Open access peer-reviewed chapter

Chronic Mild Stress and COVID-19 Sequelae

Written By

Dragana Komnenov

Submitted: 29 June 2022 Reviewed: 14 July 2022 Published: 01 September 2022

DOI: 10.5772/intechopen.106578

From the Edited Volume

COVID-19 Pandemic, Mental Health and Neuroscience - New Scenarios for Understanding and Treatment

Edited by Sara Palermo and Berend Olivier

Chapter metrics overview

136 Chapter Downloads

View Full Metrics

Abstract

Although COVID-19 clinical presentation primarily involves the respiratory system causing bilateral pneumonia, it is becoming increasingly recognized that COVID-19 is in fact a systemic disease. Neurological presentations have been reported in patients with both mild and severe COVID-19 symptoms. As such, elderly individuals are at a significantly higher risk of developing severe COVID-19 as well as neurocognitive consequences due to the presence of comorbidities associated with aging and the direct consequences of infection. Several neurological disorders that have been described in the literature include insomnia, depression, anxiety, post-traumatic stress disorder and cognitive insufficiencies. The potential underlying mechanisms are still incompletely understood but are likely multifaceted, involving both direct neurotrophic effect of SARS-CoV-2 and the indirect consequences related to social isolation in long intensive care units, the use of mechanical ventilation and sedation and the resultant brain hypoxia, systemic inflammation and secondary effects of medications used in treatment of COVID-19. Furthermore, neuro-cardiovascular adaptations resulting from the chronic stress and depression milieu of COVID-19 is expected to contribute negatively to the cardiovascular health of the survivors. It is thus imperative to implement a rigorous monitoring program for COVID-19 survivors, particularly among the elderly population, to assess potential neuro-cognitive and cardiovascular deteriorations.

Keywords

  • COVID-19
  • neuroinflammation
  • chronic stress
  • depression
  • cardiovascular disease

1. Introduction

Since the unfolding of COVID-19 pandemic starting in early 2020 it has become increasingly apparent that the disease has evolved from primarily affecting the respiratory system to being a systemic disease. A common manifestation of the latter involves the neurologic system, ranging from headache and myalgia to neuroinflammation and encephalopathies. Additionally, neuropsychiatric manifestations such as anxiety, stress, depression and post-traumatic stress disorder (PTSD) have been reported [1, 2, 3, 4]. Another common manifestation affects the cardiovascular system, with pathologies ranging from pericarditis, myocarditis, right-hearted dysfunction, enodothelialitis and prothrombotic state (as reviewed in [5]). Any potential long-term effects of these disorders are yet to manifest in the coming months and years. Based on the available data, the interactions among chronic mild stress, neurological consequences and cardiovascular manifestations due to COVID-19 pandemic are likely to contribute to a significant public health problem worldwide. These interactions are explored below throughout the chapter.

Stress, depression and anxiety are being recognized as risk factors for the development of cardiovascular disease (CVD). COVID-19 pandemic has induced many stressors on everyday life, including fear of infection, lack of social interactions due to quarantine, helplessness due to inevitability, loss of income, misinformation spread mostly by social media, and food and household item shortage. Furthermore, the viral infection itself can cause detriment to the cardiovascular system via cerebrovascular ischemia, coagulopathy and endothelial dysfunction. Therefore, both individuals who become infected and those who do not, but are exposed to the chronic mild stress (CMS) of COVID-19 pandemic may be at risk of developing neurologic and cardiovascular consequences (please see a model in Figure 1). In any event, the stressors of the pandemic can be modeled by the CMS rodent model of depression. The CMS paradigm is typically conducted for 4 weeks and consists of the exposure of rodents to mild stressors such as exposure to strobe light and white noise, acute withdrawal of water, damp bedding and social isolation [6, 7, 8, 9]. This procedure causes depression as evidenced by anhedonia (in rodents manifested as reduced 1–2% sucrose solution consumption and spontaneous wheel running), circadian rhythm disturbances and demeanor. This rodent model of human depression was used extensively to demonstrate the cardiovascular dysfunction following 4 weeks of exposure to the mild stressors, characterized by increased mean arterial pressure and sympathetic nervous system activity and decreased heart rate variability [6, 7, 9]. One study found that the 4 week period of stress exposure followed by 4 week period of stress reduction (i.e. no exposure to stressors) recovered the behavioral manifestations of depressions, such as sucrose consumption and spontaneous wheel running, but did not result in the reversal of the cardiovascular dysfunction measured by heart rate variability [9]. Such long term effects of stress exposure, as is seen in the COVID-19 pandemic, could therefore be detrimental in the post-pandemic era, highlighting the importance of cardiovascular health monitoring in all individuals.

Figure 1.

Direct and indirect effects of COVID-19 on the central nervous and cardiovascular systems. Individuals who became infected with SARS-CoV2 virus could experience neurologic and cardiovascular dysfunction due to the direct neurotrophic effects of the virus as well as indirectly, via experiencing chronic stress associated with the pandemic (i.e. loss of loved ones, loss of income, lack of exercise, poor nutrition etc.). Individual who evaded infection, although spared from the direct effects of the virus, are also at an increased risk of developing neuropsychiatric and cardiovascular diseases via experiencing chronic stress associated with the pandemic. Therefore, those who survived COVID-19 and those who never got the disease but lived through the pandemic should be monitored both for mental health and cardiovascular health changes in the years to come.

Advertisement

2. Cognitive and neuropsychiatric manifestations of COVID-19

Pandemics are considered to be one of the most devastating disaster types, since they have global consequences that particularly affect mental health. Although not of pandemic proportions, previous outbreaks of viral infections involving coronaviruses, SARS (SARS-CoV) and MERS (MERS-CoV), and other viruses such as Ebola and Zika, provided valuable insights into the potential devastating effects on mental health status [10, 11].

Multiple reports and meta-analyses described that most common neuropsychiatric manifestations of COVID-19 are insomnia, depression and anxiety, PTSD, and various psychoses. In terms of prevalence, almost one quarter (22.5%) of those infected were found to have experienced some neurological and/or psychiatric episodes among 40,469 patients of whom majority was in the United States [3]. Subsequent studies from Europe reported similar outcomes [12, 13, 14]. Critically ill individuals who required intensive care unit (ICU) admissions are particularly at risk of developing cognitive and neuropsychiatric manifestations, due to sedation, intubation, presence of comorbidities and older age [15, 16, 17, 18, 19]. One study from France reported that in a small cohort of 45 patients, 15 of them reported cognitive disturbances in form of dysexecutive syndrome (dysregulated movement patterns and lack of attention) at discharge from ICU [14]. Furthermore, almost half of them presented with confusion upon admission that was accompanied by brain hypoperfusion in several individuals revealed after brain imaging [14]. Depressive symptoms were also prevalent among individuals who recovered from COVID-19 in China [20, 21, 22]. Moreover, immune system suppression was evidenced in those with depressive symptoms indicated by increased white blood cell count and pro-inflammatory markers [21].

The impact of COVID-19 on one’s neuropsychological well-being can be a direct result of SARS-CoV2 viral infection of the central nervous system (CNS) and/or an indirect result of endured psychological stress due to the devastating elements of the pandemic, such as fear of infection, social isolation, loss of income etc. The characteristic, mechanisms and implications of the first are detailed below throughout Section 2 of this Chapter, and those of the latter are elaborated on in Section 3 of this Chapter.

2.1 Mechanisms of cognitive and neuropsychiatric sequelae of COVID-19

Initially in early 2020, it was speculated but not confirmed that SARS-CoV2 is indeed a neurotrophic virus [23]. Shortly thereafter, the first case of viral encephalitis was reported in May of 2020 [24], making it obvious that the virus is capable of invading the CNS. Similar to other coronaviruses, SARS-CoV1 and MERS-CoV, SARS-CoV2 infection can cause CNS issues that range from mild such as loss of taste and smell (ageusia and anosmia, respectively), headache and dizziness, to very serious such as stroke, seizures, loss of balance and mental status alterations [25]. These consequences could result from: (i) the direct infection of neuronal cells by the virus, (ii) immune system dysregulation, (iii) autoimmunity resulting from the infection itself and/or (iv) any combination of the above three [26]. In addition to the direct invasion of neuronal cells, a secondary systemic mechanism could also be at play. This mechanism involves acute respiratory distress syndrome (ARDS). ARDS is accompanied by hypoxemia, oxidative stress and uremia resulting from multi-organ failure, including the cardiovascular system derangements and such complications could lead to encephalopathy. Immune system dysregulation may involve the cytokine storm and subsequent breakdown of the blood brain barrier (BBB) allowing entry of SARS-CoV2 into the CNS. This model is further supported by the fact that the virus binds ACE2 receptor that is present on capillary endothelium, thus leading to BBB damage and entry of the virus into the CNS [27]. Furthermore, immune system involvement could occur at the level of toll like receptors (TLRs), of which there are 10 identified members in humans, numbered 1–10 [28]. In particular, TLR 7/8 is recruited in response to single stranded RNA viruses, such as SARS-CoV2, leading to the production of pro-inflammatory mediators such as interleukin (IL)-1, IL-6, tumor necrosis factor alpha and interferon gamma [29]. Uncontrolled production of the pro-inflammatory mediators may lead to cytokine storm which causes ARDS, leading to encephalopathy secondary to an inflammatory response [30]. In another model, it was suggested that SRAS-CoV2 may operate similar to HIV in that HIV causes encephalopathy via a mixed approach: both directly via neuronal cell invasion and indirectly via the above discussed inflammatory mechanisms [26].

