lysosomal properties, whose dysfunction has been implicated in neurodegenerative diseases.
  • Neuroinflammatory alterations due to COVID-19 such as elevated IL-6 levels or activation of NF-κB might trigger/accelerate the development of PD.
  • Direct CNS invasion by SARS-CoV-2 might also lead to induction of neurodegenerative cascades in strategic areas.
  • COVID-19 can lead to acute and persisting cognitive deficits, although some of those might be due to ARDS.
  • Dementia and Apoε4 genotype are strong risk factors for COVID-19 and its associated mortality.

6. Chapter 4

Parkinson’s Disease and COVID-19: Effects on PD Symptoms, Psychological and Social Aspects

The impact of the pandemic on aspects of daily life was massive for the entire world population, and patients with chronic diseases in need for regular care were especially affected. An extensive analysis of worldwide studies (210,419 participants total) showed that acute care for neurological disorders in general was disrupted due to the pandemic in 47.1% of cases [140]. The impact on PD patients was differentially described, as specific problems occurred in relation to pandemic-associated restrictions. Psychological issues as well as aspects regarding care and supply of medication were found to be most burdensome in this cohort [140].
COVID-19 has the ability to alter the pharmacodynamics of levodopa also due to diarrhea, which is a common symptom of COVID-19 [141]. This leads to motor fluctuations in infected PD patients [141]. PD patients suffering from COVID-19 often developed a post-COVID syndrome (85.2%) consisting of worsened motor functions, increased daily levodopa dose requirement, fatigue, loss of concentration and sleep disturbances [142].
However, subjective worsening of motor and non-motor symptoms of uninfected PD patients during the time of the pandemic was also recorded in different studies [143,144]. New behavioral symptoms were observed in 26% of PD patients in an Italian cross-sectional study [144]. PD patients reported feeling lonely and deprived of the support and communication with their physician [143].
It was hypothesized that dopamine-dependent adaptation is a requirement for successful coping; thus, PD patients are cognitively less flexible and can have more difficulties to adapt to new environments [145,146]. Therefore, the pandemic may lead to a relevant amount of stress in PD patients who are forced to adapt to a new environment quickly. Psychological stress was shown to worsen PD symptoms as well as the efficacy of dopaminergic medication especially on the tremor [145,147]. This could be an explanation for symptom exacerbation in PD patients during the pandemic.
It was found that 103 PD patients reported four main problems in the first Italian lockdown: 1. fear of contracting corona, 2. reduction of physical activity, 3. not being able to access support services and clinics and 4. reduction of socialization [148]. There was an objective reduction of physical activity, measured by a smartphone application, as most PD patients failed to meet 30 min of activity per day [149]. This was aggravated further in 44% during confinement [149]. It is well known that physical activity and training is an important treatment strategy in PD to maintain motor functions and independence, so the deprivation of physical activity during a lockdown can be suspected to lead to symptom progression and loss of independence [149].
Moreover, 66% of PD patients in a large cohort at Columbia University reported mood and sleep disturbances in the face of the pandemic; depression and insomnia were the most frequently reported psychiatric symptoms in multiple other studies as well [150,151,152,153,154]. A Chinese study revealed that PD patients had more sleep disturbances and anxiety than healthy controls and that these symptoms were independently associated with an exacerbation of other PD symptoms [154]. Sleep problems were also associated with a poorer quality of life [153].
Mindfulness-based interventions were shown to reduce depression and anxiety, improve motor function and strengthen resilience [145]. As this can be accomplished virtually, it appears to be a useful treatment strategy now and for the future [145].
The hours of caregiving increased dramatically during the pandemic. Care was mostly provided by family members [155,156]. Caregiver burden was increased during the COVID-19 era [144]. Interestingly, Montanaro et al. and others showed that anxiety and depression were frequent in both PD patients and their caregivers [157,158]. Depression was observed in 35% of PD patients and 21.7% of caregivers; 39% of PD patients and 40% of caregivers suffered from anxiety [158]. Therefore, caregivers should receive more support particularly during this pandemic to cope with their own burden and the neuropsychiatric symptoms of their relatives [159].
However, COVID-19 does not only have an impact on the PD symptoms; it was also discussed that pre-existing PD can elevate the risk of mortality or case fatality when an infection with SARS-CoV-2 occurs. The data on this topic are controversial (subsumed in Table 1).
A detailed review by Fearon et al. summed up that COVID-19 mortality is probably not increased in PD patients who tended to experience less dyspnea during the infection, were completely asymptomatic more often and less likely to require hospitalization [141,160,165,166,167]. The duration of ICU stays/hospitalization and ventilation did also not differ in PD and non-PD COVID-19 patients in a large analysis of German inpatients [165]. An Italian study compared COVID-19 patients with PD to COVID-19 patients without PD and found no difference in mortality (5.7% of PD COVID-19 patients died vs. 7.6% of non-PD COVID-19 patients) [160].
This trend could be supported by the hypothesis that amantadine and entacapone might have protective value against COVID-19, which was suggested by different studies [172,173,174]. However, a systematic review about a total of 1061 PD patients with confirmed COVID-19 showed a higher hospitalization rate, case fatality and mortality for these patients than that for non-PD COVID-19 patients [175]. A limitation of this study was the missing age matching, which is likely to influence the result, as age is one of the most established risk factors for case fatality and mortality of COVID-19 [175,176]. An American study compared 78,355 non-PD COVID-19 patients to 694 COVID-19 patients with PD and found an increased mortality even after adjusting and matching to age and sex [169]. A multicentric German study showed that prevalence and mortality of COVID-19 was higher in PD than in non-PD inpatients [168].
Apparently, these data are inconclusive and a definite suggestion on whether PD patients are more at risk for a (severe) COVID-19 infection cannot be made at this point. It should be noted that PD patients who suffer from COVID-19 infection are likely to present with atypical symptoms such as mood changes, fatigue, joint pain, flushing and exacerbation of PD-symptoms, which can complicate the diagnosis of SARS-CoV-2 infection [177].
Take home messages of Chapter 4 (Section 6):
  • Post-COVID-syndrome, altered pharmacodynamics of levodopa and worsening of motor-symptoms are common in PD patients with COVID-19.
  • Even uninfected PD patients often suffered from subjective worsening of motor and non-motor symptoms, reduced physical activity, as well as increased stress, anxiety and depression.
  • Whether PD elevates the risk of COVID-19 mortality is not clear yet, since data on this topic is inconclusive.

7. Concluding Remarks

The overview presented in this review underlines that COVID-19 has a cerebral/neurological impact. Whether SARS-CoV-2 can enter the CNS and impose direct neuronal damage or whether neurological symptoms are rather due to secondary effects cannot be differentiated with certainty. The influence of the infection on neurodegenerative diseases is also not clear yet, but various pathophysiological theories exist linking COVID-19 to neurodegeneration and making it appear likely that the pandemic can have an (accelerating) influence on neurodegenerative diseases such as AD and PD. The same was proposed for other viral infections in the past, though a clear triggering/accelerating influence of viral infections on neurodegeneration could only be demonstrated for few viruses such as HCV and HIV so far. Monitoring recovered COVID-19 patients and especially patients with neurodegenerative diseases and COVID-19 will hopefully answer some of these questions in the future. Such a prospective investigation also withholds the potential to learn more about the neurodegenerative pathophysiology and develop new strategies for disease-modifying treatments.

Author Contributions

Conceptualization: L.K.; writing—original draft preparation: L.K.; writing—review and editing: F.W., G.U.H. and M.K.H.; visualization: M.K.H. All authors have read and agreed to the published version of the manuscript.

