Skip to main content

SYSTEMATIC REVIEW article

Front. Neurol., 28 January 2021
Sec. Neurorehabilitation
This article is part of the Research Topic COVID-19: The Neurorehabilitation Perspective View all 26 articles

COVID-19 Pathophysiology Predicts That Ischemic Stroke Occurrence Is an Expectation, Not an Exception—A Systematic Review

  • 1School of Psychology and Public Health, La Trobe University, Melbourne, VIC, Australia
  • 2Department of Neurology, Western Health and University Melbourne, Australian Institute of Muscular Skeletal Sciences (AIMSS), Level Three, Western Health Centre for Research and Education (WHCRE), Sunshine Hospital, Melbourne, VIC, Australia
  • 3Department of Medicine, Faculty of Medicine, University of Rajarata, Anuradhapura, Sri Lanka
  • 4Faculty of Social and Political Sciences, Ivane Javakhishvili Tbilisi State University, Tbilisi, Georgia

Clinical reports of neurological manifestations associated with severe coronavirus disease 2019 (COVID-19), such as acute ischemic stroke (AIS), encephalopathy, seizures, headaches, acute necrotizing encephalitis, cerebral microbleeds, posterior reversible leukoencephalopathy syndrome, hemophagocytic lymphohistiocytosis, peripheral neuropathy, cranial nerve palsies, transverse myelitis, and demyelinating disorders, are increasing rapidly. However, there are comparatively few studies investigating the potential impact of immunological responses secondary to hypoxia, oxidative stress, and excessive platelet-induced aggregation on the brain. This scoping review has focused on the pathophysiological mechanisms associated with peripheral and consequential neural (central) inflammation leading to COVID-19-related ischemic strokes. It also highlights the common biological processes shared between AIS and COVID-19 infection and the importance of the recognition that severe respiratory dysfunction and neurological impairments associated with COVID and chronic inflammation [post-COVID-19 neurological syndrome (PCNS)] may significantly impact recovery and ability to benefit from neurorehabilitation. This study provides a comprehensive review of the pathobiology of COVID-19 and ischemic stroke. It also affirms that the immunological contribution to the pathophysiology of COVID-19 is predictive of the neurological sequelae particularly ischemic stroke, which makes it the expectation rather than the exception. This work is of fundamental significance to the neurorehabilitation community given the increasing number of COVID-related ischemic strokes, the current limited knowledge regarding the risk of reinfection, and recent reports of a PCNS. It further highlights the need for global collaboration and research into new pathobiology-based neurorehabilitation treatment strategies and more integrated evidence-based care.

Introduction

Since the first case of coronavirus disease 2019 (COVID-19) infection was reported in Wuhan, China in December 2019, a significant number of thrombotic complications affecting the venous and arterial systems have been published in the literature (13), with the World Stroke Organization now recognizing that acute ischemic stroke (AIS) increases the severity of severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) viral infection by 2.5-fold (4, 5). Similar associations between systemic infections, inflammation, and AIS are longstanding (6). However, to date, few reports are reviewing the molecular bases of the peripheral and central mechanisms induced by SARS-CoV2 infection and potential neurological manifestations with focused attention on AIS.

Neurological manifestations of COVID-19 infection were first described by Mao et al. who observed six cases of acute cerebrovascular disease (2). Subsequently, 19 cases of COVID-19-related strokes particularly affecting the young and involving medium to large arteries were reported from a tertiary center in New York (3), highlighting the need for better understanding of the potential mechanisms linking COVID-19 and AIS.

Stroke has been associated with other earlier coronavirus infections. In 2002, AIS was first detailed by Umapathi et al., in 4 of 206 patients who presented with large vessel occlusion associated with SARS-CoV infection in Singapore in 2002 (7). Two other patients with AIS were also described in Middle East respiratory syndrome corona virus (MERS-CoV)-infected patients in Saudi Arabia during the 2015 epidemic (8).

SARS-CoV2 is known to initially bind to the angiotensin-converting enzyme 2 (ACE2) receptors of epithelial and endothelial cells where an immediate immunological activation occurs that can, in severe cases, eventually lead to hypercoagulability or thrombophilia and increased tendency of clots forming in the blood and potentially AIS (9). However, ŧhere is limited information on the physiological abnormalities and mechanisms linking COVID-19 and AIS, although a number of mechanisms have been proposed. The major mechanisms that have been proposed to date include systemic innate immunity-mediated hyperinflammation, neurovascular endothelial dysfunction, endotheliitis, central nervous system renin–angiotensin–aldosterone system (RAAS) dysregulation, oxidative stress, and excessive platelet aggregation (1012). Thus, this scoping review aimed to elucidate potential pathophysiological mechanisms predisposing patients with COVID-19 to a higher risk for neurovascular events.

Methodology

The authors of this scoping review used the Arksey and O'Malley methodology to identify and extract useful literature (13). The steps undertaken include (1) research questions identification; (2) relevant literature identification; (3) screening and selection of relevant literature; (4) data charting; and (5) analyzing, summarizing, and reporting the results.

Identification and Development of the Research Question

This focused on the general research question: “Are there mechanisms associated with COVID-19 infection that are likely to predispose patients to ischemic stroke?”

The following are the specific areas of interest:

a. Can current understanding of the immunological mechanisms associated with the inflammatory responses to severe COVID-19 potentially predispose a patient to AIS?

b. Are there other potential pathophysiological mechanisms predisposing COVID-19 patients to upregulate procoagulable mechanisms leading to thrombi formation and potentially AIS?

Relevant Literature Identification

A literature review of articles regarding cases and mechanisms underlying the co-occurrence of COVID-19-and AIS, published in English between January 2000 and August 12th, was carried out up to August 15th, 2020. Only studies (qualitative and quantitative studies, systematic reviews, metanalysis, case reports, and case series) that directly or indirectly link pathophysiological mechanisms to ischemic stroke and COVID-19 infection have been included in the final review (see Table 1 detailing the studies).

TABLE 1
www.frontiersin.org

Table 1. Summary of the main studies on the mechanisms of COVID-19-related strokes.

Screening and Selection of Relevant Literature

In the first step, MEDLINE, Cochrane, and Cumulative Index to Nursing and Allied Health Literature (CINAHL) databases were searched to identify useful keywords. Subsequently, the identified keywords were used to search the same databases for relevant studies. The literature was first screened at the title and the abstract level; then, full-text articles were assessed for eligibility. The manual bibliographic search of identified studies was also done in the last step of the literature search. The following keywords were used: COVID-19, coronavirus, mechanism, inflammation, thrombosis, embolism, endotheliitis, arteritis, neuroinflammation, and ACE2 receptors. Two researchers (CS and TW) reviewed relevant articles independently. Any disagreement for inclusion was resolved by a third author (LK).

Data Charting and Analysis

The included studies were charted, and the following parameters were taken into account: publication year, type of study, aims and objectives of the study, and study findings and conclusion. Included studies were analyzed extensively and are listed in Table 1.

Results and Analysis

A total of 1,539 studies were identified in the electronic search. After removal of duplicates and screening at the title level, 230 articles were further reviewed at the abstract level, yielding 88 articles requiring assessment. Fifty-eight further articles were excluded, as these papers were not addressing the predefined research questions. A thorough review by two authors (CS and TW) deemed these papers to contain secondary information with repetition rather than an original contribution to the research questions. A total of 30 articles were included in the study. The details of the said studies are outlined in Table 1. The search process is summarized in a Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) in Figure 1.

FIGURE 1
www.frontiersin.org

Figure 1. Prisma chart.

At the time of this submission (as of September 5th, 2020, WHO situation report), the global number of confirmed SARS-CoV2 cases were 26,171,112 with 865,154 confirmed deaths with a mortality rate of ~3.3%. It is also important to note that only a small proportion of COVID-19-infected individuals progress to severe disease, and of these, a smaller number experience stroke and/or death. The most likely predisposing factors to serious COVID-19 disease states are age, sex, and immune system inability to deal with environmental infection together with genetic factors, and associated cardiovascular risk comorbidities such as hypertension, diabetes, obesity, neurological disorders, and medication interactions. Indeed, it appears that exaggerated immune responses to infection and chronic inflammation may be the key factor in the pathogenesis of severe COVID-19 and associated cerebrovascular complications.

Inflammatory Mechanisms of COVID-19

Initial Inflammatory Responses (Key References Are in Table 1; at the End of the Paper)

It is currently accepted that the SARS-CoV2 virus, like other coronaviruses, attacks host cells by binding to ACE2 (12, 4143). Initial virus recognition occurs via the epithelial cells of the olfactory and respiratory tract of the infected person (39, 43) and would be expected to immediately activate the innate immune system of individual cells and the associated vascular supply (44). The cells within the respiratory epithelia are assumed to release the first set of cytokines such as tumor necrosis factor (TNF) and interleukin-6 (IL-6) (45) that then activate further proinflammatory macrophages and monocytes to accumulate in the alveoli.

The recruited monocytes and macrophages are a further rich source of cytokines and monocyte chemoattractant protein-1 with an additional army of immune-related cells coming to play a major component (45), as described in Tables 2, 3. At this stage, the majority of individuals will be asymptomatic, while some may experience flu-like symptoms, sore throat, myalgia, diarrhea, fever and headache, etc.

TABLE 2
www.frontiersin.org

Table 2. Pathophysiology of COVID-19 and acute ischemic stroke.

TABLE 3
www.frontiersin.org

Table 3. Summary of Pathophysiological mechanisms of COVDI-19 related Acute Ischemic Strokes.

The intrinsic pathogenicity of severe coronavirus infection in vulnerable people ensures that the acute severe inflammatory response to the COVID-induced respiratory distress results in decreased levels of circulating lymphocytes, secondary hemophagocytic lymphohistiocytosis (HLH) (4650) shifts immune defenses toward natural killer (NK) and circulating macrophages (macrophage activation syndrome), and increased neutrophils. An early and important mediator to this phenomenon is the significant elevation of proinflammatory cytokines that fuel various processes in cerebrovascular ischemia (Tables 2, 3). Indeed, sepsis-induced stimulation of the immune system leads to a clonal expansion of antigen-specific T lymphocytes, which result in the further release of proinflammatory cytokines and activation of cells such as macrophages and cytotoxic T lymphocytes, which are otherwise known as the HLH (51, 52), and are thought to be caused by a dysfunction of the normal regulatory mechanisms (52). As a result of the build-up of proinflammatory immune responses, an imbalance favoring the mass increase in neutrophil to leukocyte ratio, and inflammatory cytokines such as TNF, interferon-γ, IL-1, IL-6, IL-18, and IL-33 propels global inflammatory activity (5255), which is known to lead to multiorgan dysfunction among critically ill patients (45).