2.2 Potential long-term neuro-psychiatric effects: long haul COVID-19

The syndrome of persistent symptoms associated with COVID-19 that extend beyond the period of initial infection was originally termed long haul (LH) COVID-19 by a patient [31], the features of which have been identified in individuals irrespective of the initial illness severity [32]. One characteristic of LH COVID-19 is that the symptoms may either persist past the 3–4 week mark and/or new symptoms may develop after the 3–4 week mark. In fact, the National Institute for Health and Care Excellence has defined the LH or post-COVID-19 syndrome as “signs and symptoms that develop during or after an infection consistent with COVID-19, continue for more than 12 weeks ( 3 months) and are not explained by an alternative diagnosis” [33]. LH COVID-19 appears in the literature under several synonyms: post-COVID-19 condition (WHO defined), post-acute COVID-19 syndrome (PACS) [34] and post-acute sequelae of COVID-19 [32].

Neurological symptoms discussed above may contribute individually or synergistically to the persistence of neurological pathophysiology past the acute phase. The loss of BBB integrity and neuroinflammation [35, 36, 37], coupled with coagulopathy and the development of micro-emboli in the CNS [38, 39] may lead to the progression of LH COVID-19. Additionally, factors associated with hospital stay (i.e. intubation, mechanical ventilation and the use of sedatives) may further exacerbate the clinical course in those with severe acute symptoms. Neuro-psychiatric features of LH COVID-19 are likely related to prolonged stress associated with the pandemic and loss of family members/friends [40].

A recently published meta-analysis aimed to evaluate the neurological and neuro-psychiatric features of LH COVID-19 in three cohorts: outpatient (community), non-ICU hospitalized an ICU hospitalized and at two different time points: 3–6 months and past the 6 month mark after the initial infection [41]. Primary outcomes included neurologic and psychiatric symptoms. Neurologic symptoms included: anosmia, dysgeusia, headache, cognitive dysfunction, fatigue, chronic fatigue syndrome, post exertional malaise, pain, peripheral nervous system symptoms. Neuropsychological symptoms included: anxiety, depression, sleep disturbances and/or insomnia and PTSD. A total number of full text articles screened were 80, and 18 studies, including 10,530 patients met the inclusion criteria. The most frequent neurological symptom of LH COVID-19 was fatigue (37%), followed by brain fog (32%), sleep disturbances (31%) and memory issues (28%). The prevalence of these symptoms tended to be higher in non-hospitalized individuals. Similarly, the neuropsychiatric symptoms of anxiety (31%) and depression (27%) were higher in the community cohort compared to the hospitalized patients (6% and 12%, respectively). Additionally, the neuropsychiatric symptoms substantially increased from mid-term follow-up to long term follow-up (i.e. past 6 months), suggesting that LH COVID-19 patients may experience increasing neuropsychiatric burden well past the initial infection. It is however not known when and whether it tapers off. On the other hand, the neurological symptoms in this cohort appear to progress from mid- to long-term highlighting that this may be the critical period of LH COVID-19 during which patients should be screen for neurological pathophysiological events and represents the critical therapeutic window. Other large retrospective cohort studies reported similar timelines [40, 42]. This may be explained by the chronic aspect of neuro-inflammation that ensues secondary to initial infection, leading to neuronal loss in that critical time period.

Taken together, the existing large cohort studies [40, 42] and meta-analyses [41] indicate that neurological and neuropsychiatric symptoms are a common feature of LH COVID-19, with specific symptoms occurring in as much as a third of the individuals who were infected with SARS-CoV2. Future research should be directed towards identifying the therapeutic strategies during the critical window, which appears to be 3–6 months post-acute illness, in an effort to decrease neuroinflammation, restore the blood brain barrier and prevent neuronal loss.

Advertisement

3. Chronic stress associated with COVID-19: multifaceted attack on the cardiovascular system

The cardiovascular system is one of the direct targets of SARS-CoV2 made possible by the viral entry into the cells via ACE2 receptors expressed on endothelial cells and cardiomyocytes. The myocardial injury associated with the acute COVID-19 illness has been well documented [43, 44, 45, 46]. The responsible mechanisms include direct cytotoxicity [47] and/or dysregulation of the renin angiotensin system (RAS) [33] and the immune response [33, 34]. The initial insights into myocardial injury came from autopsy studies which reported the presence of SARS-CoV2 pools located not in the cardiomyocytes but rather in interstitial cells and resident macrophages [48]. Perseverance of these viral reservoirs is still debatable and potentially insidious as they could play an important role in myocardial and vascular sequel in LH COVID-19. Furthermore, psychological or mental stress-related consequences of the COVID-19 pandemic are expected to contribute to the rising cases of cardiovascular disease. The link between mental stress and coronary artery disease, atrial fibrillation and stroke has been reported in various studies [49, 50, 51, 52]. A large multicenter, multinational study, INTERHEART, reported after adjustment for covariates a more than 2-fold increase in the risk of myocardial infarction as a consequence of mental stress [53]. Although the long-term implications of COVID-19 pandemic on cardiovascular health are yet to be realized, previous work done in this area foreshadows a significant uptick of CVD globally, and independently of other comorbidities.

3.1 Chronic stress-associated effects on the central nervous and the cardiovascular systems

In order to study stress as a risk factor, a proper definition must be set in place. The first distinction that must be made is between the stressor and the response of an individual to that stressor (i.e. how well they can cope with it). A stressor is not necessarily perilous per se, neither physiologically or psychologically, as there are many stressors that contribute to desirable outcomes. Much like physical exercise represents a stressor that leads to improved cardiovascular and musculoskeletal health, some psychological stress increases readiness and attention resulting in better outcomes in scholarly activities or sporting competitions. It is when individuals have unique perceptions of the stress and their inability to cope with it that creates a favorable milieu for psychological disturbance and new onset CVD. In fact, different personality types have been reported to be more susceptible to CVD as a consequence of mental stress, including Type A (hostile and angry outlook) [54] and Type D (tendency for pessimism and social inhibition) personalities [55]. Given that the COVID-19 pandemic has brought groups of stressors globally, the impact on psychological and cardiovascular well-being remains to be described.

The CMS rodent model is perhaps the most used in studying mental stress that humans endure and has the highest constructive, face and predicative validity [56]. It consists of exposing the animals to a series of mild, yet unpredictable stressors for at least 4 weeks. One could argue that this is a high-fidelity model of the pattern of stressful events that people experienced consistently during the COVID-19 pandemic, and in that view the data generated in CMS rats may necessitate a closer examination in terms of the CVD comorbidities after the chronic stress to inform future treatment strategies. Namely, the data has shown that rodents exposed to CMS develop depressive-like symptoms and behaviors with adverse cardiovascular symptoms including reduced heart rate variability, elevated resting heart rate, reduced baroreceptor function and increased sympathetic nervous system activity [6, 7, 8, 57]. The sympathetic drive has been shown to be mediated at least in part by the paraventricular nucleus (PVN), and via the vasopressinergic system rather than oxytocin [7, 8]. The CMS rats have also been shown to have increased expression of vasopressin receptors V1a and V1b in the PVN and that the simultaneous inhibition of both V1a and V1b receptors produced maximal inhibition of the neurocardiovascular responses to the exogenous vasopressin administration [7].

Stress can be categorized as acute, lasting seconds to weeks, and chronic, in the months to years range. COVID-19 pandemic-associated stress thus falls into the latter category, and further can be described as CMS. Chronic stressors associated with work and life related issues, such as injustice, effort-reward imbalance, marital stress at home, lack of life partnership, financial stress have all been shown to increase the risk of CVD [58, 59, 60, 61]. Studies in humans have relied on measuring several parameters of the cardiovascular system function to assess the impact of mental stress, including cardiovascular reactivity, levels of catecholamines and inflammatory markers, heart and brain imaging, Holter monitoring and measures of endothelial function with flow-mediated dilatation [62, 63, 64, 65]. It has been suggested that it is not the cardiac function but rather the vasculature, endothelium in particular, where the mental stress translates into CVD. Studies done in monkeys where they were exposed to a novel social environment showed increased endothelial damage in the thoracic aorta and coronary arteries [66]. Other studies in mice reported that both acute and chronic stress reduce the expression of nitric oxide synthase [67, 68], which is responsible for the synthesis of the vasodilatory molecule nitric oxide, leading to endothelial dysfunction. Stimulation of the sympathetic nervous system further increases local norepinephrine production and increase in the expression of adhesion molecules on the endothelium, and cytokine and chemokine production by macrophages and vascular smooth muscle cells. These feed forward cycles ultimately lead to leukocyte adhesion, vascular inflammation, atherosclerotic plaques instability, precipitating a cardiovascular event. Therefore, it is apparent that CMS endured during COVID-19 pandemic may cause similar vascular and endothelial dysfunction in humans as was shown in the above-described animal CMS models.