Funding

L.K. was supported by PRACTIS—Clinician Scientist Program of Hannover Medical School, funded by the German Research Foundation (DFG, ME 3696/3-1). Otherwise, the research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Arbour, N.; Côté, G.; Lachance, C.; Tardieu, M.; Cashman, N.R.; Talbot, P.J. Acute and persistent infection of human neural cell lines by human coronavirus OC43. J. Virol. 1999, 73, 3338–3350. [Google Scholar] [CrossRef] [Green Version]
  2. Arbour, N.; Day, R.; Newcombe, J.; Talbot, P.J. Neuroinvasion by Human Respiratory Coronaviruses. J. Virol. 2000, 74, 8913–8921. [Google Scholar] [CrossRef] [Green Version]
  3. Desforges, M.; le Coupanec, A.; Brison, É.; Meessen-Pinard, M.; Talbot, P.J. Neuroinvasive and neurotropic human respiratory coronaviruses: Potential neurovirulent agents in humans. Adv. Exp. Med. Biol. 2014, 807, 75–96. [Google Scholar] [CrossRef] [Green Version]
  4. Jacomy, H.; Fragoso, G.; Almazan, G.; Mushynski, W.E.; Talbot, P.J. Human coronavirus OC43 infection induces chronic encephalitis leading to disabilities in BALB/C mice. Virology 2006, 349, 335–346. [Google Scholar] [CrossRef] [PubMed]
  5. Netland, J.; Meyerholz, D.K.; Moore, S.; Cassell, M.; Perlman, S. Severe Acute respiratory syndrome coronavirus infection causes neuronal death in the absence of encephalitis in mice transgenic for human ACE2. J. Virol. 2008, 82, 7264–7275. [Google Scholar] [CrossRef] [Green Version]
  6. Desforges, M.; le Coupanec, A.; Dubeau, P.; Bourgouin, A.; Lajoie, L.; Dubé, M.; Talbot, P.J. Human coronaviruses and other respiratory viruses: Underestimated opportunistic pathogens of the central nervous system? Viruses 2019, 12, 14. [Google Scholar] [CrossRef] [Green Version]
  7. Li, M.Y.; Li, L.; Zhang, Y.; Wang, X.S. Expression of the SARS-CoV-2 cell receptor gene ACE2 in a wide variety of human tissues. Infect. Dis. Poverty 2020, 9, 45. [Google Scholar] [CrossRef]
  8. Baig, A.M.; Khaleeq, A.; Ali, U.; Syeda, H. Evidence of the COVID-19 virus targeting the CNS: Tissue distribution, host-virus interaction, and proposed neurotropic mechanisms. ACS Chem. Neurosci. 2020, 11, 995–998. [Google Scholar] [CrossRef] [Green Version]
  9. Singh, H.O.; Singh, A.; Khan, A.A.; Gupta, V. Immune mediating molecules and pathogenesis of COVID-19-associated neurological disease. Microb. Pathog. 2021, 158, 105023. [Google Scholar] [CrossRef] [PubMed]
  10. Briguglio, M.; Bona, A.; Porta, M.; Dell’Osso, B.; Pregliasco, F.E.; Banfi, G. Disentangling the hypothesis of host dysosmia and SARS-CoV-2: The bait symptom that hides neglected neurophysiological routes. Front. Physiol. 2020, 11, 671. [Google Scholar] [CrossRef] [PubMed]
  11. Erickson, M.A.; Rhea, E.M.; Knopp, R.C.; Banks, W.A. Interactions of sars-cov-2 with the blood–brain barrier. Int. J. Mol. Sci. 2021, 22, 2681. [Google Scholar] [CrossRef]
  12. Wang, C.; Zhang, M.; Garcia, G.; Tian, E.; Cui, Q.; Chen, X.; Sun, G.; Wang, J.; Arumugaswami, V.; Shi, Y. ApoE-isoform-dependent SARS-CoV-2 neurotropism and cellular response. Cell Stem Cell 2021, 28, 331–342.e5. [Google Scholar] [CrossRef]
  13. Zhou, Y.; Xu, J.; Hou, Y.; Leverenz, J.B.; Kallianpur, A.; Mehra, R.; Liu, Y.; Yu, H.; Pieper, A.A.; Jehi, L.; et al. Network medicine links SARS-CoV-2/COVID-19 infection to brain microvascular injury and neuroinflammation in dementia-like cognitive impairment. Alzheimer’s Res. Ther. 2021, 13, 110. [Google Scholar] [CrossRef] [PubMed]
  14. Achar, A.; Ghosh, C. COVID-19-associated neurological disorders: The potential route of CNS invasion and blood-brain relevance. Cells 2020, 9, 2360. [Google Scholar] [CrossRef] [PubMed]
  15. Chaudhury, S.S.; Sinha, K.; Majumder, R.; Biswas, A.; das Mukhopadhyay, C. COVID-19 and central nervous system interplay: A big picture beyond clinical manifestation. J. Biosci. 2021, 46, 47. [Google Scholar] [CrossRef]
  16. Kaundal, R.K.; Kalvala, A.K.; Kumar, A. Neurological implications of COVID-19: Role of redox imbalance and mitochondrial dysfunction. Mol. Neurobiol. 2021, 58, 4575–4587. [Google Scholar] [CrossRef]
  17. Tavčar, P.; Potokar, M.; Kolenc, M.; Korva, M.; Avšič-Županc, T.; Zorec, R.; Jorgačevski, J. Neurotropic viruses, astrocytes, and COVID-19. Front. Cell. Neurosci. 2021, 15, 662578. [Google Scholar] [CrossRef]
  18. Welcome, M.O.; Mastorakis, N.E. Neuropathophysiology of coronavirus disease 2019: Neuroinflammation and blood brain barrier disruption are critical pathophysiological processes that contribute to the clinical symptoms of SARS-CoV-2 infection. Inflammopharmacology 2021, 29, 939–963. [Google Scholar] [CrossRef] [PubMed]
  19. Wang, F.; Kream, R.M.; Stefano, G.B. Long-term respiratory and neurological sequelae of COVID-19. Med. Sci. Monit. 2020, 26, e928996. [Google Scholar] [CrossRef]
  20. McCray, P.B.; Pewe, L.; Wohlford-Lenane, C.; Hickey, M.; Manzel, L.; Shi, L.; Netland, J.; Jia, H.P.; Halabi, C.; Sigmund, C.D.; et al. Lethal infection of K18- hACE2 mice infected with severe acute respiratory syndrome coronavirus. J. Virol. 2007, 81, 813–821. [Google Scholar] [CrossRef] [Green Version]
  21. Barnett, E.M.; Cassell, M.D.; Perlman, S. Two neurotropic viruses, herpes simplex virus type 1 and mouse hepatitis virus, spread along different neural pathways from the main olfactory bulb. Neuroscience 1993, 57, 1007. [Google Scholar] [CrossRef]
  22. Mullol, J.; Alobid, I.; Mariño-Sánchez, F.; Izquierdo-Domínguez, A.; Marin, C.; Klimek, L.; Wang, D.Y.; Liu, Z. The loss of smell and taste in the COVID-19 outbreak: A tale of many countries. Curr. Allergy Asthma Rep. 2020, 20, 61. [Google Scholar] [CrossRef]
  23. Moein, S.T.; Hashemian, S.M.R.; Mansourafshar, B.; Khorram-Tousi, A.; Tabarsi, P.; Doty, R.L. Smell dysfunction: A biomarker for COVID-19. Int. Forum Allergy Rhinol. 2020, 10, 944–950. [Google Scholar] [CrossRef] [PubMed]
  24. Rebholz, H.; Braun, R.J.; Ladage, D.; Knoll, W.; Kleber, C.; Hassel, A.W. Loss of olfactory function—Early indicator for COVID-19, Other viral infections and neurodegenerative disorders. Front. Neurol. 2020, 11, 569333. [Google Scholar] [CrossRef] [PubMed]
  25. Desai, M.; Oppenheimer, J. The importance of considering olfactory dysfunction during the COVID-19 pandemic and in clinical practice. J. Allergy Clin. Immunol. Pract. 2021, 9, 7–12. [Google Scholar] [CrossRef] [PubMed]
  26. Lechien, J.R.; Chiesa-Estomba, C.M.; de Siati, D.R.; Horoi, M.; le Bon, S.D.; Rodriguez, A.; Dequanter, D.; Blecic, S.; el Afia, F.; Distinguin, L.; et al. Olfactory and gustatory dysfunctions as a clinical presentation of mild-to-moderate forms of the coronavirus disease (COVID-19): A multicenter European study. Eur. Arch. Oto-Rhino-Laryngol. 2020, 277, 2251–2261. [Google Scholar] [CrossRef] [PubMed]
  27. Kandemirli, S.G.; Altundag, A.; Yildirim, D.; Tekcan Sanli, D.E.; Saatci, O. Olfactory bulb MRI and paranasal sinus CT findings in persistent COVID-19 anosmia. Acad. Radiol. 2021, 28, 28–35. [Google Scholar] [CrossRef]
  28. Brann, D.H.; Tsukahara, T.; Weinreb, C.; Lipovsek, M.; van den Berge, K.; Gong, B.; Chance, R.; Macaulay, I.C.; Chou, H.-J.; Fletcher, R.B.; et al. Non-neuronal expression of SARS-CoV-2 entry genes in the olfactory system suggests mechanisms underlying COVID-19-associated anosmia. Sci. Adv. 2020, 6, eabc5801. [Google Scholar] [CrossRef] [PubMed]