Nod-like receptor family, pyrin domain-containing 3 (NLRP3) inflammasomes play an important role in innate immunity and are directly activated by the virus itself via the small viral protein, viroporin protein 3a, that is known to modify cellular membranes and facilitate virus release from host-infected cells (5658). This multiprotein complex in the cytosol drives a cascade of reactions resulting in the formation of proinflammatory cytokines such as interleukin-1β (IL-1β), interleukin-18 (IL-18), pathogen-associated molecular patterns (PAMPs), and damage-associated molecular patterns (DAMPs), which are usually tightly regulated in patients with normal immune systems (56). Animal studies related to SARS-CoV infection have demonstrated that the overactivation of the NLRP3 inflammasome contributes to the significantly increased virulence and the high incidence of acute lung injury (59, 60). This is also one of the rationales behind the Greek Study in the Effects of Colchicine in COVID-19 Complications Prevention (GRECCO) trial, which is investigating the utility of colchicine, as a non-specific inhibitor of the NLRP3 inflammasome, in the treatment of COVID-19 infection (61, 62). By inhibiting this pathway along with nuclear factor kappa B (NF-KB) inhibition of the innate immune response (63), statins have also been shown to be potentially beneficial for patients with COVID19 infection via intervention through this pathway (64).

In patients with stroke, it has been shown that the NLRP3 inflammasome plays a significant role in cerebral atherogenesis by similar activation of the immune system and increase in macrophages, neutrophils, lymphocytes, and vascular smooth muscle cell, which also play an important role in plaque instability (6567). The neuronal cell death, which can be attributed to the NLRP3 inflammasome activation, has also been shown to be reversible with immunoglobulin in 3-month-old mice stroke models (65). However, in patients with impaired immune responses, such as the elderly, there is an unprecedented activation of the NLRP3 inflammasome coupled with mitochondrial dysfunction and increased proportions of the mitochondrial reactive oxygen species, which further worsen tissue damage and initiate cell death (66). In patients with obesity and diabetes in which NLRP3 inflammasomes are already basally activated, and the immune responses are suboptimal, viral-induced activation of the former may worsen a preexisting chronic inflammatory state (67). Hence, this factor may be related to the poor outcomes of elderly patients with COVID-19 and stroke and with traditional cardiovascular risk factors such as obesity and diabetes (10).

Inflammation-Induced Plaque Progression and Vulnerability

COVID-19 inflammation induces a prothrombotic milieu among patients who are at risk of vascular events and those with the pre-existing atheromatous disease. Among patients with coronary disease, evidence suggests that virally induced inflammatory infiltrates such as T cells, macrophages, and neutrophils populate the atheromatous plaque leading to a cascade of events including vascular permeability, endothelial disruption, and exposure of prothrombotic elements such as collagen, tissue factor, and platelet adhesion molecules, which all play a role in thrombogenesis (68). Furthermore, it is known that carotid artery plaques with features of a thin fibrous cap, large core lipid, intraplaque bleeding, and the abundance of monocyte-derived macrophages and activated smooth muscle cells cause instability and vulnerability to plaque rupture (69). The presence of proteolytic enzymes such as metalloproteinases (MMPs) and cathepsin cysteine proteases (CCPs), which are secreted by activated macrophages, promote degradation of the extracellular matrix and plaque fragility and potentially result in thromboembolism (16, 34) while making acute ischemic stroke a likely event in the potential trajectories of patients with systemic COVID-19 infection.

Indeed Mohamud et al. have described five cases of acute ischemic stroke associated with an intraluminal carotid artery thrombus with concomitant COVID-19 symptoms 0–14 days before the onset of stroke (70). A proposed mechanism of this co-occurrence is inflammation-related plaque rupture as manifested by the elevated levels of inflammatory biomarkers such as lactate dehydrogenase, C-reactive protein, ferritin, D-dimer, and interleukin (70). Another study reported a case of symptomatic intracranial stenosis in a confirmed COVID-19 patient in a hyperinflammatory state, as evidenced by elevated acute inflammatory biomarkers (71). A similar hyperimmune trend was described in another patient with symptomatic posterior circulation stenosis and concomitant stroke (72). While these biomarkers are not specific for plaque rupture, there is evidence to suggest tha tplaque-rupture-specific MMPs implicate temporal association with the onset of acute respiratory failure among COVID-19 patients (73). Moreover, it has been proposed that aprotinin, a protease inhibitor that inhibits MMPs, may provide benefit for patients with COVID-19-related acute respiratory distress syndrome in experimental studies (74). However, at least in severe cases, the use of aprotinin to inhibit clot breakdown may need to be cautioned if the likelihood of COVID-19 inducing a proinflammatory hypercoagulation response to the SARS-CoV2 virus is considered.

Another putative mechanism that can lead to plaque progression in patients with COVID-19 infection is the disruption of the receptor-mediated uptake of oxidized low-density lipoproteins (oxLDL) by the monocyte-derived macrophages with exposure to proinflammatory stimuli (19). Indeed, animal and human studies indicate that increased expression of lectin-like oxidized lipoprotein 1 receptor (LOX-1) is a risk factor for stroke and promotes restenosis among patients with preexisting cardiovascular disease (75). It is also evident that COVID-19 infection results in a disproportionate increase in reactive oxygen species, which translate to lipid oxidation and increased oxLDL levels (20). Thus, this is presumably another one of the reasons why elderly patients with preexisting cardiovascular risk have a higher propensity of developing severe COVID-19 illness (76) and AIS.

The Role of Peripheral Biomarkers to COVID-19 That Are Also Characteristic of Ischemic Stroke

The pathognomonic features of sepsis-induced inflammation among patients with coronavirus infection is manifested peripherally as neutrophilia in combination with marked lymphopenia (77). This is characterized by the marked increase in various inflammatory biomarkers such as the neutrophil to lymphocyte ratio (NLR), C-reactive protein, and serum ferritin (2123, 7884). In a prospective cohort study involving patients with concomitant COVID-19 and ischemic stroke, 9 of the 10 patients have elevated NLR (85). Similarly, the majority of AIS patients presented recently (86, 87) and in various COVID-19 and AIS case series has reported increased NLR values (72, 8891). A significant degree of lymphopenia coupled with viral-induced neutrophilia and the migration of the neutrophils to the ischemic core may explain the elevated NLR among patients with ischemic stroke (92).

CRP is another inflammatory biomarker that is disproportionately increased among patients with coronavirus infection and AIS. The largest cohort study involving 32 ischemic stroke patients with concomitant COVID-19 infection reported that elevated CRP occurred in more than 90% of the patients (93, 94). All of the six ischemic stroke patients reported by Beyroutti et al. and three of the four patients observed by Tunc et al. showed elevated CRP levels (72, 89). AIS patients described in various case reports and case series with COVID-19 infection have also been reported to have raised CRP levels (16, 38, 91, 9598) While CRP is a marker of inflammation, a meta-analysis likewise confirms its role in thromboembolic events (98). On the other hand, serum ferritin, an acute phase reactant and an inflammatory biomarker has also been reported to be elevated among patients with COVID-19 and AIS (85, 88, 89, 99, 100). Apart from its role in systemic inflammation, serum ferritin likewise predicts the degree of neural damage among patients with AIS as characterized by its correlation with markers of blood–brain barrier damage such as glutamate, interleukin-6, matrix metalloproteinase-9, and cellular fibronectin (35, 82).

Neutrophil Increase, Lymphocyte Decrease, and Neutrophil Extracellular Traps in Thrombin in General and Previously Associated With Acute Ischemic Stroke

As alluded to above, COVID-19 infection is associated with reduced leukocyte number and increased neutrophil to leukocyte ratio and so makes the recent evidence of dysregulated neutrophil extracellular traps (NETs) predictable (101103). NETs are neutrophil-produced extracellular networks of chromatin, proteins, and oxidant enzymes that protrude from membranes of activated neutrophil and mediate infection containment (101). NETs also play a role in thrombus formation. Evidence indicates that, in patients with COVID-19 infection, there is an upsurge of NET production that further propagates inflammation and thrombosis (101103). The NET formation is characterized by elevated levels of cell-free DNA, myeloperoxidase-DNA (MPO-DNA), and citrullinated histone H3 (Cit-H3) (25, 101). A study of normal subjects compared with patients with COVID-19 infection has shown that the latter have higher levels of cell-free DNA, MPO-DNA, and Cit-H3 with both MPO-DNA and Cit-H3 being significantly elevated in mechanically ventilated patients (101). Pulmonary autopsies of patients with confirmed COVID-19 patients also confirm the presence of NET-containing microthrombi with neutrophil–platelet infiltration (103, 104).

The evidence of the role of NETs in AIS is well-described (17, 18, 105, 106, 114). Laridan et al. examined specimens of thrombi of patients undergoing endovascular thrombectomy and found that Cit-H3, the hallmark for NETs, was observed in the majority of the samples (17). Interestingly, patients with cardioembolic etiology have been shown to have a higher burden of NETs (102). Another study revealed that the presence of NETs may convey reperfusion resistance to mechanical and systemic revascularization procedures (17). Whether the role of neutrophils and NET in coronavirus-related stroke is secondary to micro- or macrothrombosis needs further elucidation.

Viral-Induced Coagulation Dysfunction

Another important mechanism resulting in life-threatening systemic thromboembolic events among patients with coronavirus infection is the disruption of the coagulation pathways. Pro- and hypercoagulation affecting macro- and microvascular systems have also been implicated in various vascular events such as ischemic strokes. The International Society of Thrombosis and Hemostasis (ISTH) recognizes this unique phenomenon of sepsis-induced coagulopathy and proposed monitoring of platelet, D-dimer, PT, and fibrinogen among COVID-19 patients needing admission (115).

Fibrinolytic Shutdown

The imbalance between coagulation and fibrinolysis favoring the former manifests hematologically as an increase in D-dimer levels (115, 116). This is an immunological defense for the body to contain the viral infection, which results in thrombi formation (116). Furthermore, this implies a shutdown in the fibrinolytic system to clear the necrotic debris leading to massive fibrin formation, which also orchestrates coagulopathy (117). This shutdown has been further analyzed among critically ill COVID-19 patients where it has been demonstrated that, apart from the elevation D-dimer levels, more than 50% of the patients failed to lyse clots on thromboelastography (118120) and hence increasing the likelihood of AIS as a further manifestation of COVID-19.

The tendency for 5% (this is still a gross underestimation, as we may never know the exact number strokes among deaths and even the mild strokes that might not present to the hospitals, as almost 40% of the stroke patients are not attending the hospitals during the pandemic time) of patients with COVID-19-related strokes to show spectacularly elevated D-dimer levels has been reported in several studies (91, 121, 122). Among the 32 cases reported by Yaghi et al., almost 50% showed D-dimer levels elevated to more than five times the normal range (94). Five of the six cases described by Beyrouti et al. also demonstrated D-dimer levels elevated more than 5–150 times above the normal limit (72). Avula et al., Lodigiani et al., Oxley et al., Berekashvili et al., and Wijeratne et al. also observed similar trends in the patients they described (3, 85, 88, 91, 123).

