Next Article in Journal
Geranylgeraniol and Green Tea Polyphenols Mitigate Negative Effects of a High-Fat Diet on Skeletal Muscle and the Gut Microbiome in Male C57BL/6J Mice
Next Article in Special Issue
A New Concept of Associations between Gut Microbiota, Immunity and Central Nervous System for the Innovative Treatment of Neurodegenerative Disorders
Previous Article in Journal
Intersection of Diet and Exercise with the Gut Microbiome and Circulating Metabolites in Male Bodybuilders: A Pilot Study
Previous Article in Special Issue
Probiotic Ameliorating Effects of Altered GABA/Glutamate Signaling in a Rodent Model of Autism
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Gut Microbiota Dynamics in Relation to Long-COVID-19 Syndrome: Role of Probiotics to Combat Psychiatric Complications

1
Department of Physiology, College of Medicine, King Saud University, Riyadh 11682, Saudi Arabia
2
Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11495, Saudi Arabia
3
Department of Radiological Sciences, College of Applied Medical Science, King Saud University, Riyadh 11595, Saudi Arabia
4
Department of Zoology, College of Science, King Saud University, Riyadh 11495, Saudi Arabia
5
Department of Biochemistry, College of Science, King Saud University, Riyadh 11495, Saudi Arabia
6
General Administration of Nutrition, Ministry of Health, Riyadh 11595, Saudi Arabia
7
Senior Scientist, Central Research Laboratory, Female Campus, King Saud University, Riyadh 11595, Saudi Arabia
8
Department of Restorative Dental Sciences, College of Dentistry, King Saud University, P.O. Box 62645, Riyadh 11595, Saudi Arabia
*
Author to whom correspondence should be addressed.
Metabolites 2022, 12(10), 912; https://doi.org/10.3390/metabo12100912
Submission received: 13 August 2022 / Revised: 11 September 2022 / Accepted: 19 September 2022 / Published: 27 September 2022

Abstract

:
Increasing numbers of patients who recover from COVID-19 report lasting symptoms, such as fatigue, muscle weakness, dementia, and insomnia, known collectively as post-acute COVID syndrome or long COVID. These lasting symptoms have been examined in different studies and found to influence multiple organs, sometimes resulting in life-threating conditions. In this review, these symptoms are discussed in connection to the COVID-19 and long-COVID-19 immune changes, highlighting oral and psychiatric health, as this work focuses on the gut microbiota’s link to long-COVID-19 manifestations in the liver, heart, kidney, brain, and spleen. A model of this is presented to show the biological and clinical implications of gut microbiota in SARS-CoV-2 infection and how they could possibly affect the therapeutic aspects of the disease. Probiotics can support the body’s systems in fighting viral infections. This review focuses on current knowledge about the use of probiotics as adjuvant therapies for COVID-19 patients that might help to prevent long-COVID-19 complications.

Graphical Abstract

1. Introduction

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the pathogen causing coronavirus disease 2019 (COVID-19), has produced morbidity and mortality at an extraordinary rate worldwide [1]. Scientific and clinical proof is evolving on the sub-acute and lasting effects of COVID-19, which can disturb various organ systems [2]. Primary reports propose long-term effects of SARS-CoV-2 infection, such as fatigue, cognitive disturbances, chest pain, dyspnea, arthralgia, and decline in quality of life [3,4]. In 2021, the finding of an online survey conducted from 6 September 2020 to 25 November 2020 of 3762 participants with confirmed, and 2742 participants with suspected, infection suggests that the morbidities of COVID-19 have been greatly overlooked. They reported that after seven months, many patients had not yet recovered, still suffered from multisystem disabilities, and had not returned to their daily functioning and baseline health [5]. Cellular injury, a tough innate immune response with remarkably high pro-inflammatory cytokine production, and a pro-coagulant state triggered by SARS-CoV-2 infection may contribute to these complications [6,7].
Liu et al. [8] classified the complications of long COVID-19 into respiratory (runny nose, nasal congestion cough, sputum, and shortness of breath), neurological (loss of taste, loss of smell, dizziness, anxiety, headache, cognitive problems, sleep disturbance, poor memory, depression, and blurred eye), gastrointestinal (diarrhoea, nausea, abdominal, and epigastric pain), dermatological (hair loss), musculoskeletal (muscle and joint pain), and fatigue [8].
Cumulative evidence has revealed that gut dysbiosis is associated with the severity of COVID-19 infection and long-lasting multisystem complications after disease recovery [9]. Several studies have made significant contributions to understanding the gastrointestinal (GI) tract in the context of long-COVID-19 syndrome. This is related to the much higher expression of SARS-CoV receptors (ACE2) in the GI tract and the impairment of gut microbiota in subjects infected with SARS-CoV-2 [10,11]. Patients with COVID-19 showed significant alterations in fecal microbiomes compared with non-COVID-19 controls, characterized by a higher abundance of opportunistic pathogens and lower growth rates of healthy or good bacterial strains [11,12]. Several gut commensals with known immunomodulatory effects, such as Faecalibacterium prausnitzii, Eubacterium rectale, and bifidobacteria, were found to be lower in the fecal samples of COVID-19 patients and remained low up to 1 month after disease recovery [13].
Zuo et al. [12] reported that the SARS-CoV-2 virus load in fecal samples of patients is inversely correlated with Bacteroides dorei, B. thetaiotaomicron, B. massiliensis, and B. ovatus [12]. In this context, it is exciting that Bacteroides species can downregulate intestinal ACE2 expression when monocolonized in the intestines of germ-free mice [14]. In particular, B. thetaiotaomicron, a dominant anaerobe commensal bacterium in the human gut, is well-known to have anti-inflammatory properties and ameliorate mucosal barrier function in rats with chemically induced colitis [15,16]. Collectively, a reduction in B. thetaiotaomicron could induce ACE2 expression and reduce anti-inflammatory activity, which may enhance SARS-CoV-2 infectivity and intestinal/systemic inflammatory tone.

2. Oral Health as Contributor to Long COVID-19

It was observed that the severity of COVID-19 symptoms significantly increased in patients with poor oral health and decreased in those with good oral health status. Moreover, the correlation of oral health with recovery period and C-reactive protein (CRP) values is significantly inverse, showing that poor oral health is correlated with increased values of CRP and delayed recovery period [17]. Promoting optimal oral health and raising oral (self) care awareness among the public via oral health professionals is, thus, encouraged [18,19].
After recovering from the acute phase of COVID-19 and being discharged from hospital care, it is more important to emphasize how to achieve oral health for people in an outpatient setting in a way that is both applicable and will not result in care-related complications [20]. Gherlone et al. (2021) [21] noted the development of changed taste and smell, salivary gland ectasia, white tongue, dry mouth, facial muscle weakness and dysesthesia, oral ulcers, temporomandibular disorder, and other new abnormalities in a population of people who had had COVID-19 [21]. According to many observations, the symptoms of long COVID-19 in the oral cavity are associated with a decrease in the body’s immunity, higher stress, and the lower general health of the patient [22,23].
Patients with COVID-19 often undergo intubation, aided exterior ventilation, and tracheostomy [24]. These processes result in hyposalivation, which worsens various pre-existing grievances in the oral cavity and can induce bacterial secondary pneumonia [25]. SARS-CoV-2 appears to cause tropism for nerves and damage to sensory neurons has been postulated in the repeated occurrence of anosmia [26]. Neuronal damage may also disturb facial muscle tone and weaken the secretory function of salivary glands. Gherlone et al. (2021) [21] reported that the examination of the oral cavity of COVID-19 patients after effective viral clearance proved the direct cytopathic action of the virus on infected cells of the oral tissues. It is more probable that oral associations arise as a consequence of the patient’s inflammatory response, which is responsible for most morbidity and mortality in COVID-19 [21]. In agreement with a role of the early innate immune response, Zangrillo et al. (2020) [24] found a highly significant relationship between salivary gland ectasia, the levels of CRP, a marker of systemic inflammation, and lactate dehydrogenase (LDH), a biomarker of general necrosis, at the beginning of clinical symptoms. LDH also acted as an independent predictor of salivary gland ectasia in multivariate analysis. LDH and CRP have been suggested as measures of COVID-19 severity [24]. Therefore, it is interesting to highlight the association of salivary gland abnormalities with long-COVID-19 complications. Interestingly, anti-SARS-CoV-2 antibodies were found to be still detectable in COVID-19 patients’ saliva for at least three months after symptom onset [27,28], indicating the involvement of the oral cavity as an immune site during COVID-19. Therefore, it is recommended to perform an extensive intraoral examination in recovered COVID-19 patients to find any related oral manifestations.

3. Role of Gut Microbiota–Organ Axis in Long-COVID-19 Multi-Organ Dysfunction

3.1. Gut–Liver Axis and Long COVID-19

The gut–liver axis refers to the bidirectional pathway between the gut and its microflora and the liver, which is based on the integration of signals produced by genetic, dietary, and environmental influences. This mutual interaction is established by the portal vein, through which gut-derived products are directly transported to the liver and the liver feedback rout of bile and antibody discharge to the gut (Figure 1).
Two recent studies suggested that metabolic-dysfunction-associated fatty liver disease (MAFLD) is a major risk factor for progression to severe and long-lasting COVID-19. Both studies prove that patients with signs of MAFLD had a higher risk of respiratory disease progression than patients without MAFLD, with a much higher risk in younger than older COVID-19 patients [29,30]. Assante et al. (2021) [31] suggested that the increased risk observed in patients with MAFLD could be related to the effect of SARS CoV-2 infection on the gut, which worsens intestinal permeability and mucosal inflammation, thus, exacerbating systemic immune dysfunction as a feature of severe COVID-19 [31]. Certainly, this process may also clarify the higher risk for COVID-19 progression in obesity, T2D, and even IBD, which are associated with altered gut microbiota, mucosal inflammation, and increased intestinal permeability.
Numerous studies have stated that GI symptoms, such as diarrhea, vomiting, and abdominal pain, are common in patients with COVID-19 and that the severity of GI symptoms increases in coincidence with respiratory problems and liver dysfunction [32,33]. Additionally, ACE-2 SARS CoV-2 receptors have been found to be expressed on enterocyte cells, as the high levels of SARS CoV-2 viruses in feces suggest that the gut is a plausible site of viral infection and inflammation. The trans-membrane serine protease 2 that is used for SARS-CoV-2 entry is also widely expressed in gut cells [34,35]. Based on this, the increased expression of viral entry receptors in the GI tract, as well as the early onset of GI symptoms, implies that GI abnormalities may result from the direct viral worsening of leaky gut, rather than being a consequence of a secondary immune–pathogenic response to the upper respiratory tract infection.
The clinical presentation of GI symptoms that are positively correlated with biomarkers of liver dysfunction supports the concept of the increased transmission of pathogen-associated molecular patterns (PAMPs) to the liver [32].
SARS-CoV-2 infection disrupts the gut barrier and leads to the elevation of systemic bacterial lipopolysaccharide and peptidoglycan and it serves to enhance systemic inflammation. Therefore, leaky gut and microbial dysbiosis could contribute to the development of cytokine storm in patients severely ill with COVID-19.
Based on this, treatments that have been developed to treat leaky gut, such as probiotics and prebiotics for gut mucosal protection/regeneration, could minimize the number of patients with MAFLD/obesity/T2D that progresses to severe and long-lasting COVID-19. Moreover, drugs that disturb intestinal microbiota, such as antibiotics, should be avoided during SARS CoV-2 viral infection.