  29. Porta-Etessam, J.; Núñez-Gil, I.J.; González García, N.; Fernandez-Perez, C.; Viana-Llamas, M.C.; Eid, C.M.; Romero, R.; Molina, M.; Uribarri, A.; Becerra-Muñoz, V.M.; et al. COVID-19 anosmia and gustatory symptoms as a prognosis factor: A subanalysis of the HOPE COVID-19 (Health Outcome Predictive Evaluation for COVID-19) registry. Infection 2021, 49, 677–684. [Google Scholar] [CrossRef] [PubMed]
  30. Pouga, L. Encephalitic syndrome and anosmia in COVID-19: Do these clinical presentations really reflect SARS-CoV-2 neurotropism? A theory based on the review of 25 COVID-19 cases. J. Med. Virol. 2021, 93, 550–558. [Google Scholar] [CrossRef]
  31. Buzhdygan, T.P.; DeOre, B.J.; Baldwin-Leclair, A.; Bullock, T.A.; McGary, H.M.; Khan, J.A.; Razmpour, R.; Hale, J.F.; Galie, P.A.; Potula, R.; et al. The SARS-CoV-2 spike protein alters barrier function in 2D static and 3D microfluidic in-vitro models of the human blood–brain barrier. Neurobiol. Dis. 2020, 146, 105131. [Google Scholar] [CrossRef]
  32. Rhea, E.M.; Logsdon, A.F.; Hansen, K.M.; Williams, L.M.; Reed, M.J.; Baumann, K.K.; Holden, S.J.; Raber, J.; Banks, W.A.; Erickson, M.A. The S1 protein of SARS-CoV-2 crosses the blood–brain barrier in mice. Nat. Neurosci. 2021, 24, 368–378. [Google Scholar] [CrossRef]
  33. Eldeeb, M.A.; Hussain, F.S.; Siddiqi, Z.A. COVID-19 infection may increase the risk of parkinsonism—Remember the Spanish flu? Cytokine Growth Factor Rev. 2020, 54, 6–7. [Google Scholar] [CrossRef] [PubMed]
  34. Caramaschi, S.; Kapp, M.E.; Miller, S.E.; Eisenberg, R.; Johnson, J.; Epperly, G.; Maiorana, A.; Silvestri, G.; Giannico, G.A. Histopathological findings and clinicopathologic correlation in COVID-19: A systematic review. Mod. Pathol. 2021, 34, 1614–1633. [Google Scholar] [CrossRef] [PubMed]
  35. Pajo, A.T.; Espiritu, A.I.; Apor, A.D.A.O.; Jamora, R.D.G. Neuropathologic findings of patients with COVID-19: A systematic review. Neurol. Sci. 2021, 42, 1255–1266. [Google Scholar] [CrossRef]
  36. Boroujeni, M.E.; Simani, L.; Bluyssen, H.A.R.; Samadikhah, H.R.; Zamanlui Benisi, S.; Hassani, S.; Akbari Dilmaghani, N.; Fathi, M.; Vakili, K.; Mahmoudiasl, G.R.; et al. Inflammatory response leads to neuronal death in human post-mortem cerebral cortex in patients with COVID-19. ACS Chem. Neurosci. 2021, 12, 2143–2150. [Google Scholar] [CrossRef] [PubMed]
  37. Paniz-Mondolfi, A.; Bryce, C.; Grimes, Z.; Gordon, R.E.; Reidy, J.; Lednicky, J.; Sordillo, E.M.; Fowkes, M. Central nervous system involvement by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). J. Med. Virol. 2020, 92, 699–702. [Google Scholar] [CrossRef] [Green Version]
  38. Pacheco-Herrero, M.; Soto-Rojas, L.O.; Harrington, C.R.; Flores-Martinez, Y.M.; Villegas-Rojas, M.M.; León-Aguilar, A.M.; Martínez-Gómez, P.A.; Campa-Córdoba, B.B.; Apátiga-Pérez, R.; Corniel-Taveras, C.N.; et al. Elucidating the neuropathologic mechanisms of SARS-CoV-2 infection. Front. Neurol. 2021, 12, 660087. [Google Scholar] [CrossRef] [PubMed]
  39. Poloni, T.E.; Medici, V.; Moretti, M.; Visonà, S.D.; Cirrincione, A.; Carlos, A.F.; Davin, A.; Gagliardi, S.; Pansarasa, O.; Cereda, C.; et al. COVID-19-related neuropathology and microglial activation in elderly with and without dementia. Brain Pathol. 2021, 31, e12997. [Google Scholar] [CrossRef]
  40. Serrano, G.E.; Walker, J.E.; Arce, R.; Glass, M.J.; Vargas, D.; Sue, L.I.; Intorcia, A.J.; Nelson, C.M.; Oliver, J.; Papa, J.; et al. Mapping of SARS-CoV-2 Brain INVASION AND HISTOPATHOLOGY in COVID-19 Disease. medRxiv 2021. the preprint server for health sciences. [Google Scholar] [CrossRef]
  41. Garcia, M.A.; Barreras, P.V.; Lewis, A.; Pinilla, G.; Sokoll, L.J.; Kickler, T.; Mostafa, H.; Caturegli, M.; Moghekar, A.; Fitzgerald, K.C.; et al. Cerebrospinal fluid in COVID-19 neurological complications: Neuroaxonal damage, anti-SARS-Cov2 antibodies but no evidence of cytokine storm. J. Neurol. Sci. 2021, 427, 117517. [Google Scholar] [CrossRef]
  42. Sun, B.; Tang, N.; Peluso, M.J.; Iyer, N.S.; Torres, L.; Donatelli, J.L.; Munter, S.E.; Nixon, C.C.; Rutishauser, R.L.; Rodriguez-Barraquer, I.; et al. Characterization and biomarker analyses of post-covid-19 complications and neurological manifestations. Cells 2021, 10, 386. [Google Scholar] [CrossRef]
  43. Trigo, J.; García-Azorín, D.; Sierra-Mencía, Á.; Tamayo-Velasco, Á.; Martínez-Paz, P.; Tamayo, E.; Guerrero, A.L.; Gonzalo-Benito, H. Cytokine and interleukin profile in patients with headache and COVID-19: A pilot, CASE-control, study on 104 patients. J. Headache Pain 2021, 22, 51. [Google Scholar] [CrossRef]
  44. Alexopoulos, H.; Magira, E.; Bitzogli, K.; Kafasi, N.; Vlachoyiannopoulos, P.; Tzioufas, A.; Kotanidou, A.; Dalakas, M.C. Anti-SARS-CoV-2 antibodies in the CSF, blood-brain barrier dysfunction, and neurological outcome: Studies in 8 stuporous and comatose patients. Neurol. Neuroimmunol. Neuroinflamm. 2020, 7, e893. [Google Scholar] [CrossRef]
  45. Hosp, J.A.; Dressing, A.; Blazhenets, G.; Bormann, T.; Rau, A.; Schwabenland, M.; Thurow, J.; Wagner, D.; Waller, C.; Niesen, W.D.; et al. Cognitive impairment and altered cerebral glucose metabolism in the subacute stage of COVID-19. Brain 2021, 144, 1263–1276. [Google Scholar] [CrossRef] [PubMed]
  46. Huang, Y.H.; Jiang, D.; Huang, J.T. SARS-CoV-2 detected in cerebrospinal fluid by PCR in a case of COVID-19 encephalitis. Brain Behav. Immun. 2020, 87, 149. [Google Scholar] [CrossRef]
  47. Luis, M.B.; Liguori, N.F.; López, P.A.; Alonso, R. SARS-CoV-2 RNA detection in cerebrospinal fluid: Presentation of two cases and review of literature. Brain Behav. Immun. Health 2021, 15, 100282. [Google Scholar] [CrossRef] [PubMed]
  48. Moriguchi, T.; Harii, N.; Goto, J.; Harada, D.; Sugawara, H.; Takamino, J.; Ueno, M.; Sakata, H.; Kondo, K.; Myose, N.; et al. A first case of meningitis/encephalitis associated with SARS-Coronavirus-2. Int. J. Infect. Dis. 2020, 94, 55–58. [Google Scholar] [CrossRef] [PubMed]
  49. Kanberg, N.; Ashton, N.J.; Andersson, L.M.; Yilmaz, A.; Lindh, M.; Nilsson, S.; Price, R.W.; Blennow, K.; Zetterberg, H.; Gisslén, M. Neurochemical evidence of astrocytic and neuronal injury commonly found in COVID-19. Neurology 2020, 95, e1754–e1759. [Google Scholar] [CrossRef] [PubMed]
  50. Lewis, A.; Frontera, J.; Placantonakis, D.G.; Lighter, J.; Galetta, S.; Balcer, L.; Melmed, K.R. Cerebrospinal fluid in COVID-19: A systematic review of the literature. J. Neurol. Sci. 2021, 421, 117316. [Google Scholar] [CrossRef] [PubMed]
  51. Placantonakis, D.G.; Aguero-Rosenfeld, M.; Flaifel, A.; Colavito, J.; Inglima, K.; Zagzag, D.; Snuderl, M.; Louie, E.; Frontera, J.A.; Lewis, A. SARS-CoV-2 Is not detected in the cerebrospinal fluid of encephalopathic COVID-19 patients. Front. Neurol. 2020, 11, 587384. [Google Scholar] [CrossRef] [PubMed]
  52. Garcia, M.A.; Barreras, P.V.; Lewis, A.; Pinilla, G.; Sokoll, L.J.; Kickler, T.; Mostafa, H.; Caturegli, M.; Moghekar, A.; Fitzgerald, K.C.; et al. Cerebrospinal fluid in COVID-19 neurological complications: No cytokine storm or neuroinflammation. medRxiv 2021. the preprint server for health sciences. [Google Scholar] [CrossRef]