Venous thrombosis has also been described in a number of cases of COVID-19 infection resulting in fatal outcomes (107). Unsurprisingly, D-dimer levels were disproportionately elevated among patients with cerebral venous thrombosis (107, 108, 124). Another biomarker of fibrinolytic shutdown is the accumulation of fibrinogen and fibrin degradation products (119), and again, such retrospective analysis comparing COVID-19 and non-COVID-19 patients revealed higher fibrinogen levels in the former (123). Similarly, patients described in the literature with stroke and active COVID-19 infection have elevated serum fibrinogen levels (3, 72, 85).

The abnormalities of the coagulation biomarkers associated with COVID infection have been used to justify the use of systemic anticoagulation even in the acute stroke period. However, the outcomes for COVID patients are inconsistent (89, 123). Two of the five patients described by Lodigiani et al. who received Nadroparin died, and two of the four patients in Tunc's case series remain bedridden despite heparinization at the time of the publication (89, 123). Among the 32 COVID-19-related AIS described by Yaghi et al., 25 of whom received anticoagulation, more than 80% died or remained critically ill at the time of the publication (94). While the reason for poor neurological outcomes among these patients is multifactorial, a potential contributor is heparin resistance, especially among critically ill COVID-19 patients (24, 125).

For the small number of cases of COVID-19 patients who have also suffered AIS and received reperfusion therapy, consisting of intravenous thrombolysis or endovascular thrombectomy or a combination of both, the outcomes have not been particularly positive. In a case series regarding four patients with COVID-19-related AIS who also received systemic thrombolysis, all died (126). Similarly, in COVID-19-infected stroke patients reported in a healthcare system in New York who received alteplase and mechanical thrombectomy, the neurological outcomes were also poor (94). Currently, there is inadequate data to determine whether patients with COVID-19-related strokes may have resistance to tPA; however, it is known that the activation of the renin–angiotensin–aldosterone system results in the formation of plasminogen activator inhibitor (PAI-1), an inhibitor of tPA (127). Thus, it remains to be investigated whether these poor outcomes are related to counteractive mechanisms of PAI-I, increased thrombus burden, or resistance to tPA (128).

The Role of Natural Anticoagulants and Antiphospholipid Antibodies

Hypercoagulability is another sequela of COVID-19, especially in critically ill patients. One of the putative mechanisms, to which this is attributed, is the depletion of physiological anticoagulants and increased levels of coagulant factors and antiphospholipid antibodies (129131). In a cross-sectional study of COVID-19-positive critically ill patients in China, it has been shown that the plasma levels of natural anticoagulants such as protein C, protein S, and antithrombin were below physiologically normal levels (127). The same study revealed that more than 50% of the patients developed antiphospholipid antibodies (127) that have been suggested to play a role in the pathogenesis of COVID-19-related strokes. Zhang et al. described three patients with multiple cerebral infarctions who tested positive for anticardiolipin immunoglobulin A (IgA) antibodies as well as anti-β2-glycoprotein I IgA and IgG antibodies (130). On the other hand, five of the six COVID-19-associated stroke patients described by Beyroutti et al. did not show antiphospholipid antibodies, but all of them were positive for lupus anticoagulant (LA) (72). Two other severe stroke patients with poor neurological outcome and concomitant COVID-19 infection have also been reported as testing negative for both antiphospholipid antibodies and LA (99, 100). Thus, while there is inconsistency in the trend of these procoagulant factors in ischemic stroke patients, Zhang has proposed a possible secondary antiphospholipid antibody syndrome especially in patients who have thrombotic manifestations and positive serum antibodies (130). If this was the case, then the initiation of therapeutic anticoagulation would be required for these patients (130). However, it has not translated to screening of all COVID-19 patients, as a transient increase in these parameters is expected in viral infections (129).

The Role of the Endothelium

A component of Virchow's triad, endothelial dysfunction is another major contributor to SARS-CoV-associated thrombosis. The virus has been shown to demonstrate a predilection to invade vascular endothelial cells by attaching to the ACE2 protein, which facilitates subsequent invasion (9, 132, 133).

Endothelium-Driven Activation of the Extrinsic Coagulation System

COVID-19 promotes an imbalance between ACE2 and angiotensin II (AT-II) receptors leading to the upregulation of the latter, which is responsible for the tonic production of platelet tissue factor (TF or factor111) in platelets and macrophages, and further interaction with factor VII to activate the extrinsic coagulation system (134, 135). TF is essential for fibrin formation at the site of vascular injury and may also be an important factor contributing to thrombogenesis, especially with the induction of inflammatory cells originating from the subendothelium (26).

Increased expression of TF has previously also been shown to be associated with thrombus formation in patients with ischemic stroke (109, 110). The EPICOR study in 2015 revealed that patients with elevated TF doubled the risk of stroke (109). Experimental studies also demonstrated that the inhibition of TNF-α-induced TF significantly reduces atheromatous plaque formation (30). Whether inflammation-induced tissue factor formation has a role in intracranial thrombosis in COVID patients particularly remains speculative.

Endothelium-Driven Nitrous Oxide Deficiency

Healthy endothelium is necessary to produce nitric oxide (NO), which is known as a vasodilator and plays a role in preventing thrombosis (27). SARS-CoV2-related endotheliopathy results in the suppression of nitric oxide synthase (NOS), resulting in nitric oxide deficiency (133). In vitro studies performed in 2009 on SARS-CoV also revealed that nitrous oxide inhibited viral entry replication by decreasing spike (S) protein expression and via its effects on the cysteine proteases encoded in the SARS-CoV (133).

Endothelium-derived nitrous oxide affects the cerebral circulation by its tonic vasodilatory effect and the prevention of adhesion of platelets and leukocytes to the vessel wall, both of which have an obligatory role in stroke pathogenesis (28). A study comparing stroke patients and healthy controls revealed that the levels of asymmetrical dimethylarginine (ADMA), a NOS inhibitor, is increased in the former, making it a risk factor for stroke (29). To date, few studies, if any, are looking into the potential role of NO in the treatment and prevention of COVID-19-related strokes; however, there is evidence to suggest that NO may also have a potential benefit for pulmonary-related COVID-19 complications (136).

The Role of the Renin–Angiotensin–Aldosterone System and Ace2 Deficiency

A unique characteristic of the SARS-CoV2 infection is its avid interaction with the ACE2 receptors, which affect the RAAS to a significant degree (137). It is known that viral-induced ACE2 deficiency results in significant alterations in the balance between the classical RAAS and the counterregulatory ACE2, which is essential for maintaining homeostatic mechanisms in the body (137). In particular, ACE2, which is ubiquitous in the brain, heart, and the vascular systems protects patients from the organ-damaging effects of the classical RAAS (138). This COVID-19-related disruption of the RAAS axes is likely to contribute to stroke pathogenesis, as it promotes inflammation, vasoconstriction, and end-organ damage (139, 140).

Various COVID-19 registries have shown that there is an increased fatality rate among patients with preexisting cardiovascular diseases such as hypertension and diabetes (140). Similarly, a majority of patients with AIS and COVID-19 report hypertension as a classical risk factor (111). The contribution of hypertension in the outcomes of patients with COVID-19 infection is likely explained in various mechanisms. Still, overactivity of the classical RAAS along with viral-induced endothelial vasoconstriction may be factors adding to the neurological exacerbation of COVID infection (140). Worse neurological outcomes among patients with COVID and ischemic strokes may be attributable to the unimpeded activity of the classical RAAS pathway in the brain, which has also been implicated in higher brain functions such as memory and cognition (141). Furthermore, upregulation of the classical RAAS pathway is likely to result in the overactivity of the sympathetic nervous system, which may exacerbate preexisting traditional risk factors for stroke such as hypertension, diabetes, and cardiac dysrhythmias (142).

Covid, Neurological Manifestations and Implications for Neurorehabilitation

Neurorehabilitation for patients recovering from severe COVID and AIS is set to be extra challenging in times of pandemic with its requirements for social distancing and, in many cases, need for online delivery in the immediate post-COVID stages. Currently, there are little reliable data about how well individuals with severe respiratory system and lung damage will recover or whether scarring will make such patients vulnerable to other viral infections (31). Furthermore, questions remain regarding whether the hypoxia engendered during the acute hospitalization phase will have lasting effects on the brain and nerve tissue especially on high metabolic demand areas including the sympathetic system and the fast visual attention and eye movements pathways (112) and long motor pathways.

Helms et al. first described the occurrence of dysexecutive syndrome in 36% of patients of critically ill COVID survivors in a French cohort characterized by lack of attention and disorganized movement with commands (143). Various case reports likewise described cases of patients who had severe impairments in executive function in the postacute stage who had significant recovery after aggressive neurorehabilitation (37, 144). In the same manner, varying degrees of depression and anxiety have also been identified after the acute stage of infection among COVID survivors (36, 145, 146). Interestingly, a meta-analysis of observational studies also notes that, among patients with previous SARS and MERS-CoV infection, neuropsychiatric manifestations such as depression, anxiety, irritability, memory impairment, sleep deprivation, and posttraumatic disorder were strikingly prevalent (147). One would also expect that psychological anxiety and the physiologically high immune responses (psychoneuroimmunological outcomes) would also lead to various psychiatric sequelae that have adverse effects on mental health of patients (32, 148). The neurological sequelae of SARS-CoV2 infection have gained recent publicity in social and print media (Facebook “Long haul COVID support groups”). Symptoms include persistent “brain fog,” fatigue, breathlessness, anxiety, depressive mood, and motor weakness, affecting subsets of young, middle aged, and older patients including a significant proportion who showed only mild symptoms, during the infected stage (40, 149). We have suggested that these symptoms comprise aspects of PCNS and hypothesized that such a syndrome is due to persistent inflammation following COVID-19 infection (113, 150). These complications are undeniably posing significant challenges ahead for the neurorehabilitation fraternity.

Mathew et al. identified a subgroup of patients with T-cell activation characteristic of acute viral response plasmablast responses reaching over 30% of the circulating cells with three different immunotypes associated with poor clinical trajectories supporting the current understanding of the pathobiology (151).

Fridman et al. found the frequency of stroke to be high among patients with severe COVID-19 with a very high inpatient mortality across all age groups demonstrating further evidence to the shared pathobiology as one go through the cluster analysis of this paper (143).

Overall, our review of the pathobiological basis of COVID-19 demonstrates that it is a multisystem illness with high likelihood of long-term physical, cognitive, psychological, and social sequalae in those who survive the illness. The scale of the burden of the disease and impact is enormous globally.

Furthermore, there is also no doubt that the current pandemic is overwhelming the neurorehabilitation sector globally. While it is too early to comment about the potential pandemic of long-term neurological issues for the millions of infected patients worldwide, the current review predicts the high likelihood of this possibility. Similarly, Boldrini et al. has noted that the two main factors already impacting the neurorehabilitation sector during the first phase of the pandemic (144) are the following:

(i) increased pressure from the acute care services to transfer the patients to rehabilitation medicine services;

(ii) increased difficulties without patient rehabilitation activities and home-based rehabilitation due to the restrictions enforced by the local and national authorities (144).