3.2. Gut–Heart Axis in Long COVID-19

It is well documented that gut microbiota alterations and reductions in gut bacterial diversity are common in patients with heart failure and coronary artery disease [36].
A dysfunctional gut barrier could ease the passive leakage of bacterial products, among which is the passage of pro-inflammatory lipopolysaccharides (LPSs) to the blood, which could contribute to systemic inflammation through inflammasome activation. This has been proven through the remarkable increase in plasma levels of LPS binding protein (LBP) as a predictive biomarker of high cardiovascular risk in old-aged men [37]. Interestingly, gut leakage and inflammasome activation have been found to be positively correlated with troponin as a marker of myocardial damage.
In an attempt to understand the role of the gut microbiota–heart axis in long-COVID-19 syndrome, it is interesting to mention that a considerable percentage of hospitalized COVID-19 patients have cardiac problems [38]. Earlier cardiovascular disease (CVD) and risk factors for CVD, such as obesity, appear to be key risk factors for developing severe and long-lasting COVID-19 complications [38,39]. Yet, a high proportion of COVID-19 patients have cardiac involvement without previous CVD [40].
Cardiac issues have also appeared as a substantial and life-threatening problem in COVID-19 patients, ranging from myocardial infarction (MI) and myocarditis to pulmonary hypertension with cardiac stress [41,42]. The mechanisms underlying this cardiac involvement are not clear. Angiotensin converting enzyme (ACE2) is expressed in several organs and, in addition to the lung, heart, and kidney tissues, ACE2 is also expressed in the gut, where ACE2 expression areas in enterocytes may serve as sites for SARS-CoV-2 entry and prompt gut infection [43]. The downregulation of the anti-inflammatory and cardio-protective angiotensin (AT)-1-7 pathway, secondary to the downregulation of ACE2, the SARSCoV-2 receptor, directs the infection of the myocardium through ACE2-expressing cardiac cells, leading to cardiac inflammation [44] (Figure 2).
Hoel et al. (2021) [45] suggested that long-term follow-up with cardiac imaging, in combination with microbiota analyses from the gut compartment, represent the necessary next steps to further test the potential impact of the gut–heart axis in long-COVID-19 patients [45].

3.3. Gut–Kidney Axis in Long COVID-19

The pathogenic interconnection between the gut microbiota and kidney diseases is referred to as the gut–kidney axis and it appears to be involved in a wide range of clinical manifestations, such as chronic kidney disease (CKD), acute kidney injury (AKI), and hypertension. In the case of leaky gut, the passage of viable bacteria frequently occurs from the gut to other extra-intestinal locations, such as the kidneys. This bacterial translocation may be concomitant with dysbiosis, the overgrowth of pathogenic bacteria, and a low host immune system [46,47]. The gut microbiota produces many toxins and uremic solutes, such as p-cresyl sulfate (PCS), indoxyl sulfate, and trimethylamine (TMA) N-oxide, in the case of chronic kidney disease (CKD). On the other hand, increasing urea levels could lead to alterations in the gut microbiota [48] (Figure 3). Uremic toxins may cause fatigue, mineral bone disorders, neurological disorders, and cardiovascular impairment in CKD patients [48].
In an attempt to understand the role of gut dysbiosis in long-lasting kidney problems associated with COVID-19, acute kidney injury (AKI) is commonly addressed as a complication among patients with COVID-19. In addition to pre-existing CKD being associated with severe illness or death in COVID-19 [49], it is noteworthy to address the different pathways through which SARS CoV-2 can access the kidney through the AEC2 receptors and induce clinical manifestations. It is broadly accepted that the virus can directly enter the kidneys and replicate, leading to dysfunction [50], and that it affects the kidneys through local disturbance in the renin–angiotensin–aldosterone system’s (RAAS) homeostasis [51]. Kunutsor and Laukkanen (2020) [52] reported that groups with higher prevalence of pre-existing CKD might be prone to higher incidences of AKI [52]. Emerging evidence also suggests that renal manifestations of COVID-19 are associated with increased risks of long-lasting severe COVID-19-related kidney complications [53].
In dysbiotic COVID-19 patients, the beneficial microbiota (primarily Bifidobacterium and Lactobacillus) disappear gradually and a drop in SCFAs and bile acid levels is observed due to the microbiota alteration and pathogen domination [9,54]. SCFAs and, specifically, butyrate are important energy sources for colonocytes [55] and also play an important role in epithelial integrity. Moreover, the activation of the SCFA receptor GPR109A is linked to the suppression of several proinflammatory mediators [56]. This might explain the remarkable long-lasting complications seen in COVID-19 patients. The authors of [57] reported a decrease in the anaerobic microflora in patients with CKD, while an increase in the aerobic microflora background [58] can be observed, with a predominance of Enterobacteriaceae [59]. All these mechanisms could explain the long-lasting kidney complications in some COVID-19 patients.
Monitoring kidney function during hospitalization for COVID-19 could help in the identification of patients at high risk for worse consequences, aiding in early and more effective intervention.

3.4. Gut–Brain Axis in Long COVID-19

Although the major clinical presentations of COVID-19 are related to the respiratory system, they can also distress the brain, initiating acute cerebrovascular and intracranial infections. About 35% of patients and up to 85% of those who become severely ill report neurological symptoms, including headache, dizziness, myalgia, or loss of taste and smell [60].
There are numerous mechanisms through which COVID-19 infection may lead to neurological disorders, as well as structural and functional alterations in the brain (Figure 4). Cognitive troubles are amongst the most commonly reported symptoms, affecting between 10 and 25% of COVID-19 patients and presenting as chronic illness post-SARS-CoV infection [61]. The authors found a consistent pattern of memory deficits in those that had experienced COVID-19 infection, with deficits increasing with the severity of self-reported ongoing symptoms. Moreover, they reported that fatigue/mixed symptoms during the initial illness and ongoing neurological symptoms were predictive of cognitive performance.
There is accumulating evidence that COVID-19 is associated with neural damage, mostly in the presence of neurological symptoms [55,62]. Post-mortem studies of patients who have died from COVID-19 show indications of ischemic injury and evidence of neuro-inflammation as the etiological mechanism [63]. Numerous studies have recorded functional as well as structural deformities, such as hemorrhagic injuries and epileptiform discharges, in different brain areas [64,65].
In relation to neuro-inflammation, the role of glutamate excitotoxicity should be considered as a contributor to long-lasting COVID-19-assocated neurological symptoms. Ahmed et al. (2020) [66] reported that SARS-CoV infection induces a significant increase in the production of pro-inflammatory cytokines and neuronal degeneration as an outcome of glutamate excitotoxicity [66]. Simply, glutamate, as a primary excitatory neurotransmitter in the nervous system, is mainly produced by neurons and discharged in the synaptic cleft, after which it binds to the ligand-dependent AMPA receptor (alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionoc acid receptor). This helps the access of sodium ions and the passage of the nerve impulse through the post-synaptic neuron, leading to the activation of the N-methyl-D-aspartate receptor (NMDA), which induces the entrance of calcium ions. During the infection of neurons caused by coronavirus, microglial cells produce pro-inflammatory cytokines (TNF-α, IL-1β, and IL-6), which downregulate the glutamate transporter 1 (GLT-1) on astrocytes and pre-synaptic neurons. This will decrease the rate of the efficient re-uptake of glutamate and cause an imbalance in glutamate/GABA neurotransmitters and the overstimulation of NMDA receptors. These events disturb the control of glutamate homeostasis and the overproduction of glutamate in the synaptic cleft induces neuronal excitotoxicity with a significant entrance of calcium, which eventually leads to nerve cell degeneration and loss (Figure 4).

Neuropsychiatric Complications and Gut–Brain Axis

Recent studies have reported that more than one-third of positive patients develop neurological and neuropsychiatric symptoms in the early stages of infection, but other reported cases show such symptoms even after the resolution of COVID-19 [67]. White matter and brain abnormalities, such as hyperintensities and hypodensities, hemorrhages, and infarcts, were found in 34% of the COVID-19 population [55,68]. Severe infections can contribute to delayed neurological central nervous system (CNS) complications. Recent studies have suggested the possible neuroinvasive mechanisms of COVID-19 leading to a neurotropically induced “cytokine storm”, which releases a large number of inflammatory markers [69] and could reactivate immune-mediated processes [70]. Neuroinflammation and any axonal damage are due to viral replication, which causes delayed self-reactive T-cell suppression [71,72]. Different mechanisms suggest that the first approach is a direct viral injury to the CNS via blood circulation or the cribriform plate [73], leading to encephalitis [73,74]. The second stage of CNS damage results from peripheral demyelination [75], which is a host immune response that follows acute infection. Indirect injury is the third mechanism, resulting from systemic circulatory dissemination [76]. The last suggested mechanism is cytokine release as a result of immune response overactivation [77]. These mechanisms could induce delayed nervous system damage and neurological complications, known as long COVID-19 [78]. More reports showing delayed neuro-demyelination following COVID-19 infection [79], confirmed by animal studies, support the theory that SARS-CoV-2 infection crosses the blood–brain barrier and causes acute or delayed CNS demyelination and/or axonal damage [80]. Brain structural abnormalities in postmortem COVID-19 suggest hemorrhagic and posterior reversible encephalopathy syndrome (PRES) [81]. Acute necrotizing encephalopathy (ANE) is a rare COVID-19 complication that has been related to intracranial cytokine storms [82].
Systematic follow-ups in published studies have identified that post-COVID-19 patients after hospitalization suffer from physical and psychological symptom burdens [83]. Psychiatric symptoms are common after a coronavirus infection; many reports have found SARS-CoV-2 infection to be associated with a high prevalence of anxiety [84,85] and patients with these symptoms tend to have more somatic and pain complaints [86]. More recently, attention has focused on many other mental disorders, such as fatigue [67,83], panic disorders, and pain and depressive disorders [87]. Fatigue prevalence in post-COVID-19 patients, at 3–5 months, was reported in 64.2% of cases [88]. Central factors influencing post-COVID-19 fatigue include the levels of dopamine and serotonin [89], which act as neurotransmitters, and many other factors, such as severe changes in axonal conduction velocity or neuronal excitability [90]. Longitudinal studies have found that dysgeusia and hyposmia symptoms are associated with the development of cognitive impairment (CI) after COVID-19 [91,92], with the main involvement of the executive functions, such as flexible thinking, planning, and information processing.
A considerable amount of literature has been published on the neuroimaging findings of multiple brain regions involved in both acute and long COVID-19. 18F-FDG-PET imaging following COVID-19 revealed hypometabolism in the insula, para-hippocampal, fusiform gyri [93], olfactory gyrus, and connected limbic/paralimbic regions [94], as well as frontoparietal hypometabolism [95]. Cortical hypometabolism could be a consequence of white matter (WM) or brainstem damage [95]. Frontoparietal hypometabolism has also been associated with multiple neurodegenerative disorders, which may cause a decline in cognitive function in long COVID-19 [96].
Structural MRI studies investigated corticospinal tract and corpus callosum hyperintensities following COVID-19 and found that these are more suggestive of Neuromyelitis-Optica spectrum disorder (NMOSD) [97,98]. The most prominent observation was deep WM abnormalities [99] with diffuse subcortical changes [79,100]. A grey matter (GM) loss in brain regions directly linked to the primary olfactory and gustatory systems can explain the cerebral spread of SARS-CoV-2 [92,101]. Another study yielded opposing results, with higher GM volumes in olfactory cortices, hippocampi, insulas, some regions of the secondary somatosensory and primary auditory cortices, and cingulate gyrus, as well as a general decline in white matter seen via diffusion tensor imaging (DTI), but an increase via fractional anisotropy (FA). The case studies have revealed many other neurological manifestations, such as acute necrotizing encephalomyelitis (ANE) following COVID-19, as a case of an acute CNS injury involving hemorrhage and cavitation [82]. The MRI characteristics of ANE include hyperintensities, with internal hemorrhage on T2-weighted fluid-attenuated inversion recovery (FLAIR) and a ring of contrast enhancement on enhanced images [82].
However, there have also been reports of neuroplasticity, where acute neurological symptoms and CI were resolved at some point from a matter of days to after 10 months [102,103]. There is an urgent need to follow-up COVID-19 survival in order to understand the potential and real long-term consequences, especially for extrapulmonary sequelae.

3.5. Gut–Spleen Axis in Long COVID-19

Recently, interest has emerged in the role of spleen function during COVID-19 infection. At the beginning of the epidemic, asplenia as the anatomic absence of the spleen, or spleen dysfunction secondary to disease states, was found to confer a mortality risk comparable to other known risk factors. SARS-CoV-2 was shown to induce a particular tropism for the spleen, possibly through the ACE-2 receptor. Spleen dysfunction was thought to contribute, along with other mechanisms, to B and T cell lymphopenia, which is a typical feature in COVID-19 post-infection [104,105]. Based on the higher abundance of lipopolysaccharides (LPSs) as a product of the Gram-negative bacteria in splenectomized or spleen-dysfunctional patients compared with healthy controls, an altered gut microbiota composition as a major cause of elevated plasma LPS could be related to long-COVID-19 complications [106].

4. Immune Perspective on Long COVID-19 and Proposed Mechanisms

There have been increasing numbers of case reports and cited observations documenting chronic symptoms lingering past the actual acute stage of COVID-19 infection [3]. These symptoms are multi-systematic and can be respiratory, cardiovascular, neurological, or hormonal [3]. There are several definitions for long COVID-19 that take into consideration the time it takes to manifest (Table 1). Nonetheless, the general consensus is that long COVID-19 is confirmed when symptoms persist about three months after the clearance of the acute COVID-19 infection [107]. These chronic symptoms of long COVID-19 have been found to include: dyspnea as a common complaint, alongside fatigue, chest pain, palpitations, tachycardia, orthostatic intolerance, headache, depression, insomnia, cognitive impairment, memory loss, changes in taste and smell, joint pain, myalgia, and gastrointestinal symptoms [104,105]. Several studies have investigated the causes of such chronic symptoms of COVID-19 in order to assess the pathophysiology. Mechanisms have been proposed for long COVID-19, but this is still a growing field of interest. More work is needed to clarify its seemingly complicated pathophysiology, as different systems are usually involved based on the clinical symptoms presented.
Examining long-COVID-19′s pathophysiology, immune dysregulation is implicated as the trigger causing other systems to be involved, as the innate immune response is overactivated. This not only disturbs other immune parameters, but also other systems and organs [106,108]. For instance, an overactivated immune response dysregulates the RAAS (renin angiotensin aldosterone system), which has a direct influence on overall body homeostasis [108,109]. This overactivated innate immune response is limited to thrombi formation primarily in micro vessels [108,109].
That said, despite other studies that focused on other causes of long COVID-19, considering the neurological and gut microbiota links, this can only be comprehensively understood once it is considered in the immune context, since it is an immune-based dysregulation condition. Examples of such studies that have analyzed other factors include those that have investigated the neurological symptoms of long COVID-19, in order to understand their cause [105]. One suggested cause is the slow regeneration of neurons as the damage to the brain stem can be long-lasting, leading to the neurological symptoms observed in long COVID-19 [105].
Nonetheless, many studies have investigated the immunological pathophysiology of long COVID-19, given that it is a viral infection compromising the immune system [110]. For instance, CD8 T cells, which are cytotoxic in nature and mediate adaptive immunity, have been found infiltrating the lungs of long-COVID-19 patients, which supports the diagnosis of T cell dysfunction [110,111]. A mechanism called bystander activation has been proposed for cases in which antigen-presenting cells present antigens to auto-reactive T cells [110,111]. Another example of this auto-reactive T cell dysfunction is the presentation of autoimmune thyroid dysfunction in 15–20% of patients with COVID-19 [112,113]. Another immune cause linked to long COVID-19 is B cell activity dysfunction, as 52% of samples had more severe clinical outcomes, with anti-phospholipid autoantibodies presenting signaling neutrophil hyperactivity [114]. Autoantibodies were also found against interferon, cyclic citrullinated peptides, neutrophils, connective tissues, and the cell nucleus in 10–50% of COVID-19 patients exhibiting long-term symptoms [115]. These findings implicate B cell involvement in long COVID-19’s pathophysiology. This increase in autoantibodies in chronic COVID-19 has been linked to an increased incidence of autoimmune diseases post-COVID-19 infection, such as Sjogren syndromes, lupus erythematosus, and rheumatoid arthritis [116]. More studies have investigated this link and one proposed mechanism is the lymphocytopenia present in severe COVID-19. Nevertheless, many reports have not found such a link between COVID-19 severity and long-COVID-19 symptoms, which further complicates the understanding of the immune pathophysiology of the condition [3,107].
More studies have investigated the association between long COVID-19 and immune cellular activity [117,118]. Some reports may appear contradictive but taking into consideration the different definitions for long COVID-19, which entail different timelines, can explain the variation and help us to understand the immune pathophysiology. For instance, some studies report that long COVID-19 is linked to the unresolved hyperinflammation exerted by the renewed B and T lymphocytes during infection [117]. This hyperinflammation contributes to long-COVID-19 manifestation [117]. Another hypothesis that has been proposed is that the continuous shedding of the SARS-CoV2 virus is due to the diminished B and T cell count and function as the condition lingers [119]. This inflammatory dysfunction has been observed to take place about 2–6 weeks post infection, which certainly suggests adaptive immunity dysregulation [120]. Such inflammatory symptoms may present along with increased levels of pro-inflammatory markers (e.g., interleukin-6 (IL-6), C reactive protein (CRP), ferritin, and D-dimer), with no respiratory problems but with neurological and cardiovascular as well as gastrointestinal symptoms [119,120]. To confirm this persistent inflammation in long COVID-19, some researchers have performed radiological assessments of the implicated tissues and bones for Fluorine 18 fluorodeoxyglucose (FDG). FDG uptake identifies the foci of infection and correlates with the metabolic rate of the cells [121]. FDG positron emission tomography (PET) can indicate disease severity and spread and help correlate findings with treatment response [121]. Patients with long COVID-19 have shown increased and chronic inflammation in bones and vessels at least 4 weeks post-infection [119,121]. That said, it is important to note that although different studies have found increased levels of pro-inflammatory markers in long-COVID-19 patients, others have not found the same correlation [119,121]. Such findings explain the current lack of a resolved understanding of long COVID-19, which varies in terms of timeline, manifestation, symptoms, and, therefore, treatment [122]. This variation in clinical presentations has not been found to be significantly correlated with age, acute infection severity, or gender and race, which presents health practitioners with problems when predicting the condition or delivering a prognosis [123].
Nevertheless, attempts have been made to find out about associations with long COVID-19 that can predict a chronic pathology. The Institute for Systems Biology conducted a longitudinal mutli-omic systems biology investigation of 200 subjects 2–3 months post-acute COVID-19 infection [124]. The findings of this study reveal some factors significantly associated with long COVID-19, which include the following: pre-existing diabetes mellitus, high-level SARS-CoV-2 viremia, autoantibodies (including those targeting the interferons), and Epstein–Barr virus (EBV) reactivation during acute infection [124]. This study found that if these factors are present, then it is highly likely that COVID-19 patients will develop long COVID-19. Among the most devastating long-COVID-19 symptoms are the cardiovascular problems that develop following acute infection. One report cited that immunothrombosis and venous thromboemboli are critically dangerous in long-COVID-19 patients. This certainly makes examining predictive parameters important to preventing possible deaths due to such symptoms. The immune system and the hemostatic system are linked, allowing multiple factors to cause immunothromobosis in long COVID-19 [3,107,110,111]. In the occurrence of this immunothrombosis, the following factors have been implicated: endothelial inflammation, microthrombi formation, and the disruption of the intercellular junction [125]. Increasing levels of cytokines during the infection, along with activated platelets, are found to be associated with coagulopathy in long COVID-19, as it creates the proinflammatory environment of immunothrombosis, which is primarily a host defense mechanism that becomes altered, forming immunologically mediated thrombi that influence the microvasculature [125]. Dysfunctional endothelial cells (ECs) disturb the control of coagulation and anti-coagulation systems, which ultimately results in the increased coagulation seen in COVID-19 and long-COVID-19 conditions [126]. In addition to this, tissue factor (TF) is found to be released by ECs under inflammatory conditions, which, once in the blood stream, activates the coagulation system [127,128]. The same has been found in monocytes during inflammation and both result in increased mortality [129]. The different factors implicated in the immune dysregulation causing long COVID-19 call for more studies to investigate the stages at which each factor is important and when it combines with other parameters to manifest the pathology. Immune pathophysiology is essential to not only understanding the condition, but also to managing it efficiently.
Table 1. Proposed definitions and categories of long COVID-19 (2).
Table 1. Proposed definitions and categories of long COVID-19 (2).
DescriptionTerms References
Symptoms lasting 4 weeks post-acute infection Long COVID[104]
Symptoms 3 months post-acute infections Post COVID[130]
Long-term COVID-19 is said to be cyclical, progressive, and multiphasicLong COVID[131]
Multi-organ indications that continue for months after acute COVID-19Long-hauler COVID-19[132,133]
Chronic COVID syndromeLong-COVID
Symptoms lasting more than 100 daysLong-haul COVID[134]
Long-tail COVID
Symptoms lasting more than 2 monthsLong COVID[135,136]
Symptoms to last more than 4 weeksLate sequelae of SARS-CoV-2 infection[137,138]
Long-haulers
Long-COVID
Symptoms persist more than 4 weeks after acute infection diagnosisPost-acute COVID-19 syndrome[130]
Symptoms continue for 5–12 weeksAcute post-COVID symptoms[139]
Symptoms continue for 12–24 weeksLong post-COVID symptoms
Symptoms continue for >24 weeksPersistent post-COVID symptoms
Symptoms continue for 1–3 months Post-acute COVID-19[106,140]
On-going symptomatic COVID-19
Symptoms continue more than 3 months Chronic COVID-19
Long COVID
Post-COVID-19 syndrome

5. Potential Preventive and/or Therapeutic Effects of Prebiotic- and Probiotic-Related Strategies in COVID-19

Probiotics are live microbes that confer beneficial effects on the host, when administered in appropriate quantities [141]. Prebiotics are defined as “substrates that are selectively utilized by host microorganisms conferring a health benefit [142]”. Interventions targeting the gut microbiome may have systemic beneficial effects in patients with COVID-19 [143]. Evidence indicates that viral infections in the respiratory tract initiate a disturbance in the gut microbiota; in patients with COVID-19, the gut microbiota is transformed via severe hypoxemia [144,145].
Probiotics and prebiotics are the two components in our diet that can affect the microbiome. Nutritional status and diet play a crucial role in COVID-19, predominantly owing to the bidirectional interaction between the lungs and the gut microbiota [146]. Figure 5 depicts the interactions between the human gut and lungs and the potential positive immune responses triggered by probiotics and prebiotics. Both can enhance the phagocytic activity of macrophage cells [147], balance T cell immunity in favor of a more regulatory status [148], increase the activity of salivary IgA [149], and exert immunoregulatory extracellular and intracellular functions through the production of SCFAs as important signaling molecules [150]. The manipulation of the gut microbiota using prebiotics and probiotics represents a promising therapeutic approach to lung diseases in clinical research [147] (Figure 5).
The China National Health Commission and National Administration of Traditional Chinese Medicine Guidelines have recommended the consumption of probiotics along with conventional therapies in patients with severe COVID-19 infection, in order to improve the balance of intestinal flora and prevent secondary bacterial infections [148].
Probiotics may alter the composition of the gut microbiota and play a crucial role in maintaining the ecosystem of the gut microbiota [149]. Although the immune responses caused by bacteria are relatively different from those caused by the virus, numerous clinical studies have concluded that probiotics contribute to the fight against COVID-19 [150]. Apart from the improvements in the intestinal microbial balance, recent evidence indicates that probiotics can also confer beneficial effects on the host through modulating host immune functions [151]. Some studies have reported on the potential of probiotics to interact with angiotensin converting enzyme II, a host entry receptor of severe acute respiratory syndrome coronavirus 2 (SARS-COV 2) [143]. For example, several probiotics (mainly probiotic lactic acid bacteria) have been reported to release peptides with high affinity for the angiotensin-converting enzyme during milk fermentation [152]. Similarly, probiotics may also improve respiratory tract infections through the angiotensin converting enzyme II pathway [153]. Probiotics also improve the levels of natural killer (NK) cells, type I interferons, T and B lymphocytes, and antigen-presenting cells (APC) in the lung immune system [144]. NK cells play an important role in the early immune response against viral infections, predominantly via the clearance of viral infections. A previous study demonstrated that probiotics alter the expression of interleukin-10 and reduce the expression of inflammatory cytokines [154].
A recently published study in China revealed that probiotics have the ability to moderate immunity and reduce the incidence of secondary infection in patients with COVID-19 [153]. Baud and colleagues in 2020 [144] reported the following probiotics as potentially able to reduce the burden of COVID-19: Bifidobacterium bifidum, Lactobacillus plantarum, Pediococcus pentosaceus, Leuconostoc mesenteroides, Bifidobacterium longum, Lactobacillus rhamnosus, Lactobacillus gasseri, Bifidobacterium breve, and Lactobacillus casei [144]. A previous study in China concluded that COVID-19 infection affects the intestinal microbiota balance, based on the observation of reduced counts of Bifidobacterium and Lactobacillus strains in COVID-19 patients [155]. Probiotic strains restore the gut microbiota, decrease the translocation of pathogenic bacteria across the gut mucosa, maintain a healthy gut–lung axis, and lessen the incidence of secondary bacterial infection [156]. Enterococcus sp., Bacillus sp., Bifdibacterium sp., actobacillus sp., Streptocococus sp., and Pediococcus sp. are the most frequently used species in the preparation of probiotics [157,158].
Prebiotics include polyunsaturated fatty acids, resistant starch, arabinooligosaccharides, oligosaccharides, fructans, oligosaccharides, galactomannan, psyllium, lactosucrose, lactobionic acid, and polyphenols [142,159]. Foods that contain prebiotics, such as fiber, oligosaccharides, and polyphenols, can improve the growth of bacteria [160,161]. Prebiotics modulate the gut microbiota in a similar manner to probiotics, thus, inhibiting pathogens and stimulating the immune system. Likewise, prebiotics, via direct and indirect mechanisms, confer beneficial alterations upon the immune system and the host’s health [162]. In addition, they selectively stimulate the favorable growth and enhance the activities of probiotic bacteria [163]. Prebiotics have potential effects against COVID-19 infection by enhancing the growth and survivability of probiotics. Moreover, prebiotics provide energy for the growth of probiotics [143]. Prebiotics may also have a potential effect on gastrointestinal symptoms caused by COVID-19 by blocking angiotensin-converting enzymes [145].
Prebiotics apparently reduce the levels of the proinflammatory interleukin 6, which seems to be the leading cause of the hitherto described grave prognosis of COVID-19 and improve the levels of anti-inflammatory interleukin 10 [164]. The concurrent use of prebiotics and probiotics is crucial for the treatment of COVID-19 infection [165]. However, there is limited evidence regarding the effectiveness of prebiotics in COVID-19 infection, unlike the case for probiotics.

6. Conclusions

According to the previous findings, it can be concluded that a patient’s chance of acquiring “long COVID-19” after infection with SARS-CoV-2 may be influenced by the composition of their gut flora. Thus, an altered gut microbiota or dysbiosis can act as modulators of systemic inflammatory activity and can affect different organs through the multiple gut–organ axis. Increased gut permeability, or leaky gut, allows the entrance of bacterial metabolites and toxins into the circulatory system and further worsens the systemic inflammatory response, leading to different COVID-19 complications. Thus, the adjustment of the gut microbiome with probiotics could be an alternative strategy for boosting immunity, treating COVID-19, and protecting against the development of post-acute COVID-19 syndrome.

7. Future Direction

Based on the reviewed literature, a healthy gut microbiota composition during hospitalization is associated with a more favorable clinical presentation of COVID-19. Further studies are required to explore the direct connection between gut bacterial profiles and long-COVID-19 complications and to consider microbial configurations for prognostic and therapeutic strategies in clinical practice.

Author Contributions

Conceptualization, M.F.A. and A.K.E.-A.; funding acquisition, H.A.B.; review and editing, H.A.B., H.I.A., A.R.A., M.H.D., M.A.A. and R.S.L.; writing—original draft preparation M.F.A., A.K.E.-A., H.A.B., H.I.A., A.R.A., M.H.D., M.A.A. and R.S.L.; review and editing H.A.B. All authors have read and agreed to the published version of the manuscript.

Funding

This work is funded by Research Supporting Project number (RSP2022R179) King Saud University, Riyadh, Saudi Arabia.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Dong, E.; Du, H.; Gardner, L. An interactive web-based dashboard to track COVID-19 in real time. Lancet Infect. Dis. 2020, 20, 533–534. [Google Scholar] [CrossRef]
  2. Gupta, A.; Madhavan, M.V.; Sehgal, K.; Nair, N.; Mahajan, S.; Sehrawat, T.S.; Bikdeli, B.; Ahluwalia, N.; Ausiello, J.C.; Wan, E.Y.; et al. Extrapulmonary manifestations of COVID-19. Nat. Med. 2020, 26, 1017–1032. [Google Scholar] [CrossRef] [PubMed]
  3. Carfì, A.; Bernabei, R.; Landi, F. Persistent symptoms in patients after Acute COVID-19. JAMA 2020, 324, 603–6052020. [Google Scholar] [PubMed]
  4. Huang, L.; Wang, Y.; Li, X.; Ren, L.; Gu, X. 6-month consequences of COVID-19 in patients discharged from hospital: A cohort study. Lancet 2021, 397, 220–232. [Google Scholar] [PubMed]
  5. Davis, H.E.; Assaf, G.S.; McCorkell, L.; Wei, H.; Low, R.J.; Re’em, Y.; Redfield, S.; Austin, J.P.; Akrami, A. Characterizing long COVID in an international cohort: 7 months of symptoms and their impact. EClinicalMedicine 2021, 38, 101019. [Google Scholar] [CrossRef] [PubMed]
  6. McElvaney, O.J.; McEvoy, N.L.; McElvaney, O.F.; Carroll, T.P.; Murphy, M.P.; Dunlea, D.M.; Ní Choileáin, O.; Clarke, J.; O’Connor, E.; Hogan, G.; et al. Characterization of the inflammatory response to severe COVID-19 Illness. Am. J. Respir. Crit. Care Med. 2020, 202, 812–821. [Google Scholar] [CrossRef] [PubMed]
  7. Tang, N.; Li, D.; Wang, X.; Sun, Z. Abnormal coagulation parameters are associated with poor prognosis in patients with novel coronavirus pneumonia. J. Thromb. Haemost. 2020, 18, 844–847. [Google Scholar] [CrossRef] [PubMed]
  8. Liu, Q.; Mak, J.W.Y.; Su, Q.; Yeoh, Y.K.; Lui, G.C.Y.; Ng, S.S.S.; Zhang, F.; Li, A.Y.L.; Lu, W.; Hui, D.S.; et al. Gut microbiota dynamics in a prospective cohort of patients with post-acute COVID-19 syndrome. Gut 2022, 71, 544–552. [Google Scholar] [CrossRef]
  9. Chen, Y.; Gu, S.; Chen, Y.; Lu, H.; Shi, D.; Guo, J.; Wu, W.R.; Yang, Y.; Li, Y.; Xu, K.J.; et al. Six-month follow-up of gut microbiota richness in patients with COVID-19. Gut 2022, 71, 222. [Google Scholar] [CrossRef]
  10. Ng, S.C.; Tilg, H. COVID-19 and the gastrointestinal tract: More than meets the eye. Gut 2020, 69, 973. [Google Scholar] [CrossRef] [Green Version]
  11. Zuo, T.; Zhang, F.; Lui, G.C.; Yeoh, Y.K.; Li, A.Y.; Zhan, H.; Wan, Y.; Chung, A.C.K.; Cheung, C.P.; Chen, N.; et al. Alterations in gut microbiota of patients with COVID-19 during time of hospitalization. Gastroenterology 2020, 159, 944–955.e8. [Google Scholar] [CrossRef]
  12. Zuo, T.; Liu, Q.; Zhang, F.; Lui, G.C.Y.; Tso, E.Y.; Yeoh, Y.K.; Chen, Z.; Boon, S.S.; Chan, F.K.L.; Chan, P.K.S.; et al. Depicting SARS-CoV-2 faecal viral activity in association with gut microbiota composition in patients with COVID-19. Gut 2021, 70, 276–284. [Google Scholar] [CrossRef]
  13. Yeoh, Y.K.; Zuo, T.; Lui, G.C.; Zhang, F.; Liu, Q.; Li, A.Y.; Chung, A.C.; Cheung, C.P.; Tso, E.Y.; Fung, K.S.; et al. Gut microbiota composition reflects disease severity and dysfunctional immune responses in patients with COVID-19. Gut 2021, 70, 698–706. [Google Scholar] [CrossRef]
  14. Geva-Zatorsky, N.; Sefik, E.; Kua, L.; Pasman, L.; Tan, T.G.; Ortiz-Lopez, A.; Yanortsang, T.B.; Yang, L.; Jupp, R.; Mathis, D.; et al. Mining the human gut microbiota for immunomodulatory organisms. Cell 2017, 168, 928–943. [Google Scholar] [CrossRef]
  15. Kelly, D.; Campbell, J.I.; King, T.P.; Grant, G.; Jansson, E.A.; Coutts, A.G.; Pettersson, S.; Conway, S. Commensal anaerobic gut bacteria attenuate inflammation by regulating nuclear-cytoplasmic shutting of PPAR-γ and ReIA. Nat. Immunol. 2004, 5, 104–112. [Google Scholar] [CrossRef]
  16. Wrzosek, L.; Miquel, S.; Noordine, M.L.; Bouet, S.; Chevalier-Curt, M.J.; Robert, V.; Philippe, C.; Bridonneau, C.; Cherbuy, C.; Robbe-Masselot, C.; et al. Bacteroides thetaiotaomicron and Faecalibacterium prausnitzii influence the production of mucus glycans and the development of goblet cells in the colonic epithelium of a gnotobiotic model rodent. BMC Biol. 2013, 11, 61, PMCID:PMC3673873. [Google Scholar] [CrossRef] [PubMed]
  17. Kamel, A.H.M.; Basuoni, A.; Salem, Z.A.; AbuBakr, N. The impact of oral health status on COVID-19 severity, recovery period and C-reactive protein values. Br. Dent. J. 2021. [Google Scholar] [CrossRef]
  18. Franziska, B. Good oral health may prevent severe COVID-19 progression. Ger. Soc. Dent. Oral Med. GSDOM 2020, 22, 6527. [Google Scholar]
  19. Buunk-Werkhoven, Y.A.B.; Reyerse, E. What is the impact of oral (Public) health promotion, and of interventions for oral (self) care awareness raising and behavior change? J. Dent. Oral Disord. Ther. 2020, 8, 1–4. [Google Scholar] [CrossRef]
  20. France, K.; Glick, M. Long COVID and oral health care considerations. J. Am. Dent. Assoc. 2022, 153, 167–174. [Google Scholar] [CrossRef]
  21. Gherlone, E.F.; Polizzi, E.; Tetè, G.; De Lorenzo, R.; Magnaghi, C.; Rovere Querini, P.; Ciceri, F. Frequent and persistent salivary gland ectasia and oral disease after COVID-19. J. Dent. Res. 2021, 100, 464–471. [Google Scholar] [CrossRef]
  22. Neto, C.L.D.M.M.; Bannwart, L.C.; de Melo Moreno, A.L.; Goiato, M.C. SARS-CoV-2 and Dentistry-Review. Eur. J. Dent. 2020, 14, S130–S139. [Google Scholar] [CrossRef]
  23. Paradowska-Stolarz, A.M. Oral manifestations of COVID-19: Brief review. Dent. Med. Probl. 2021, 58, 123–126. [Google Scholar] [CrossRef]
  24. Zangrillo, A.; Beretta, L.; Scandroglio, A.M.; Monti, G.; Fominskiy, E.; Colombo, S.; Morselli, F.; Belletti, A.; Silvani, P.; Crivellari, M.; et al. Characteristics, treatment, outcomes and cause of death of invasively ventilated patients with COVID-19 ARDS in Milan, Italy. Crit. Care Resusc. 2020, 22, 200–211. [Google Scholar]
  25. Wu, C.; Chen, X.; Cai, Y.; Zhou, X.; Xu, S.; Huan, H.; Zhang, L.; Zhou, X.; Du, C.; Zhang, Y.; et al. Risk factors associated with acute respiratory distress syndrome and death in patients with Coronavirus Disease 2019 Pneumonia in Wuhan, China. JAMA Intern. Med 2020, 180, 934–943. [Google Scholar] [CrossRef]
  26. Vaira, L.A.; Salzano, G.; Deiana, G.; de Riu, G. Anosmia and Ageusia: Common Findings in COVID-19 Patients. Laryngoscope 2020, 130, 1787. [Google Scholar] [CrossRef]
  27. Isho, B.; Abe, K.T.; Zuo, M.; Jamal, A.J.; Rathod, B.; Wang, J.H.; Li, Z.; Chao, G.; Rojas, O.L.; Bang, Y.M.; et al. Persistence of serum and saliva antibody responses to SARS-CoV-2 spike antigens in COVID-19 patients. Sci. Immunol. 2020, 5, eabe5511. [Google Scholar] [CrossRef]
  28. Pisanic, N.; Randad, P.R.; Kruczynski, K.; Manabe, Y.C.; Thomas, D.L.; Pekosz, A.; Klein, S.L.; Betenbaugh, M.J.; Clarke, W.A.; Laeyendecker, O.; et al. COVID-19 serology at population scale: SARS-CoV-2-specific antibody responses in saliva. J. Clin. Microbiol. 2020, 59, e02204-20. [Google Scholar] [CrossRef]
  29. Ji, D.; Qin, E.; Xu, J.; Zhang, D.; Cheng, G.; Wang, Y.; Lau, G. Non-alcoholic fatty liver diseases in patients with COVID-19: A retrospective study. J. Hepatol. 2020, 73, 451–453. [Google Scholar] [CrossRef]
  30. Zhou, Y.J.; Zheng, K.I.; Wang, X.B.; Yan, H.D.; Sun, Q.F.; Pan, K.H.; Wang, T.Y.; Ma, H.L.; Chen, Y.P.; George, J.; et al. Younger patients with MAFLD are at increased risk of severe COVID-19 illness: A multicenter preliminary analysis. J. Hepatol. 2020, 73, 719–721. [Google Scholar] [CrossRef]
  31. Assante, G.; Williams, R.; Youngson, N.A. Is the increased risk for MAFLD patients to develop severe COVID-19 linked to perturbation of the gut-liver axis? J. Hepatol. 2021, 74, 487–488. [Google Scholar] [CrossRef] [PubMed]
  32. Pan, L.; Mu, M.I.; Yang, P.; Sun, Y.; Wang, R.; Yan, J.; Li, P.; Hu, B.; Wang, J.; Hu, C.; et al. Clinical characteristics of COVID-19 patients with digestive symptoms in Hubei, China: A descriptive, cross-sectional, multicenter study. Am. J. Gastroenterol. 2020, 115, 766–773. [Google Scholar] [CrossRef] [PubMed]
  33. Xiao, F.; Tang, M.; Zheng, X.; Liu, Y.; Li, X.; Shan, H. Evidence for gastrointestinal infection of SARS-CoV-2. Gastroenterology 2020, 158, 1831–1833.e3. [Google Scholar] [CrossRef] [PubMed]
  34. Hoffmann, M.; Kleine-Weber, H.; Schroeder, S.; Krüger, N.; Herrler, T.; Erichsen, S.; Schiergens, T.S.; Herrler, G.; Wu, N.H.; Nitsche, A.; et al. SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell 2020, 181, 271–280.e8. [Google Scholar] [CrossRef]
  35. Bertram, S.; Heurich, A.; Lavender, H.; Gierer, S.; Danisch, S.; Perin, P.; Lucas, J.M.; Nelson, P.S.; Pöhlmann, S.; Soilleux, E.J. Influenza and SARS-coronavirus activating proteases TMPRSS2 and HAT are expressed at multiple sites in human respiratory and gastrointestinal tracts. PLoS ONE 2012, 7, e35876. [Google Scholar] [CrossRef]
  36. Trøseid, M.; Andersen, G.Ø.; Broch, K.; Hov, J.R. The gut microbiome in coronary artery disease and heart failure: Current knowledge and future directions. EBioMedicine 2020, 52, 102649. [Google Scholar] [CrossRef]
  37. Roberts, L.M.; Buford, T.W. Lipopolysaccharide binding protein is associated with CVD risk in older adults. Aging Clin. Exp. Res. 2021, 33, 1651–1658. [Google Scholar] [CrossRef]
  38. Shi, Y.; Wang, G.; Cai, X.P.; Deng, J.W.; Zheng, L.; Zhu, H.H.; Yang, B.; Zheng, M.; Chen, Z. An overview of COVID-19. J. Zhejiang Univ. Sci. B 2020, 21, 343–360. [Google Scholar] [CrossRef]
  39. Wu, Y.C.; Chen, C.S.; Chan, Y.J. The outbreak of COVID-19: An overview. J. Chin. Med. Assoc. 2020, 83, 217–220. [Google Scholar] [CrossRef]
  40. Inciardi, R.M.; Lupi, L.; Zaccone, G.; Italia, L.; Raffo, M.; Tomasoni, D.; Cani, D.S.; Cerini, M.; Farina, D.; Gavazzim, E. Cardiac involvement in a patient with coronavirus disease 2019 (COVID-19). JAMA Cardiol 2020, 5, 819–824. [Google Scholar] [CrossRef]
  41. Sarfraz, Z.; Sarfraz, A.; Barrios, A.; Garimella, R.; Dominari, A.; Kc, M.; Pandav, K.; Pantoja, J.C.; Retnakumar, V.; Cherrez-Ojeda, I. Cardio-Pulmonary sequelae in recovered COVID-19 patients: Considerations for primary care. J. Prim. Care Community Health 2021, 12, 21501327211023726. [Google Scholar] [CrossRef]
  42. Stefanini, A.M.; Fidelis, T.O.; Penna, G.M.; Pessanha, G.R.G.; Marques, R.A.G.; Oliveira, D.C.D. Tomographic identification and evaluation of pulmonary involvement due to SARS-CoV-2 infection using artificial intelligence and image segmentation technique. In Bioengineering and Biomedical Signal and Image Processing; Rojas, I., Castillo-Secilla, D., Eds.; Springer International Publishing: Berlin/Heidelberg, Germany, 2021; pp. 405–416. [Google Scholar]
  43. Gheblawi, M.; Wang, K.; Viveiros, A.; Nguyen, Q.; Zhong, J.C.; Turner, A.J.; Raizada, M.K.; Grant, M.B.; Oudit, G.Y. Angiotensin-Converting enzyme 2: SARS-CoV-2 receptor and regulator of the renin-angiotensin system: Celebrating the 20th anniversary of the discovery of ACE2. Circ. Res. 2020, 126, 1456–1474. [Google Scholar] [CrossRef]
  44. Verdecchia, P.; Angeli, F.; Reboldi, G. Angiotensin-converting enzyme inhibitors, angiotensin II receptor blockers and coronavirus. Cell 2020, 38, 1190–1191. [Google Scholar] [CrossRef]
  45. Hoel, H.; Heggelund, L.; Reikvam, D.H.; Stiksrud, B.; Ueland, T.; Michelsen, A.E.; Otterdal, K.; Muller, K.E.; Lind, A.; Muller, F.; et al. Elevated markers of gut leakage and inflammasome activation in COVID-19 patients with cardiac involvement. J. Intern. Med. 2021, 289, 523–531. [Google Scholar] [CrossRef]
  46. Mielcarek, C.; Romond, P.C.; Romond, M.B.; Bezirtzoglou, E. Modulation of bacterial translocation in mice mediated through lactose and human milk oligosaccharides. Anaerobe 2011, 17, 361–366. [Google Scholar] [CrossRef]
  47. Gebbers, J.-O.; Laissue, J.-A. Bacterial Translocation in the Normal Human Appendix Parallels the Development of the Local Immune System. Ann. N. Y. Acad. Sci. 2004, 1029, 337–343. [Google Scholar] [CrossRef]
  48. Hobby, G.P.; Karaduta, O.; Dusio, G.F.; Singh, M.; Zybailov, B.L.; Arthur, J.M. Chronic kidney disease and the gut microbiome. Am. J. Physiol. Ren. Physiol. 2019, 316, F1211–F1217. [Google Scholar] [CrossRef]
  49. Flythe, J.E.; Assimon, M.M.; Tugman, M.J.; Chang, E.H.; Gupta, S.; Shah, J.; Sosa, M.A.; Renaghan, A.D.; Melamed, M.L.; Wilson, F.P.; et al. Characteristics and outcomes of individuals with pre-existing kidney disease and covid-19 admitted to intensive care units in the United States. Am. J. Kidney Dis. 2021, 77, 190–203.e1. [Google Scholar] [CrossRef]
  50. Silva, M.A.; da Silva, A.R.P.A.; do Amaral, M.A.; Fragas, M.G.; Câmara, N.O.S. Metabolic Alterations in SARS-CoV-2 Infection and Its Implication in Kidney Dysfunction. Front. Physiol. 2021, 12, 624698. [Google Scholar] [CrossRef]
  51. Robinson, F.A.; Mihealsick, R.P.; Wagener, B.M.; Hanna, P.; Poston, M.D.; Efimov, I.R.; Shivkumar, K.; Hoover, D.B. Role of angiotensin-converting enzyme 2 and pericytes in cardiac complications of COVID-19 infection. Am. J. Physiology. Heart Circ. Physiol. 2020, 319, H1059–H1068. [Google Scholar] [CrossRef]
  52. Kunutsor, S.K.; Laukkanen, J.A. Renal complications in COVID-19: A systematic review and meta-analysis. Ann. Med. 2020, 52, 345–353. [Google Scholar] [CrossRef]
  53. Voorend, C.G.N.; Van Oevelen, M.; Nieberg, M.; Meuleman, Y.; Franssen, C.F.M.; Joosten, H.; Berkhout-Byrne, N.C.; Abrahams, A.C.; Mooijaart, S.P.; Bos, W.J.W.; et al. Impact of the COVID-19 pandemic on symptoms of anxiety and depression and health-related quality of life in older patients with chronic kidney disease. BMC Geriatr. 2021, 21, 650. [Google Scholar] [CrossRef]
  54. Luo, Y.; Wu, J.; Lu, J.; Xu, X.; Long, W.; Yan, G.; Tang, M.; Zou, L.; Xu, Z.; Zhuo, P.; et al. Investigation of COVID-19-related symptoms based on factor analysis. Ann Palliat Med. 2020, 9, 1851–1858. [Google Scholar] [CrossRef]
  55. Helms, J.; Kremer, S.; Merdji, H.; Clere-Jehl, R.; Schenck, M.; Kummerlen, C.; Collange, O.; Boulay, C.; Fafi-Kremer, S.; Ohanam, M.; et al. Neurologic Features in Severe SARS-CoV-2 Infection. New Engl. J. Med. 2020, 382, 2268–2270. [Google Scholar] [CrossRef]
  56. Richards, J.L.; Yap, Y.A.; McLeod, K.H.; MacKay, C.R.; Marinõ, E. Dietary metabolites and the gut microbiota: An alternative approach to control inflammatory and autoimmune diseases. Clin. Transl. Immunol. 2016, 5, e82. [Google Scholar] [CrossRef]
  57. Ranganathan, N.; Ranganathan, P.; Friedman, E.A.; Joseph, A.; Delano, B.; Goldfarb, D.S.; Tam, P.; Rao, A.V.; Anteyi, E.; Musso, C.G. Pilot study of probiotic dietary supplementation for promoting healthy kidney function in patients with chronic kidney disease. Adv. Ther. 2010, 27, 634–647. [Google Scholar] [CrossRef]
  58. Fukuuchi, F.; Hida, M.; Aiba, Y.; Koga, Y.; Endoh, M.; Kurokawa, K.; Sakai, H. Intestinal bacteria-derived putrefactants in chronic renal failure. Clin. Exp. Nephrol. 2002, 6, 99–104. [Google Scholar] [CrossRef]
  59. Nalewajska, M.; Przybyciński, J.; Marchelek-Myśliwiec, M.; Dziedziejko, V.; Ciechanowski, K. Gut Microbiota In Chronic Kidney Disease. Postępy Mikrobiol. Adv. Microbiol. 2019, 58, 237–245. [Google Scholar] [CrossRef]
  60. Mao, L.; Jin, H.; Wang, M.; Hu, Y.; Chen, S.; He, Q.; Chang, J.; Hong, C.; Zhou, Y.; Wang, D.; et al. Neurological Manifestations of Hospitalized Patients with COVID-19 in Wuhan, China. JAMA Neurol. 2020, 77, 683–690. [Google Scholar] [CrossRef]
  61. Guo, J.; Huang, Z.; Lin, L.; Lv, J. Coronavirus disease 2019 (COVID-19) and cardiovascular disease: A viewpoint on the potential influence of angiotensin-converting enzyme inhibitors/angiotensin receptor blockers on onset and severity of severe acute respiratory syndrome coronavirus 2 infection. J. Am. Heart Assoc. 2020, 9, e016219. [Google Scholar] [CrossRef]
  62. Kandemirli, S.G.; Dogan, L.; Sarikaya, Z.T.; Kara, S.; Akinci, C.; Kaya, D.; Kaya, Y.; Yildirim, D.; Tuzuner, F.; Yildirim, M.S.; et al. Brain MRI findings in patients in the intensive care unit with COVID-19 infection. Radiology 2020, 297, E232–E235. [Google Scholar] [CrossRef] [PubMed]
  63. Matschke, J.; Lütgehetmann, M.; Hagel, C.; Sperhake, J.P.; Schröder, A.S.; Edler, C.; Mushumba, H.; Fitzek, A.; Allweiss, L.; Dandri, M.; et al. Neuropathology of patients with COVID-19 in Germany: A post-mortem case series. Lancet Neurol. 2020, 19, 919–929. [Google Scholar] [CrossRef]
  64. Le Guennec, L.; Devianne, J.; Jalin, L.; Cao, A.; Galanaud, D.; Navarro, V.; Boutolleau, D.; Rohaut, B.; Weiss, N.; Demeret, S. Orbitofrontal involvement in a neuroCOVID-19 patient. Epilepsia 2020, 61, e90–e94. [Google Scholar] [CrossRef] [PubMed]
  65. Xiong, W.; Kwan, P.; Zhou, D.; Del Felice, A.; Duncan, J.S.; Sander, J.W. Acute and late neurological complications of COVID19: The quest for evidence. Brain 2020, 143, e99. [Google Scholar] [CrossRef]
  66. Ahmed, M.U.; Hanif, M.; Ali, M.J.; Haider, M.A.; Kherani, D.; Memon, G.M.; Karim, A.H.; Sattar, A. Neurological Manifestations of COVID-19 (SARS-CoV-2): A Review. Front. Neurol. 2020, 11, 518. [Google Scholar] [CrossRef]
  67. Rudroff, T.; Kamholz, J.; Fietsam, A.C.; Deters, J.R.; Bryant, A.D. Post-covid-19 fatigue: Potential contributing factors. Brain Sci. 2020, 10, 1012. [Google Scholar] [CrossRef]
  68. Egbert, A.R.; Cankurtaran, S.; Karpiak, S. Brain abnormalities in COVID-19 acute/subacute phase: A rapid systematic review. Brain Behav. Immun. 2020, 89, 543–554. [Google Scholar] [CrossRef]
  69. Bohmwald, K.; Gálvez, N.M.S.; Ríos, M.; Kalergis, A.M. Neurologic alterations due to respiratory virus infections. Front. Cell. Neurosci. 2018, 12, 386. [Google Scholar] [CrossRef]
  70. Kim, J.E.; Heo, J.H.; Kim, H.O.; Song, S.H.; Park, S.S.; Park, T.H.; Ahn, J.Y.; Kim, M.K.; Choi, J.P. Neurological complications during treatment of middle east respiratory syndrome. J. Clin. Neurol. 2017, 13, 227–233. [Google Scholar] [CrossRef]
  71. Savarin, C.; Bergmann, C.C. Viral-induced suppression of self-reactive T cells: Lessons from neurotropic coronavirus-induced demyelination. J. Neuroimmunol. 2017, 308, 12–16. [Google Scholar] [CrossRef]
  72. Cheng, Y.; Skinner, D.D.; Lane, T.E. Innate immune responses and viral-induced neurologic disease. J. Clin. Med. 2019, 8, 3. [Google Scholar] [CrossRef]
  73. Wu, Y.; Xu, X.; Chen, Z.; Duan, J.; Hashimoto, K.; Yang, L.; Cunming, L.; Yang, C. Nervous system involvement after infection with COVID-19 and other coronaviruses. Brain Behav. Immun. 2020, 87, 18–22. [Google Scholar] [CrossRef]
  74. Ye, M.; Ren, Y.; Lv, T. Encephalitis as a clinical manifestation of COVID-19. Brain Behav. Immun. 2020, 88, 945–946. [Google Scholar] [CrossRef]
  75. Toscano, G.; Palmerini, F.; Ravaglia, S.; Ruiz, L.; Invernizzi, P.; Cuzzoni, M.G.; Franciotta, D.; Baldanti, F.; Daturi, R.; Postorino, P.; et al. Guillain–Barré Syndrome associated with SARS-CoV-2. New Engl. J. Med. 2020, 382, 2574–2576. [Google Scholar] [CrossRef]
  76. Pincherle, A.; Jöhr, J.; Pancini, L.; Leocani, L.; Dalla Vecchia, L.; Ryvlin, P.; Schiff, N.D.; Diserens, K. Intensive Care admission and early Neuro-Rehabilitation. Lessons for COVID-19? Front. Neurol. 2020, 11, 880. [Google Scholar] [CrossRef]
  77. Yin, C.H.; Wang, C.; Tang, Z.; Wen, Y.; Zhang, S.W.; Wang, B.E. Clinical analysis of multiple organ dysfunction syndrome in patients suffering from SARS. Zhongguo Wei Zhong Bing Ji Jiu Yi Xue. 2004, 16, 646–650. [Google Scholar]
  78. Klein, R.S.; Garber, C.; Howard, N. Infectious immunity in the central nervous system and brain function. Nat. Immunol. 2017, 18, 132–141. [Google Scholar] [CrossRef]
  79. Zanin, L.; Saraceno, G.; Panciani, P.P.; Renisi, G.; Signorini, L.; Migliorati, K.; Fontanella, M.M. SARS-CoV-2 can induce brain and spine demyelinating lesions. Acta Neurochir. 2020, 162, 1491–1494. [Google Scholar] [CrossRef]
  80. Desforges, M.; Le Coupanec, A.; Dubeau, P.; Bourgouin, A.; Lajoie, L.; Dubé, M.; Talbot, P.J. Human coronaviruses and other respiratory viruses: Underestimated opportunistic pathogens of the central nervous system? Viruses 2019, 12, 14. [Google Scholar] [CrossRef]
  81. Coolen, T.; Lolli, V.; Sadeghi, N.; Rovai, A.; Trotta, N.; Taccone, F.S.; Creteur, J.; Henrard, S.; Goffard, J.C.; Dewitte, O.; et al. Early postmortem brain MRI findings in COVID-19 non-survivors. Neurology 2020, 95, e2016–e2027. [Google Scholar] [CrossRef]
  82. Poyiadji, N.; Shahin, G.; Noujaim, D.; Stone, M.; Patel, S.C.; Griffith, B. COVID-19–associated Acute Hemorrhagic Necrotizing Encephalopathy: CT and MRI Features. Cureus 2020, 296, E119–E120. [Google Scholar]
  83. Mandal, S.; Barnett, J.; Brill, S.E.; Brown, J.S.; Denneny, E.K.; Hare, S.S.; Heightman, M.; Hillman, T.E.; Jacob, J.; Jarvis, H.; et al. Long-COVID’: A cross-sectional study of persisting symptoms, biomarker and imaging abnormalities following hospitalisation for COVID-19. Thorax 2021, 76, 396–398. [Google Scholar] [CrossRef] [PubMed]
  84. Mazza, M.G.; De Lorenzo, R.; Conte, C.; Poletti, S.; Vai, B.; Bollettini, I.; Melloni, E.M.T.; Furlan, R.; Ciceri, F.; Rovere-Querini, P.; et al. Anxiety and depression in COVID-19 survivors: Role of inflammatory and clinical predictors. Brain Behav. Immun. 2020, 89, 594–600. [Google Scholar] [CrossRef] [PubMed]
  85. Bottemanne, H.; Delaigue, F.; Lemogne, C. SARS-CoV-2 Psychiatric Sequelae: An Urgent Need of Prevention. Front. Psychiatry 2021, 2, 1479. [Google Scholar] [CrossRef]
  86. Grover, S.; Kumar, V.; Chakrabarti, S.; Hollikatti, P.; Singh, P.; Tyagi, S.; Kulhara, P.; Avasthi, A. Prevalence and Type of Functional Somatic Complaints in Patients with First-episode Depression. East Asian Arch Psychiatry. East Asian Arch. Psychiatry 2012, 22, 146–153. [Google Scholar]
  87. Perna, G.; Caldirola, D. COVID-19 and panic disorder: Clinical considerations for the most physical of mental disorders. Braz. J. Psychiatry 2021, 43, 110–111. [Google Scholar] [CrossRef]
  88. Ezzat, M.M.; Elsherif, A.A. Prevalence of Fatigue in Patients Post COVID-19. Eur. J. Mol. Clin. Med. 2021, 08, 1330–1340. [Google Scholar]
  89. Katafuchi, T.; Kondo, T.; Take, S.; Yoshimura, M. Brain Cytokines and the 5-HT System during Poly I:C-Induced Fatigue. Ann. N. Y. Acad. Sci. 2006, 1088, 230–237. [Google Scholar] [CrossRef]
  90. Costa, L.H.A.; Santos, B.M.; Branco, L.G.S. Can selective serotonin reuptake inhibitors have a neuroprotective effect during COVID-19? Eur. J. Pharmacol. 2020, 889, 173629. [Google Scholar] [CrossRef]
  91. Douaud, G.; Lee, S.; Alfaro-Almagro, F.; Arthofer, C.; Wang, C.; McCarthy, P.; Lange, F.; Andersson, J.; Griffanti, L.; Duff, E.; et al. SARS-CoV-2 is associated with changes in brain structure in UK Biobank. Nature 2022, 604, 697–707. [Google Scholar] [CrossRef]
  92. Al-Sarraj, S.; Troakes, C.; Hanley, B.; Osborn, M.; Richardson, M.P.; Hotopf, M.; Bullmore, E.; Everall, I. Invited Review: The spectrum of neuropathology in COVID-19. Neuropathol. Appl. Neurobiol. 2021, 47, 3–16. [Google Scholar] [CrossRef]
  93. Donegani, M.I.; Miceli, A.; Pardini, M.; Bauckneht, M.; Chiola, S.; Pennone, M.; Marini, C.; Massa, F.; Raffa, S.; Ferrarazzo, G.; et al. Brain metabolic correlates of persistent olfactory dysfunction after SARS-CoV-2 infection. Biomedicines 2021, 9, 287. [Google Scholar] [CrossRef]
  94. Guedj, E.; Campion, J.Y.; Dudouet, P.; Kaphan, E.; Bregeon, F.; Tissot-Dupont, H.; Guis, S.; Barthelemy, F.; Habert, P.; Ceccaldi, M.; et al. 18F-FDG brain PET hypometabolism in patients with long COVID. Eur. J. Nucl. Med. Mol. Imaging 2021, 48, 2823–2833. [Google Scholar] [CrossRef]
  95. Hosp, J.A.; Dressing, A.; Blazhenets, G.; Bormann, T.; Rau, A.; Schwabenland, M.; Thurow, J.; Wagner, D.; Waller, C.; Niesen, W.D.; et al. Cognitive impairment and altered cerebral glucose metabolism in the subacute stage of COVID-19. Brain 2021, 144, 1263–1276. [Google Scholar] [CrossRef]
  96. Mosconi, L. Brain glucose metabolism in the early and specific diagnosis of Alzheimer’s disease: FDG-PET studies in MCI and AD. Eur. J. Nucl. Med. Mol. Imaging 2005, 32, 486–510. [Google Scholar] [CrossRef]
  97. Zoghi, A.; Ramezani, M.; Roozbeh, M.; Darazam, I.A.; Sahraian, M.A. A case of possible atypical demyelinating event of the central nervous system following COVID-19. Mult. Scler. Relat. Disord. 2020, 44, 102324. [Google Scholar] [CrossRef]
  98. Wingerchuk, D.M.; Banwell, B.; Bennett, J.L.; Cabre, P.; Carroll, W.; Chitnis, T.; de Seze, J.; Fujihara, K.; Greenberg, B.; Jacob, A.; et al. International consensus diagnostic criteria for neuromyelitis optica spectrum disorders. Neurology 2015, 85, 177–189. [Google Scholar] [CrossRef]
  99. Hepburn, M.; Mullaguri, N.; George, P.; Hantus, S.; Punia, V.; Bhimraj, A.; Newey, C.R. Acute Symptomatic Seizures in Critically Ill Patients with COVID-19: Is There an Association? Neurocrit. Care 2021, 34, 139–143. [Google Scholar] [CrossRef]
  100. Afshar, H.; Yassin, Z.; Kalantari, S.; Aloosh, O.; Lotfi, T.; Moghaddasi, M.; Sadeghipour, A.; Emamikhah, M. Evolution and resolution of brain involvement associated with SARS-CoV-2 infection: A close Clinical—Paraclinical follow up study of a case. Mult. Scler. Relat. Disord. 2020, 43, 102216. [Google Scholar] [CrossRef]
  101. Benedetti, F.; Palladini, M.; Paolini, M.; Melloni, E.; Vai, B.; De Lorenzo, R.; Furlan, R.; Rovere-Querini, P.; Falini, A.; Mazza, M.G. Brain correlates of depression, post-traumatic distress, and inflammatory biomarkers in COVID-19 survivors: A multimodal magnetic resonance imaging study. Brain Behav. Immun. Health 2021, 18, 100387. [Google Scholar] [CrossRef]
  102. Cecchetti, G.; Agosta, F.; Canu, E.; Basaia, S.; Barbieri, A.; Cardamone, R.; Bernasconi, M.P.; Castelnovo, V.; Cividini, C.; Cursi, M.; et al. Cognitive, EEG, and MRI features of COVID-19 survivors: A 10-month study. J. Neurol. 2022, 269, 3400–3412. [Google Scholar] [CrossRef]
  103. Anzalone, N.; Castellano, A.; Scotti, R.; Scandroglio, A.M.; Filippi, M.; Ciceri, F.; Tresoldi, M.; Falini, A. Multifocal laminar cortical brain lesions: A consistent MRI finding in neuro-COVID-19 patients. J. Neurol. 2020, 267, 2806–2809. [Google Scholar] [CrossRef]
  104. Naeije, R.; Caravita, S. Phenotyping long COVID. Eur. Respir. J. 2021, 58, 2101763. [Google Scholar] [CrossRef]
  105. Lukiw, W.J.; Pogue, A.; Hill, J.M. SARS-CoV-2 Infectivity and Neurological Targets in the Brain. Cell Mol. Neurobiol. 2022, 42, 42,217–224. [Google Scholar] [CrossRef]
  106. Greenhalgh, T.; Knight, M.; A’Court, C.; Buxton, M.; Husain, L. Management of post-acute covid-19 in primary care. The BMJ 2020, 370, m3026. [Google Scholar] [CrossRef]
  107. Yong, S.J. Long COVID or post-COVID-19 syndrome: Putative pathophysiology, risk factors, and treatments. Infect. Dis. 2021, 53, 737–754. [Google Scholar] [CrossRef]
  108. Vaduganathan, M.; Vardeny, O.; Michel, T.; McMurray, J.J.V.; Pfeffer, M.A.; Solomon, S.D. Renin-Angiotensin-Aldosterone system inhibitors in patients with COVID-19. N. Engl. J. Med. 2020, 382, 1653–1659. [Google Scholar] [CrossRef]
  109. Kim, K.D.; Zhao, J.; Auh, S.; Yang, X.; Du, P.; Tang, H.; Fu, Y.X. Adaptive immune cells temper initial innate responses. Nat. Med. 2007, 13, 1248–1252. [Google Scholar] [CrossRef]
  110. Karlsson, A.C.; Humbert, M.; Buggert, M. The known unknowns of T cell immunity to COVID-19. Sci. Immunol. 2020, 8063, 5. [Google Scholar] [CrossRef]
  111. Ehrenfeld, M.; Tincani, A.; Andreoli, L.; Cattalini, M.; Greenbaum, A.; Kanduc, D.; Alijotas-Reig, J.; Zinserling, V.; Semenov, N.; Howard Amital, H.; et al. COVID-19 and autoimmunity. Autoimmun. Rev. 2020, 19, 102597. [Google Scholar] [CrossRef]
  112. Lui, D.T.W.; Lee, C.H.; Chow, W.S.; Lee, A.C.H.; Tam, A.R.; Fong, C.H.Y.; Law, C.Y.; Leung, E.K.H.; To, K.K.W.; Kathryn Choon Beng Tan, K.C.B.; et al. Thyroid dysfunction in relation to immune profile, disease status and outcome in 191 patients with COVID-19. J. Clin. Endocrinol. Metab. 2021, 106, e926–e935. [Google Scholar] [CrossRef] [PubMed]
  113. Muller, I.; Cannavaro, D.; Dazzi, D.; Covelli, D.; Mantovani, G.; Muscatello, A.; Ferrante, E.; Orsi, E.; Resi, V.; Longari, V.; et al. SARS-CoV-2-related atypical thyroiditis. Lancet Diabetes Endocrinol. 2020, 8, 739–741. [Google Scholar] [CrossRef]
  114. Zuo, Y.; Estes, S.K.; Ali, R.A.; Gandhi, A.A.; Yalavarthi, S.; Shi, H.; Sule, G.; Gockman, K.; Jacqueline A Madison, J.A.; Zuo, M.; et al. Prothrombotic autoantibodies in serum from patients hospitalized with COVID-19. Sci. Transl. Med. 2020, 12, 3876. [Google Scholar] [CrossRef] [PubMed]
  115. Bastard, P.; Rosen, L.B.; Zhang, Q.; Michailidis, E.; Hoffmann, H.H.; Zhang, Y.; Dorgham, k.; Philippot, Q.; Rosain, J.; Béziat, V.; et al. Autoantibodies against type I IFNs in patients with life-threatening COVID-19. Science 2020, 1979, 370. [Google Scholar] [CrossRef] [PubMed]
  116. Elkon, K.; Casali, P. Nature and functions of autoantibodies. Nat. Clin. Pract. Rheumatol. 2008, 4, 491–498. [Google Scholar] [CrossRef] [PubMed]
  117. Kong, M.; Zhang, H.; Cao, X.; Mao, X.; Lu, Z. Higher level of Neutrophil-to-Lymphocyte is associated with severe COVID-19. Epidemiol. Infect. 2020, 148, e139. [Google Scholar] [CrossRef] [PubMed]
  118. Fathi, N.; Rezaei, N. Lymphopenia in COVID-19: Therapeutic opportunities. Cell Biol. Int. 2020, 44, 1792–1797. [Google Scholar] [CrossRef] [PubMed]
  119. Hu, F.; Chen, F.; Ou, Z.; Fan, O.; Tan, X.; Wang, Y.; Pan, Y.; Ke, B.; Li, L.; Guan, Y.; et al. A compromised specific humoral immune response against the SARS-CoV-2 receptor-binding domain is related to viral persistence and periodic shedding in the gastrointestinal tract. Cell Mol. Immunol. 2020, 17, 1119–1125. [Google Scholar] [CrossRef]
  120. Amato, M.K.; Hennessy, C.; Shah, K.; Mayer, J. Multisystem inflammatory syndrome in an adult. J. Emerg. Med. 2021, 61, e1–e3. [Google Scholar] [CrossRef]
  121. Love, C.; Tomas, M.B.; Bronco, G.G.; Palestro, C.J. FDG PET of infection and inflammation. Radiographics 2005, 25, 1357–1368. [Google Scholar] [CrossRef]
  122. Sollini, M.; Ciccarelli, M.; Cecconi, M.; Aghemo, A.; Morelli, P.; Gelardi, F.; Chiti, A. Vasculitis changes in COVID-19 survivors with persistent symptoms: An [18 F] FDG-PET/CT study. Eur. J. Nucl. Med. Mol. Imaging 2020, 48, 1460–1466. [Google Scholar] [CrossRef]
  123. Kucuk, A.; Cumhur, C.M.; Cure, E. Can COVID-19 cause myalgia with a completely different mechanism? A hypothesis. Clin. Rheumatol. 2020, 39, 2103–2104. [Google Scholar] [CrossRef]
  124. Su, Y.; Yuan, D.; Chen, D.G.; Ng, R.H.; Wang, K.; Choi, J.; Li, S.; Hong, S.; Zhang, R.; Xie, J.; et al. Multiple early factors anticipate post-acute COVID-19 sequelae. Cell 2022, 185, 881–895.e20. [Google Scholar] [CrossRef]
  125. Congqing Wu, C.; Wei Lu, W.; Zhang, Y.; Guoying Zhang, G.; Shi, X.; Hisada, Y.; Grover, S.P.; Xinyi Zhang, X.; Li, L.; Xiang, B.; et al. Inflammasome activation triggers blood clotting and host death through pyroptosis. Immunity 2019, 50, 1401–1411.e4. [Google Scholar]
  126. Bochenek, M.L.; Schäfer, K. Role of endothelial cells in acute and chronic thrombosis. Hamostaseologie 2019, 39, 128–139. [Google Scholar] [CrossRef]
  127. Rauch, U.; Nemerson, Y. Tissue Factor, the Blood, and the Arterial Wall. Trends. Cardiovasc. Med. 2000, 10, 139–143. [Google Scholar] [CrossRef]
  128. Hottz, E.D.; Azevedo-Quintanilha, I.G.; Palhinha, L.; Teixeira, L.; Barreto, E.A.; Pão, C.R.; Righy, C.; Franco, S.; Souza, T.M.; Pedro Kurtz, P.; et al. Platelet activation and platelet-monocyte aggregate formation trigger tissue factor expression in patients with severe COVID-19. Blood 2020, 136, 1330–1341. [Google Scholar] [CrossRef]
  129. Al-Ani, F.; Chehade, S.; Lazo-Langner, A. Thrombosis risk associated with COVID-19 infection. A scoping review. Thromb. Res. 2020, 192, 152–160. [Google Scholar] [CrossRef]
  130. Nalbandian, A.; Sehgal, K.; Gupta, A.; Madhavan, M.V.; McGroder, C.; Stevens, J.S.; Cook, J.R.; Nordvig, A.S.; Shalev, D.; Sehrawat, T.S.; et al. Post-acute COVID-19 syndrome. Nat. Med. 2021, 27, 601–615. [Google Scholar] [CrossRef]
  131. Callard, F.; Perego, E. How and why patients made Long Covid. Soc. Sci. Med. 2021, 268, 113426. [Google Scholar] [CrossRef]
  132. Altmann, D.M.; Boyton, R.J. Decoding the unknowns in long covid. The BMJ 2021, 372, n132. [Google Scholar] [CrossRef]
  133. Venkatesan, P. NICE guideline on long COVID. Lancet Respir. Med. 2021, 9, 129. [Google Scholar] [CrossRef]
  134. Nath, A. Long-Haul COVID. Neurology 2020, 95, 559–560. [Google Scholar] [CrossRef]
  135. Brodin, P. Immune determinants of COVID-19 disease presentation and severity. Nat. Med. 2021, 27, 28–33. [Google Scholar] [CrossRef]
  136. Davido, B.; Seang, S.; Tubiana, R.; de Truchis, P. Post–COVID-19 chronic symptoms: A postinfectious entity? Clin. Microbiol. Infect. 2020, 26, 1448–1449. [Google Scholar] [CrossRef]
  137. Datta, S.D.; Talwar, A.; Lee, J.T. A Proposed framework and timeline of the spectrum of disease due to SARS-CoV-2 Infection: Illness beyond acute infection and public health implications. JAMA J. Am. Med. Assoc. 2020, 324, 2251–2252. [Google Scholar] [CrossRef]
  138. Sivan, M.; Taylor, S. NICE guideline on long COVID: Research must be done urgently to fill the many gaps in this new ‘living guideline’. BMJ 2020, 371, m4938. [Google Scholar] [CrossRef]
  139. Fernández-De-las-peñas, C.; Palacios-Ceña, D.; Gómez-Mayordomo, V.; Cuadrado, M.L.; Florencio, L.L. Defining post-covid symptoms (Post-acute covid, long covid, persistent post-covid): An integrative classification. Int. J. Environ. Res. Public Health 2021, 18, 2621. [Google Scholar] [CrossRef]
  140. Shah, W.; Hillman, T.; Playford, E.D.; Hishmeh, L. Managing the long-term effects of COVID-19: Summary of NICE, SIGN, and RCGP rapid guideline. BMJ 2021, 372, n136, Erratum in BMJ 2022, 376, o126. [Google Scholar] [CrossRef] [PubMed]
  141. Corcionivoschi, N.; Drinceanu, D.; Stef, L.; Luca, I.; Julean, C. Probiotics-identification and ways of action. Innov. Rom. Food Biotechnol. 2010, 6, 1. [Google Scholar]
  142. Gibson, G.R.; Hutkins, R.; Sanders, M.E.; Prescott, S.L.; Reimer, R.A.; Salminen, S.J.; Scott, K.; Stanton, C.; Swanson, K.S.; Cani, P.D.; et al. Expert consensus document: The International scientific association for probiotics and prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 491–502. [Google Scholar] [CrossRef] [PubMed]
  143. Hu, J.; Zhang, L.; Lin, W.; Tang, W.; Chan, F.K.; Ng, S.C. Review article: Probiotics, prebiotics and dietary approaches during COVID-19 pandemic. Trends Food Sci. Technol. 2021, 108, 187–196. [Google Scholar] [CrossRef] [PubMed]
  144. Baud, D.; Agri, V.D.; Gibson, G.R.; Reid, G.; Giannoni, E. Using probiotics to flatten the curve of coronavirus disease COVID-2019 Pandemic. Front. Public Health 2020, 8, 186. [Google Scholar] [CrossRef] [PubMed]
  145. Olaimat, A.N.; Aolymat, I.; Al-Holy, M.; Ayyash, M.; Abu Ghoush, M.; Al-Nabulsi, A.A.; Osaili, T.; Apostolopoulos, V.; Liu, S.Q.; Shah, N.P. The potential application of probiotics and prebiotics for the prevention and treatment of COVID-19. NPJ Sci. Food 2020, 4, 1–7. [Google Scholar] [CrossRef]
  146. Batista, K.S.; de Albuquerque, J.G.; Vasconcelos, M.; Bezerra, M.; da Silva Barbalho, M.B.; Pinheiro, R.O.; Aquino, J.S. Probiotics and prebiotics: Potential prevention and therapeutic target for nutritional management of COVID-19? Nutr. Res. Rev. 2021, 1–18, Epub ahead of print.. PMCID:PMC8593414. [Google Scholar] [CrossRef] [PubMed]
  147. Zhang, D.; Li, S.; Wang, N.; Tan, H.Y.; Zhang, Z.; Feng, Y. The Crosstalk between gut microbiota and lungs in common lung diseases. Front. Microbiol. 2020, 11, 301. [Google Scholar] [CrossRef]
  148. National Health Committee of the People’s Republic of China. National Administration of Traditional Chinese Medicine Diagnostic and Therapeutic Guidance for 2019 Novel Coronavirus Disease (Version 5). 2019. Available online: http://www.nhc.gov.cn/yzygj/s7653p/202002/d4b895337e19445f8d728fcaf1e3e13a/files/ab6bec7f93e64e7f998d802991203cd6.pdf (accessed on 5 June 2022).
  149. Liang, Y.; Liang, S.; Zhang, Y.; Deng, Y.; He, Y.; Chen, Y.; Liu, C.; Lin, C.; Yang, Q. Oral administration of compound probiotics ameliorates HFD-Induced gut microbe dysbiosis and chronic metabolic inflammation via the G Protein-Coupled Receptor 43 in Non-alcoholic fatty liver disease rats. Probiotics Antimicrob Proteins 2019, 11, 175–185. [Google Scholar] [CrossRef]
  150. Zhao, S.; Feng, P.; Meng, W.; Jin, W.; Li, X.; Li, X. Modulated gut microbiota for potential COVID-19 prevention and treatment. Front. Med. 2022, 9, 811176, PMCID:PMC8927624. [Google Scholar] [CrossRef] [PubMed]
  151. Klaenhammer, T.R.; Kleerebezem, M.; Kopp, M.V.; Rescigno, M. The impact of probiotics and prebiotics on the immune system. Nat. Rev. Immunol. 2012, 12, 728–734. [Google Scholar] [CrossRef]
  152. Ettinger, G.; MacDonald, K.; Reid, G.; Burton, J.P. The influence of the human microbiome and probiotics on cardiovascular health. Gut Microbes 2015, 5, 719–728. [Google Scholar] [CrossRef]
  153. Li, Q.; Cheng, F.; Xu, Q.; Su, Y.; Cai, X.; Zeng, F.; Zhang, Y. The role of probiotics in coronavirus disease-19 infection in Wuhan: A retrospective study of 311 severe patients. Int. Immunopharmacol. 2021, 95, 107531. [Google Scholar] [CrossRef]
  154. Oliva, S.; Di Nardo, G.; Ferrari, F.; Mallardo, S.; Rossi, P.; Patrizi, G.; Cucchiara, S.; Stronati, L. Randomised clinical trial: The effectiveness of Lactobacillus reuteri ATCC 55730 rectal enema in children with active distal ulcerative colitis. Aliment. Pharmacol. Ther. 2012, 35, 327–334. [Google Scholar] [CrossRef]
  155. Xu, K.; Cai, H.; Shen, Y.; Ni, Q.; Chen, Y.; Hu, S.; Li, J.; Wang, H.; Yu, L.; Huang, H.; et al. Management of COVID-19: The Zhejiang experience the: Zhejiang experience. J. Zhejiang University. Med. Sci. 2020, 49, 147–157. [Google Scholar]
  156. Dhar, D.; Mohanty, A. Gut microbiota and COVID-19- possible link and implications. Virus Res. 2020, 285, 198018. [Google Scholar] [CrossRef]
  157. Gogineni, V.K.; Morrow, L.E.; Malesker, M.A. Probiotics: Mechanisms of action and clinical applications. J. Probiotics Health 2013, 1, 1–11. [Google Scholar] [CrossRef]
  158. Amer, M.; Nadeem, M.; Nazir, S.; Fakhar, M.; Abid, F.; Ain, Q.U.; Asif, E. Probiotics and their use in inflammatory bowel disease. Altern. Ther. Health Med. 2018, 24, 16–23. [Google Scholar]
  159. Davani-Davari, D.; Negahdaripour, M.; Karimzadeh, I.; Seifan, M.; Mohkam, M.; Masoumi, S.J.; Berenjian, A.; Ghasemi, Y. Prebiotics: Definition, types, sources, mechanisms, and clinical applications. Foods 2019, 92, 8. [Google Scholar] [CrossRef]
  160. Parnell, J.A.; Reimer, R.A. Prebiotic fiber modulation of the gut microbiota improves risk factors for obesity and the metabolic syndrome. Gut Microbes 2012, 3, 29–34. [Google Scholar] [CrossRef]
  161. Wiciński, M.; Gębalski, J.; Mazurek, E.; Podhorecka, M.; Śniegocki, M.; Szychta, P.; Sawicka, E.; Malinowski, B. The influence of polyphenol compounds on human gastrointestinal tract microbiota. Nutrients 2020, 350, 12. [Google Scholar] [CrossRef]
  162. Ranucci, G.; Buccigrossi, V.; Borgia, E.; Piacentini, D.; Visentin, F.; Cantarutti, L.; Baiardi, P.; Felisi, M.; Spagnuolo, M.I.; Zanconato, S.; et al. Galacto-oligosaccharide/polidextrose enriched formula protects against respiratory infections in infants at high risk of atopy: A randomized clinical trial. Nutrients 2018, 10, 286. [Google Scholar] [CrossRef] [Green Version]
  163. Wang, S.; Xiao, Y.; Tian, F.; Zhao, J.; Zhang, H.; Zhai, Q.; Chen, W. Rational use of prebiotics for gut microbiota alterations: Specific bacterial phylotypes and related mechanisms. J. Funct. Foods 2020, 66, 103838. [Google Scholar] [CrossRef]
  164. West, C.E.; Dzidic, M.; Prescott, S.L.; Jenmalm, M.C. Bugging allergy; role of pre-, pro- and synbiotics in allergy prevention. Allergol. Int. 2017, 66, 529–538. [Google Scholar] [CrossRef]
  165. Din, A.U.; Mazhar, M.; Waseem, M.; Ahmad, W.; Bibi, A.; Hassan, A.; Ali, N.; Gang, W.; Qian, G.; Ullah, R.; et al. SARS-CoV-2 microbiome dysbiosis linked disorders and possible probiotics role. Biomed. Pharmacother. 2021, 133, 110947. [Google Scholar] [CrossRef]
Figure 1. Gut–microbiota–liver axis in COVID-19 patients: role of dysbiosis and leaky gut in long-lasting liver complication through PAMPs, secondary bile acids, toxins, and pathogenic bacteria metabolites.
Figure 1. Gut–microbiota–liver axis in COVID-19 patients: role of dysbiosis and leaky gut in long-lasting liver complication through PAMPs, secondary bile acids, toxins, and pathogenic bacteria metabolites.
Metabolites 12 00912 g001
Figure 2. Potential impact of the gut–heart axis in long-COVID-19 patients.
Figure 2. Potential impact of the gut–heart axis in long-COVID-19 patients.
Metabolites 12 00912 g002
Figure 3. Role of leaky gut and dysbiosis in chronic kidney damage as a long-COVID-19 complication.
Figure 3. Role of leaky gut and dysbiosis in chronic kidney damage as a long-COVID-19 complication.
Metabolites 12 00912 g003
Figure 4. Role of COVID-19 infection in glutamate excitotoxicity as an etiological mechanism of psychological complications.
Figure 4. Role of COVID-19 infection in glutamate excitotoxicity as an etiological mechanism of psychological complications.
Metabolites 12 00912 g004
Figure 5. Interactions between the human gut and lung and potential positive immune responses triggered by probiotics and prebiotics.
Figure 5. Interactions between the human gut and lung and potential positive immune responses triggered by probiotics and prebiotics.
Metabolites 12 00912 g005
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Alenazy, M.F.; Aljohar, H.I.; Alruwaili, A.R.; Daghestani, M.H.; Alonazi, M.A.; Labban, R.S.; El-Ansary, A.K.; Balto, H.A. Gut Microbiota Dynamics in Relation to Long-COVID-19 Syndrome: Role of Probiotics to Combat Psychiatric Complications. Metabolites 2022, 12, 912. https://doi.org/10.3390/metabo12100912

AMA Style

Alenazy MF, Aljohar HI, Alruwaili AR, Daghestani MH, Alonazi MA, Labban RS, El-Ansary AK, Balto HA. Gut Microbiota Dynamics in Relation to Long-COVID-19 Syndrome: Role of Probiotics to Combat Psychiatric Complications. Metabolites. 2022; 12(10):912. https://doi.org/10.3390/metabo12100912

Chicago/Turabian Style

Alenazy, Maha F., Haya I. Aljohar, Ashwag R. Alruwaili, Maha H. Daghestani, Mona A. Alonazi, Ranyah S. Labban, Afaf K. El-Ansary, and Hanan A. Balto. 2022. "Gut Microbiota Dynamics in Relation to Long-COVID-19 Syndrome: Role of Probiotics to Combat Psychiatric Complications" Metabolites 12, no. 10: 912. https://doi.org/10.3390/metabo12100912

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

Article Metrics

Back to TopTop