  53. Hoffman, L.A.; Vilensky, J.A. Encephalitis lethargica: 100 years after the epidemic. Brain 2017, 140, 2246–2251. [Google Scholar] [CrossRef]
  54. Meng, L.; Shen, L.; Ji, H.F. Impact of infection on risk of Parkinson’s disease: A quantitative assessment of case-control and cohort studies. J. NeuroVirology 2019, 25, 221–228. [Google Scholar] [CrossRef] [PubMed]
  55. Bu, X.-L.; Wang, X.; Xiang, Y.; Shen, L.L.; Wang, Q.H.; Liu, Y.H.; Jiao, S.S.; Wang, Y.R.; Cao, H.Y.; Yi, X.; et al. The association between infectious burden and Parkinson’s disease: Acase-control study. Parkinsonism Relat. Disord. 2015, 21, 877–881. [Google Scholar] [CrossRef] [PubMed]
  56. Lai, S.W.; Lin, C.H.; Lin, H.F.; Lin, C.L.; Lin, C.C.; Liao, K.F. Herpes zoster correlates with increased risk of Parkinson’s disease in older people A population-based cohort study in Taiwan. Medicine 2017, 96, e6075. [Google Scholar] [CrossRef]
  57. Limphaibool, N.; Iwanowski, P.; Holstad, M.J.V.; Kobylarek, D.; Kozubski, W. Infectious etiologies of Parkinsonism: Pathomechanisms and clinical implications. Front. Neurol. 2019, 10, 652. [Google Scholar] [CrossRef]
  58. Lin, W.Y.; Lin, M.S.; Weng, Y.H.; Yeh, T.H.; Lin, Y.S.; Fong, P.Y.; Wu, Y.R.; Lu, C.S.; Chen, R.S.; Huang, Y.Z. Association of antiviral therapy with risk of parkinson disease in patients with chronic Hepatitis C virus infection. JAMA Neurol. 2019, 76, 1019–1027. [Google Scholar] [CrossRef] [Green Version]
  59. Wijarnpreecha, K.; Chesdachai, S.; Jaruvongvanich, V.; Ungprasert, P. Hepatitis C virus infection and risk of Parkinson’s disease: A systematic review and meta-analysis. Eur. J. Gastroenterol. Hepatol. 2018, 30, 9–13. [Google Scholar] [CrossRef]
  60. Tzeng, N.S.; Chung, C.H.; Lin, F.H.; Chiang, C.P.; Yeh, C.-B.; Huang, S.Y.; Lu, R.B.; Chang, H.A.; Kao, Y.C.; Yeh, H.W.; et al. Anti-herpetic Medications and reduced risk of dementia in patients with herpes simplex virus infections—A nationwide, population-based cohort study in Taiwan. Neurotherapeutics 2018, 15, 417–429. [Google Scholar] [CrossRef] [Green Version]
  61. Wu, W.Y.Y.; Kang, K.H.; Chen, S.L.S.; Chiu, S.Y.H.; Yen, A.M.F.; Fann, J.C.Y.; Su, C.W.; Liu, H.C.; Lee, C.Z.; Fu, W.M.; et al. Hepatitis C virus infection: A risk factor for Parkinson’s disease. J. Viral Hepat. 2015, 22, 784–791. [Google Scholar] [CrossRef]
  62. Sadasivan, S.; Zanin, M.; O’Brien, K.; Schultz-Cherry, S.; Smeyne, R.J. Induction of microglia activation after infection with the non-neurotropic A/CA/04/2009 H1N1 influenza virus. PLoS ONE 2015, 10, e0124047. [Google Scholar] [CrossRef] [Green Version]
  63. Jang, H.; Boltz, D.; Sturm-Ramirez, K.; Shepherd, K.R.; Jiao, Y.; Webster, R.; Smeyne, R.J. Highly pathogenic H5N1 influenza virus can enter the central nervous system and induce neuroinflammation and neurodegeneration. Proc. Natl. Acad. Sci. USA 2009, 106, 14063–14068. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Rohn, T.T.; Catlin, L.W. Immunolocalization of influenza a virus and markers of inflammation in the human Parkinson’s disease brain. PLoS ONE 2011, 6, e20495. [Google Scholar] [CrossRef] [PubMed]
  65. Cocoros, N.M.; Svensson, E.; Szépligeti, S.K.; Vestergaard, S.V.; Szentkúti, P.; Thomsen, R.W.; Borghammer, P.; Sørensen, H.T.; Henderson, V.W. Long-term risk of Parkinson disease following influenza and other infections. JAMA Neurol. 2021, 78, 1461–1470. [Google Scholar] [CrossRef]
  66. Murgod, U.A.; Muthane, U.B.; Ravi, V.; Radhesh, S.; Desai, A. Persistent movement disorders following Japanese encephalitis. Neurology 2001, 57, 2313–2315. [Google Scholar] [CrossRef]
  67. Beatman, E.L.; Massey, A.; Shives, K.D.; Burrack, K.S.; Chamanian, M.; Morrison, T.E.; Beckham, J.D. Alpha-synuclein expression restricts RNA viral infections in the brain. J. Virol. 2016, 90, 2767–2782. [Google Scholar] [CrossRef] [Green Version]
  68. Sejvar, J.J. Clinical manifestations and outcomes of West Nile virus infection. Viruses 2014, 6, 606–623. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Eimer, W.A.; Vijaya Kumar, D.K.; Navalpur Shanmugam, N.K.; Rodriguez, A.S.; Mitchell, T.; Washicosky, K.J.; György, B.; Breakefield, X.O.; Tanzi, R.E.; Moir, R.D. Alzheimer’s disease-associated β-Amyloid is rapidly seeded by herpesviridae to protect against brain infection. Neuron 2018, 99, 56–63.e3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Wainberg, M.; Luquez, T.; Koelle, D.M.; Readhead, B.; Johnston, C.; Darvas, M.; Funk, C.C. The viral hypothesis: How herpesviruses may contribute to Alzheimer’s disease. Mol. Psychiatry 2021. online ahead of print. [Google Scholar] [CrossRef]
  71. Duarte, L.F.; Farías, M.A.; Álvarez, D.M.; Bueno, S.M.; Riedel, C.A.; González, P.A. Herpes simplex virus type 1 infection of the central nervous system: Insights into proposed interrelationships with neurodegenerative disorders. Front. Cell. Neurosci. 2019, 13, 46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Caggiu, E.; Paulus, K.; Arru, G.; Piredda, R.; Sechi, G.P.; Sechi, L.A. Humoral cross reactivity between α-synuclein and herpes simplex-1 epitope in Parkinson’s disease, a triggering role in the disease? J. Neuroimmunol. 2016, 291, 110–114. [Google Scholar] [CrossRef] [PubMed]
  73. Sulzer, D.; Alcalay, R.N.; Garretti, F.; Cote, L.; Kanter, E.; Agin-Liebes, J.; Liong, C.; McMurtrey, C.; Hildebrand, W.H.; Mao, X.; et al. T cells from patients with Parkinson’s disease recognize α-synuclein peptides. Nature 2017, 546, 656–661. [Google Scholar] [CrossRef] [Green Version]
  74. Sommer, A.; Maxreiter, F.; Krach, F.; Fadler, T.; Grosch, J.; Maroni, M.; Graef, D.; Eberhardt, E.; Riemenschneider, M.J.; Yeo, G.W.; et al. Th17 lymphocytes induce neuronal cell death in a human iPSC-based model of Parkinson’s disease. Cell Stem Cell 2018, 23, 123–131.e6. [Google Scholar] [CrossRef] [Green Version]
  75. Gate, D.; Tapp, E.; Leventhal, O.; Shahid, M.; Nonninger, T.J.; Yang, A.C.; Strempfl, K.; Unger, M.S.; Fehlmann, T.; Oh, H.; et al. CD4+ T cells contribute to neurodegeneration in Lewy body dementia. Science 2021, 374, 868–874. [Google Scholar] [CrossRef]
  76. Idrees, D.; Kumar, V. SARS-CoV-2 spike protein interactions with amyloidogenic proteins: Potential clues to neurodegeneration. Biochem. Biophys. Res. Commun. 2021, 554, 94–98. [Google Scholar] [CrossRef]
  77. Tavassoly, O.; Safavi, F.; Tavassoly, I. Seeding brain protein aggregation by SARS-CoV-2 as a possible long-term complication of COVID-19 infection. ACS Chem. Neurosci. 2020, 11, 3704–3706. [Google Scholar] [CrossRef]
  78. Ezzat, K.; Pernemalm, M.; Pålsson, S.; Roberts, T.C.; Järver, P.; Dondalska, A.; Bestas, B.; Sobkowiak, M.J.; Levänen, B.; Sköld, M.; et al. The viral protein corona directs viral pathogenesis and amyloid aggregation. Nat. Commun. 2019, 10, 2331. [Google Scholar] [CrossRef] [Green Version]
  79. Liu, S.; Hossinger, A.; Heumüller, S.-E.; Hornberger, A.; Buravlova, O.; Konstantoulea, K.; Müller, S.A.; Paulsen, L.; Rousseau, F.; Schymkowitz, J.; et al. Highly efficient intercellular spreading of protein misfolding mediated by viral ligand-receptor interactions. Nat. Commun. 2021, 12, 5739. [Google Scholar] [CrossRef] [PubMed]