Indeed, the challenges described by Boldrini et al. above make the situation even more complex. It is critical to create an efficient healthcare delivery system tailored to each individual, each community, and each country. It is equally important to understand the biological underpinning behind the illness and post-COVID-19 complications. Tailor-made, individualized, targeted neurorehabilitation program should be developed with the available resources (e.g., telemedicine can be tested with easily available smart phone-based deliverables such as WhatsApp, Viber, Facetime, and Google Talk where professional personnel are not always available and sophisticated technology are not around). Family members can be a really useful resource in these challenging circumstances (36, 37, 145, 146).

To this end, we recommend considerations of Mantovani et al. (152) who recently collected and reviewed relevant evidence-based recommendations on the efficacy of cognitive rehabilitation, based on a comprehensive evaluation of 491 papers presenting the latest evidence-based practice for neurorehabiliation of traumatic brain injury (TBI) patients whose pathobiology bears many similarities to that presented in this review in the context of COVID-19 and systemic involvement (147, 152, 153).

Mantovani et al. as well offer a road map and a comprehensive perspective on the role of tele-neurorehabilitation and virtual rehabilitation to gain adequate cognitive stimulation in the era of physical distancing and lockdowns related to COVID-19 pandemic, reminding us of the potential opportunities globally (152). For further details, see Mantovani et al. and the 29 recommendations on evidence-based cognitive rehabilitation in the context of stroke and TBI nominated by the Cognitive Rehabilitation Task Force (CRTF), which suggests that a more comprehensive biological understanding of the various systems affected by TBI should lead to more collaborative translational research utilizing artificial intelligence and novel technology to find better individually designed precision solutions (e.g., video-consults, tablet-based measurements, and use of wearable devices). Lastly, the integration of services, including private and public partnerships such as university–public hospital partnerships and partnerships with community organizations and volunteers, are likely to create excellent opportunities for the future (112, 154, 155) and better evidence-based neurorehabilitation pathways.

In support of this more generalized theoretical approach, our group has demonstrated here the expected neurological impact of shared pathobiology between AIS and COVID-19 here, and the debilitating condition that characterizes PCNS (150) manifests as chronic fatigue, impaired thinking, depression, anxiety, breathing difficulties, and muscle weakness (40, 40, 149, 150). Thus, we suggest that treatment with known Food and Drug Administration (FDA)-approved immunomodulatory hormones such as melatonin (14, 15, 33, 156162) and nutraceuticals such as curcumin (163) might be expected to enhance the likelihood of PCNS patients benefitting from any neurorehabilitation interventions as a matter of priority.

Conclusion

Our review has shown here that severe COVID-19 infection has the potential to lead to the disruption of most physiological systems and results in various multisystemic thrombotic phenomena including acute ischemic stroke. While inflammation orchestrates its pathogenesis, the further perturbation of the coagulation system resulting in fibrinolytic shutdown likewise contributes to neurological manifestation. Furthermore, the predilection of the virus to attach to ACE2 receptors in various cells, including the vascular endothelial system, may also disrupt the renin–angiotensin system, which further contributes to stroke pathogenesis. Clearly, there is a gap in the understanding of this phenomenon, and large-scale human and animal studies are necessary, as this co-occurrence results in deleterious outcomes.

The impact of COVID-19 in the human brain is as such that millions of patients are likely to experience problems with normal functioning requiring far more psychoneuroimmunological evidence-based understanding of neurorehabilitation services that are able to overcome incipient recovery problems.

Data Availability Statement

The original contributions generated in the study are included in the article/supplementary material, further inquiries can be directed to the corresponding author.

Author Contributions

TW conceived the idea. TW and CS performed the literature search independently. TW, CS, SC, and LK contributed to the manuscript writing with critical revisions. All authors read the final manuscript and agreed.

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

1. Al-Ani F, Chehade S, Lazo-Langner A. Thrombosis risk associated with COVID-19 infection. A scoping review. Thromb Res. (2020) 192:152–60. doi: 10.1016/j.thromres.2020.05.039

CrossRef Full Text | Google Scholar

2. Mao L, Jin H, Wang M, Hu Y, Chen S, He Q, et al. Neurologic manifestations of hospitalized patients with coronavirus disease 2019 in Wuhan, China. JAMA Neurol. (2020) 77:683–90. doi: 10.1001/jamaneurol.2020.1127

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Oxley TJ, Mocco J, Majidi S, Kellner CP, Shoirah H, Singh IP, et al. Large-vessel stroke as a presenting feature of covid-19 in the Young. N Engl J Med. (2020) 382:e60. doi: 10.1056/NEJMc2009787

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Markus HS, Brainin M. COVID-19 and stroke-a global world stroke organization perspective. Int J Stroke. (2020) 15:361–4. doi: 10.1177/1747493020923472

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Aggarwal G, Lippi G, Michael Henry B. Cerebrovascular disease is associated with an increased disease severity in patients with Coronavirus Disease 2019 (COVID-19): a pooled analysis of published literature. Int J Stroke. (2020) 15:385–9. doi: 10.1177/1747493020921664

PubMed Abstract | CrossRef Full Text | Google Scholar

6. McColl BW, Allan SM, Rothwell NJ. Systemic infection, inflammation and acute ischemic stroke. Neuroscience. (2009) 158:1049–61. doi: 10.1016/j.neuroscience.2008.08.019

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Umapathi T, Kor AC, Venketasubramanian N, Lim CC, Pang BC, Yeo TT, et al. Large artery ischaemic stroke in severe acute respiratory syndrome (SARS). J Neurol. (2004) 251:1227–31. doi: 10.1007/s00415-004-0519-8

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Kim JE, Heo JH, Kim HO, Song SH, Park SS, Park TH, et al. Neurological complications during treatment of middle east respiratory syndrome. J Clin Neurol. (2017) 13:227–33. doi: 10.3988/jcn.2017.13.3.227

PubMed Abstract | CrossRef Full Text

9. Ni W, Yang X, Yang D, Bao J, Li R, Xiao Y, et al. Role of angiotensin-converting enzyme 2 (ACE2) in COVID-19. Crit Care. (2020) 24:422. doi: 10.1186/s13054-020-03120-0

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Najjar S, Najjar A, Chong DJ, Pramanik BK, Kirsch C, Kuzniecky RI, et al. Central nervous system complications associated with SARS-CoV-2 infection: integrative concepts of pathophysiology and case reports. J Neuroinflammation. (2020) 17:231. doi: 10.1186/s12974-020-01896-0

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Spence JD, de Freitas GR, Pettigrew LC, Ay H, Liebeskind DS, Kase CS, et al. Mechanisms of Stroke in COVID-19. Cerebrovasc Dis. (2020) 49:451–8. doi: 10.1159/000509581

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Gupta A, Madhavan MV, Sehgal K, Nair N, Mahajan S, Sehrawat TS, et al. Extrapulmonary manifestations of COVID-19. Nat Med. (2020) 26:1017–32. doi: 10.1038/s41591-020-0968-3

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Arksey H, O'Malley L. Scoping studies: towards a methodological framework. Int J Soc Res Methodol. (2005) 8:19–32. doi: 10.1080/1364557032000119616

CrossRef Full Text | Google Scholar

14. Anderson G, Reiter RJ. Melatonin: roles in influenza, Covid-19, and other viral infections. Rev Med Virol. (2020) 30:e2109. doi: 10.1002/rmv.2109

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Carrillo-Vico A, Lardone PJ, Alvarez-Sánchez N, Rodríguez-Rodríguez A, Guerrero JM. Melatonin: buffering the immune system. Int J Mol Sci. (2013) 14:8638–83. doi: 10.3390/ijms14048638

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Camaré C, Pucelle M, Nègre-Salvayre A, Salvayre R. Angiogenesis in the atherosclerotic plaque. Redox Biol. (2017) 12:18–34. doi: 10.1016/j.redox.2017.01.007

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Laridan E, Denorme F, Desender L, François O, Andersson T, Deckmyn H, et al. Neutrophil extracellular traps in ischemic stroke thrombi. Ann Neurol. (2017) 82:223–32. doi: 10.1002/ana.24993

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Lim HH, Jeong IH, An GD, Woo KS, Kim KH, Kim JM, et al. Evaluation of neutrophil extracellular traps as the circulating marker for patients with acute coronary syndrome and acute ischemic stroke. J Clin Lab Anal. (2020) 34:e23190. doi: 10.1002/jcla.23190

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Pirillo A, Norata GD, Catapano AL. LOX-1, OxLDL, and atherosclerosis. Mediators Inflamm. (2013) 2013:152786. doi: 10.1155/2013/152786

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Schonrich G, Raftery MJ, Samstag Y. Devilishly radical NETwork in COVID-19: oxidative stress, neutrophil extracellular traps (NETs), and T cell suppression. Adv Biol Regul. (2020) 77:100741. doi: 10.1016/j.jbior.2020.100741

CrossRef Full Text | Google Scholar

21. Liu Y, Du X, Chen J, Jin Y, Peng L, Wang HHX, et al. Neutrophil-to-lymphocyte ratio as an independent risk factor for mortality in hospitalized patients with COVID-19. J Infect. (2020) 81:e6–12. doi: 10.1016/j.jinf.2020.04.002

CrossRef Full Text | Google Scholar

22. Liu J, Li S, Liu J, Liang B, Wang X, Wang H, et al. Longitudinal characteristics of lymphocyte responses and cytokine profiles in the peripheral blood of SARS-CoV-2 infected patients. EBioMedicine. (2020) 55:102763. doi: 10.1016/j.ebiom.2020.102763

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Accinelli RA, Zhang Xu CM, Ju Wang JD, Yachachin-Chavez JM, Caceres-Pizarro JA, Tafur-Bances KB, et al. COVID-19: la pandemia por el nuevo virus SARS-CoV-2. Rev Peru Med Exp Salud Publica. (2020) 37:302–11. doi: 10.17843/rpmesp.2020.372.5411

PubMed Abstract | CrossRef Full Text | Google Scholar

24. White D, MacDonald S, Bull T, Hayman M, de Monteverde-Robb R, Sapsford D, et al. Heparin resistance in COVID-19 patients in the intensive care unit. J Thromb Thrombolysis. (2020) 50:287–91. doi: 10.1007/s11239-020-02145-0

CrossRef Full Text | Google Scholar

25. Hirose T, Hamaguchi S, Matsumoto N, Irisawa T, Seki M, Tasaki O, et al. Presence of neutrophil extracellular traps and citrullinated histone H3 in the bloodstream of critically ill patients. PLoS ONE. (2014) 9:e111755. doi: 10.1371/journal.pone.0111755

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Saha D, Saha S, Sergeeva EG, Ionova ZI, Gorbach AV. Tissue factor and atherothrombosis. Curr Pharm Des. (2015) 21:1152–7. doi: 10.2174/1381612820666141013154946

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Tousoulis D, Kampoli AM, Tentolouris C, Papageorgiou N, Stefanadis C. The role of nitric oxide on endothelial function. Curr Vasc Pharmacol. (2012) 10:4–18. doi: 10.2174/157016112798829760

CrossRef Full Text | Google Scholar

28. Cosentino F, Rubattu S, Savoia C, Venturelli V, Pagannonne E, Volpe M. Endothelial dysfunction and stroke. J Cardiovasc Pharmacol. (2001) 38(Suppl. 2):S75–8. doi: 10.1097/00005344-200111002-00018

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Ercan M, Mungan S, Güzel I, Celik HT, Bal C, Abusoglu S, et al. Serum asymmetric dimethylarginine and nitric oxide levels in Turkish patients with acute ischemic stroke. Adv Clin Exp Med. (2019) 28:693–8. doi: 10.17219/acem/78360

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Hao E, Pang G, Du Z, Lai Y-H, Chen J-R, Xie J, et al. Peach kernel oil downregulates expression of tissue factor and reduces atherosclerosis in ApoE knockout mice. Int J Mol Sci. (2019) 20:405. doi: 10.3390/ijms20020405

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Lee C-H, Giuliani F. The role of inflammation in depression and fatigue. Front Immunol. (2019) 10:1696. doi: 10.3389/fimmu.2019.01696

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Slavich GM, Auerbach, RP. Stress and its Sequelae: Depression, Suicide, Inflammation, and Physical Illness. In: Hooley JM, Butcher JN, editor. Washington DC: American Psychological Association (2018).