Chronic mild stress that individuals worldwide have endured during the COVID-19 pandemic has put them at a higher risk of developing anxiety and depression [69]. Lockdown policies instituted across the world resulted in isolation from human contact, worsening dementia and anxiety in individuals in long-term care facilities, exacerbation of conflict due to confinement and fear and confusion resulting from continuous bombardment with reporting information on all media, many of which were unreliable. Additionally, physical activity decreased partly due to the closure of fitness facilities as well as the lack of motivation and fear of SARS-CoV2 infection when leaving outside to obtain exercise. Some of the examples of reduced physical activity can be appreciated from the data from 30 million Fitbit activity tracker users, which showed a significant reduction in daily step counts by as much as 38% in Spain [70]. Similar data was obtained from analyzing step count trends from the app Argus in almost half a million users- a mean reduction in activity by 27.4% [71]. Some implied outcomes from these reductions in daily activity include exacerbation of hypertension. Several cross sectional studies indicated that reductions in step counts led to an increase of systolic blood pressure (SBP) of up to 7 mmHg [72, 73] and an increase of 4.5 mmHg for every additional hour of sitting every day [74]. Other behaviors during the pandemic that could have deleterious effects on blood pressure management include increase in body weight [75], increased sodium and decreased potassium intake [76, 77] which is particularly detrimental in the western countries where dietary intake of sodium is already high [78, 79, 80, 81], and increase in alcohol consumption [82, 83, 84].

Most notably, CMS associated with the pandemic is expected to have adverse consequences on BP in both normotensive and hypertensive individuals. Although no study to date has reported direct associations related to COVID-19, published data indicate that chronic stress leads to an increase in the sympathetic drive as assessed with norepinephrine levels, changes in heart rate as well as via direct neurography [85, 86, 87, 88]. Published clinical evidence repeatedly shows that depressed patients are at a higher risk of developing CVD which persists for a decade following the initial onset of depression [89, 90, 91]. This relationship is not unipolar, as patients with CVD have been shown to develop depressive symptoms [90, 91, 92, 93]. Some of the mechanisms explaining the co-occurrence of depression and CVD include neuroinflammation [94] and autonomic dysfunction [95], but they are by no means an exhaustive list (Figure 1).

Important knowledge has been gleaned from reliable, validated rodent models of CMS [96], which are still utilized to tease apart the mechanistic links between CVD and stress/depression. Importantly, the new and ongoing investigations have been focusing on explaining the difference in vulnerability of individual animals to stress-associated CVD development [97, 98, 99, 100], much like occurs in humans. Rodents exhibit two distinct coping styles when exposed to stress: [1] the proactive coping, which is characterized by more offensive, aggressive and impulsive behavior; and [2] and reactive coping, which is characterized by more cautious and fearful behavior [99, 101]. In addition to the behavioral differences, physiologically the two differ as well, where the proactive (active) copers exhibit heightened sympathetic activity and low HPA axis reactivity and the reactive (passive) copers show the opposite trends [99, 102]. The passive coping rats were also shown to have persistently elevated levels of pro-inflammatory cytokine IL-1β and oxidative stress [103], and it is thus plausible that neuroinflammation is at the intersection of depressive symptoms and CVD.

The sex-based dichotomy in the prevalence and severity of depression has been well-characterized [104, 105]. Furthermore, the efficacy of antidepressant pharmacotherapeutic agents also differs between men and women [106, 107]. Likewise, women are more likely to develop CVD that co-occurs with depression [108]. A growing body of evidence has emerged indicating that COVID-19 pandemic has increased the incidence of depression, with the meta-analysis of 12 community-based studies worldwide highlighting a prevalence of depression of 25% [109], with female gender emerging as a significant risk factor [110, 111, 112]. One study reported that women under 50 persist more devastating symptoms such as fatigue, myalgia, brain fog and fatigue after being hospitalized for COVID-19 [113]. Animal model studies of CMS that address this disparity in males and females are scarce, and some have shown differences in behavioral and hormonal profiles. Anhedonia associated with depressive-like state in CMS rodents is typically measured by an intake of 1–2% sucrose solution, and has been found to be more pronounced in females than in males [114]. The same study found no differences in the corticosterone levels however, indicating similar stress hormonal profiles. These findings are the extend of our understanding of sex-based differences in CVD susceptibility as a function of chronic stress thus representing a large gap in knowledge that future preclinical studies should address. Developing treatments that will target both the depressive symptomatology and the cardiovascular pathology, while also being titratable, will be of utmost importance since there may be a difference in the magnitude of effects caused by chronic stress associated with the pandemic between women than men.

3.2 Long haul COVID-19: emerging effects on the brain, heart and vasculature

During the acute phase of SARS-CoV2 infection, the viral entry into the CNS can be accomplished either directly or indirectly (via neuroinflammation) [115]. The direct viral entry, as mentioned previously, can occur via the olfactory [116] or terminal cranial nerves [117]. ACE2 expression has been recognized on endothelial cells, pericytes and astrocytes, allowing the viral invasion of the CNS via compromised BBB. Alternatively or even additionally, the virus could traverse the microvascular endothelial cells, as has been shown [118]. Consequently, the BBB leakage would allow the influx of the circulating pro-inflammatory cytokines, chemokines and mediators, further perpetuating neuroinflammation. In vitro studies also described the capabilities of SARS-CoV2 to initiate activation of astrocytes and microglia via its structural protein subunit (S1) [119]. This has been confirmed in autopsy studies of COVID-19 patients showing enlarged astrocytes and activated microglia [37]. Under normal physiological conditions, astrocytes play a crucial role in neurotransmission, as they control the synthesis of most essential neurotransmitters glutamate and GABA [120]. Additionally, astrocytes are involved in maintenance of synaptic plasticity via reuptake and recycling of neurotransmitters [121]. Under inflammatory conditions (i.e. SARS-CoV2 infection) astrocytes become reactive, which disrupts the glutamatergic balance, leading to excess extracellular glutamate contributing to dysfunction in both the CNS [58] and the cardiovascular systems [122]. Reactive astrocytosis is further supported by microglia via the NFkB pathway [123]. In the absence of the mechanisms that will shut down reactive astrocytosis (i.e. during COVID-19) the process could lead to the formation of astrocytic scars and in the long term neuronal death and neurodegeneration. Data describing the contribution of reactive astrocytosis in LH COVID-19 is lacking. One study so far has been published that measured plasma biomarkers of CNS injury in 100 COVID-19 survivors in Sweden. The biomarkers included nuerofilament light chain, glial fibrillary acidic protein (GFAP) and differentiation factor 15. In the acute phase, patients with severe symptoms had elevated neurfilament light chain compared to both age-matched controls and mild and moderate COVID-19, as well as higher GFAP than controls. However, after the median follow up of 225 days all CNS injury markers normalized and were indistinguishable from those found in healthy controls [124]. Since emerging data are pointing towards increased neuropathological manifestations one-year out compared to 6 months out [7], more studies are urgently needed to explain the mechanistic details and thus inform appropriate therapeutic strategies.

Data from the prospective post-acute follow up studies focusing on cardiac events have been more abundant in the literature compared to those on the CNS abnormalities. Several large (n > 400) observational studies are still ongoing in 2022. Transthoracic echocardiography and cardiac magnetic resonance are the gold standard techniques used in the diagnosis of cardiac pathologies [125, 126]. The vast majority of the studies have reported the presence of pericarditis, right ventricular dysfunction and myocardial infarctions [127, 128, 129, 130, 131, 132, 133]. Persistent myocarditis was reported in a cohort of 100 patients [127], while in another study of healthcare workers matched for comorbidities and severity of infection showed no difference in cardiac abnormalities 6 months post-infection [128]. Studies in athletes [129, 130, 131, 132] were undertaken within 1–2 months of infection, and the prevalence of myocarditis is generally considered to be low (0–3%), albeit studies beyond the 2 month mark are lacking. Echocardiographic studies have consistently reported right heart abnormalities [133, 134, 135] while the left systolic function is significantly less impaired [133, 136], even in patients with severe acute symptomatology. On the other hand, perhaps the emerging trend that will have to be closely monitored in LH COVID-19 is the diastolic dysfunction, as it was shown to be common in up to 60% of hospitalized patients [137]. It is thus plausible to speculate that the pathologic changes in diastology could manifest during LH COVID-19, given the time lapse from the initial infection. In terms of vasculature, one angiography study reported an association between vascular inflammation caused by the variant B1.1.7 (WHO label Alpha) and increased mortality risk [138]. Although multiple studies have been published so far that have highlighted or implied the development of cardiovascular pathologies in LH COVID-19, one common denominator of limitations in most of them is that the comparator groups were either healthy individuals or individuals unmatched for comorbidities. Perhaps the most important consideration should be the lack of pre-COVID cardiac imaging studies, which makes it difficult to discern whether the pathologies observed were due to COVID-19 or other comorbidities, or perhaps both. Nevertheless, given the non-invasive nature of echocardiography, it likely behooves the cardiac clinicians to implement cardiac imaging in LH COVID-19 given the evidence from prospective studies and the available therapeutics for ensuing cardiac pathologies.