  80. Lippi, A.; Domingues, R.; Setz, C.; Outeiro, T.F.; Krisko, A. SARS-CoV-2: At the crossroad between aging and neurodegeneration. Mov. Disord. 2020, 35, 716–720. [Google Scholar] [CrossRef] [Green Version]
  81. Singh, K.; Chen, Y.C.; Judy, J.T.; Seifuddin, F.; Tunc, I.; Pirooznia, M. Network analysis and transcriptome profiling identify autophagic and mitochondrial dysfunctions in SARS-CoV-2 infection. bioRxiv 2020. [Google Scholar] [CrossRef]
  82. Sfera, A.; Osorio, C.; Maguire, G.; Rahman, L.; Afzaal, J.; Cummings, M.; Maldonado, J.C. COVID-19, ferrosenescence and neurodegeneration, a mini-review. Prog. Neuro Psychopharmacol. Biol. Psychiatry 2021, 109, 110230. [Google Scholar] [CrossRef] [PubMed]
  83. Fishman, P.S.; Gass, J.S.; Swoveland, P.T.; Lavi, E.; Highkin, M.K.; Weiss, S.R. Infection of the basal ganglia by a murine coronavirus. Science 1985, 229, 877–879. [Google Scholar] [CrossRef] [PubMed]
  84. Fazzini, E.; Fleming, J.; Fahn, S. Cerebrospinal fluid antibodies to coronavirus in patients with Parkinson’s disease. Movement Disorders 1992, 7, 153. [Google Scholar] [CrossRef] [PubMed]
  85. Merello, M.; Bhatia, K.P.; Obeso, J.A. SARS-CoV-2 and the risk of Parkinson’s disease: Facts and fantasy. Lancet Neurol. 2021, 20, 94–95. [Google Scholar] [CrossRef]
  86. Morassi, M.; Palmerini, F.; Nici, S.; Magni, E.; Savelli, G.; Guerra, U.P.; Chieregato, M.; Morbelli, S.; Vogrig, A. SARS-CoV-2-related encephalitis with prominent parkinsonism: Clinical and FDG-PET correlates in two patients. J. Neurol. 2021, 268, 3980–3987. [Google Scholar] [CrossRef]
  87. Brundin, P.; Nath, A.; Beckham, J.D. Is COVID-19 a perfect storm for Parkinson’s disease? Trends Neurosci. 2020, 43, 931–933. [Google Scholar] [CrossRef]
  88. Chen, H.; O’Reilly, E.J.; Schwarzschild, M.A.; Ascherio, A. Peripheral inflammatory biomarkers and risk of Parkinson’s disease. Am. J. Epidemiol. 2008, 167, 90–95. [Google Scholar] [CrossRef] [Green Version]
  89. Yang, L.; Cornell, W.; Kim, M.T.; Nair, M.; Harschnitz, O.; Wang, P.; Koo, S.Y.; Lacko, L.; Bram, Y.; Medicine, W.C.; et al. SARS-CoV-2 infection causes dopaminergic neuron senescence. Res. Sq. 2021. preprint. [Google Scholar] [CrossRef]
  90. Ramani, A.; Müller, L.; Ostermann, P.N.; Gabriel, E.; Abida-Islam, P.; Müller-Schiffmann, A.; Mariappan, A.; Goureau, O.; Gruell, H.; Walker, A.; et al. SARS -CoV-2 targets neurons of 3D human brain organoids. EMBO J. 2020, 39, e106230. [Google Scholar] [CrossRef]
  91. Ghosh, A.; Roy, A.; Liu, X.; Kordower, J.H.; Mufson, E.J.; Hartley, D.M.; Ghosh, S.; Lee Mosley, R.; Gendelman, H.E.; Pahan, K. Selective inhibition of NF-B activation prevents dopaminergic neuronal loss in a mouse model of Parkinson’s disease. Proc. Natl. Acad. Sci. USA 2007, 104, 18754–18759. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Meredith, G.E.; Rademacher, D.J. MPTP mouse models of Parkinson’s disease: An update. J. Parkinson’s Dis. 2011, 1, 19–33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Chaudhry, Z.L.; Klenja, D.; Janjua, N.; Cami-Kobeci, G.; Ahmed, B.Y. COVID-19 and Parkinson’s disease: Shared inflammatory pathways under oxidative stress. Brain Sci. 2020, 10, 807. [Google Scholar] [CrossRef]
  94. Milsted, A.; Barnaf, B.P.; Ransohoff, R.M.; Bridget Brosnihan, K.; Ferrario, C.M. Astrocyte cultures derived from human brain tissue express angiotensinogen mRNA (gene expression/renin-angiotensin system/central nervous system/angiotensin). Proc. Natl. Acad. Sci. USA 1990, 87, 5720–5723. [Google Scholar] [CrossRef] [Green Version]
  95. Stornetta, R.L.; Hawelu-Johnson, C.L.; Guyenet, P.G.; Lynch, K.R. Astrocytes synthesize angiotensinogen in brain. Science 1988, 242, 1444–1446. [Google Scholar] [CrossRef]
  96. Labandeira-Garcia, J.L.; Rodriguez-Pallares, J.; Dominguez-Meijide, A.; Valenzuela, R.; Villar-Cheda, B.; Rodríguez-Perez, A.I. Dopamine-Angiotensin interactions in the basal ganglia and their relevance for Parkinson’s disease. Mov. Disord. 2013, 28, 1337–1342. [Google Scholar] [CrossRef] [PubMed]
  97. Kehoe, P.G.; Wilcock, G.K. Personal View Is inhibition of the renin-angiotensin system a new treatment option for Alzheimer’s disease? Lancet Neurol. 2007, 6, 373–378. [Google Scholar] [CrossRef]
  98. Marreiros, R.; Müller-Schiffmann, A.; Trossbach, S.V.; Prikulis, I.; Hänsch, S.; Weidtkamp-Peters, S.; Moreira, A.R.; Sahu, S.; Soloviev, I.; Selvarajah, S.; et al. Disruption of cellular proteostasis by H1N1 influenza A virus causes α-synuclein aggregation. Proc. Natl. Acad. Sci. USA 2020, 117, 6741–6751. [Google Scholar] [CrossRef]
  99. Braak, H.; del Tredici, K.; Rüb, U.; de Vos, R.A.I.; Jansen Steur, E.N.H.; Braak, E. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol. Aging 2003, 24, 197–211. [Google Scholar] [CrossRef]
  100. Braak, H.; del Tredici, K. Neuropathological staging of brain pathology in sporadic Parkinson’s disease: Separating the wheat from the chaff. J. Parkinson’s Dis. 2017, 7, S73–S87. [Google Scholar] [CrossRef] [Green Version]
  101. Heidbreder, A.; Sonnweber, T.; Stefani, A.; Ibrahim, A.; Cesari, M.; Bergmann, M.; Brandauer, E.; Tancevski, I.; Löffler-Ragg, J.; Högl, B. Video-polysomnographic findings after acute COVID-19: REM sleep without atonia as sign of CNS pathology? Sleep Med. 2021, 80, 92–95. [Google Scholar] [CrossRef]
  102. Romano, S.; Savva, G.M.; Bedarf, J.R.; Charles, I.G.; Hildebrand, F.; Narbad, A. Meta-analysis of the Parkinson’s disease gut microbiome suggests alterations linked to intestinal inflammation. npj Parkinson’s Dis. 2021, 7, 27. [Google Scholar] [CrossRef] [PubMed]
  103. Follmer, C. Gut microbiome imbalance and neuroinflammation: Impact of COVID-19 on Parkinson’s disease. Mov. Disord. 2020, 35, 1495–1496. [Google Scholar] [CrossRef]
  104. Effenberger, M.; Grabherr, F.; Mayr, L.; Schwaerzler, J.; Nairz, M.; Seifert, M.; Hilbe, R.; Seiwald, S.; Scholl-Buergi, S.; Fritsche, G.; et al. Faecal calprotectin indicates intestinal inflammation in COVID-19. Gut 2020, 69, 1543–1544. [Google Scholar] [CrossRef] [Green Version]
  105. Xiao, F.; Tang, M.; Zheng, X.; Liu, Y.; Li, X.; Shan, H. Evidence for gastrointestinal infection of SARS-CoV-2. Gastroenterology 2020, 158, 1831–1833.e3. [Google Scholar] [CrossRef]
  106. Estrada, E. Cascading from SARS-CoV-2 to parkinson’s disease through protein-protein interactions. Viruses 2021, 13, 897. [Google Scholar] [CrossRef]
  107. Vavougios, G.D. Human coronaviruses in idiopathic Parkinson’s disease: Implications of SARS-CoV-2′s modulation of the host’s transcriptome. Infect. Genet. Evol. 2021, 89, 104733. [Google Scholar] [CrossRef]
  108. Rosen, B.; Kurtishi, A.; Vazquez-Jimenez, G.R.; Møller, S.G. The intersection of Parkinson’s disease, viral infections, and COVID-19. Mol. Neurobiol. 2021, 58, 4477–4486. [Google Scholar] [CrossRef] [PubMed]
  109. Ait Wahmane, S.; Achbani, A.; Ouhaz, Z.; Elatiqi, M.; Belmouden, A.; Nejmeddine, M. The possible protective role of α-synuclein against severe acute respiratory syndrome coronavirus 2 infections in patients with Parkinson’s disease. Mov. Disord. 2020, 35, 1293–1294. [Google Scholar] [CrossRef] [PubMed]