Google Scholar

33. Chitimus DM, Popescu MR, Voiculescu SE, Panaitescu AM, Pavel B, Zagrean L, et al. Melatonin's impact on antioxidative and anti-inflammatory reprogramming in homeostasis and disease. Biomolecules. (2020) 10:1211. doi: 10.3390/biom10091211

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Hermus L, Lefrandt JD, Tio RA, Breek JC, Zeebregts CJ. Carotid plaque formation and serum biomarkers. Atherosclerosis. (2010) 213:21–9. doi: 10.1016/j.atherosclerosis.2010.05.013

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Millán M, Sobrino T, Arenillas JF, Rodríguez-Yáñez M, García M, Nombela F, et al. Biological signatures of brain damage associated with high serum ferritin levels in patients with acute ischemic stroke and thrombolytic treatment. Dis Markers. (2008) 25:181–8. doi: 10.1155/2008/380356

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Bermúdez i Badia S, Cameirão MS. The neurorehabilitation training toolkit (NTT): a novel worldwide accessible motor training approach for at-home rehabilitation after stroke. Stroke Res Treat. (2012) 2012:802157. doi: 10.1155/2012/802157

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Foster AM, Armstrong J, Buckley A, Sherry J, Young T, Foliaki S, et al. Encouraging family engagement in the rehabilitation process: a rehabilitation provider's development of support strategies for family members of people with traumatic brain injury. Disabil Rehabili. (2012) 34:1855–62. doi: 10.3109/09638288.2012.670028

PubMed Abstract | CrossRef Full Text | Google Scholar

38. González-Pinto T, Luna-Rodríguez A, Moreno-Estébanez A, Agirre-Beitia G, Rodríguez-Antigüedad A, Ruiz-Lopez M. Emergency room neurology in times of COVID-19: malignant ischaemic stroke and SARS-CoV-2 infection. Eur J Neurol. (2020) 27:e35–6. doi: 10.1111/ene.14286

PubMed Abstract | CrossRef Full Text | Google Scholar

39. South K, McCulloch L, McColl BW, Elkind MS, Allan SM, Smith CJ. Preceding infection and risk of stroke: an old concept revived by the COVID-19 pandemic. Int J Stroke. 2020 15:722–32. doi: 10.1177/1747493020943815

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Goërtz YMJ, Van Herck M, Delbressine JM, Vaes AW, Meys R, Machado FVC, et al. Persistent symptoms 3 months after a SARS-CoV-2 infection: the post-COVID-19 syndrome? Spruit ERJ Open Res. (2020) 6:00542–2020. doi: 10.1183/23120541.00542-2020

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Adamczyk-Sowa M, Niedziela N, Kubicka-Baczyk K, Wierzbicki K, Jaroszewicz J, Sowa P. Neurological symptoms as a clinical manifestation of COVID-19: implications for internists. Pol Arch Intern Med. (2020). doi: 10.20452/pamw.15575. [Epub ahead of print].

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Mankad K, Perry MD, Mirsky DM, Rossi A. COVID-19: a primer for Neuroradiologists. Neuroradiology. (2020) 62:647–8. doi: 10.1007/s00234-020-02437-5

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Zhou P, Yang X-L, Wang X-G, Hu B, Zhang L, Zhang W, et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature. (2020) 579:270–3. doi: 10.1038/s41586-020-2012-7

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Yang M, Li CK, Li K, Hon KLE, Ng MHL, Chan PKS, et al. Hematological findings in SARS patients and possible mechanisms (review). Int J Mol Med. (2004) 14:311–5. doi: 10.3892/ijmm.14.2.311

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Soy M, Keser G, Atagündüz P, Tabak F, Atagündüz I, Kayhan S. Cytokine storm in COVID-19: pathogenesis and overview of anti-inflammatory agents used in treatment. Clin Rheumatol. (2020) 39:2085–94. doi: 10.1007/s10067-020-05190-5

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Agbuduwe C, Basu S. Haematological manifestations of COVID-19: from cytopenia to coagulopathy. Eur J Haematol. (2020) 105:540–6. doi: 10.1111/ejh.13491

PubMed Abstract | CrossRef Full Text | Google Scholar

47. An PJ, Zhu YZ, Yang LP. Biochemical indicators of coronavirus disease 2019 exacerbation and the clinical implications. Pharmacol Res. (2020) 159:104946. doi: 10.1016/j.phrs.2020.104946

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Bindoli S, Felicetti M, Sfriso P, Doria A. The amount of cytokine-release defines different shades of Sars-Cov2 infection. Exp Biol Med (Maywood). (2020) 245:970–6. doi: 10.1177/1535370220928964

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Misra DP, Agarwal V, Gasparyan AY, Zimba O. Rheumatologists' perspective on coronavirus disease 19 (COVID-19) and potential therapeutic targets. Clin Rheumatol. (2020) 39:2055–62. doi: 10.1007/s10067-020-05073-9

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Soy M, Atagündüz P, Atagündüz I, Sucak GT. Hemophagocytic lymphohistiocytosis: a review inspired by the COVID-19 pandemic. Rheumatol Int. (2020) 6:1–12. doi: 10.1007/s00296-020-04636-y

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Crayne CB, Albeituni S, Nichols KE, Cron RQ. The immunology of macrophage activation syndrome. Front Immunol. (2019) 10:119. doi: 10.3389/fimmu.2019.00119

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Raschke RA, Garcia-Orr R. Hemophagocytic lymphohistiocytosis: a potentially underrecognized association with systemic inflammatory response syndrome, severe sepsis, and septic shock in adults. CHEST. (2011) 140:933–8. doi: 10.1378/chest.11-0619

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Filipovich AH. Hemophagocytic lymphohistiocytosis and other hemophagocytic disorders. Immunol Allergy Clin. (2008) 28:293–313. doi: 10.1016/j.iac.2008.01.010

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Emmenegger U, Schaer DJ, Larroche C, Neftel KA. Haemophagocytic syndromes in adults: current concepts and challenges ahead. Swiss Med Wkly. (2005) 135:299–314.

PubMed Abstract | Google Scholar

55. Castillo L, Carcillo J. Secondary hemophagocytic lymphohistiocytosis and severe sepsis/ systemic inflammatory response syndrome/multiorgan dysfunction syndrome/macrophage activation syndrome share common intermediate phenotypes on a spectrum of inflammation. Pediatr Crit Care Med. (2009) 10:387–92. doi: 10.1097/PCC.0b013e3181a1ae08

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Chen I-Y, Moriyama M, Chang M-F, Ichinohe T. Severe acute respiratory syndrome coronavirus viroporin 3a activates the NLRP3 inflammasome. Front Microbiol. (2019) 10:50. doi: 10.3389/fmicb.2019.00050

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Tong Y, Ding Z-H, Zhan F-X, Cai L, Yin X, Ling J-L, et al. The NLRP3 inflammasome and stroke. Int J Clin Exp Med. (2015) 8:4787–94.

PubMed Abstract | Google Scholar

58. Yang Y, Wang H, Kouadir M, Song H, Shi F. Recent advances in the mechanisms of NLRP3 inflammasome activation and its inhibitors. Cell Death Dis. (2019) 10:128. doi: 10.1038/s41419-019-1413-8

PubMed Abstract | CrossRef Full Text

59. Shah A. Novel coronavirus-induced NLRP3 inflammasome activation: a potential drug target in the treatment of COVID-19. Front Immunol. (2020) 11:1021. doi: 10.3389/fimmu.2020.01021

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Freeman TL, Swartz TH. Targeting the NLRP3 inflammasome in severe COVID-19. Front Immunol. (2020) 11:1518. doi: 10.3389/fimmu.2020.01518

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Toldo S, Abbate A. The NLRP3 inflammasome in acute myocardial infarction. Nat Rev Cardiol. (2018) 15:203–14. doi: 10.1038/nrcardio.2017.161

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Deftereos SG, Giannopoulos G, Vrachatis DA, Siasos GD, Giotaki SG, Gargalianos P, et al. Effect of colchicine vs standard care on cardiac and inflammatory biomarkers and clinical outcomes in patients hospitalized with coronavirus disease 2019: the GRECCO-19 randomized clinical trial. JAMA Netw Open. (2020) 3:e2013136. doi: 10.1001/jamanetworkopen.2020.13136

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Albensi BC. What is nuclear factor kappa B (NF-κB) doing in and to the mitochondrion? Front Cell Dev Biol. (2019) 7:154. doi: 10.3389/fcell.2019.00154

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Rodrigues-Diez RR, Tejera-Muñoz A, Marquez-Exposito L, Rayego-Mateos S, Santos Sanchez L, Marchant V, et al. Statins: could an old friend help in the fight against COVID-19? Br J Pharmacol. (2020) 177:4873–86. doi: 10.1111/bph.15166

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Fann DY, Lee SY, Manzanero S, Tang SC, Gelderblom M, Chunduri P, et al. Intravenous immunoglobulin suppresses NLRP1 and NLRP3 inflammasome-mediated neuronal death in ischemic stroke. Cell Death Dis. (2013) 4:e790. doi: 10.1038/cddis.2013.326

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Lara PC, Macías-Verde D, Burgos-Burgos J. Age-induced NLRP3 inflammasome over-activation increases lethality of SARS-CoV-2 pneumonia in elderly patients. Aging Dis. (2020) 11:756–62. doi: 10.14336/AD.2020.0601

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Bertocchi I, Foglietta F, Collotta D, Eva C, Brancaleone V, Thiemermann C, et al. The hidden role of NLRP3 inflammasome in obesity-related COVID-19 exacerbations: lessons for drug repurposing. Br J Pharmacol. (2020) 177:4921–30.