The concerning aspect of the known and implied consequences of COVID-19 discussed above and LH COVID-19 is that these pathological processes do not exist in isolation and ultimately lead to multisystem dysfunction. A multi-organ magnetic resonance imaging study on a small cohort of recovered COVID-19 patients and matched controls revealed some level of abnormalities in the lung (60%), heart (26%), liver (10%), kidneys (29%) and brain (11%) [18]. Recovery from multisystem damage has been shown to be impeded by the persistent pro-inflammatory state [139] as well as endothelial dysfunction [140, 141, 142] and perpetual prothrombotic state [143]. These studies highlight the importance of approaching treatment strategies from the multisystem perspective rather than treating isolated pathologies. For example, antithrombotic therapy may be beneficial for individuals who present with the prothrombotic phenotype and have persistent inflammation in their LH COVID-19 phase.

Advertisement

4. Conclusions

According to the above discussed evidence and implications, the potential neurologic and cardiovascular consequences (Figure 1), coupled with the ensuing healthcare burden of COVID-19 necessitate a careful crafting of the clinical approach in the coming months and years. Both individuals who survived the infection and those who survived the pandemic without becoming infected with SARS-CoV2 (but were exposed to CMS) are at an appreciable risk of developing neurogenic and neuropsychiatric disturbances (Figure 1), and thus mental health checks for virtually all individuals on an ongoing basis are warranted. Furthermore, female gender may add another layer of risk for developing depression and CVD as a consequence of pandemic-associated CMS exposure. Studies conducted so far underscore the utility of echocardiography in revealing COVID-19-associated pericarditis, myocarditis and right heart dysfunction. Furthermore, resolution of inflammation should be at the forefront of treatment strategies, since prolonged inflammatory state is associated with poorer outcomes and LH COVID-19 symptomatology. Ultimately, clinical vigilance in monitoring individuals’ mental and cardiovascular health will be of utmost importance in the post-pandemic years and research strategies aimed at mitigating the defunct mechanisms at the intersection of neurological and cardiovascular pathologies are merited.

References

  1. 1. Rogers JP, Chesney E, Oliver D, Pollak TA, McGuire P, Fusar-Poli P, et al. 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(7):611-627. DOI: 10.1016/S2215-0366(20)30203-0
  2. 2. Liguori C, Pierantozzi M, Spanetta M, Sarmati L, Cesta N, Iannetta M, et al. Subjective neurological symptoms frequently occur in patients with SARS-CoV2 infection. Brain, Behavior, and Immunity. 2020;88:11-16. DOI: 10.1016/j.bbi.2020.05.037
  3. 3. Nalleballe K, Reddy Onteddu S, Sharma R, Dandu V, Brown A, Jasti M, et al. Spectrum of neuropsychiatric manifestations in COVID-19. Brain, Behavior, and Immunity. 2020;88:71-74. DOI: 10.1016/j.bbi.2020.06.020
  4. 4. Dinakaran D, Manjunatha N, Naveen Kumar C, Suresh BM. Neuropsychiatric aspects of COVID-19 pandemic: A selective review. Asian Journal of Psychiatry. 2020;53:102188. DOI: 10.1016/j.ajp.2020.102188
  5. 5. Raman B, Bluemke DA, Luscher TF, Neubauer S. Long COVID: Post-acute sequelae of COVID-19 with a cardiovascular focus. European Heart Journal. 2022;43(11):1157-1172. DOI: 10.1093/eurheartj/ehac031
  6. 6. Grippo AJ, Moffitt JA, Johnson AK. Evaluation of baroreceptor reflex function in the chronic mild stress rodent model of depression. Psychosomatic Medicine. 2008;70(4):435-443. DOI: 10.1097/PSY.0b013e31816ff7dd
  7. 7. Komnenov D, Quaal H, Rossi NF. V-1a and V-1b vasopressin receptors within the paraventricular nucleus contribute to hypertension in male rats exposed to chronic mild unpredictable stress. American Journal of Physiology-Regulatory, Integrative and Comparative Physiology. 2021;320(3):R213-RR25. DOI: 10.1152/ajpregu.00245.2020
  8. 8. Das S, Komnenov D, Newhouse L, Rishi AK, Rossi NF. Paraventricular nucleus V1a receptor knockdown blunts neurocardiovascular responses to acute stress in male rats after chronic mild unpredictable stress. Physiology & Behavior. 2022;253:113867. DOI: 10.1016/j.physbeh.2022.113867
  9. 9. Grippo AJ, Beltz TG, Johnson AK. Behavioral and cardiovascular changes in the chronic mild stress model of depression. Physiology & Behavior. 2003;78(4-5):703-710. DOI: 10.1016/s0031-9384(03)00050-7
  10. 10. Esterwood E, Saeed SA. Past epidemics, natural disasters, COVID19, and mental health: Learning from history as we Deal with the present and prepare for the future. Psychiatric Quarterly. 2020;91(4):1121-1133. DOI: 10.1007/s11126-020-09808-4
  11. 11. Morganstein JC, Fullerton CS, Ursano RJ, Donato D, Holloway HC. Pandemics: Health Care Emergencies. Textbook of Disaster Psychiatry. 2nd ed. Cambridge, UK: Cambridge University Press; 2017. pp. 270-284. DOI: 10.1017/ 9781316481424
  12. 12. Varatharaj A, Thomas N, Ellul MA, Davies NWS, Pollak TA, Tenorio EL, et al. Neurological and neuropsychiatric complications of COVID-19 in 153 patients: A UK-wide surveillance study. Lancet Psychiatry. 2020;7(10):875-882. DOI: 10.1016/S2215-0366(20)30287-X
  13. 13. Parra A, Juanes A, Losada CP, Alvarez-Sesmero S, Santana VD, Marti I, et al. Psychotic symptoms in COVID-19 patients. A retrospective descriptive study. Psychiatry Research. 2020;291:113254. DOI: 10.1016/j.psychres.2020.113254
  14. 14. Helms J, Meziani F. Neurologic features in severe SARS-CoV-2 infection. New England Journal of Medicine. 2020;382(23):2268-2270. DOI: 10.1056/NEJMc2008597
  15. 15. Taquet M, Luciano S, Geddes JR, Harrison PJ. Bidirectional associations between COVID-19 and psychiatric disorder: Retrospective cohort studies of 62 354 COVID-19 cases in the USA. Lancet Psychiatry. 2020;8(1):E1-EE. DOI: 10.1016/S2215-0366(20)30462-4
  16. 16. Kremer S, Lersy F, Anheim M, Merdji H, Schenck M, Oesterle H, et al. Neurologic and neuroimaging findings in patients with COVID-19 a retrospective multicenter study. Neurology. 2020;95(13):E1868-E1E82. DOI: 10.1212/Wnl.0000000000010112
  17. 17. Pezzini A, Padovani A. Lifting the mask on neurological manifestations of COVID-19. Nature Reviews. Neurology. 2020;16(11):636-644. DOI: 10.1038/s41582-020-0398-3
  18. 18. Raman B, Cassar MP, Tunnicliffe EM, Filippini N, Griffanti L, Alfaro-Almagro F, et al. Medium-term effects of SARS-CoV-2 infection on multiple vital organs, exercise capacity, cognition, quality of life and mental health, post-hospital discharge. Eclinicalmedicine. 2021;31. DOI: 10.1016/j.eclinm.2020.100683
  19. 19. Cipriani G, Di Fiorino M. Access to care for dementia patients suffering from COVID-19. American Journal of Geriatric Psychiatry. 2020;28(7):796-797
  20. 20. Cai X, Hu XP, Ekumi IO, Wang JC, An YW, Li ZW, et al. Psychological distress and its correlates among COVID-19 survivors during early convalescence across age groups. American Journal of Geriatric Psychiatry. 2020;28(10):1030-1039. DOI: 10.1016/j.jagp.2020.07.003
  21. 21. Yuan B, Li WX, Liu HQ , Cai X, Song S, Zhao J, et al. Correlation between immune response and self-reported depression during convalescence from COVID-19. Brain Behavior and Immunity. 2020;88:39-43. DOI: 10.1016/j.bbi.2020.05.062
  22. 22. Zhang J, Lu HP, Zeng HP, Zhang SN, Du QF, Jiang TY, et al. The differential psychological distress of populations affected by the COVID-19 pandemic. Brain Behavior and Immunity. 2020;87:49-50. DOI: 10.1016/j.bbi.2020.04.031
  23. 23. Kabbani N, Olds JL. Does COVID19 infect the brain? If so, smokers might be at a higher risk. Molecular Pharmacology. 2020;97(5):351-353. DOI: 10.1124/molpharm.120.000014
  24. 24. Moriguchi T, Harii N, Goto J, Harada D, Sugawara H, Takamino J, et al. A first case of meningitis/encephalitis associated with SARS-Coronavirus-2. International Journal of Infectious Diseases. 2020;94:55-58. DOI: 10.1016/j.ijid.2020.03.062
  25. 25. Mirfazeli FS, Sarabi-Jamab A, Jahanbakhshi A, Kordi A, Javadnia P, Shariat SV, et al. Neuropsychiatric manifestations of COVID-19 can be clustered in three distinct symptom categories. Scientific Reports. 2020;10(1). DOI: 10.1038/s41598-020-78050-6
  26. 26. Troyer EA, Kohn JN, Hong SZ. Are we facing a crashing wave of neuropsychiatric sequelae of COVID-19? Neuropsychiatric symptoms and potential immunologic mechanisms. Brain Behavior and Immunity. 2020;87:34-39. DOI: 10.1016/j.bbi.2020.04.027
  27. 27. Baig AM, 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 Chemical Neuroscience. 2020;11(7):995-998. DOI: 10.1021/acschemneuro.0c00122
  28. 28. Onofrio L, Caraglia M, Facchini G, Margherita V, De Placido S, Buonerba C. Toll-like receptors and COVID-19: A two-faced story with an exciting ending. Future Science OA. 2020;6(8). DOI: 10.2144/fsoa-2020-0091
  29. 29. van Vuren EJ, Steyn SF, Brink CB, Moller M, Viljoen FP, Harvey BH. The neuropsychiatric manifestations of COVID-19: Interactions with psychiatric illness and pharmacological treatment. Biomedicine & Pharmacotherapy. 2021;135. DOI: 10.1016/j.biopha.2020.111200
  30. 30. Wang YX, Wang YY, Chen Y, Qin QS. Unique epidemiological and clinical features of the emerging 2019 novel coronavirus pneumonia (COVID-19) implicate special control measures. Journal of Medical Virology. 2020;92(6):568-576. DOI: 10.1002/jmv.25748
  31. 31. Long COVID: Let patients help define long-lasting COVID symptoms. Nature. 2020;586(7828):170. DOI: 10.1038/d41586-020-02796-2
  32. 32. Al-Aly Z, Xie Y, Bowe B. High-dimensional characterization of post-acute sequelae of COVID-19. Nature. 2021;594(7862):259-264. DOI: 10.1038/s41586-021-03553-9
  33. 33. Shah W, Hillman T, Playford ED, Hishmeh L. Managing the long term effects of covid-19: Summary of NICE, SIGN, and RCGP rapid guideline. BMJ: British Medical Journal. 2021;372. DOI: 10.1136/bmj.n136
  34. 34. Nalbandian A, Sehgal K, Gupta A, Madhavan MV, McGroder C, Stevens JS, et al. Post-acute COVID-19 syndrome. Nature Medicine. 2021;27(4):601-615. DOI: 10.1038/s41591-021-01283-z
  35. 35. Buzhdygan TP, DeOre BJ, Baldwin-Leclair A, Bullock TA, McGary HM, Khan JA, 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. Neurobiology of Disease. 2020;146. DOI: 10.1016/j.nbd.2020.105131
  36. 36. Crook H, Raza S, Nowell J, Young M, Edison P. Long covid-mechanisms, risk factors, and management. BMJ: British Medical Journal. 2021;374. DOI: 10.1136/bmj.n1648
  37. 37. Perl DP, Folkerth RD, Nath A. Microvascular injury in the brains of patients with Covid-19. New England Journal of Medicine. 2021;384(5):481-483. DOI: 10.1056/NEJMc2033369
  38. 38. Dixon L, McNamara C, Gaur P, Mallon D, Coughlan C, Tona F, et al. Cerebral microhaemorrhage in COVID-19: A critical illness related phenomenon? Stroke and Vascular Neurology. 2020;5(4):315-322. DOI: 10.1136/svn-2020-000652
  39. 39. Sawlani V, Scotton S, Nader K, Jen JP, Patel M, Gokani K, et al. COVID-19-related intracranial imaging findings: A large single-centre experience. Clinical Radiology. 2021;76(2):108-116. DOI: 10.1016/j.crad.2020.09.002
  40. 40. Taquet M, Geddes JR, Husain M, Luciano S, Harrison PJ. 6-month neurological and psychiatric outcomes in 236 379 survivors of COVID-19: A retrospective cohort study using electronic health records. Lancet Psychiatry. 2021;8(5):416-427. DOI: 10.1016/S2215-0366(21)00084-5
  41. 41. Premraj L, Kannapadi NV, Briggs J, Seal SM, Battaglini D, Fanning J, et al. Mid and long-term neurological and neuropsychiatric manifestations of post-COVID-19 syndrome: A meta-analysis. Journal of the Neurological Sciences. 2022;434. DOI: 10.1016/j.jns.2022.120162
  42. 42. Estiri H, Strasser ZH, Brat GA, Semenov YR, Patel CJ, Murphy SN, et al. Evolving phenotypes of non-hospitalized patients that indicate long COVID. BMC Medicine. 2021;19(1). DOI: 10.1186/s12916-021-02115-0
  43. 43. Lala A, Johnson KW, Januzzi JL, Russak AJ, Paranjpe I, Richter F, et al. Prevalence and impact of myocardial injury in patients hospitalized with COVID-19 infection. Journal of the American College of Cardiology. 2020;76(5):533-546. DOI: 10.1016/j.jacc.2020.06.007
  44. 44. Shi S, Qin M, Shen B, Cai Y, Liu T, Yang F, et al. Association of cardiac injury with mortality in hospitalized patients with COVID-19 in Wuhan, China. JAMA Cardiology. 2020;5(7):802-810. DOI: 10.1001/jamacardio.2020.0950
  45. 45. Shi S, Qin M, Cai Y, Liu T, Shen B, Yang F, et al. Characteristics and clinical significance of myocardial injury in patients with severe coronavirus disease 2019. European Heart Journal. 2020;41(22):2070-2079. DOI: 10.1093/eurheartj/ehaa408
  46. 46. Wang D, Hu B, Hu C, Zhu F, Liu X, Zhang J, et al. Clinical characteristics of 138 hospitalized patients with 2019 novel coronavirus-infected pneumonia in Wuhan, China. JAMA. 2020;323(11):1061-1069. DOI: 10.1001/jama.2020.1585
  47. 47. Dong E, Du H, Gardner L. An interactive web-based dashboard to track COVID-19 in real time. The Lancet Infectious Diseases. 2020;20(5):533-534. DOI: 10.1016/S1473-3099(20)30120-1
  48. 48. Kawakami R, Sakamoto A, Kawai K, Gianatti A, Pellegrini D, Nasr A, et al. Pathological evidence for SARS-CoV-2 as a cause of myocarditis JACC review topic of the week. Journal of the American College of Cardiology. 2021;77(3):314-325. DOI: 10.1016/j.jacc.2020.11.031
  49. 49. Dragano N, Siegrist J, Nyberg ST, Lunau T, Fransson EI, Alfredsson L, et al. Effort-reward imbalance at work and incident coronary heart disease a multicohort study of 90,164 individuals. Epidemiology. 2017;28(4):619-626. DOI: 10.1097/Ede.0000000000000666
  50. 50. Huang YL, Xu SX, Hua JH, Zhu DJ, Liu CH, Hu YZ, et al. Association between job strain and risk of incident stroke a meta-analysis. Neurology. 2015;85(19):1648-1654. DOI: 10.1212/Wnl.0000000000002098
  51. 51. Kivimaki M, Kawachi I. Work stress as a risk factor for cardiovascular disease. Current Cardiology Reports. 2015;17(9). DOI: 10.1007/s11886-015-0630-8
  52. 52. Kivimaki M, Jokela M, Nyberg ST, Singh-Manoux A, Fransson EI, Alfredsson L, et al. Long working hours and risk of coronary heart disease and stroke: A systematic review and meta-analysis of published and unpublished data for 603 838 individuals. Lancet. 2015;386(10005):1739-1746. DOI: 10.1016/S0140-6736(15)60295-1
  53. 53. Rosengren A, Hawken S, Ounpuu S, Sliwa K, Zubaid M, Almahmeed WA, et al. Association of psychosocial risk factors with risk of acute myocardial infarction in 11119 cases and 13 648 controls from 52 countries (the INTERHEART study): Case-control study. Lancet. 2004;364(9438):953-962. DOI: 10.1016/S0140-6736(04)17019-0
  54. 54. Chida Y, Steptoe A. The association of anger and hostility with future coronary heart disease a meta-analytic review of prospective evidence. Journal of the American College of Cardiology. 2009;53(11):936-946. DOI: 10.1016/j.jacc.2008.11.044
  55. 55. Grande G, Romppel M, Barth J. Association between type D personality and prognosis in patients with cardiovascular diseases: A systematic review and meta-analysis. Annals of Behavioral Medicine. 2012;43(3):299-310. DOI: 10.1007/s12160-011-9339-0
  56. 56. Willner P. Chronic mild stress (CMS) revisited: Consistency and behavioural-neurobiological concordance in the effects of CMS. Neuropsychobiology. 2005;52(2):90-110. DOI: 10.1159/000087097
  57. 57. Bouzinova EV, Moller-Nielsen N, Boedtkjer DB, Broegger T, Wiborg O, Aalkjaer C, et al. Chronic mild stress-induced depression-like symptoms in rats and abnormalities in catecholamine uptake in small arteries. Psychosomatic Medicine. 2012;74(3):278-287. DOI: 10.1097/PSY.0b013e31824c40a9
  58. 58. Sara JD, Prasad M, Eleid MF, Zhang M, Widmer RJ, Lerman A. Association between work-related stress and coronary heart disease: A review of prospective studies through the job strain, effort-reward balance, and organizational justice models. Journal of the American Heart Association. 2018;7(9):e008073. DOI: 10.1161/JAHA.117.008073
  59. 59. Orth-Gomer K, Wamala SP, Horsten M, Schenck-Gustafsson K, Schneiderman N, Mittleman MA. Marital stress worsens prognosis in women with coronary heart disease—the Stockholm female coronary risk study. JAMA—Journal of the American Medical Association. 2000;284(23):3008-3014. DOI: 10.1001/jama.284.23.3008
  60. 60. Albert MA, Durazo EM, Slopen N, Zaslavsky AM, Buring JE, Silva T, et al. Cumulative psychological stress and cardiovascular disease risk in middle aged and older women: Rationale, design, and baseline characteristics. American Heart Journal. 2017;192:1-12. DOI: 10.1016/j.ahj.2017.06.012
  61. 61. Carlsson AC, Starrin B, Gigante B, Leander K, Hellenius ML, de Faire U. Financial stress in late adulthood and diverse risks of incident cardiovascular disease and all-cause mortality in women and men. BMC Public Health. 2014;14. DOI: 10.1186/1471-2458-14-17
  62. 62. Widmer RJ, Prasad M, Gomaa M, Sara JDS, Reriani MK, Lerman LO, et al. Vascular reactivity to mental stress is associated with poor cardiovascular disease outcomes in females following acute coronary syndrome. Coronary Artery Disease. 2020;31(3):300-305. DOI: 10.1097/Mca.0000000000000831
  63. 63. Bandodkar AJ, Wang J. Non-invasive wearable electrochemical sensors: A review. Trends in Biotechnology. 2014;32(7):363-371. DOI: 10.1016/j.tibtech.2014.04.005
  64. 64. Mitchell JS, Lowe TE, Ingram JR. Rapid ultrasensitive measurement of salivary cortisol using nano-linker chemistry coupled with surface plasmon resonance detection. The Analyst. 2009;134(2):380-386. DOI: 10.1039/b817083p
  65. 65. Rozanski A, Bairey CN, Krantz DS, Friedman J, Resser KJ, Morell M, et al. Mental stress and the induction of silent myocardial ischemia in patients with coronary-artery disease. New England Journal of Medicine. 1988;318(16):1005-1012. DOI: 10.1056/Nejm198804213181601
  66. 66. Strawn WB, Bondjers G, Kaplan JR, Manuck SB, Schwenke DC, Hansson GK, et al. Endothelial dysfunction in response to psychosocial stress in monkeys. Circulation Research. 1991;68(5):1270-1279. DOI: 10.1161/01.Res.68.5.1270
  67. 67. Custodis F, Gertz K, Balkaya M, Prinz V, Mathar I, Stamm C, et al. Heart rate contributes to the vascular effects of chronic mental stress effects on endothelial function and ischemic brain injury in mice. Stroke. 2011;42(6):1742-1749. DOI: 10.1161/Strokeaha.110.598607
  68. 68. Chung IM, Kim YM, Yoo MH, Shin MK, Kim CK, Suh SH. Immobilization stress induces endothelial dysfunction by oxidative stress via the activation of the angiotensin II/its type I receptor pathway. Atherosclerosis. 2010;213(1):109-114. DOI: 10.1016/j.atherosclerosis.2010.08.052
  69. 69. Brooks SK, Webster RK, Smith LE, Woodland L, Wessely S, Greenberg N, et al. The psychological impact of quarantine and how to reduce it: Rapid review of the evidence. Lancet. 2020;395(10227):912-920. DOI: 10.1016/S0140-6736(20)30460-8
  70. 70. Kreutz R, Dobrowolski P, Prejbisz A, Algharably EAEH, Bilo G, Creutzig F, et al. Lifestyle, psychological, socioeconomic and environmental factors and their impact on hypertension during the coronavirus disease 2019 pandemic. Journal of Hypertension. 2021;39(6):1077-1089. DOI: 10.1097/Hjh.0000000000002770
  71. 71. Tison GH, Avram R, Kuhar P, Abreau S, Marcus GM, Pletcher MJ, et al. Worldwide effect of COVID-19 on physical activity: A descriptive study. Annals of Internal Medicine. 2020;173(9):767. DOI: 10.7326/M20-2665
  72. 72. Menai M, Brouard B, Vegreville M, Chieh A, Schmidt N, Oppert JM, et al. Cross-sectional and longitudinal associations of objectively-measured physical activity on blood pressure: Evaluation in 37 countries. Health Promotion Perspectives. 2017;7(4):190-196. DOI: 10.15171/hpp.2017.34
  73. 73. Zucatti ATN, de Paula TP, Viana LV, DallAgnol R, Cureau FV, Azevedo MJ, et al. Low levels of usual physical activity are associated with higher 24 h blood pressure in type 2 diabetes mellitus in a cross-sectional study. Journal Diabetes Research. 2017;2017:6232674. DOI: 10.1155/2017/6232674
  74. 74. Maasakkers CM, de Heus RAA, Thijssen DHJ, Melis RJF, Gardiner PA, Claassen J. Objectively-measured activity patterns are associated with home blood pressure in memory clinic patients. Journal of Alzheimer's Disease. 2020;74(2):691-697. DOI: 10.3233/JAD-191310
  75. 75. Sidor A, Rzymski P. Dietary choices and habits during COVID-19 lockdown: Experience from Poland. Nutrients. 2020;12(6). DOI: 10.3390/nu12061657
  76. 76. Yilmaz C, Gokmen V. Neuroactive compounds in foods: Occurrence, mechanism and potential health effects. Food Research International. 2020;128:108744. DOI: 10.1016/j.foodres.2019.108744
  77. 77. Rodriguez-Martin BC, Meule A. Food craving: New contributions on its assessment, moderators, and consequences. Frontiers in Psychology. 2015;6:21. DOI: 10.3389/fpsyg.2015.00021
  78. 78. Aburto NJ, Hanson S, Gutierrez H, Hooper L, Elliott P, Cappuccio FP. Effect of increased potassium intake on cardiovascular risk factors and disease: Systematic review and meta-analyses. BMJ. 2013;346:f1378. DOI: 10.1136/bmj.f1378
  79. 79. Adler AJ, Taylor F, Martin N, Gottlieb S, Taylor RS, Ebrahim S. Reduced dietary salt for the prevention of cardiovascular disease. Cochrane Database of Systematic Reviews. 2014;(12):CD009217. DOI: 10.1002/14651858.CD009217.pub3
  80. 80. Binia A, Jaeger J, Hu Y, Singh A, Zimmermann D. Daily potassium intake and sodium-to-potassium ratio in the reduction of blood pressure: A meta-analysis of randomized controlled trials. Journal of Hypertension. 2015;33(8):1509-1520. DOI: 10.1097/HJH.0000000000000611
  81. 81. He FJ, MacGregor GA. Fortnightly review: Beneficial effects of potassium. BMJ. 2001;323(7311):497-501. DOI: 10.1136/bmj.323.7311.497
  82. 82. Hamer M, Kivimaki M, Gale CR, Batty GD. Lifestyle risk factors, inflammatory mechanisms, and COVID-19 hospitalization: A community-based cohort study of 387,109 adults in UK. Brain, Behavior, and Immunity. 2020;87:184-187. DOI: 10.1016/j.bbi.2020.05.059
  83. 83. Colbert S, Wilkinson C, Thornton L, Richmond R. COVID-19 and alcohol in Australia: Industry changes and public health impacts. Drug and Alcohol Review. 2020;39(5):435-440. DOI: 10.1111/dar.13092
  84. 84. The lancet gastroenterology H. drinking alone: COVID-19, lockdown, and alcohol-related harm. Lancet Gastroenterología y Hepatología. 2020;5(7):625. DOI: 10.1016/S2468-1253(20)30159-X
  85. 85. Mancia G, Grassi G, Pomidossi G, Gregorini L, Bertinieri G, Parati G, et al. Effects of blood-pressure measurement by the doctor on patients blood-pressure and heart-rate. Lancet. 1983;2(8352):695-698
  86. 86. Grassi G, Turri C, Vailati S, Dell'Oro R, Mancia G. Muscle and skin sympathetic nerve traffic during the "white-coat" effect. Circulation. 1999;100(3):222-225. DOI: 10.1161/01.Cir.100.3.222
  87. 87. Esler M, Eikelis N, Schlaich M, Lambert G, Alvarenga M, Dawood T, et al. Chronic mental stress is a cause of essential hypertension: Presence of biological markers of stress. Clinical and Experimental Pharmacology and Physiology. 2008;35(4):498-502. DOI: 10.1111/j.1440-1681.2008.04904.x
  88. 88. Esler M, Eikelis N, Schlaich M, Lambert G, Alvarenga M, Kaye D, et al. Human sympathetic nerve biology parallel influences of stress and epigenetics in essential hypertension and panic disorder. Annals of the New York Academy of Sciences. 2008;1148:338-348. DOI: 10.1196/annals.1410.064
  89. 89. Ford DE, Mead LA, Chang PP, Cooper-Patrick L, Wang NY, Klag MJ. Depression is a risk factor for coronary artery disease in men—The precursors study. Archives of Internal Medicine. 1998;158(13):1422-1426. DOI: 10.1001/archinte.158.13.1422
  90. 90. Cohen BE, Edmondson D, Kronish IM. State of the art review: Depression, stress, anxiety, and cardiovascular disease. American Journal of Hypertension. 2015;28(11):1295-1302. DOI: 10.1093/ajh/hpv047
  91. 91. Hare DL, Toukhsati SR, Johansson P, Jaarsma T. Depression and cardiovascular disease: A clinical review. European Heart Journal. 2014;35(21):1365-1372. DOI: 10.1093/eurheartj/eht462
  92. 92. Whooley MA, de Jonge P, Vittinghoff E, Otte C, Moos R, Carney RM, et al. Depressive symptoms, health behaviors, and risk of cardiovascular events in patients with coronary heart disease. Journal of the American Medical Association. 2008;300(20):2379-2388. DOI: 10.1001/jama.2008.711
  93. 93. Wu JR, Frazier SK, Rayens MK, Lennie TA, Chung ML, Moser DK. Medication adherence, social support, and event-free survival in patients with heart failure. Health Psychology. 2013;32(6):637-646. DOI: 10.1037/a0028527
  94. 94. Slavich GM, Irwin MR. From stress to inflammation and major depressive disorder: A social signal transduction theory of depression. Psychological Bulletin. 2014;140(3):774-815. DOI: 10.1037/a0035302
  95. 95. Sgoifo A, Carnevali L, Alfonso Mde L, Amore M. Autonomic dysfunction and heart rate variability in depression. Stress. 2015;18(3):343-352. DOI: 10.3109/10253890.2015.1045868
  96. 96. Carnevali L, Montano N, Statello R, Sgoifo A. Rodent models of depression-cardiovascular comorbidity: Bridging the known to the new. Neuroscience and Biobehavioral Reviews. 2017;76(Pt A):144-153. DOI: 10.1016/j.neubiorev.2016.11.006
  97. 97. Armario A, Nadal R. Individual differences and the characterization of animal models of psychopathology: A strong challenge and a good opportunity. Frontiers in Pharmacology. 2013;4:137. DOI: 10.3389/fphar.2013.00137
  98. 98. de Boer SF, Buwalda B, Koolhaas JM. Untangling the neurobiology of coping styles in rodents: Towards neural mechanisms underlying individual differences in disease susceptibility. Neuroscience and Biobehavioral Reviews. 2017;74(Pt B):401-422. DOI: 10.1016/j.neubiorev.2016.07.008
  99. 99. Koolhaas JM, Korte SM, De Boer SF, Van Der Vegt BJ, Van Reenen CG, Hopster H, et al. Coping styles in animals: Current status in behavior and stress-physiology. Neuroscience and Biobehavioral Reviews. 1999;23(7):925-935. DOI: 10.1016/s0149-7634(99)00026-3
  100. 100. Krishnan V, Han MH, Graham DL, Berton O, Renthal W, Russo SJ, et al. Molecular adaptations underlying susceptibility and resistance to social defeat in brain reward regions. Cell. 2007;131(2):391-404. DOI: 10.1016/j.cell.2007.09.018
  101. 101. Coppens CM, de Boer SF, Koolhaas JM. Coping styles and behavioural flexibility: Towards underlying mechanisms. Philosophical Transactions of the Royal Society of London. Series B, Biological Sciences. 2010;365(1560):4021-4028. DOI: 10.1098/rstb.2010.0217
  102. 102. Wood SK. Individual differences in the neurobiology of social stress: Implications for depression-cardiovascular disease comorbidity. Current Neuropharmacology. 2014;12(2):205-211. DOI: 10.2174/1570159X11666131120224413
  103. 103. Wood CS, Valentino RJ, Wood SK. Individual differences in the locus coeruleus-norepinephrine system: Relevance to stress-induced cardiovascular vulnerability. Physiology & Behavior. 2017;172:40-48. DOI: 10.1016/j.physbeh.2016.07.008
  104. 104. Hyde JS, Mezulis AH. Gender differences in depression: Biological, affective, cognitive, and sociocultural factors. Harvard Review of Psychiatry. 2020;28(1):4-13. DOI: 10.1097/Hrp.0000000000000230
  105. 105. Kuehner C. Why is depression more common among women than among men? Lancet Psychiatry. 2017;4(2):146-158. DOI: 10.1016/S2215-0366(16)30263-2
  106. 106. Marcus SM, Flynn HA, Blow F, Barry K. A screening study of antidepressant treatment rates and mood symptoms in pregnancy. Archives of Women's Mental Health. 2005;8(1):25-27. DOI: 10.1007/s00737-005-0072-1
  107. 107. Sloan DM, Kornstein SG. Gender differences in depression and response to antidepressant treatment. The Psychiatric Clinics of North America. 2003;26(3):581-594. DOI: 10.1016/s0193-953x(03)00044-3
  108. 108. Moller-Leimkuhler AM. Gender differences in cardiovascular disease and comorbid depression. Dialogues in Clinical Neuroscience. 2007;9(1):71-83
  109. 109. Bueno-Notivol J, Gracia-Garcia P, Olaya B, Lasheras I, Lopez-Anton R, Santabarbara J. Prevalence of depression during the COVID-19 outbreak: A meta-analysis of community-based studies. International Journal of Clinical and Health Psychology. 2021;21(1). DOI: 10.1016/j.ijchp.2020.07.007
  110. 110. Sriharan A, Ratnapalan S, Tricco AC, Lupea D, Ayala AP, Pang H, et al. Occupational stress, burnout, and depression in women in healthcare during COVID-19 pandemic: Rapid scoping review. Frontiers in Global Women's Health. 2020;1:596690. DOI: 10.3389/fgwh.2020.596690
  111. 111. Almeida M, Shrestha AD, Stojanac D, Miller LJ. The impact of the COVID-19 pandemic on women's mental health. Archives of Women's Mental Health. 2020;23(6):741-748. DOI: 10.1007/s00737-020-01092-2
  112. 112. Solomou I, Constantinidou F. Prevalence and predictors of anxiety and depression symptoms during the COVID-19 pandemic and compliance with precautionary measures: Age and sex matter. International Journal of Environmental Research and Public Health. 2020;17(14). DOI: 10.3390/ijerph17144924
  113. 113. Torjesen I. Covid-19: Middle aged women face greater risk of debilitating long term symptoms. BMJ. 2021;372:n829. DOI: 10.1136/bmj.n829
  114. 114. Page GG, Opp MR, Kozachik SL. Sex differences in sleep, anhedonia, and HPA axis activity in a rat model of chronic social defeat. Neurobiology of Stress. 2016;3:105-113. DOI: 10.1016/j.ynstr.2016.03.002
  115. 115. Heneka MT, Golenbock D, Latz E, Morgan D, Brown R. Immediate and long-term consequences of COVID-19 infections for the development of neurological disease. Alzheimer's Research & Therapy. 2020;12(1):69. DOI: 10.1186/s13195-020-00640-3
  116. 116. Meinhardt J, Radke J, Dittmayer C, Franz J, Thomas C, Mothes R, et al. Olfactory transmucosal SARS-CoV-2 invasion as a port of central nervous system entry in individuals with COVID-19. Nature Neuroscience. 2021;24(2):168-175. DOI: 10.1038/s41593-020-00758-5
  117. 117. Bilinska K, von Bartheld CS, Butowt R. Expression of the ACE2 virus entry protein in the nervus terminalis reveals the potential for an alternative route to brain infection in COVID-19. Frontiers in Cellular Neuroscience. 2021;15. DOI: 10.3389/fncel.2021.674123
  118. 118. Zhang L, Zhou L, Bao LL, Liu JN, Zhu H, Lv Q , et al. SARS-CoV-2 crosses the blood-brain barrier accompanied with basement membrane disruption without tight junctions alteration. Signal Transduction and Targeted Therapy. 2021;6(1). DOI: 10.1038/s41392-021-00719-9
  119. 119. Frank MG, Nguyen KH, Ball JB, Hopkins S, Kelley T, Baratta MV, et al. SARS-CoV-2 spike S1 subunit induces neuroinflammatory, microglial and behavioral sickness responses: Evidence of PAMP-like properties. Brain Behavior and Immunity. 2022;100:267-277. DOI: 10.1016/j.bbi.2021.12.007
  120. 120. Schousboe A. Metabolic signaling in the brain and the role of astrocytes in control of glutamate and GABA neurotransmission. Neuroscience Letters. 2019;689:11-13. DOI: 10.1016/j.neulet.2018.01.038
  121. 121. De Pitta M, Brunel N, Volterra A. Astrocytes: Orchestrating synaptic plasticity? Neuroscience. 2016;323:43-61. DOI: 10.1016/j.neuroscience.2015.04.001
  122. 122. Matott MP, Kline DD, Hasser EM. Glial EAAT2 regulation of extracellular nTS glutamate critically controls neuronal activity and cardiorespiratory reflexes. Journal of Physiology London. 2017;595(17):6045-6063. DOI: 10.1113/Jp274620
  123. 123. Villarreal A, Vidos C, Busso MM, Cieri MB, Ramos AJ. Pathological neuroinflammatory conversion of reactive astrocytes is induced by microglia and involves chromatin remodeling. Frontiers in Pharmacology. 2021;12. DOI: 10.3389/fphar.2021.689346
  124. 124. Kanberg N, Simren J, Eden A, Andersson LM, Nilsson S, Ashton NJ, et al. Neurochemical signs of astrocytic and neuronal injury in acute COVID-19 normalizes during long-term follow-up. Ebiomedicine. 2021;70. DOI: 10.1016/j.ebiom.2021.103512
  125. 125. Flachskampf FA. Cardiac imaging to evaluate left ventricular diastolic function. JACC: Cardiovascular Imaging. 2015;8(9):1072-1093
  126. 126. Pennell DJ. Cardiovascular magnetic resonance. Circulation. 2010;121(5):692-705. DOI: 10.1161/Circulationaha.108.811547
  127. 127. Puntmann VO, Carerj ML, Wieters I, Fahim M, Arendt C, Hoffmann J, et al. Outcomes of cardiovascular magnetic resonance imaging in patients recently recovered from coronavirus disease 2019 (COVID-19). JAMA Cardiology. 2020;5(11):1265-1273. DOI: 10.1001/jamacardio.2020.3557
  128. 128. Joy G, Artico J, Kurdi H, Seraphim A, Lau C, Thornton GD, et al. Prospective case-control study of cardiovascular abnormalities 6 months following mild COVID-19 in healthcare workers. JACC: Cardiovascular Imaging. 2021;14(11):2155-2166. DOI: 10.1016/j.jcmg.2021.04.011
  129. 129. Vago H, Szabo L, Dohy Z, Merkely B. Cardiac magnetic resonance findings in patients recovered from COVID-19 initial experiences in elite athletes. JACC: Cardiovascular Imaging. 2021;14(6):1279-1281. DOI: 10.1016/j.jcmg.2020.11.014
  130. 130. Daniels CJ, Rajpal S, Greenshields JT, Rosenthal GL, Chung EH, Terrin M, et al. Prevalence of clinical and subclinical myocarditis in competitive athletes with recent SARS-CoV-2 infection results from the big ten COVID-19 cardiac registry. JAMA Cardiology. 2021;6(9):1078-1087. DOI: 10.1001/jamacardio.2021.2065
  131. 131. Martinez MW, Tucker AM, Bloom OJ, Green G, DiFiori JP, Solomon G, et al. Prevalence of inflammatory heart disease among professional athletes with prior COVID-19 infection who received systematic return-to-play cardiac screening. JAMA Cardiology. 2021;6(7):745-752. DOI: 10.1001/jamacardio.2021.0565
  132. 132. Moulson N, Petek BJ, Drezner JA, Harmon KG, Kliethermes SA, Patel MR, et al. SARS-CoV-2 cardiac involvement in Young competitive athletes. Circulation. 2021;144(4):256-266. DOI: 10.1161/Circulationaha.121.054824
  133. 133. Moody WE, Liu BY, Mahmoud-Elsayed HM, Senior J, Lalla SS, Khan-Kheil AM, et al. Persisting adverse ventricular Remodeling in COVID-19 survivors: A longitudinal echocardiographic study. Journal of the American Society of Echocardiography. 2021;34(5):562-566. DOI: 10.1016/j.echo.2021.01.020
  134. 134. Argulian E, Sud K, Vogel B, Bohra C, Garg VP, Talebi S, et al. Right ventricular dilation in hospitalized patients with COVID-19 infection. JACC: Cardiovascular Imaging. 2020;13(11):2459-2461. DOI: 10.1016/j.jcmg.2020.05.010
  135. 135. Kim J, Volodarskiy A, Sultana R, Pollie MP, Yum B, Nambiar L, et al. Prognostic utility of right ventricular remodeling over conventional risk stratification in patients with COVID-19. Journal of the American College of Cardiology. 2020;76(17):1965-1977. DOI: 10.1016/j.jacc.2020.08.066
  136. 136. Kotecha T, Knight DS, Razvi Y, Kumar K, Vimalesvaran K, Thornton G, et al. Patterns of myocardial injury in recovered troponin-positive COVID-19 patients assessed by cardiovascular magnetic resonance. European Heart Journal. 2021;42(19):1866-1878. DOI: 10.1093/eurheartj/ehab075
  137. 137. Sonnweber T, Sahanic S, Pizzini A, Luger A, Schwabl C, Sonnweber B, et al. Cardiopulmonary recovery after COVID-19: An observational prospective multicentre trial. The European Respiratory Journal. 2021;57(4). DOI: 10.1183/13993003.03481-2020
  138. 138. Kotanidis CP, Xie C, Adlam D, Rodrigues JC, Siddique M, Lockstone H, et al. Radiotranscriptomic analysis of perivascular adipose tissue quantifies vascular inflammation in Covid-19 from routine Ct angiograms: Stratification of "new Uk variant" infection and prediction of in-hospital outcomes. Heart. 2021;107:A177-A1A8. DOI: 10.1136/heartjnl-2021-BCS.238
  139. 139. Evans RA, McAuley H, Harrison EM, Shikotra A, Singapuri A, Sereno M, et al. Physical, cognitive, and mental health impacts of COVID-19 after hospitalisation (PHOSP-COVID): A UK multicentre, prospective cohort study. Lancet Respiratory Medicine. 2021;9(11):1275-1287. DOI: 10.1016/S2213-2600(21)00383-0
  140. 140. Fogarty H, Townsend L, Morrin H, Ahmad A, Comerford C, Karampini E, et al. Persistent endotheliopathy in the pathogenesis of long COVID syndrome. Journal of Thrombosis and Haemostasis. 2021;19(10):2546-2553. DOI: 10.1111/jth.15490
  141. 141. Chioh FWJ, Fong SW, Young BE, Wu KX, Siau A, Krishnan S, et al. Convalescent COVID-19 patients are susceptible to endothelial dysfunction due to persistent immune activation. eLife. 2021;10:e64909. DOI: 10.7554/eLife.64909
  142. 142. Gavriilaki E, Eftychidis I, Papassotiriou I. Update on endothelial dysfunction in COVID-19: Severe disease, long COVID-19 and pediatric characteristics. Journal of Laboratory Medicine. 2021;45(6):293-302. DOI: 10.1515/labmed-2021-0134
  143. 143. Ho FK, Man KKC, Toshner M, Church C, Celis-Morales C, Wong ICK, et al. Thromboembolic risk in hospitalized and nonhospitalized COVID-19 patients: A self-controlled case series analysis of a nationwide cohort. Mayo Clinic Proceedings. 2021;96(10):2587-2597. DOI: 10.1016/j.mayocp.2021.07.002

Written By

Dragana Komnenov

Submitted: 29 June 2022 Reviewed: 14 July 2022 Published: 01 September 2022