  110. Alster, P.; Madetko, N.; Koziorowski, D.; Friedman, A. Microglial activation and inflammation as a factor in the pathogenesis of progressive supranuclear palsy (PSP). Front. Neurosci. 2020, 14, 893. [Google Scholar] [CrossRef] [PubMed]
  111. Malpetti, M.; Passamonti, L.; Jones, P.S.; Street, D.; Rittman, T.; Fryer, T.D.; Hong, Y.T.; Vàsquez Rodriguez, P.; Bevan-Jones, W.R.; Aigbirhio, F.I.; et al. Neuroinflammation predicts disease progression in progressive supranuclear palsy. J. Neurol. Neurosurg. Psychiatry 2021, 92, 769–775. [Google Scholar] [CrossRef]
  112. Kübler, D.; Wächter, T.; Cabanel, N.; Su, Z.; Turkheimer, F.E.; Dodel, R.; Brooks, D.J.; Oertel, W.H.; Gerhard, A. Widespread microglial activation in multiple system atrophy. Mov. Disord. 2019, 34, 564–568. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Xiang, X.; Wind, K.; Wiedemann, T.; Blume, T.; Shi, Y.; Briel, N.; Beyer, L.; Biechele, G.; Eckenweber, F.; Zatcepin, A.; et al. Microglial activation states drive glucose uptake and FDG-PET alterations in neurodegenerative diseases. Sci. Transl. Med. 2021, 13, eabe5640. [Google Scholar] [CrossRef] [PubMed]
  114. Palleis, C.; Sauerbeck, J.; Beyer, L.; Harris, S.; Schmitt, J.; Morenas-Rodriguez, E.; Finze, A.; Nitschmann, A.; Ruch-Rubinstein, F.; Eckenweber, F.; et al. In Vivo assessment of neuroinflammation in 4-Repeat tauopathies. Mov. Disord. 2021, 36, 883–894. [Google Scholar] [CrossRef]
  115. Del Brutto, O.H.; Wu, S.; Mera, R.M.; Costa, A.F.; Recalde, B.Y.; Issa, N.P. Cognitive decline among individuals with history of mild symptomatic SARS-CoV-2 infection: A longitudinal prospective study nested to a population cohort. Eur. J. Neurol. 2021, 28, 3245–3253. [Google Scholar] [CrossRef]
  116. Pistarini, C.; Fiabane, E.; Houdayer, E.; Vassallo, C.; Manera, M.R.; Alemanno, F. Cognitive and emotional disturbances due to COVID-19: An exploratory study in the rehabilitation setting. Front. Neurol. 2021, 12, 643646. [Google Scholar] [CrossRef]
  117. Alemanno, F.; Houdayer, E.; Parma, A.; Spina, A.; del Forno, A.; Scatolini, A.; Angelone, S.; Brugliera, L.; Tettamanti, A.; Beretta, L.; et al. COVID-19 cognitive deficits after respiratory assistance in the subacute phase: A COVID rehabilitation unit experience. PLoS ONE 2021, 16, e0246590. [Google Scholar] [CrossRef] [PubMed]
  118. Jaywant, A.; Vanderlind, W.M.; Alexopoulos, G.S.; Fridman, C.B.; Perlis, R.H.; Gunning, F.M. Frequency and profile of objective cognitive deficits in hospitalized patients recovering from COVID-19. Neuropsychopharmacology 2021, 46, 2235–2240. [Google Scholar] [CrossRef]
  119. Rogers, J.P.; Chesney, E.; Oliver, D.; Pollak, T.A.; McGuire, P.; Fusar-Poli, P.; Zandi, M.S.; Lewis, G.; David, A.S. Psychiatric and neuropsychiatric presentations associated with severe coronavirus infections: A systematic review and meta-analysis with comparison to the COVID-19 pandemic. Lancet Psychiatry 2020, 7, 611–627. [Google Scholar] [CrossRef]
  120. Versace, V.; Sebastianelli, L.; Ferrazzoli, D.; Romanello, R.; Ortelli, P.; Saltuari, L.; D’Acunto, A.; Porrazzini, F.; Ajello, V.; Oliviero, A.; et al. Intracortical GABAergic dysfunction in patients with fatigue and dysexecutive syndrome after COVID-19. Clin. Neurophysiol. 2021, 132, 1138–1143. [Google Scholar] [CrossRef]
  121. Benussi, A.; di Lorenzo, F.; Dell’era, V.; Cosseddu, M.; Alberici, A.; Caratozzolo, S.; Cotelli, M.S.; Micheli, A.; Rozzini, L.; Depari, A.; et al. Transcranial magnetic stimulation distinguishes Alzheimer disease from frontotemporal dementia. Neurology 2017, 89, 665–672. [Google Scholar] [CrossRef]
  122. Wolters, A.E.; Slooter, A.J.C.; Van Der Kooi, A.W.; Van Dijk, D. Cognitive impairment after intensive care unit admission: A systematic review. Intensive Care Med. 2013, 39, 376–386. [Google Scholar] [CrossRef] [PubMed]
  123. Ciaccio, M.; lo Sasso, B.; Scazzone, C.; Gambino, C.M.; Ciaccio, A.M.; Bivona, G.; Piccoli, T.; Giglio, R.V.; Agnello, L. COVID-19 and Alzheimer’s disease. Brain Sci. 2021, 11, 305. [Google Scholar] [CrossRef] [PubMed]
  124. Xia, X.; Wang, Y.; Zheng, J. COVID-19 and Alzheimer’s disease: How one crisis worsens the other. Transl. Neurodegener. 2021, 10, 15. [Google Scholar] [CrossRef]
  125. Mikkelsen, M.E.; Christie, J.; Lanken, P.N.; Biester, R.C.; Thompson, B.T.; Bellamy, S.L.; Localio, A.R.; Demissie, E.; Hopkins, R.O.; Angus, D.C. The adult respiratory distress syndrome cognitive outcomes study: Long-term neuropsychological function in survivors of acute lung injury. Am. J. Respir. Crit. Care Med. 2012, 185, 1307–1315. [Google Scholar] [CrossRef] [Green Version]
  126. Atkins, J.L.; Masoli, J.A.H.; Delgado, J.; Pilling, L.C.; Kuo, C.L.; Kuchel, G.A.; Melzer, D. Preexisting Comorbidities predicting COVID-19 and mortality in the UK biobank community cohort. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 2020, 75, 2224–2230. [Google Scholar] [CrossRef] [PubMed]
  127. Rangon, C.-M.; Krantic, S.; Moyse, E.; Fougère, B. The Vagal Autonomic Pathway of COVID-19 at the Crossroad of Alzheimer’s Disease and Aging: A Review of Knowledge. J. Alzheimer’s Dis. Rep. 2020, 4, 537–551. [Google Scholar] [CrossRef]
  128. Saragih, I.D.; Saragih, I.S.; Batubara, S.O.; Lin, C.J. Dementia as a mortality predictor among older adults with COVID-19: A systematic review and meta-analysis of observational study. Geriatr. Nurs. 2021, 42, 1230–1239. [Google Scholar] [CrossRef]
  129. Vrillon, A.; Mhanna, E.; Aveneau, C.; Lebozec, M.; Grosset, L.; Nankam, D.; Albuquerque, F.; Razou Feroldi, R.; Maakaroun, B.; Pissareva, I.; et al. COVID-19 in adults with dementia: Clinical features and risk factors of mortality—A clinical cohort study on 125 patients. Alzheimer’s Res. Ther. 2021, 13, 77. [Google Scholar] [CrossRef]
  130. Yu, Y.; Travaglio, M.; Popovic, R.; Leal, N.S.; Martins, L.M. Alzheimer’s and Parkinson’s diseases predict different COVID-19 outcomes: A UK biobank study. Geriatrics 2021, 6, 10. [Google Scholar] [CrossRef]
  131. Brown, E.E.; Kumar, S.; Rajji, T.K.; Pollock, B.G.; Mulsant, B.H. Anticipating and Mitigating the Impact of the COVID-19 Pandemic on Alzheimer’s Disease and Related Dementias. Am. J. Geriatr. Psychiatry 2020, 28, 712–721. [Google Scholar] [CrossRef]
  132. McAlpine, L.S.; Fesharaki-Zadeh, A.; Spudich, S. Coronavirus disease 2019 and neurodegenerative disease: What will the future bring? Curr. Opin. Psychiatry 2021, 34, 177–185. [Google Scholar] [CrossRef]
  133. Ding, Q.; Shults, N.v.; Harris, B.T.; Suzuki, Y.J. Angiotensin-converting enzyme 2 (ACE2) is upregulated in Alzheimer’s disease brain. bioRxiv 2020. the preprint server for biology. [Google Scholar] [CrossRef]
  134. Lee, S.; Viqar, F.; Zimmerman, M.E.; Narkhede, A.; Tosto, G.; Benzinger, T.L.S.; Marcus, D.S.; Fagan, A.M.; Goate, A.; Fox, N.C.; et al. White matter hyperintensities are a core feature of Alzheimer’s disease: Evidence from the dominantly inherited Alzheimer network. Ann. Neurol. 2016, 79, 929–939. [Google Scholar] [CrossRef] [PubMed]