PubMed Abstract | Google Scholar

68. Pereira MdP, Lima EG, Serrano Junior CV. Viral infections and atherothrombosis: another caution in the wake of COVID-19? Revista da Associação Médica Brasileira. (2020) 66:366–9. doi: 10.1590/1806-9282.66.3.366

PubMed Abstract | CrossRef Full Text | Google Scholar

69. Hansson GK, Libby P, Tabas I. Inflammation and plaque vulnerability. J Intern Med. (2015) 278:483–93. doi: 10.1111/joim.12406

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Mohamud AY, Griffith B, Rehman M, Miller D, Chebl A, Patel SC, et al. Intraluminal carotid artery thrombus in COVID-19: another danger of cytokine storm? Am J Neuroradiol. (2020) 41:1677–82. doi: 10.3174/ajnr.A6674

PubMed Abstract | CrossRef Full Text | Google Scholar

71. Co COC, Yu JRT, Laxamana LC, David-Ona DIA. Intravenous thrombolysis for stroke in a COVID-19 positive filipino patient, a case report. J Clin Neurosci. (2020) 77:234–6. doi: 10.1016/j.jocn.2020.05.006

PubMed Abstract | CrossRef Full Text | Google Scholar

72. Beyrouti R, Adams ME, Benjamin L, Cohen H, Farmer SF, Goh YY, et al. Characteristics of ischaemic stroke associated with COVID-19. J Neurol Neurosurg Psychiatry. (2020) 91:889–91. doi: 10.1136/jnnp-2020-323586

PubMed Abstract | CrossRef Full Text | Google Scholar

73. Ueland T, Holter JC, Holten AR, Müller KE, Lind A, Bekken GK, et al. Distinct and early increase in circulating MMP-9 in COVID-19 patients with respiratory failure. J Infect. (2020) 81:e41–3. doi: 10.1016/j.jinf.2020.06.061

PubMed Abstract | CrossRef Full Text | Google Scholar

74. Solun B, Shoenfeld Y. Inhibition of metalloproteinases in therapy for severe lung injury due to COVID-19. Med Drug Discov. (2020) 7:100052. doi: 10.1016/j.medidd.2020.100052

PubMed Abstract | CrossRef Full Text | Google Scholar

75. Sawamura T, Fujita Y, Horiuchi S, Kakino A. LOX-1 in ischemic stroke. J Atheroscler Thromb. (2017) 24:566–8. doi: 10.5551/jat.ED071

PubMed Abstract | CrossRef Full Text | Google Scholar

76. Erol A. Role of oxidized LDL-induced “trained macrophages” in the pathogenesis of COVID-19 and benefits of pioglitazone: a hypothesis. Diabetes Metab Syndr. (2020) 14:713–4. doi: 10.1016/j.dsx.2020.05.007

PubMed Abstract | CrossRef Full Text | Google Scholar

77. Zhao Q, Meng M, Kumar R, Wu Y, Huang J, Deng Y, et al. Lymphopenia is associated with severe coronavirus disease 2019 (COVID-19) infections: a systemic review and meta-analysis. Int J Infect Dis. (2020) 96:131–5. doi: 10.1016/j.ijid.2020.04.086

PubMed Abstract | CrossRef Full Text | Google Scholar

78. Xia X, Wen M, Zhan S, He J, Chen W. An increased neutrophil/lymphocyte ratio is an early warning signal of severe COVID-19. Nan Fang Yi Ke Da Xue Xue Bao. (2020) 40:333–6. doi: 10.12122/j.issn.1673-4254.2020.03.06

PubMed Abstract | CrossRef Full Text | Google Scholar

79. Liu Y-P, Li G-M, He J, Liu Y, Li M, Zhang R, et al. Combined use of the neutrophil-to-lymphocyte ratio and CRP to predict 7-day disease severity in 84 hospitalized patients with COVID-19 pneumonia: a retrospective cohort study. Ann Transl Med. (2020) 8:635. doi: 10.21037/atm-20-2372

PubMed Abstract | CrossRef Full Text | Google Scholar

80. Wang C, Deng R, Gou L, Fu Z, Zhang X, Shao F, et al. Preliminary study to identify severe from moderate cases of COVID-19 using combined hematology parameters. Ann Transl Med. (2020) 8:593. doi: 10.21037/atm-20-3391

PubMed Abstract | CrossRef Full Text | Google Scholar

81. Shang W, Dong J, Ren Y, Tian M, Li W, Hu J, et al. The value of clinical parameters in predicting the severity of COVID-19. J Med Virol. (2020) 92:2188–92. doi: 10.1002/jmv.26031

PubMed Abstract | CrossRef Full Text | Google Scholar

82. Zhang ZL, Hou YL, Li DT, Li FZ. Laboratory findings of COVID-19: a systematic review and meta-analysis. Scand J Clin Lab Invest. (2020) 80:441–7. doi: 10.1080/00365513.2020.1768587

CrossRef Full Text | Google Scholar

83. Herold T, Jurinovic V, Arnreich C, Lipworth BJ, Hellmuth JC, von Bergwelt-Baildon M, et al. Elevated levels of IL-6 and CRP predict the need for mechanical ventilation in COVID-19. J Allergy Clin Immunol. (2020) 146:128–36.e4. doi: 10.1016/j.jaci.2020.05.008

PubMed Abstract | CrossRef Full Text | Google Scholar

84. Toniati P, Piva S, Cattalini M, Garrafa E, Regola F, Castelli F, et al. Tocilizumab for the treatment of severe COVID-19 pneumonia with hyperinflammatory syndrome and acute respiratory failure: a single center study of 100 patients in Brescia, Italy. Autoimmun Rev. (2020) 19:102568. doi: 10.1016/j.autrev.2020.102568

PubMed Abstract | CrossRef Full Text | Google Scholar

85. Berekashvili K, Dmytriw AA, Vulkanov V, Agarwal S, Khaneja A, Turkel-Parella D, et al. Etiologic subtypes of ischemic stroke in SARS-COV-2 virus patients. medRxiv. (2020) doi: 10.1101/2020.05.03.20077206. [Epub ahead of print].

PubMed Abstract | CrossRef Full Text | Google Scholar

86. Zhang J, Ren Q, Song Y, He M, Zeng Y, Liu Z, et al. Prognostic role of neutrophil-lymphocyte ratio in patients with acute ischemic stroke. Medicine. (2017) 96:e8624. doi: 10.1097/MD.0000000000008624

PubMed Abstract | CrossRef Full Text | Google Scholar

87. Nguyen VA, Crewther SG, Howells DW, Wijeratne T, Ma H, Hankey GJ, et al. Acute routine leukocyte and neutrophil counts are predictive of poststroke recovery at 3 and 12 months poststroke: an exploratory study. Neurorehabil Neural Repair. (2020) 34:844–55. doi: 10.1177/1545968320948607

PubMed Abstract | CrossRef Full Text | Google Scholar

88. Avula A, Nalleballe K, Narula N, Sapozhnikov S, Dandu V, Toom S, et al. COVID-19 presenting as stroke. Brain Behav Immun. (2020) 87:115–9. doi: 10.1016/j.bbi.2020.04.077

PubMed Abstract | CrossRef Full Text | Google Scholar

89. TunÇ A, ÜnlÜbaS Y, Alemdar M, AkyÜz E. Coexistence of COVID-19 and acute ischemic stroke report of four cases. J Clin Neurosci. (2020) 77:227–9. doi: 10.1016/j.jocn.2020.05.018

PubMed Abstract | CrossRef Full Text | Google Scholar

90. Tissa Wijeratne CS, Karimi L, Crewther SG. Acute ischemic stroke in COVID-19: a case-based systematic review. Front Neurol. (2020) 11:1031. doi: 10.3389/fneur.2020.01031

PubMed Abstract | CrossRef Full Text | Google Scholar

91. Wijeratne T, Sales CA, Crewther SG, Nguyen V, Karimi L. First Australian case of good recovery of a COVID-19 patient with severe neurological symptoms post prolonged hospitalization. Cureus. (2020) 12:10366–77. doi: 10.7759/cureus.10366

PubMed Abstract | CrossRef Full Text | Google Scholar

92. Kolaczkowska E, Kubes P. Neutrophil recruitment and function in health and inflammation. Nat Rev Immunol. (2013) 13:159–75. doi: 10.1038/nri3399

PubMed Abstract | CrossRef Full Text | Google Scholar

93. Agarwal S, Scher E, Rossan-Raghunath N, Marolia D, Butnar M, Torres J, et al. Acute stroke care in a New York City comprehensive stroke center during the COVID-19 pandemic. J Stroke Cerebrovasc Dis. (2020) 29:105068. doi: 10.1016/j.jstrokecerebrovasdis.2020.105068

PubMed Abstract | CrossRef Full Text | Google Scholar

94. Yaghi S, Ishida K, Torres J, Grory BM, Raz E, Humbert K, et al. SARS-CoV-2 and stroke in a New York healthcare system. Stroke. (2020) 51:2002–11. doi: 10.1161/STROKEAHA.120.030335

CrossRef Full Text | Google Scholar

95. Wijeratne T, Menon R, Sales C, Karimi L, Crewther S. Carotid artery stenosis and inflammatory biomarkers: the role of inflammation-induced immunological responses affecting the vascular systems. Ann Transl Med. (2020) 8:1276. doi: 10.21037/atm-20-4388

PubMed Abstract | CrossRef Full Text | Google Scholar

96. Morassi M, Bagatto D, Cobelli M, D'Agostini S, Gigli GL, Bnà C, et al. Stroke in patients with SARS-CoV-2 infection: case series. J Neurol. (2020) 267:2185–92. doi: 10.1007/s00415-020-09885-2

PubMed Abstract | CrossRef Full Text | Google Scholar

97. Valderrama EV, Humbert K, Lord A, Frontera J, Yaghi S. Severe acute respiratory syndrome coronavirus 2 infection and ischemic stroke. Stroke. (2020) 51:e124–7. doi: 10.1161/STROKEAHA.120.030153

PubMed Abstract | CrossRef Full Text | Google Scholar

98. Kunutsor SK, Seidu S, Blom AW, Khunti K, Laukkanen JA. Serum C-reactive protein increases the risk of venous thromboembolism: a prospective study and meta-analysis of published prospective evidence. Eur J Epidemiol. (2017) 32:657–67. doi: 10.1007/s10654-017-0277-4

PubMed Abstract | CrossRef Full Text | Google Scholar

99. Deliwala S, Abdulhamid S, Abusalih MF, Al-Qasmi MM, Bachuwa G. Encephalopathy as the sentinel sign of a cortical stroke in a patient infected with coronavirus disease-19 (COVID-19). Cureus. (2020) 12:e8121. doi: 10.7759/cureus.8121

PubMed Abstract | CrossRef Full Text | Google Scholar

100. Gunasekaran K, Amoah K, Rajasurya V, Buscher MG. Stroke in a young COVID-19 patient. QJM Int J Med. (2020) 113:573–4. doi: 10.1093/qjmed/hcaa177

PubMed Abstract | CrossRef Full Text | Google Scholar

101. Zuo Y, Yalavarthi S, Shi H, Gockman K, Zuo M, Madison JA, et al. Neutrophil extracellular traps (NETs) as markers of disease severity in COVID-19. medRxiv. (2020) doi: 10.1101/2020.04.09.20059626. [Epub ahead of print].

CrossRef Full Text | Google Scholar

102. Tomar B, Anders H-J, Desai J, Mulay SR. Neutrophils and neutrophil extracellular traps drive necroinflammation in COVID-19. Cells. (2020) 9:1383. doi: 10.3390/cells9061383

PubMed Abstract | CrossRef Full Text | Google Scholar

103. Middleton EA, He XY, Denorme F, Campbell RA, Ng D, Salvatore SP, et al. Neutrophil extracellular traps contribute to immunothrombosis in COVID-19 acute respiratory distress syndrome. Blood. (2020) 136:1169–79. doi: 10.1182/blood.2020007008

PubMed Abstract | CrossRef Full Text | Google Scholar

104. Urban CF, Ermert D, Schmid M, Abu-Abed U, Goosmann C, Nacken W, et al. Neutrophil extracellular traps contain calprotectin, a cytosolic protein complex involved in host defense against Candida albicans. PLoS Pathog. (2009) 5:e1000639. doi: 10.1371/journal.ppat.1000639

PubMed Abstract | CrossRef Full Text | Google Scholar

105. Zhou P, Li T, Jin J, Liu Y, Li B, Sun Q, et al. Interactions between neutrophil extracellular traps and activated platelets enhance procoagulant activity in acute stroke patients with ICA occlusion. EBioMedicine. (2020) 53:102671. doi: 10.1016/j.ebiom.2020.102671

PubMed Abstract | CrossRef Full Text | Google Scholar

106. Kang L, Yu H, Yang X, Zhu Y, Bai X, Wang R, et al. Neutrophil extracellular traps released by neutrophils impair revascularization and vascular remodeling after stroke. Nat Commun. (2020) 11:2488. doi: 10.1038/s41467-020-16191-y

PubMed Abstract | CrossRef Full Text | Google Scholar

107. Cavalcanti DD, Raz E, Shapiro M, Dehkharghani S, Yaghi S, Lillemoe K, et al. Cerebral venous thrombosis associated with COVID-19. Am J Neuroradiol. (2020) 41:1370–6. doi: 10.3174/ajnr.A6644

PubMed Abstract | CrossRef Full Text | Google Scholar

108. Garaci F, Di Giuliano F, Picchi E, Da Ros V, Floris R. Venous cerebral thrombosis in COVID-19 patient. J Neurol Sci. (2020) 414:116871. doi: 10.1016/j.jns.2020.116871

PubMed Abstract | CrossRef Full Text | Google Scholar

109. Iacoviello L, Castelnuovo AD, Curtis Ad, Agnoli C, Frasca G, Mattiello A, et al. Circulating tissue factor levels and risk of stroke. Stroke. (2015) 46:1501–7. doi: 10.1161/STROKEAHA.115.008678

PubMed Abstract | CrossRef Full Text | Google Scholar

110. Previtali E, Bucciarelli P, Passamonti SM, Martinelli I. Risk factors for venous and arterial thrombosis. Blood Transfus. (2011) 9:120–38. doi: 10.2450/2010.0066-10

PubMed Abstract | CrossRef Full Text | Google Scholar

111. Tan Y-K, Goh C, Leow AST, Tambyah PA, Ang A, Yap E-S, et al. COVID-19 and ischemic stroke: a systematic review and meta-summary of the literature. J Thromb Thrombolysis. (2020) 50:587–95. doi: 10.1007/s11239-020-02228-y

PubMed Abstract | CrossRef Full Text | Google Scholar

112. Wijesundera C, Vingrys AJ, Wijeratne T, Crewther SG. Acquired visual deficits independent of lesion site in acute stroke. Front Neurol. (2020) 11:705. doi: 10.3389/fneur.2020.00705

PubMed Abstract | CrossRef Full Text | Google Scholar

113. Tissa Wijeratne SGC, Wijeratne T. What is Post Covid 19 Neurological Syndrome? University of Melbourne (2020). Available online at: https://pursuit.unimelb.edu.au/articles/what-is-post-covid-19-neurological-syndrome?fbclid=IwAR1H5RVh7Fmr9orgJYst2c-NSdhjFz1Ybxmu-hEhTP4M5AUSo_hbXVjo-1w (accessed January 8, 2020).

114. Peña-Martínez C, Durán-Laforet V, García-Culebras A, Ostos F, Hernández-Jiménez M, Bravo-Ferrer I, et al. Pharmacological modulation of neutrophil extracellular traps reverses thrombotic stroke tPA (tissue-type plasminogen activator) resistance. Stroke. (2019) 50:3228–37. doi: 10.1161/STROKEAHA.119.026848

PubMed Abstract | CrossRef Full Text | Google Scholar

115. Thachil J, Tang N, Gando S, Falanga A, Cattaneo M, Levi M, et al. ISTH interim guidance on recognition and management of coagulopathy in COVID-19. J Thromb Haemost. (2020) 18:1023–6. doi: 10.1111/jth.14810

PubMed Abstract | CrossRef Full Text | Google Scholar

116. Thachil J. All those D-dimers in COVID-19. J Thromb Haemost. (2020) 18:2075–6. doi: 10.1111/jth.14939

CrossRef Full Text | Google Scholar

117. Medcalf RL, Keragala CB, Myles PS. Fibrinolysis and COVID-19: a plasmin paradox. J Thromb Haemost. (2020) 18:2118–22. doi: 10.1111/jth.14960

PubMed Abstract | CrossRef Full Text | Google Scholar

118. Creel-Bulos C, Auld SC, Caridi-Scheible M, Barker N, Friend S, Gaddh M, et al. Fibrinolysis shutdown and thrombosis in a COVID-19 ICU. Shock. (2020). doi: 10.1097/SHK.0000000000001635

PubMed Abstract | CrossRef Full Text | Google Scholar

119. Wright FL, Vogler TO, Moore EE, Moore HB, Wohlauer MV, Urban S, et al. Fibrinolysis shutdown correlation with thromboembolic events in severe COVID-19 infection. J Am Coll Surg. (2020) 231:193–203.e1. doi: 10.1016/j.jamcollsurg.2020.05.007

PubMed Abstract | CrossRef Full Text | Google Scholar

120. Shaydakov ME, Sigmon DF, Blebea J. Thromboelastography (TEG). In: StatPearls. Treasure Island, FL: StatPearls Publishing (2020). Available online at: https://www.ncbi.nlm.nih.gov/books/NBK537061/ (accessed April 24, 2020).

PubMed Abstract | Google Scholar

121. Requena M, Olivé-Gadea M, Muchada M, García-Tornel Á, Deck M, Juega J, et al. COVID-19 and stroke: incidence and etiological description in a high-volume center. J Stroke Cerebrovasc Dis. (2020) 29:105225. doi: 10.1016/j.jstrokecerebrovasdis.2020.105225

PubMed Abstract | CrossRef Full Text | Google Scholar

122. Yao Y, Cao J, Wang Q, Shi Q, Liu K, Luo Z, et al. D-dimer as a biomarker for disease severity and mortality in COVID-19 patients: a case control study. J Intensive Care. (2020) 8:49. doi: 10.1186/s40560-020-00466-z

PubMed Abstract | CrossRef Full Text | Google Scholar

123. Lodigiani C, Iapichino G, Carenzo L, Cecconi M, Ferrazzi P, Sebastian T, et al. Venous and arterial thromboembolic complications in COVID-19 patients admitted to an academic hospital in Milan, Italy. Thromb Res. (2020) 191:9–14. doi: 10.1016/j.thromres.2020.04.024

PubMed Abstract | CrossRef Full Text | Google Scholar

124. Klein DE, Libman R, Kirsch C, Arora R. Cerebral venous thrombosis: a typical presentation of COVID-19 in the young. J Stroke Cerebrovasc Dis. (2020) 29:104989. doi: 10.1016/j.jstrokecerebrovasdis.2020.104989

PubMed Abstract | CrossRef Full Text | Google Scholar

125. Beun R, Kusadasi N, Sikma M, Westerink J, Huisman A. Thromboembolic events and apparent heparin resistance in patients infected with SARS-CoV-2. Int J Lab Hematol. (2020) 42(Suppl. 1):19–20. doi: 10.1111/ijlh.13230

PubMed Abstract | CrossRef Full Text | Google Scholar

126. Sangalli D, Polonia V, Colombo D, Mantero V, Filizzolo M, Scaccabarozzi C, et al. A single-centre experience of intravenous thrombolysis for stroke in COVID-19 patients. Neurol Sci. (2020) 41:2325–9. doi: 10.1007/s10072-020-04591-3

PubMed Abstract | CrossRef Full Text | Google Scholar

127. Ahmed S, Zimba O, Gasparyan AY. Thrombosis in coronavirus disease 2019 (COVID-19) through the prism of Virchow's triad. Clin Rheumatol. (2020) 39:2529–43. doi: 10.1007/s10067-020-05275-1

PubMed Abstract | CrossRef Full Text | Google Scholar

128. Setia G, Tyler J, Kwan A, Faguet J, Sharma S, Singh S, et al. High thrombus burden despite thrombolytic therapy in ST-elevation myocardial infarction in a patient with COVID-19. Rev Cardiovasc Med. (2020) 21:289–95. doi: 10.31083/j.rcm.2020.02.92

PubMed Abstract | CrossRef Full Text | Google Scholar

129. Zhang Y, Cao W, Jiang W, Xiao M, Li Y, Tang N, et al. Profile of natural anticoagulant, coagulant factor and anti-phospholipid antibody in critically ill COVID-19 patients. J Thromb Thrombolysis. (2020) 50:1–7. doi: 10.1007/s11239-020-02182-9

PubMed Abstract | CrossRef Full Text | Google Scholar

130. Zhang Y, Xiao M, Zhang S, Xia P, Cao W, Jiang W, et al. Coagulopathy and antiphospholipid antibodies in patients with covid-19. N Engl J Med. (2020) 382:e38. doi: 10.1056/NEJMc2007575

PubMed Abstract | CrossRef Full Text | Google Scholar

131. Aubignat M, Godefroy O. COVID-19 and ischemic stroke: should we systematically look for lupus anticoagulant and antiphospholipid antibodies? Rev Neurol (Paris). (2020) 176:505–6. doi: 10.1016/j.neurol.2020.05.001

PubMed Abstract | CrossRef Full Text | Google Scholar

132. Libby P, Lüscher T. COVID-19 is, in the end, an endothelial disease. Eur Heart J. (2020) 41:3038–44. doi: 10.1093/eurheartj/ehaa623

PubMed Abstract | CrossRef Full Text | Google Scholar

133. Akerström S, Gunalan V, Keng CT, Tan Y-J, Mirazimi A. Dual effect of nitric oxide on SARS-CoV replication: viral RNA production and palmitoylation of the S protein are affected. Virology. (2009) 395:1–9. doi: 10.1016/j.virol.2009.09.007

PubMed Abstract | CrossRef Full Text | Google Scholar

134. DiNicolantonio JJ, McCarty M. Thrombotic complications of COVID-19 may reflect an upregulation of endothelial tissue factor expression that is contingent on activation of endosomal NADPH oxidase. Open Heart. (2020) 7:e001337. doi: 10.1136/openhrt-2020-001337

PubMed Abstract | CrossRef Full Text | Google Scholar

135. Bautista-Vargas M, Bonilla-Abadía F, Cañas CA. Potential role for tissue factor in the pathogenesis of hypercoagulability associated with in COVID-19. J Thromb Thrombolysis. (2020) 50:1–5. doi: 10.1007/s11239-020-02172-x

PubMed Abstract | CrossRef Full Text | Google Scholar

136. Martel J, Ko Y-F, Young JD, Ojcius DM. Could nasal nitric oxide help to mitigate the severity of COVID-19? Microbes Infect. (2020) 22:168–71. doi: 10.1016/j.micinf.2020.05.002

PubMed Abstract | CrossRef Full Text | Google Scholar

137. Cheng H, Wang Y, Wang GQ. Organ-protective effect of angiotensin-converting enzyme 2 and its effect on the prognosis of COVID-19. J Med Virol. (2020) 92:726–30. doi: 10.1002/jmv.25785

PubMed Abstract | CrossRef Full Text | Google Scholar

138. Gan R, Rosoman NP, Henshaw DJE, Noble EP, Georgius P, Sommerfeld N. COVID-19 as a viral functional ACE2 deficiency disorder with ACE2 related multi-organ disease. Med Hypotheses. (2020) 144:110024. doi: 10.1016/j.mehy.2020.110024

PubMed Abstract | CrossRef Full Text | Google Scholar

139. Hess DC, Eldahshan W, Rutkowski E. COVID-19-related stroke. Transl Stroke Res. (2020) 11:322–5. doi: 10.1007/s12975-020-00818-9

PubMed Abstract | CrossRef Full Text

140. Huang S, Wang J, Liu F, Liu J, Cao G, Yang C, et al. COVID-19 patients with hypertension have more severe disease: a multicenter retrospective observational study. Hypertens Res. (2020) 43:824–31. doi: 10.1038/s41440-020-0485-2

PubMed Abstract | CrossRef Full Text | Google Scholar

141. Gironacci MM, Vicario A, Cerezo G, Silva MG. The depressor axis of the renin-angiotensin system and brain disorders: a translational approach. Clin Sci (Lond). (2018) 132:1021–38. doi: 10.1042/CS20180189

PubMed Abstract | CrossRef Full Text | Google Scholar

142. Tsuda K. Renin-Angiotensin system and sympathetic neurotransmitter release in the central nervous system of hypertension. Int J Hypertens. (2012) 2012:474870. doi: 10.1155/2012/474870

PubMed Abstract | CrossRef Full Text | Google Scholar

143. Fridman S, Bullrich MB, Jimenez-Ruiz A, Costantini P, Shah P, Just C, et al. Stroke risk, phenotypes, and death in COVID-19: systematic review and newly reported cases. Neurology. (2020) 95:e3373–85. doi: 10.1212/WNL.0000000000010851

PubMed Abstract | CrossRef Full Text | Google Scholar

144. Boldrini P, Bernetti A, Fiore P. Impact of COVID-19 outbreak on rehabilitation services and physical and rehabilitation medicine physicians' activities in Italy. An official document of the Italian PRM Society (SIMFER). Eur J Phys Rehabil Med. (2020) 56:316–8. doi: 10.23736/S1973-9087.20.06256-5

PubMed Abstract | CrossRef Full Text | Google Scholar

145. Giansanti D. WhatsApp in mHealth: an overview on the potentialities and the opportunities in medical imaging. Mhealth. (2020) 6:19. doi: 10.21037/mhealth.2019.11.01

PubMed Abstract | CrossRef Full Text | Google Scholar

146. Davis TL, DiClemente R, Prietula M. Taking mHealth forward: examining the core characteristics. JMIR Mhealth Uhealth. (2016)4:e97. doi: 10.2196/mhealth.5659

PubMed Abstract | CrossRef Full Text | Google Scholar

147. Cicerone KD, Goldin Y, Ganci K, Rosenbaum A, Wethe JV, Langenbahn DM, et al. Evidence-based cognitive rehabilitation: systematic review of the literature from 2009 through 2014. Arch Phys Med Rehabil. (2019) 100:1515–33. doi: 10.1016/j.apmr.2019.02.011

PubMed Abstract | CrossRef Full Text | Google Scholar

148. Slavich GM. Psychoneuroimmunology of stress and mental health. In: Harkness KL, Hayden EP, editors. The Oxford Handbook of Stress and Mental Health. Oxford: Oxford University Press (2020).

Google Scholar

149. Vaes AW, Machado FVC, Meys R, Delbressine JM, Goertz YMJ, Van Herck M, et al. Care dependency in non-hospitalized patients with COVID-19. J Clin Med. (2020) 9. doi: 10.3390/jcm9092946

PubMed Abstract | CrossRef Full Text | Google Scholar

150. Wijeratne T, Crewther S. Post-COVID 19 neurological syndrome (PCNS); a novel syndrome with challenges for the global neurology community. J Neurol Sci. (2020) 419:117179. doi: 10.1016/j.jns.2020.117179

PubMed Abstract | CrossRef Full Text | Google Scholar

151. Mathew D, Giles JR, Baxter AE, Oldridge DA, Greenplate AR, Wu JE, et al. Deep immune profiling of COVID-19 patients reveals distinct immunotypes with therapeutic implications. Science. (2020) 369:eabc8511. doi: 10.1126/science.abc8511

PubMed Abstract | CrossRef Full Text | Google Scholar

152. Mantovani E, Zucchella C, Bottiroli S, Federico A, Giugno R, Sandrini G, et al. Telemedicine and virtual reality for cognitive rehabilitation: a roadmap for the COVID-19 pandemic. Front Neurol. (2020) 11:926. doi: 10.3389/fneur.2020.00926

PubMed Abstract | CrossRef Full Text | Google Scholar

153. Cicerone KD, Dahlberg C, Kalmar K, Langenbahn DM, Malec JF, Bergquist TF, et al. Evidence-based cognitive rehabilitation: recommendations for clinical practice. Arch Phys Med Rehabil. (2000) 81:1596–615. doi: 10.1053/apmr.2000.19240

PubMed Abstract | CrossRef Full Text | Google Scholar

154. Sivan M, Halpin S, Hollingworth L, Snook N, Hickman K, Clifton IJ. Development of an integrated rehabilitation pathway for individuals recovering from COVID-19 in the community. J Rehabil Med. (2020) 52:jrm00089. doi: 10.2340/16501977-2727

PubMed Abstract | CrossRef Full Text | Google Scholar

155. Wade DT. Rehabilitation after COVID-19: an evidence-based approach. Clin Med (Lond). (2020) 20:359–65. doi: 10.7861/clinmed.2020-0353

PubMed Abstract | CrossRef Full Text | Google Scholar

156. Garcia IG, Rodriguez-Rubio M, Mariblanca AR, de Soto LM, Garcia LD, Villatoro JM, et al. A randomized multicenter clinical trial to evaluate the efficacy of melatonin in the prophylaxis of SARS-CoV-2 infection in high-risk contacts (MeCOVID Trial): a structured summary of a study protocol for a randomised controlled trial. Trials. (2020) 21:466. doi: 10.1186/s13063-020-04436-6

PubMed Abstract | CrossRef Full Text | Google Scholar

157. Galano A, Tan DX, Reiter RJ. Melatonin: a versatile protector against oxidative DNA damage. Molecules. (2018) 23. doi: 10.3390/molecules23030530

PubMed Abstract | CrossRef Full Text | Google Scholar

158. Jean-Louis G, von Gizycki H, Zizi F. Melatonin effects on sleep, mood, and cognition in elderly with mild cognitive impairment. J Pineal Res. (1998) 25:177–83. doi: 10.1111/j.1600-079X.1998.tb00557.x

PubMed Abstract | CrossRef Full Text | Google Scholar

159. Kleszczynski K, Slominski AT, Steinbrink K, Reiter RJ. Clinical trials for use of melatonin to fight against COVID-19 are urgently needed. Nutrients. (2020) 12:2561. doi: 10.3390/nu12092561

PubMed Abstract | CrossRef Full Text | Google Scholar

160. Raza H, John A, Brown EM, Benedict S, Kambal A. Alterations in mitochondrial respiratory functions, redox metabolism and apoptosis by oxidant 4-hydroxynonenal and antioxidants curcumin and melatonin in PC12 cells. Toxicol Appl Pharmacol. (2008) 226:161–8. doi: 10.1016/j.taap.2007.09.002

PubMed Abstract | CrossRef Full Text | Google Scholar

161. Reiter RJ, Tan DX, Rosales-Corral S, Galano A, Jou MJ, Acuna-Castroviejo D. Melatonin mitigates mitochondrial meltdown: interactions with SIRT3. Int J Mol Sci. (2018) 19:2439. doi: 10.3390/ijms19082439

PubMed Abstract | CrossRef Full Text | Google Scholar

162. Sadanandan N, Cozene B, Cho J, Park YJ, Saft M, Gonzales-Portillo B, et al. Melatonin-A potent therapeutic for stroke and stroke-related dementia. Antioxidants (Basel). (2020) 9:672. doi: 10.3390/antiox9080672

PubMed Abstract | CrossRef Full Text | Google Scholar

163. Manoharan Y, Haridas V, Vasanthakumar KC, Muthu S, Thavoorullah FF, Shetty P. Curcumin: a wonder drug as a preventive measure for COVID19 management. Indian J Clin Biochem. (2020) 35:373–5. doi: 10.1007/s12291-020-00902-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: COVID-19, stroke, ACE2, cytokines, pathogen associated molecular pattern, post Covid19 neurology syndrome, PCNS

Citation: Wijeratne T, Gillard Crewther S, Sales C and Karimi L (2021) COVID-19 Pathophysiology Predicts That Ischemic Stroke Occurrence Is an Expectation, Not an Exception—A Systematic Review. Front. Neurol. 11:607221. doi: 10.3389/fneur.2020.607221

Received: 16 September 2020; Accepted: 30 November 2020;
Published: 28 January 2021.

Edited by:

Thomas Platz, University of Greifswald, Germany

Reviewed by:

Monica Verduzco-Gutierrez, The University of Texas Health Science Center at San Antonio, United States
Luca Sebastianelli, Hospital of Vipiteno, Italy

Copyright © 2021 Wijeratne, Gillard Crewther, Sales and Karimi. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Tissa Wijeratne, tissa.wijeratne@wh.org.au

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.