  135. Soscia, S.J.; Kirby, J.E.; Washicosky, K.J.; Tucker, S.M.; Ingelsson, M.; Hyman, B.; Burton, M.A.; Goldstein, L.E.; Duong, S.; Tanzi, R.E.; et al. The Alzheimer’s disease-associated amyloid β-protein is an antimicrobial peptide. PLoS ONE 2010, 5, e9505. [Google Scholar] [CrossRef]
  136. Kuo, C.L.; Pilling, L.C.; Atkins, J.L.; Masoli, J.A.H.; Delgado, J.; Kuchel, G.A.; Melzer, D. APOE e4 genotype predicts severe COVID-19 in the UK biobank community cohort. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 2020, 75, 2231–2232. [Google Scholar] [CrossRef] [PubMed]
  137. Chen, X.; Zhao, B.; Qu, Y.; Chen, Y.; Xiong, J.; Feng, Y.; Men, D.; Huang, Q.; Liu, Y.; Yang, B.; et al. Detectable serum SARS-CoV-2 viral load (RNAaemia) is closely correlated with drastically elevated interleukin 6 (IL-6) level in critically ill COVID-19 patients. Clin. Infect. Dis. 2020, 71, 1937–1942. [Google Scholar] [CrossRef]
  138. Kaczmarczyk, R.; Tejera, D.; Simon, B.J.; Heneka, M.T. Microglia modulation through external vagus nerve stimulation in a murine model of Alzheimer’s disease. J. Neurochem. 2018, 146, 76–85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Naughton, S.X.; Raval, U.; Pasinetti, G.M. Potential novel role of COVID-19 in Alzheimer’s Disease and preventative mitigation strategies. J. Alzheimer’s Dis. 2020, 76, 21–25. [Google Scholar] [CrossRef] [PubMed]
  140. García-Azorín, D.; Seeher, K.M.; Newton, C.R.; Okubadejo, N.U.; Pilotto, A.; Saylor, D.; Winkler, A.S.; Charfi Triki, C.; Leonardi, M. Disruptions of neurological services, its causes and mitigation strategies during COVID-19: A global review. J. Neurol. 2021, 268, 3947–3960. [Google Scholar] [CrossRef]
  141. Fearon, C.; Fasano, A. Parkinson’s disease and the COVID-19 pandemic. J. Parkinson’s Dis. 2021, 11, 431–444. [Google Scholar] [CrossRef]
  142. Leta, V.; Rodríguez-Violante, M.; Abundes, A.; Rukavina, K.; Teo, J.T.; Falup-Pecurariu, C.; Irincu, L.; Rota, S.; Bhidayasiri, R.; Storch, A.; et al. Parkinson’s disease and post–COVID-19 syndrome: The Parkinson’s long-COVID spectrum. Mov. Disord. 2021, 36, 1287–1289. [Google Scholar] [CrossRef]
  143. Anghelescu, B.A.-M.; Bruno, V.; Martino, D.; Roach, P. Effects of the COVID-19 pandemic on Parkinson’s disease: A single-centered qualitative study. Can. J. Neurol. Sci. 2021, 12, 1–13. [Google Scholar] [CrossRef]
  144. Balci, B.; Aktar, B.; Buran, S.; Tas, M.; Donmez Colakoglu, B. Impact of the COVID-19 pandemic on physical activity, anxiety, and depression in patients with Parkinson’s disease. Int. J. Rehabil. Res. 2021, 44, 173–176. [Google Scholar] [CrossRef]
  145. Helmich, R.C.; Bloem, B.R. The Impact of the COVID-19 pandemic on Parkinson’s disease: Hidden sorrows and emerging opportunities. J. Parkinson’s Dis. 2020, 10, 351–354. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Douma, E.H.; de Kloet, E.R. Stress-induced plasticity and functioning of ventral tegmental dopamine neurons. Neurosci. Biobehav. Rev. 2020, 108, 48–77. [Google Scholar] [CrossRef]
  147. Zach, H.; Dirkx, M.F.; Pasman, J.W.; Bloem, B.R.; Helmich, R.C. Cognitive stress reduces the effect of levodopa on Parkinson’s resting tremor. CNS Neurosci. Ther. 2017, 23, 209–215. [Google Scholar] [CrossRef] [PubMed]
  148. Cavallieri, F.; Sireci, F.; Fioravanti, V.; Toschi, G.; Rispoli, V.; Antonelli, F.; Costantini, M.; Ghirotto, L.; Valzania, F. Parkinson’s disease patients’ needs during the COVID-19 pandemic in a red zone: A framework analysis of open-ended survey questions. Eur. J. Neurol. 2021, 28, 3254–3262. [Google Scholar] [CrossRef] [PubMed]
  149. Vila-Viçosa, D.; Clemente, A.; Pona-Ferreira, F.; Leitão, M.; Bouça-Machado, R.; Kauppila, L.A.; Costa, R.M.; Matias, R.; Ferreira, J.J. Unsupervised walking activity assessment reveals COVID-19 impact on Parkinson’s disease patients. Mov. Disord. Off. J. Mov. Disord. Soc. 2021, 36, 531–532. [Google Scholar] [CrossRef] [PubMed]
  150. Janiri, D.; Petracca, M.; Moccia, L.; Tricoli, L.; Piano, C.; Bove, F.; Imbimbo, I.; Simonetti, A.; di Nicola, M.; Sani, G.; et al. COVID-19 pandemic and psychiatric symptoms: The impact on Parkinson’s disease in the elderly. Front. Psychiatry 2020, 11, 581144. [Google Scholar] [CrossRef]
  151. Feeney, M.P.; Xu, Y.; Surface, M.; Shah, H.; Vanegas-Arroyave, N.; Chan, A.K.; Delaney, E.; Przedborski, S.; Beck, J.C.; Alcalay, R.N. The impact of COVID-19 and social distancing on people with Parkinson’s disease: A survey study. npj Parkinson’s Dis. 2021, 7, 10. [Google Scholar] [CrossRef]
  152. Kitani-Morii, F.; Kasai, T.; Horiguchi, G.; Teramukai, S.; Ohmichi, T.; Shinomoto, M.; Fujino, Y.; Mizuno, T. Risk factors for neuropsychiatric symptoms in patients with Parkinson’s disease during COVID-19 pandemic in Japan. PLoS ONE 2021, 16, e0245864. [Google Scholar] [CrossRef]
  153. Kumar, N.; Gupta, R.; Kumar, H.; Mehta, S.; Rajan, R.; Kumar, D.; Kandadai, R.M.; Desai, S.; Wadia, P.; Basu, P.; et al. Impact of home confinement during COVID-19 pandemic on sleep parameters in Parkinson’s disease. Sleep Med. 2021, 77, 15–22. [Google Scholar] [CrossRef] [PubMed]
  154. Xia, Y.; Kou, L.; Zhang, G.; Han, C.; Hu, J.; Wan, F.; Yin, S.; Sun, Y.; Wu, J.; Li, Y.; et al. Investigation on sleep and mental health of patients with Parkinson’s disease during the Coronavirus disease 2019 pandemic. Sleep Med. 2020, 75, 428–433. [Google Scholar] [CrossRef]
  155. Hanff, A.M.; Pauly, C.; Pauly, L.; Schröder, V.E.; Hansen, M.; Meyers, G.R.; Kaysen, A.; Hansen, L.; Wauters, F.; Krüger, R. Unmet needs of people with Parkinson’s disease and their caregivers during COVID-19-related confinement: An explorative secondary data analysis. Front. Neurol. 2021, 11, 615172. [Google Scholar] [CrossRef]
  156. Klietz, M.; von Eichel, H.; Schnur, T.; Staege, S.; Höglinger, G.U.; Wegner, F.; Stiel, S. One year trajectory of caregiver burden in parkinson’s disease and analysis of gender-specific aspects. Brain Sci. 2021, 11, 295. [Google Scholar] [CrossRef] [PubMed]
  157. Klietz, M.; Drexel, S.C.; Schnur, T.; Lange, F.; Groh, A.; Paracka, L.; Greten, S.; Dressler, D.; Höglinger, G.U.; Wegner, F. Mindfulness and psychological flexibility are inversely associated with caregiver Burden in parkinson’s disease. Brain Sci. 2020, 10, 111. [Google Scholar] [CrossRef] [Green Version]
  158. Montanaro, E.; Artusi, C.A.; Rosano, C.; Boschetto, C.; Imbalzano, G.; Romagnolo, A.; Bozzali, M.; Rizzone, M.G.; Zibetti, M.; Lopiano, L. Anxiety, depression, and worries in advanced Parkinson disease during COVID-19 pandemic. Neurol. Sci. 2021, 1–8. [Google Scholar] [CrossRef]
  159. Hu, C.; Chen, C.; Dong, X.P. Impact of COVID-19 pandemic on patients with neurodegenerative diseases. Front. Aging Neurosci. 2021, 13, 664965. [Google Scholar] [CrossRef] [PubMed]
  160. Fasano, A.; Cereda, E.; Barichella, M.; Cassani, E.; Ferri, V.; Zecchinelli, A.L.; Pezzoli, G. COVID-19 in Parkinson’s disease patients living in Lombardy, Italy. Mov. Disord. 2020, 35, 1089–1093. [Google Scholar] [CrossRef]
  161. Del Prete, E.; Francesconi, A.; Palermo, G.; Mazzucchi, S.; Frosini, D.; Morganti, R.; Coleschi, P.; Raglione, L.M.; Vanni, P.; Ramat, S.; et al. Prevalence and impact of COVID-19 in Parkinson’s disease: Evidence from a multi-center survey in Tuscany region. J. Neurol. 2021, 268, 1179–1187. [Google Scholar] [CrossRef]
  162. Fasano, A.; Elia, A.E.; Dallocchio, C.; Canesi, M.; Alimonti, D.; Sorbera, C.; Alonso-Canovas, A.; Pezzoli, G. Predictors of COVID-19 outcome in Parkinson’s disease. Parkinsonism Relat. Disord. 2020, 78, 134–137. [Google Scholar] [CrossRef]
  163. Antonini, A.; Leta, V.; Teo, J.; Chaudhuri, K.R. Outcome of Parkinson’s disease patients affected by COVID-19. Mov. Disord. 2020, 35, 905–908. [Google Scholar] [CrossRef] [PubMed]
  164. Cilia, R.; Bonvegna, S.; Straccia, G.; Andreasi, N.G.; Elia, A.E.; Romito, L.M.; Devigili, G.; Cereda, E.; Eleopra, R. Effects of COVID-19 on Parkinson’s disease clinical features: A community-based case-control study. Mov. Disord. 2020, 35, 1287–1292. [Google Scholar] [CrossRef] [PubMed]
  165. Huber, M.K.; Raichle, C.; Lingor, P.; Synofzik, M.; Borgmann, S.; Erber, J.; Tometten, L.; Rimili, W.; Dolff, S.; Wille, K.; et al. Outcomes of SARS-CoV-2 infections in patients with neurodegenerative diseases in the LEOSS cohort. Mov. Disord. 2021, 36, 791–793. [Google Scholar] [CrossRef] [PubMed]
  166. Buccafusca, M.; Micali, C.; Autunno, M.; Versace, A.G.; Nunnari, G.; Musumeci, O. Favourable course in a cohort of Parkinson’s disease patients infected by SARS-CoV-2: A single-centre experience. Neurol. Sci. 2021, 42, 811–816. [Google Scholar] [CrossRef]
  167. Vignatelli, L.; Zenesini, C.; Belotti, L.M.B.; Baldin, E.; Bonavina, G.; Calandra-Buonaura, G.; Cortelli, P.; Descovich, C.; Fabbri, G.; Giannini, G.; et al. Risk of hospitalization and death for COVID-19 in people with Parkinson’s disease or Parkinsonism. Mov. Disord. Off. J. Mov. Disord. Soc. 2021, 36, 1–10. [Google Scholar] [CrossRef]
  168. Scherbaum, R.; Kwon, E.H.; Richter, D.; Bartig, D.; Gold, R.; Krogias, C.; Tönges, L. Clinical profiles and mortality of COVID-19 inpatients with Parkinson’s disease in germany. Mov. Disord. 2021, 36, 1049–1057. [Google Scholar] [CrossRef]
  169. Zhang, Q.; Schultz, J.L.; Aldridge, G.M.; Simmering, J.E.; Narayanan, N.S. Coronavirus disease 2019 case fatality and Parkinson’s disease. Mov. Disord. 2020, 35, 1914–1915. [Google Scholar] [CrossRef]
  170. Zhai, H.; Lv, Y.; Xu, Y.; Wu, Y.; Zeng, W.; Wang, T.; Cao, X.; Xu, Y. Characteristic of Parkinson’s disease with severe COVID-19: A study of 10 cases from Wuhan. J. Neural Transm. 2021, 128, 37–48. [Google Scholar] [CrossRef]
  171. Artusi, C.A.; Romagnolo, A.; Imbalzano, G.; Marchet, A.; Zibetti, M.; Rizzone, M.G.; Lopiano, L. COVID-19 in Parkinson’s disease: Report on prevalence and outcome. Parkinsonism Relat. Disord. 2020, 80, 7–9. [Google Scholar] [CrossRef]
  172. Araújo, R.; Aranda-Martínez, J.D.; Aranda-Abreu, G.E. Amantadine treatment for people with COVID-19. Arch. Med. Res. 2020, 51, 739–740. [Google Scholar] [CrossRef] [PubMed]
  173. Gordon, D.E.; Jang, G.M.; Bouhaddou, M.; Xu, J.; Obernier, K.; White, K.M.; O’Meara, M.J.; Rezelj, V.V.; Guo, J.Z.; Swaney, D.L.; et al. A SARS-CoV-2 protein interaction map reveals targets for drug repurposing. Nature 2020, 583, 459–468. [Google Scholar] [CrossRef] [PubMed]
  174. Kamel, W.A.; Kamel, M.I.; Alhasawi, A.; Elmasry, S.; AlHamdan, F.; Al-Hashel, J.Y. Effect of pre-exposure use of amantadine on COVID-19 infection: A hospital-based cohort study in patients with Parkinson’s Disease or multiple sclerosis. Front. Neurol. 2021, 12, 704186. [Google Scholar] [CrossRef]
  175. Artusi, C.A.; Romagnolo, A.; Ledda, C.; Zibetti, M.; Rizzone, M.G.; Montanaro, E.; Bozzali, M.; Lopiano, L. COVID-19 and Parkinson’s disease: What do we know so far? J. Parkinson’s Dis. 2021, 11, 445–454. [Google Scholar] [CrossRef] [PubMed]
  176. Ferini-Strambi, L.; Salsone, M. COVID-19 and neurological disorders: Are neurodegenerative or neuroimmunological diseases more vulnerable? J. Neurol. 2021, 268, 409–419. [Google Scholar] [CrossRef]
  177. Hainque, E.; Grabli, D. Rapid worsening in Parkinson’s disease may hide COVID-19 infection. Parkinsonism Relat. Disord. 2020, 75, 126–127. [Google Scholar] [CrossRef]
Table 1. Mortality and morbidity in COVID-19 PD cases versus COVID-19 non-PD cases.
Table 1. Mortality and morbidity in COVID-19 PD cases versus COVID-19 non-PD cases.
AuthorsCountryType of StudyNo. of PatientsResults
Fasano et al., 2020 [160]ItalyCase controln = 105 PD + COVID,
n = 92 controls
-
PD/non-PD with no difference in COVID-19 prevalence (7.1% vs. 7.6%) and mortality (5.7% vs. 7.6%)
-
COVID-19 symptoms in PD milder compared to non-PD
Del Prete et al., 2020 [161]ItalyCase-controlled surveyn = 740 PD cases, 7 infected with COVID
-
Mortality 14% in COVID-infected PD-cases (n = 1)
-
Age-stratified mortality in line with national data (14%)
Fasano et al., 2020 [162]Italy, Iran, Spain, UKMulticenter cohortn = 117 PD + COVID
-
Mortality rate 19.4% (PD + COVID)
Antonini et al., 2020 [163]ItalyCase seriesn = 10 PD + COVID
-
Mortality rate 40% (n = 4)
-
Higher mortality when older/longer PD-duration/DBS/levodopa infusion therapy
Cilia et al., 2020 [164]ItalyCase controln = 12 PD + COVID,
n = 36 controls
-
Mortality 0%
-
Higher PD-associated morbidity
Huber et al., 2021 [165]GermanyProspective multicenter cohort studyn = 40 PD + COVID,
n = 600 matched controls
-
COVID-19-associated mortality (32.5% PD vs. 26.7% non-PD) and morbidity not significantly different
Buccafusca et al., 2021 [166]ItalyCase seriesn = 12 PD + COVID
-
Mortality 0%
Vignatelli et al., 2021 [167]ItalyCase controln = 696 PD + COVID,
n = 8590 controls
-
30 days case fatality rate comparable (PD 35.1% vs. controls 39%)
Scherbaum et al., 2021 [168]GermanyCross-sectional studyn = 693 PD + COVID,
control number unknown
-
COVID-19 mortality rate: PD 35.4% vs. 20.7% controls (p < 0.001)
Zhang et al., 2020 [169]USACase controln = 694 PD + COVID,
n = 78,355 controls
-
Mortality rate 21.3% PD vs. 5.5% non-PD (p < 0.001)
-
1:5 matching: PD patients with significantly higher mortality from COVID
Zhai et al., 2020 [170]ChinaSingle-center retrospective studyn = 10 PD + COVID,
n = 286 controls
-
Inclusion of severe or critical ill COVID patients
-
Mortality rate PD 30% vs. non-PD 40.2% (p > 0.05)
Artusi et al., 2020 [171]ItalyCase seriesn = 8 PD + COVID,
control “general population”
-
Higher mortality (75% PD vs. 12% controls)
-
PD symptoms worsened in all patients
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Krey, L.; Huber, M.K.; Höglinger, G.U.; Wegner, F. Can SARS-CoV-2 Infection Lead to Neurodegeneration and Parkinson’s Disease? Brain Sci. 2021, 11, 1654. https://doi.org/10.3390/brainsci11121654

AMA Style

Krey L, Huber MK, Höglinger GU, Wegner F. Can SARS-CoV-2 Infection Lead to Neurodegeneration and Parkinson’s Disease? Brain Sciences. 2021; 11(12):1654. https://doi.org/10.3390/brainsci11121654

Chicago/Turabian Style

Krey, Lea, Meret Koroni Huber, Günter U. Höglinger, and Florian Wegner. 2021. "Can SARS-CoV-2 Infection Lead to Neurodegeneration and Parkinson’s Disease?" Brain Sciences 11, no. 12: 1654. https://doi.org/10.3390/brainsci11121654

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop