Skip to main content

HYPOTHESIS AND THEORY article

Front. Neurosci., 28 September 2023
Sec. Gut-Brain Axis

Gut-brain pathogenesis of post-acute COVID-19 neurocognitive symptoms

  • 1School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, United States
  • 2The Mind Research Network/Lovelace Biomedical and Environmental Research Institute, Albuquerque, NM, United States
  • 3Division of Gastroenterology and Hepatology, University of New Mexico, Albuquerque, NM, United States
  • 4Section of Gastroenterology, New Mexico Veterans Affairs Health Care System, Albuquerque, NM, United States
  • 5Nene and Jamie Koch Comprehensive Movement Disorder Center, Department of Neurology, University of New Mexico, Albuquerque, NM, United States
  • 6Department of Psychiatry and Behavioral Sciences, University of New Mexico School of Medicine, Albuquerque, NM, United States
  • 7Division of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM, United States

Approximately one third of non-hospitalized coronavirus disease of 2019 (COVID-19) patients report chronic symptoms after recovering from the acute stage of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Some of the most persistent and common complaints of this post-acute COVID-19 syndrome (PACS) are cognitive in nature, described subjectively as “brain fog” and also objectively measured as deficits in executive function, working memory, attention, and processing speed. The mechanisms of these chronic cognitive sequelae are currently not understood. SARS-CoV-2 inflicts damage to cerebral blood vessels and the intestinal wall by binding to angiotensin-converting enzyme 2 (ACE2) receptors and also by evoking production of high levels of systemic cytokines, compromising the brain’s neurovascular unit, degrading the intestinal barrier, and potentially increasing the permeability of both to harmful substances. Such substances are hypothesized to be produced in the gut by pathogenic microbiota that, given the profound effects COVID-19 has on the gastrointestinal system, may fourish as a result of intestinal post-COVID-19 dysbiosis. COVID-19 may therefore create a scenario in which neurotoxic and neuroinflammatory substances readily proliferate from the gut lumen and encounter a weakened neurovascular unit, gaining access to the brain and subsequently producing cognitive deficits. Here, we review this proposed PACS pathogenesis along the gut-brain axis, while also identifying specific methodologies that are currently available to experimentally measure each individual component of the model.

1. Introduction

The coronavirus disease of 2019 (COVID-19) is caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and has the hallmark clinical presentation of pulmonary complications that range from mild hypoxia to pneumonia to acute respiratory distress (Johnson et al., 2020). Given the potentially life-threatening severities of COVID-19, most of the research immediately following the 2019 COVID-19 outbreak was focused on the acute disease stage. However, reports of lasting effects began to emerge shortly after the beginning of the pandemic, leading the Centers for Disease Control (CDC) to define “long COVID-19” as symptoms lasting beyond 3 months (Raman et al., 2022). Such effects were quickly found to not necessarily limit themselves to the lungs. Many of the persistent symptoms involve the central nervous and the gastrointestinal systems, including fatigue, brain fog, irritable bowel syndrome, and dyspepsia (Peiris et al., 2021; Choudhury et al., 2022). Here we discuss a multi-system constellation of residual SARS-CoV-2 effects that can remain after the acute stage and may lead to the emergence and maintenance of chronic COVID-19 symptoms. Specifically, we focus on the lasting neurocognitive sequelae that often follow COVID-19, and provide an overview of their proposed pathogenesis along the gut-brain axis.

Subsequent to the acute stage of COVID-19, up to 30% of patients report long-term symptoms, despite the initial infection being resolved and the virus no longer being detectable in the body (Davis et al., 2021). As such, while the epidemiological waves of SARS-CoV-2 have come and gone, the number of individuals suffering from its chronic sequelae has been increasing cumulatively during and after the pandemic, with the CDC estimating that approximately 1 in 13 adults in the US have long COVID-19 at the time of this writing. The pathogenesis of these residual effects of COVID-19 remains unclear, and few tools are currently available to address this looming global health crisis. As such, the described gut-brain model may motivate novel applications for existing gastrointestinal therapies in order to indirectly influence the central nervous system, potentially alleviating the neurocognitive sequelae of COVID-19. We note that the CDC recently approved an ICD-10 code for Post-Acute Sequelae of COVID-19 (PASC); however, other definitions such as Post-Acute COVID-19 Syndrome (PACS) and Post COVID-19 Conditions (PCC) are also common in clinical and scientific communications. With long COVID nomenclature remaining somewhat ambiguous, the authors of this manuscript refer to the condition as PACS.

2. Gut-brain axis model of PACS

Described initially as general symptoms and conditions that continue or develop after SARS-CoV-2 infection, the definitive clinical presentation of long COVID-19 remains elusive. Cognitive issues are frequently reported, including memory loss and brain fog (Han et al., 2022), with patients over the age of 65 reporting more severe acute- and chronic-stage symptomology (Mueller et al., 2020; Han et al., 2022). Long COVID also appears to involve developing new-onset conditions such as diabetes, thrombotic, and cerebrovascular disease (Xie et al., 2022). As discussed below in more detail, there is compelling evidence that the SARS-CoV-2 virus compromises intestinal epithelium and cerebrovascular endothelium (Buzhdygan et al., 2020; Cardinale et al., 2020) – both directly and indirectly – lending support to the PACS pathogenesis model based on dysfunctional gut-brain interactions due to damage to the neurovascular unit and intestinal barrier. Specifically, angiotensin-converting enzyme 2 (ACE2) receptors that SARS-CoV-2 relies on for cellular entry are widely expressed by cerebrovascular endothelial and smooth muscle cells (Hamming et al., 2004; Lu et al., 2020; Zhou et al., 2020), and have been implicated in both direct viral damage to the neurovascular unit (Buzhdygan et al., 2020) and indirect damage via virus-induced cytokine storms (Elyaspour et al., 2021; Nicosia et al., 2021). The intestinal wall is also rich with ACE2 receptors, making it vulnerable to viral infiltration that can damage the intestinal barrier and increase its systemic permeability (Cardinale et al., 2020). Further, common COVID-19 gastrointestinal disturbances often produce dysbiosis – a shift toward pathological, pro-inflammatory, and potentially neurotoxic gut microbiota (Kopel et al., 2020; Venzon et al., 2021; Yeoh et al., 2021). Collectively, COVID-19 may thus create a scenario in which neurotoxic and neuroinflammatory substances systemically permeate from the gut lumen through the damaged intestinal barrier and produce chronic cognitive symptoms of PACS by accessing the brain via weakened cerebral vasculature – damaged both directly by the virus as well as indirectly via dysregulated cytokine activity.

3. Neuronal and cerebrovascular components of PACS

3.1. Regarding direct SARS-CoV-2 infiltration of the brain

We note that while most investigations to-date have failed to detect widespread SARS-CoV-2 distributions in neuronal or glial cells, some evidence exists of viral entry into the central nervous system via the olfactory bulb – a path that may be related to anosmia that is common in COVID-19 (Boldrini et al., 2021; Meinhardt et al., 2021). Specifically, several autopsy studies have provided evidence for SARS-CoV-2 tropism from the olfactory bulb into the brainstem – a brain region that has a relatively high expression of ACE2 receptors (Lukiw et al., 2022). Such evidence includes brainstem neurodegeneration and presence of viral RNA in the brainstem, with higher concentrations specifically in the medullar subregion (Matschke et al., 2020; Solomon et al., 2020; von Weyhern et al., 2020). However, brainstem neuropathology has also been described in deceased COVID-19 patients’ brains with no detectable viral RNA to accompany such findings, giving credence to hypotheses of indirect neurological damage (Al-Dalahmah et al., 2020; Deigendesch et al., 2020; Fabbri et al., 2020). Direct viral pathogenesis for chronic long COVID-19 is particularly unlikely since the acute disease that precedes PACS is often mild in its severity, motivating our indirect model along the gut-brain axis. Such indirect pathogenic effects can occur due to several COVID-19 comorbidities that have well-established neurological sequelae, and include neuroinflammation, metabolic abnormalities, and cerebrovascular dysfunction (Benghanem et al., 2020; Guedj et al., 2021; Solomon, 2021; Yong, 2021; Philippens et al., 2022).

The aforementioned brainstem findings in long COVID-19 are consistent with its cognitive symptomology that largely revolves around fatigue and brain fog, reflecting prior reports of brainstem abnormalities in chronic fatigue syndrome (CFS) (Zhang et al., 2020a,b; Zhang Y. et al., 2021; Hugon et al., 2022; Thapaliya et al., 2023). Other neuroimaging findings are consistent with this clinical presentation as well, with diffusion weighted imaging (DWI) based microstructural changes reported in the thalamus – another region extensively implicated in CFS (Heine et al., 2023). Using DWI, post-COVID-19 changes have also been reported in multiple major white matter bundles that connect distant cortical parts of the brain, including the corona radiata, corticospinal tract, corpus callosum, arcuate fasciculus, cingulate, fornix, inferior fronto-occipital fasciculus, inferior longitudinal fasciculus, superior longitudinal fasciculus, and uncinate fasciculus (Bispo et al., 2022). In addition to microstructural changes, MRI-based volumetric and density effects of COVID-19 have also been reported across multiple brain regions; however, effect directions have been inconsistent (Harapan and Yoo, 2021; Kamasak et al., 2023). Nevertheless, such neuroimaging-based findings have been consistently reported following mild COVID-19 cases, and the extent of such brain changes appear to reflect the severities of acute-stage disease (Qin et al., 2021; Abbasi, 2022; Taruffi et al., 2023). However, with direct SARS-CoV-2 infection of the brain being an unlikely culprit in PACS, the process by which the virus produces its neurological sequelae remains unclear, motivating the indirect gut-brain axis hypothesis described here.

3.2. Cerebrovascular damage and inflammation

Despite its apparent multi-faceted effects on the central nervous system, it remains unknown how COVID-19 produces its neurocognitive deficits. Early post-mortem investigations of fatal COVID-19 cases suggested that direct infections may indeed occur, yet these cases followed severe and rapid disease progressions, and were accompanied by classic symptoms of brain infections such as encephalitis and cerebral hemorrhage (Siow et al., 2021; Song et al., 2021). In contrast, we argue that damage to the cerebrovascular endothelium plays a major role in underpinning the various sequelae of COVID-19 in the brain. Early histopathological studies of severe COVID-19 showed endothelial injury that co-localized with cerebrovascular expression of angiotensin-converting enzyme 2 (ACE2) receptors as well as the resulting viral interactions with the tissue (Hamming et al., 2004; Zhang L. et al., 2021). SARS-CoV-2 thus appears to directly bind to the endothelium, possibly compromising the blood brain barrier (BBB) and exposing the brain to harmful substances that may be present systemically, activating immune resident cells and leading to neuroinflammation (Almutairi et al., 2021; Theoharides, 2022). While such systemic compounds are described in further detail below, we note that the viral spike protein itself has been speculated to permeate the damaged BBB and to be involved in microglial activation, with subsequent chronic neuroinflammation likely leading to neuronal damage and contributing to PACS neurocognitive effects such as fatigue and brain fog (Boldrini et al., 2021; Theoharides, 2022).

Existing work also highlights the role of perivascular inflammation in endothelial dysfunction after COVID-19 (Huertas et al., 2020). Proinflammatory cytokines such as TNF-alpha, IL-6, and IL-4 are abundantly released in the context of systemic inflammation associated with acute-stage SARS-CoV-2 infection (Gubernatorova et al., 2020), and often remain elevated chronically as well (Gubernatorova et al., 2020; Queiroz et al., 2022; Schultheiß et al., 2022). Monocyte-derived macrophages (MDMs) help maintain such inflammatory profiles long-term, leaving pro-inflammatory imprints in monocytes that produce long-lasting, aberrant immune responses in patients who recovered from COVID-19, with such MDM responses common even in patients who experience mild COVD-19 (Bohnacker et al., 2022). Specifically, SARS-COV-2 spike protein stimulation produces profound cytokine release in macrophages derived from COVID-19 patients, but not those derived from controls who were never infected (Theobald et al., 2021). Leukotrienes (LTs) produced by white blood cells have also been reported to induce and elevate levels of chemokine ligand 2 and fatty acid synthesis in monocytes and leukocytes, suggesting that enhanced LT synthesis may drive exaggerated pro-inflammatory chemokine responses in post COVID-19 MDMs (Pacheco et al., 2007; Merad and Martin, 2020; Ranjbar et al., 2022). Indeed, this long-term presence of proinflammatory compounds in itself has been shown to inflict damage to cerebrovascular endothelium following COVID-19 as well as other infections, exposing the brain to systemic harmful substances and virions via the compromised blood–brain barrier (BBB) (Daniels et al., 2014; Bermejo-Martin et al., 2020; Elyaspour et al., 2021; Nicosia et al., 2021).

Microglial release of cytokines further compounds the inflammatory effects on cerebrovascular endothelium described above, also downregulating astrocytic and pre-synaptic expression of glutamate transporter-1. This in turn leads to disruption of glutamatergic pathways that can potentially induce neuronal excitotoxicity, degeneration, and death (Crunfli et al., 2020; Alenazy et al., 2022). The role of such glutamatergic dysfunction has recently been implicated in PACS neurocognitive symptomology specifically within the dorsolateral prefrontal cortex (Yesilkaya et al., 2021) – a brain region that is part of the executive function network and is involved in complex integration of multimodal information (Vakhtin et al., 2014). Further, the resulting endothelial dysfunction in the brain would be expected to disrupt cerebrovascular autoregulation of blood flow on a broader level, producing lingering neurocognitive effects due to neuronal malnourishment (Nicosia et al., 2021). Crucially, this notion is supported by the increased rates of other, more severe types cerebrovascular pathology associated with COVID-19 such as stroke and microhemorrhage (Fitsiori et al., 2020; Fridman et al., 2020; Sashindranath and Nandurkar, 2021). Collectively, endothelial dysfunction in PACS likely maintains its neurological sequelae via at least the following three ways: 1) exposure of the brain via compromised BBB to harmful substances that 2) accumulate due to inadequate cerebrovascular mechanical action necessary for glymphatic removal of byproducts, reflecting blood dysregulation that 3) malnourishes brain tissue and leads to neuronal dysfunction.

3.3. Neuroimaging of cerebral vasculature and Neuroinflammation

Non-invasive assessment of the neurovascular unit remains elusive. While using gadolinium enhanced imaging to assess blood–brain barrier integrity for research purposes has become controversial due to its toxicity (Rogosnitzky and Branch, 2016), novel techniques such as diffusion-prepared arterial spin labeled perfusion are emerging (Shao et al., 2019). A promising technique for assessing the integrity of the neurovascular unit indirectly is MRI-based cerebrovascular reactivity (CVR) imaging, which measures the hemodynamic response to hypercapnia – increased partial pressure of blood CO2 – that induces vasodilation-related changes in cerebral blood flow (CBF) (Lu et al., 2014). Breathing a weak gas mixture of CO2, a rapid and potent vasodilator, during neuroimaging allows for measurement of a functional vascular response, which stands in contrast to static CBF measures obtained using arterial spin labeling and transcranial Doppler ultrasound. Like conventional functional MRI, CVR also offers voxel-wise quantification of reactivity. In light of these advantages, the technique has seen increased utility in pathologies related to brain vasculature, such as stroke (Fierstra et al., 2018), small vessel disease (Smith and Beaudin, 2018), traumatic brain injury (Dodd et al., 2020), and age-related cognitive impairments (Sur et al., 2020). Given the profound multi-organ vascular effects of COVID-19, CVR is an ideal tool for examining cerebrovascular function in PACS patients. Quantifying systemic Glial Fibrillary Acidic Protein (GFAP) may complement neuroimaging methods such as CVR in assessing the neurovascular unit, specifically with regard to astrocytic roles in BBB integrity. Astrocytes contribute necessary, non-redundant roles in neurovascular unit maintenance by regulating intercellular endothelial associations in BBB tight junction formations and by linking cerebrovascular endothelial blood flux with neurons (Alvarez et al., 2013; Heithoff et al., 2021). Astrocytic activation responds to multiple pathologies involving BBB disruption, and upregulation of astrocytic cytoskeletal protein GFAP is the hallmark of such reactive astrogliosis (Pekny and Nilsson, 2005).

Despite the great utility of positron emission tomography (PET) for measuring neuroinflammation in-vivo for clinical purposes, concerns about its invasive nature as well as high costs often preclude the use of PET in research settings. Non-invasive assessment of neuroinflammation thus remains difficult. Recent MRI-based methods, however, show promise for quantifying regional microglial activation – a part of the innate immune response. Specifically, diffusion weighted magnetic resonance spectroscopy (DW-MRS) can be used to quantify the diffusivity of individual metabolites in the brain (Genovese et al., 2021). The sequence detects microglial activation using apparent diffusion coefficient of cholinergic compounds (ADCCho), which reside in microglial cells in concentrations 3x higher than other brain cells (Urenjak et al., 1993). As microglia undergo morphological changes during activation, changing from a highly branched state to a more spherical one, cholinergic metabolites are able to diffuse more freely within these cells, resulting in higher ADCCho values detected in microglia-rich brain regions such as the thalamus (Schubert et al., 2021). Importantly, injections of lipopolysaccharide – a potent inflammatory endotoxin – have been demonstrated to induce elevated ADCCho in the thalamus (de Marco et al., 2022). DW-MRS may thus be an attractive tool for measuring neuroinflammation in the context of this PACS model along the gut-brain axis.

4. Gastrointestinal components of PACS

4.1. Dysbiosis

The gastrointestinal tract is the largest immune organ within the body, with microbiota regulating host immunity in conjunction with the intestinal mucosal layer (Toor et al., 2019). Although the main target of COVID-19 is the respiratory tract, several lines of evidence point toward substantial involvement of the gastrointestinal tract (Lamers et al., 2020; Meringer and Mehandru, 2022). Gut microbial dysbiosis – a decrease in the diversity of gut flora that allows pathogenic strains to become overrepresented – likely underpins the production of harmful byproducts in the intestine, which can subsequently accumulate within the gut lumen and proliferate systemically via a weakened intestinal barrier (Toor et al., 2019). Given that severe gastrointestinal disturbances are common in the acute stage of COVID-19, there is strong evidence for the resulting gut microbiota dysbiosis that can persist long-term (Villapol, 2020). Crucially, symptoms such as diarrhea also commonly occur in patients who have mild cases of infection and do not require hospitalization (Han et al., 2020). Indeed, microbiome examinations in PACS patients 6 months post-infection revealed decreases in diversity, suggesting that COVID-19-induced dysbiosis may persist chronically and maintain inflammation long-term (Chen et al., 2022; Giannos and Prokopidis, 2022).

Compelling evidence for the role of gut dysbiosis in PACS has been emerging via both observational and interventional studies, partially motivated by its relatively established contributions to chronic fatigue syndrome – a condition that shares several of its hallmark symptoms with PACS (Guo et al., 2023; Xiong et al., 2023). Indeed, deficits of beneficial intestinal microbial strains and increased pathogen concentrations have been reported in both severe and mild COVID-19 patient groups, as well as across acute and chronic disease stages. Blooms of opportunistic and harmful bacteria, including those with antibiotic-resistance, have been reported in hospitalized COVID-19 patients, increasing their susceptibility to multi-drug resistant infections and mortality from septic shock to rates as high as 57% (Grasselli et al., 2021; Nori et al., 2021). In addition, COVID-19 has been associated with decreased microbial production of short-chain fatty acids such as butyrate – an important energy source for colonocyte metabolism and maintenance of colonic mucosal health. Deficits in butyrate-producing bacteria, such as Faecalibacterium prausnitzii, have been shown to reflect acute-stage COVID-19 severities and be associated with systemic proinflammatory cytokine concentrations (Reinold et al., 2021; Yeoh et al., 2021). This microbiome under-representation of F. prausnitzii was found to persist chronically in PACS patients as well, with PACS symptom severities being inversely associated with gut levels of F. prausnitzii and Bifidobacterium pseudocatenulatum – another butyrate-producing bacterial strain (Liu et al., 2022).

4.2. Detection of intestinal dysbiosis

While gut dysbiosis can be assessed using stool samples, this can present challenges in research settings. Lactulose breath testing offers non-invasive assessment of gut microbiota that does not require collection of stool samples, and uses hydrogen and methane concentrations in the breath to detect imbalances in gut microbial flora (Bond et al., 1971). Production of H2 via gut bacterial fermentation is abundant – about 13 L/day (Hartmann et al., 2000; Wolf et al., 2016). While 60–70% is excreted, the rest is used by other gut bacteria (Christl et al., 1992), producing CH4 by reducing CO2, CH4O, and C2H3O2 (Triantafyllou et al., 2014). In humans, H2 and CH4 are entirely microbially-derived, have no biological roles (Rezaie et al., 2017), and readily diffuse into blood via the intestinal wall, which allows them to be measured non-invasively in exhaled breath (Bond et al., 1971; Levitt, 1971). Indeed, H2 and CH4 production in response to lactulose – an indigestible starch that gets fermented in the colon in a dose-dependent manner (Bond and Levitt, 1972; Corazza et al., 1993) – can detect a form of dysbiosis called small intestinal bacterial overgrowth (SIBO) (Rezaie et al., 2017). These two gasses are routinely measured in the clinical setting to estimate gut dysbiosis using a Lactulose Breath Test. A Lactulose Breath Test (LBT) positive for H2 (>20 ppm over baseline) (Rezaie et al., 2017) indicates dysbiosis associated with diarrhea (Chen et al., 2018), while a positive CH4 result (>10 ppm over baseline) (Rezaie et al., 2017) reflects methanogen overgrowth related to constipation (Eckburg et al., 2005; Hwang et al., 2010; Pimentel et al., 2020). Lactulose breath testing may thus offer a useful measure of SIBO for identifying gut dysbiosis in PACS patients.

4.3. Gut microbiota and innate immunity

Type I and type III interferons (IFNs) play crucial roles in fighting viral diseases, and while type I IFN is the dominant phenotype in systemic infections, type III IFN appears to preferentially enforce and strengthen the antiviral response at mucosal sites (Mordstein et al., 2010; Pott et al., 2011). Type III IFN receptors are extensively expressed at these sites, and organs such as the gastrointestinal tract and the lungs respond strongly to systemic expression of type III IFN (Mordstein et al., 2010; Pulverer et al., 2010; Pott and Stockinger, 2017). The microbiome appears to mediate type III IFN production and induce the expression of antiviral IFN-stimulated genes in the intestinal epithelium (van Winkle et al., 2022). Importantly, mucosal microbiomes can modulate IFN responses both locally and remotely, either by systemically releasing metabolites that prime distal immune and epithelial cells, or by being sampled by immune cells that migrate to other organs and influence local immune response (Wirusanti et al., 2022). Microbial translocation itself can also drive the proliferation of altered immune IFN capabilities. For example, epithelial type III IFN promoters become suppressed and its receptors become cleaved in the presence of Porphyromonas gingivalis in the mouth – a strain that is capable of translocation to mucosal surfaces in the gut and respiratory tissues, where it has been associated with dysbiosis and pneumonia, respectively (Nakajima et al., 2015; Benedyk et al., 2016; Rodriguez-Hernandez et al., 2021). With growing evidence that SARS-CoV-2 increases intestinal infections, type III IFN is thus emerging as a potential therapeutic target for both acute and chronic gastrointestinal sequelae of COVID-19 (Pan et al., 2023). Importantly, type III IFN has been shown to be more potent and long-lasting than type I IFN in reducing SARS-CoV-2 viral loads in human intestinal epithelial cells (Metz-Zumaran et al., 2022).

Recently described timelines of the type I and type III IFN responses to SARS-CoV-2 infection offer insight into the mechanisms of proinflammatory cytokine storms during acute COVID-19 as well as its long-lasting effects as part of PACS. As it evolved, the SARS-CoV-2 virus developed strategies to evade and antagonize pattern recognition receptor signaling, inhibiting IFNs and preventing activation of the host innate immune response (Deng et al., 2017; Hackbart et al., 2020; Choi and Shin, 2021). Of particular interest is that this dysregulated IFN response appears to occur in the upper airways and in the early stages of COVID-19, followed by exuberant IFN production in the context of a hyperactive inflammatory response that occurs later in the lungs (Eskandarian Boroujeni et al., 2022). This reversal of the general immunity paradigm – that the IFN-mediated responses precede pro-inflammatory ones – has been shown to be specific to COVID-19, with influenza patients hospitalized for pneumonia showing traditional temporal IFN patterns (Galani et al., 2021; Kim and Shin, 2021). The delayed and exaggerated IFN activity in the acute stage of COVID-19 likely contributes its to long-term neurocognitive effects as well. Post-mortem COVID-19 analyses point to strong expression of type I IFN signatures that are specific to the choroid plexus, despite the absence of RNA and protein traces of SARS-CoV-2 in the tissue (Yang et al., 2021). Such enhanced type I IFN signaling in the choroid plexus has been reported in both healthy aging under non-infectious conditions as well as in neurodegenerative conditions such as Alzheimer’s disease – both of which are associated with decline in cognitive ability – and have been compared to PACS using other molecular signatures (Baruch et al., 2014; Stopa et al., 2018; Mavrikaki et al., 2022; Reiken et al., 2022). The choroid plexus has thus been argued as particularly susceptible to SARS-CoV-2, with the local type I IFN immune response being the possible driver of long-term cognitive deficits that are often reported in PACS (Suzzi et al., 2023). As such, this area may serve as a useful target for further inquiries into the neuropathology of PACS.

4.4. Intestinal barrier damage

In the context of the gut-brain PACS model, dysbiosis would presumably need to be coupled with increased gut permeability due to a compromised intestinal barrier, allowing for neuroinflammatory and neurotoxic substances such as LPS and PGN to permeate systemically. Consistent with the first prong of the proposed PACS pathogenesis along the gut-brain axis, SARS-CoV-2 has well-established effects on the intestinal epithelium that are analogous to those in the cerebrovascular endothelium. ACE2 receptors are extensively expressed by the intestinal enterocytes and serve as viral binding sites, promoting tissue-damaging interactions between SARS-CoV-2 and the epithelium (Cardinale et al., 2020; Alenazy et al., 2022). The resulting compromises to the intestinal barrier facilitate systemic permeability of potentially harmful byproducts produced by the intestinal microbiome, which in turn can influence both local and systemic inflammatory activities, further compounding the runaway inflammation of COVID-19 (Ruff et al., 2020; Delgado-Gonzalez et al., 2021). Butyrate also plays an important role in preserving the intestinal barrier. In addition to supporting of the intestinal mucosal layer, butyrate helps maintain acetylation of histones, affecting the molecular remodeling of chromatin toward a transcriptionally ready state (Candido et al., 1978). In endothelial cells specifically, butyrate effectively suppresses the expression of pro-inflammatory genes and their LPS-induced release (Chriett et al., 2019). As such, depletions of butyrate-producing bacteria have been suggested to exacerbate SARS-CoV-2-induced gut epithelial cell damage via insufficient downregulation of these mechanisms (Li et al., 2021). Butyrate has also been shown to enrich the innate immune viral response via the toll-like receptor signaling pathway, upregulating interleukin-1β, interferon regulatory factor-7, and interferon-alpha/beta receptor at mRNA and protein levels (Li et al., 2021). As such, butyrate appears likely to be involved in the innate immune response to SARS-CoV-2.

4.5. Markers of intestinal barrier damage

Intestinal fatty acid binding protein (FABP2) is expressed by intestinal enterocytes, largely in the absorptive parts of the intestinal epithelial villi, where it is involved in fatty acid absorption and transport (Gajda and Storch, 2015). While FABP2 is not a measure of transcellular permeability proper, such as Horseradish Peroxidase or Fluorescent Labeled Particles, it has emerged as a sensitive biomarker of intestinal epithelial dysfunction due to its physical cellular disentegration (Pelsers et al., 2003). For example, the intestinal epithelium is damaged during intestinal ischemia, and FABP2 is released into the blood, where it has been shown to reliably reflect such damage in humans (Kanda et al., 1992; Montagnana et al., 2018). Indeed, intestinal and mesenteric ischemia has recently emerged as both a presenting feature and a late complication of COVID-19 during hospitalization (Norsa et al., 2020; Kiwango et al., 2021; Singh and Kaur, 2021), with approximately one-third of such patients having ultimately succumbed to the disease (Keshavarz et al., 2021). While such severe cases warranted the computed tomography scans that ultimately revealed profound cases of gastrointestinal ischemia, increases in FABP2 have been detected in less severe COVID-19 cases as well (Prasad et al., 2021). However, there remains the question of how prevalent intestinal microinfarctions are in mild disease severities, and whether they are associated with increased FABP2 plasma levels. As such, whether the epithelial damage is sustained via direct viral infiltration or indirectly by intestinal ischemia, FABP2 may a highly useful metric for quantifying intestinal permeability in PACS.

Zonulin, on the other hand, is specific to paracellular epithelial damage, and is often accompanied by transcellular permeability as well, as detected by aforementioned FABP2. Zonulin modulates the abilities of tight junctions of intestinal epithelial cells to regulate paracellular permeability (Fasano et al., 2000). Specifically, increased concentrations of zonulin disassemble ‘zonula occludens’ proteins, structurally disrupting the tight junction complex (Fasano, 2012). Indeed, increased blood plasma concentrations of zonulin have been suggested to reflect intestinal permeability across several conditions (Sapone et al., 2006; Ganda Mall et al., 2018), and are closely linked to plasma concentrations of endotoxin Lipopolysaccharide and transcellular permeability biomarker FABP2 (Stevens et al., 2018). Zonulin release has additionally been implicated in gut permeability associated with celiac disease (Lammers et al., 2008) and proposed to play roles in various inflammatory and autoimmune disorders (Fasano, 2011; Asbjornsdottir et al., 2020). This leads us to speculate that similar chronic gastrointestinal disturbances in PACS patients may be due to elevated zonulin levels, with increased intestinal permeability allowing for dysbiosis-induced endotoxins to permeate systemically.

4.6. Systemic translocation of microbial products

Lipopolysaccharide (LPS) is a major component of the outer membrane of Gram-negative bacteria in the gut (Raetz and Whitfield, 2002), and its presence induces systemic inflammation as well as microglial activation in the brain (Dobrovolskaia and Vogel, 2002; Brown, 2019). The endotoxin has previously been found in elevated levels in hospitalized COVID-19 patients, but its presence in the post-acute disease stage and contribution to the neurocognitive PACS symptoms remains unclear (Prasad et al., 2021; Teixeira et al., 2021). Importantly, direct LPS infiltration of the brain has been demonstrated in rats, where it binds to endothelial cell receptors at blood–brain interfaces, thus further exacerbating the endothelial damage inflicted by the virus and cytokines described above (Vargas-Caraveo et al., 2017). Of particular interest is that LPS has been shown to disrupt the blood–brain barrier in some brain regions, but not in others, impacting the BBB in the thalamus, frontal cortex, cerebellum, and pons-medulla. Thalamic and frontal impairments are of particular interest in the context of PACS, as these regions are important to higher-order cognitive processes (Jung and Haier, 2007; Vakhtin et al., 2014). Systemic LPS may thus have a triple impact in post-COVID-19 pathology by stimulating pro-inflammatory cytokines and macrophage activation, directly interacting with the cerebrovascular endothelium concurrently with the virus, as well as increasing the exposure of some brain regions to neurotoxins via the compromised BBB.

Another common microbial byproduct that may contribute to systemic endotoxicity in PACS is peptidoglycan (PGN). While the term “endotoxin” has until recently been synonymous with LPS, PGN has emerged as a gram-positive counterpart of LPS in the recent years (Myhre et al., 2006). This was largely driven by the increasing rates of sepsis due to gram-positive organisms, which have recently overcome those due to gram-negative bacteria (Mayr et al., 2014). PGN concentrations in blood plasma have also been shown to reflect intestinal permeability due to ischemia, hemorrhagic shock, and ethanol-induced injury (Shimizu et al., 2002; Tabata et al., 2002; Tsunooka et al., 2004). Importantly, the presence of gut-derived PGN in brain dendritic cells and macrophages has been associated with idiopathic inflammatory and autoimmune conditions, such as multiple sclerosis (Schrijver et al., 2001; Visser et al., 2005). Further, the neurotoxicity of PGN has been demonstrated in rats, where it induced acute microglial and astrocytic nitric oxide production, which mediate neuronal cell death (Boje and Arora, 1992; Kim and Täuber, 1996; Buskila et al., 2005). Beyond innate immunity, PGN has also been implicated in pathogenesis of neurodevelopment and its associated disorders, such as autism spectrum disorder, which may be related to the potential cognitive sequelae of its neurotoxic properties (Arentsen et al., 2017; Gonzalez-Santana and Diaz Heijtz, 2020). Like LPS, concentrations of PGN in blood plasma have also been shown to be elevated in hospitalized COVID-19 patients (Prasad et al., 2021). As such, PGN concentration is a potential contributor to persistent neuroinflammation and neurotoxicity in the PACS framework along the gut-brain axis.

5. Conclusion

Here we described a comprehensive model for long COVID-19 along the gut-brain axis. In summary, neurocognitive PACS symptoms may persist because viral damage to the blood–brain and the intestinal barriers may allow for unchecked flow of harmful substances produced in the gut lumen in the context of dysbiosis. This indirect pathogenesis stands in contrast to models that ascribe the neurocognitive symptoms of PACS to direct effects of the virus, regardless of whether they occur during the acute stage or are maintained chronically by remnant viral reservoirs (Proal and VanElzakker, 2021; Swank et al., 2023). Likewise, our model also circumvents the hypothesized reactivation of existing underlying pathogens such as the Epstein–Barr virus (Peluso et al., 2023). It does, however, unify several other hypotheses for the pathogenesis of PACS and its neurocognitive symptoms, including innate immune dysregulation, impact on microbiota, as well as endothelial and epithelial damage and dysfunction that can lead to degradation of brain and intestinal barriers, respectively (Arthur et al., 2021; Haffke et al., 2022; Davis et al., 2023). Similar models along the gut-brain axis have been described previously (Vakili et al., 2022; Gareau and Barrett, 2023), and while our narrative for the initiation and maintenance of neurocognitive symptomology reflects these existing models, we also elaborate on the specific metrics that can be quantified experimentally to assess each component of the model.

The described mechanism of gut-brain pathology has been described extensively in other disorders, such as depression (Valles-Colomer et al., 2019), anxiety (Kim and Shin, 2018), neurodegenerative conditions (Ryman et al., 2023), and other chronic multi-symptom illnesses such as Gulf War illness (Alhasson et al., 2017; Bajaj et al., 2019; Keating et al., 2019). Systemic inflammation due to increased permeability in the gut has been implicated in conditions such as irritable bowel syndrome (Camilleri et al., 2012; Fukui, 2016; Moser et al., 2018), and these pathways are highly relevant to COVID-19 due to its well-established cytokine storms that can result in fatal hyper-inflammation (Cron et al., 2021). While SARS-CoV-2 infection fits particularly well into this model due to its utilization of ACE2 receptors, which are expressed extensively in the brain endothelium and the intestinal epithelium, we note that this pathogenesis may be applicable to other psychiatric as well as neurodegenerative conditions.

Multiple studies examining gut-based interventions in PACS are ongoing as of this writing, motivated by emerging data on probiotic effectiveness in alleviating COVID-19 symptoms. Blinded, randomized, and placebo-controlled probiotic treatment of symptomatic outpatient COVID-19 patients has been shown to modulate the immune response, significantly increasing SARS-CoV-2-specific IgM and IgG levels (Gutiérrez-Castrellón et al., 2022). In-patients with moderate-to-severe COVID-19 who received single-strain probiotic Bifidobacterium boosters were found to have significantly shorter hospital stays, IL-6 level reductions, radiological lung improvements, and lower mortality rates relative to the non-probiotic group (Bozkurt and Bilen, 2021). Probiotic fecal microbial transplantation (FMT) has been shown to produce rapid resolution of COVID-19 in acute-stage patient case reports as well (Biliński et al., 2022). Collectively, these interventions hold promise in alleviating the neurocognitive symptoms of PACS indirectly via the gut and in the absence of direct interventions acting on the central nervous system – as few such approaches exist currently.

Data availability statement

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

Author contributions

AP wrote the first draft of the manuscript. YM conducted literature review and organization. HL, SR, DQ, AB, and ANP contributed to the conception and design of the manuscript, and offered clinical expertise on the subject matter. AV contributed to conception and finalization of the manuscript, as well as refinement of technical and methodological sections. All authors contributed to the article and approved the submitted version.

Funding

This work was funded in part by the National Institute of Neurological Disorders and Stroke (R01NS129407) and the Winkler Bacterial Overgrowth Research Fund.

Conflict of interest

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

Publisher’s note

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

References

Abbasi, J. (2022). Even mild COVID-19 may change the brain. JAMA 327:1321. doi: 10.1001/jama.2022.4507

PubMed Abstract | CrossRef Full Text | Google Scholar

Al-Dalahmah, O., Thakur, K. T., Nordvig, A. S., Prust, M. L., Roth, W., Lignelli, A., et al. (2020). Neuronophagia and microglial nodules in a SARS-CoV-2 patient with cerebellar hemorrhage. Acta Neuropathol. Commun. 8:147. doi: 10.1186/s40478-020-01024-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Alenazy, M. F., Aljohar, H. I., Alruwaili, A. R., Daghestani, M. H., Alonazi, M. A., Labban, R. S., et al. (2022). Gut microbiota dynamics in relation to Long-COVID-19 syndrome: role of probiotics to combat psychiatric complications. Meta 12:912. doi: 10.3390/metabo12100912

PubMed Abstract | CrossRef Full Text | Google Scholar

Alhasson, F., das, S., Seth, R., Dattaroy, D., Chandrashekaran, V., Ryan, C. N., et al. (2017). Altered gut microbiome in a mouse model of gulf war illness causes neuroinflammation and intestinal injury via leaky gut and TLR4 activation. PLoS One 12:e0172914. doi: 10.1371/journal.pone.0172914

PubMed Abstract | CrossRef Full Text | Google Scholar

Almutairi, M. M., Sivandzade, F., Albekairi, T. H., Alqahtani, F., and Cucullo, L. (2021). Neuroinflammation and its impact on the pathogenesis of COVID-19. Front. Med. 8:745789. doi: 10.3389/fmed.2021.745789

PubMed Abstract | CrossRef Full Text | Google Scholar

Alvarez, J. I., Katayama, T., and Prat, A. (2013). Glial influence on the blood brain barrier. Glia 61, 1939–1958. doi: 10.1002/glia.22575

PubMed Abstract | CrossRef Full Text | Google Scholar

Arentsen, T., Qian, Y., Gkotzis, S., Femenia, T., Wang, T., Udekwu, K., et al. (2017). The bacterial peptidoglycan-sensing molecule Pglyrp2 modulates brain development and behavior. Mol. Psychiatry 22, 257–266. doi: 10.1038/mp.2016.182

PubMed Abstract | CrossRef Full Text | Google Scholar

Arthur, J. M., Forrest, J. C., Boehme, K. W., Kennedy, J. L., Owens, S., Herzog, C., et al. (2021). Development of ACE2 autoantibodies after SARS-CoV-2 infection. PLoS One 16:e0257016. doi: 10.1371/journal.pone.0257016

PubMed Abstract | CrossRef Full Text | Google Scholar

Asbjornsdottir, B., Snorradottir, H., Andresdottir, E., Fasano, A., Lauth, B., Gudmundsson, L. S., et al. (2020). Zonulin-dependent intestinal permeability in children diagnosed with mental disorders: a systematic review and Meta-analysis. Nutrients 12:1982. doi: 10.3390/nu12071982

PubMed Abstract | CrossRef Full Text | Google Scholar

Bajaj, J. S., Sharma, A., Dudeja, P. K., Iqbal, Z., Singh, A. B., Wilson, K. T., et al. (2019). Targeting gut microbiome interactions in service-related gastrointestinal and liver diseases of veterans. Gastroenterology 157, 1180–1183.e1. doi: 10.1053/j.gastro.2019.07.060

PubMed Abstract | CrossRef Full Text | Google Scholar

Baruch, K., Deczkowska, A., David, E., Castellano, J. M., Miller, O., Kertser, A., et al. (2014). Aging-induced type I interferon response at the choroid plexus negatively affects brain function. Science 346, 89–93. doi: 10.1126/science.1252945

PubMed Abstract | CrossRef Full Text | Google Scholar

Benedyk, M., Mydel, P. M., Delaleu, N., Płaza, K., Gawron, K., Milewska, A., et al. (2016). Gingipains: critical Factors in the development of aspiration pneumonia caused by Porphyromonas gingivalis. J. Innate Immun. 8, 185–198. doi: 10.1159/000441724

PubMed Abstract | CrossRef Full Text | Google Scholar

Benghanem, S., Mazeraud, A., Azabou, E., Chhor, V., Shinotsuka, C. R., Claassen, J., et al. (2020). Brainstem dysfunction in critically ill patients. Crit. Care Lond. Engl. 24:5. doi: 10.1186/s13054-019-2718-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Bermejo-Martin, J. F., Almansa, R., Torres, A., González-Rivera, M., and Kelvin, D. J. (2020). COVID-19 as a cardiovascular disease: the potential role of chronic endothelial dysfunction. Cardiovasc. Res. 116, e132–e133. doi: 10.1093/cvr/cvaa140

PubMed Abstract | CrossRef Full Text | Google Scholar

Biliński, J., Winter, K., Jasiński, M., Szczęś, A., Bilinska, N., Mullish, B. H., et al. (2022). Rapid resolution of COVID-19 after faecal microbiota transplantation. Gut 71, 230–232. doi: 10.1136/gutjnl-2021-325010

PubMed Abstract | CrossRef Full Text | Google Scholar

Bispo, D. D. D. C., Brandão, P. R. P., Pereira, D. A., Maluf, F. B., Dias, B. A., Paranhos, H. R., et al. (2022). Brain microstructural changes and fatigue after COVID-19. Front. Neurol. 13:1029302. doi: 10.3389/fneur.2022.1029302

PubMed Abstract | CrossRef Full Text | Google Scholar

Bohnacker, S., Hartung, F., Henkel, F., Quaranta, A., Kolmert, J., Priller, A., et al. (2022). Mild COVID-19 imprints a long-term inflammatory eicosanoid- and chemokine memory in monocyte-derived macrophages. Mucosal Immunol. 15, 515–524. doi: 10.1038/s41385-021-00482-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Boje, K. M., and Arora, P. K. (1992). Microglial-produced nitric oxide and reactive nitrogen oxides mediate neuronal cell death. Brain Res. 587, 250–256. doi: 10.1016/0006-8993(92)91004-X

PubMed Abstract | CrossRef Full Text | Google Scholar

Boldrini, M., Canoll, P. D., and Klein, R. S. (2021). How COVID-19 affects the brain. JAMA Psychiatry 78:682. doi: 10.1001/jamapsychiatry.2021.0500

PubMed Abstract | CrossRef Full Text | Google Scholar

Bond, J. H. Jr., Engel, R. R., and Levitt, M. D. (1971). Factors influencing pulmonary methane excretion in man: an indirect method of studying the in situ metabolism of the methane-producing colonic BACTERIA. J. Exp. Med. 133, 572–588. doi: 10.1084/jem.133.3.572

PubMed Abstract | CrossRef Full Text | Google Scholar

Bond, J. H., and Levitt, M. D. (1972). Use of pulmonary hydrogen (H2) measurements to quantitate carbohydrate absorption: study of partially gastrectomized patients. J. Clin. Invest. 51, 1219–1225. doi: 10.1172/JCI106916

PubMed Abstract | CrossRef Full Text | Google Scholar

Bozkurt, H. S., and Bilen, Ö. (2021). Oral booster probiotic bifidobacteria in SARS-COV-2 patients. Int. J. Immunopathol. Pharmacol. 35:20587384211059676. doi: 10.1177/20587384211059677

CrossRef Full Text | Google Scholar

Brown, G. C. (2019). The endotoxin hypothesis of neurodegeneration. J. Neuroinflammation 16:180. doi: 10.1186/s12974-019-1564-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Buskila, Y., Farkash, S., Hershfinkel, M., and Amitai, Y. (2005). Rapid and reactive nitric oxide production by astrocytes in mouse neocortical slices. Glia 52, 169–176. doi: 10.1002/glia.20217

PubMed Abstract | CrossRef Full Text | Google Scholar

Buzhdygan, T. P., DeOre, B. J., Baldwin-Leclair, A., Bullock, T. A., McGary, H. M., Khan, J. A., et al. (2020). The SARS-CoV-2 spike protein alters barrier function in 2D static and 3D microfluidic in-vitro models of the human blood–brain barrier. Neurobiol. Dis. 146:105131. doi: 10.1016/j.nbd.2020.105131

PubMed Abstract | CrossRef Full Text | Google Scholar

Camilleri, M., Lasch, K., and Zhou, W. (2012). Irritable bowel syndrome: methods, mechanisms, and pathophysiology. The confluence of increased permeability, inflammation, and pain in irritable bowel syndrome. Am. J. Physiol.-Gastrointest. Liver Physiol. 303, G775–G785. doi: 10.1152/ajpgi.00155.2012

PubMed Abstract | CrossRef Full Text | Google Scholar

Candido, E. P. M., Reeves, R., and Davie, J. R. (1978). Sodium butyrate inhibits histone deacetylation in cultured cells. Cells 14, 105–113. doi: 10.1016/0092-8674(78)90305-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Cardinale, V., Capurso, G., Ianiro, G., Gasbarrini, A., Arcidiacono, P. G., and Alvaro, D. (2020). Intestinal permeability changes with bacterial translocation as key events modulating systemic host immune response to SARS-CoV-2: a working hypothesis. Ital. J. Gastroenterol. Hepatol. 52, 1383–1389. doi: 10.1016/j.dld.2020.09.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, Y., Gu, S., Chen, Y., Lu, H., Shi, D., Guo, J., et al. (2022). Six-month follow-up of gut microbiota richness in patients with COVID-19. Gut 71, 222–225. doi: 10.1136/gutjnl-2021-324090

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, B., Kim, J. J.-W., Zhang, Y., Du, L., and Dai, N. (2018). Prevalence and predictors of small intestinal bacterial overgrowth in irritable bowel syndrome: a systematic review and meta-analysis. J. Gastroenterol. 53, 807–818. doi: 10.1007/s00535-018-1476-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Choi, H., and Shin, E.-C. (2021). Roles of type I and III interferons in COVID-19. Yonsei Med. J. 62, 381–390. doi: 10.3349/ymj.2021.62.5.381

PubMed Abstract | CrossRef Full Text | Google Scholar

Choudhury, A., Tariq, R., Jena, A., Vesely, E. K., Singh, S., Khanna, S., et al. (2022). Gastrointestinal manifestations of long COVID: a systematic review and meta-analysis. Ther. Adv. Gastroenterol. 15:175628482211184. doi: 10.1177/17562848221118403

PubMed Abstract | CrossRef Full Text | Google Scholar

Chriett, S., Dąbek, A., Wojtala, M., Vidal, H., Balcerczyk, A., and Pirola, L. (2019). Prominent action of butyrate over β-hydroxybutyrate as histone deacetylase inhibitor, transcriptional modulator and anti-inflammatory molecule. Sci. Rep. 9:742. doi: 10.1038/s41598-018-36941-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Christl, S. U., Murgatroyd, P. R., Gibson, G. R., and Cummings, J. H. (1992). Production, metabolism, and excretion of hydrogen in the large intestine. Gastroenterology 102, 1269–1277. doi: 10.1016/0016-5085(92)90765-Q

PubMed Abstract | CrossRef Full Text | Google Scholar

Corazza, G., Strocchi, A., Sorge, M., Benati, G., and Gasbarrini, G. (1993). Prevalence and consistency of low breath H2 excretion following lactulose ingestion. Dig. Dis. Sci. 38, 2010–2016. doi: 10.1007/BF01297077

PubMed Abstract | CrossRef Full Text | Google Scholar

Cron, R. Q., Caricchio, R., and Chatham, W. W. (2021). Calming the cytokine storm in COVID-19. Nat. Med. 27, 1674–1675. doi: 10.1038/s41591-021-01500-9

CrossRef Full Text | Google Scholar

Crunfli, F., Carregari, V. C., Veras, F. P., Silva, L. S., Nogueira, M. H., Antunes, A. S. L. M., et al. (2020). Morphological, cellular and molecular basis of brain infection in COVID-19 patients. Proc. Natl. Acad. Sci. U. S. A. 119:e2200960119. doi: 10.1101/2020.10.09.20207464

CrossRef Full Text | Google Scholar

Daniels, B. P., Holman, D. W., Cruz-Orengo, L., Jujjavarapu, H., Durrant, D. M., and Klein, R. S. (2014). Viral pathogen-associated molecular patterns regulate blood-brain barrier integrity via competing innate cytokine signals. MBio 5, e01476–e01414. doi: 10.1128/mBio.01476-14

PubMed Abstract | CrossRef Full Text | Google Scholar

Davis, H. E., Assaf, G. S., McCorkell, L., Wei, H., Low, R. J., Re'em, Y., et al. (2021). Characterizing long COVID in an international cohort: 7 months of symptoms and their impact. EClinicalMedicine 38:101019. doi: 10.1016/j.eclinm.2021.101019

CrossRef Full Text | Google Scholar

Davis, H. E., McCorkell, L., Vogel, J. M., and Topol, E. J. (2023). Long COVID: major findings, mechanisms and recommendations. Nat. Rev. Microbiol. 21, 133–146. doi: 10.1038/s41579-022-00846-2

PubMed Abstract | CrossRef Full Text | Google Scholar

de Marco, R., Ronen, I., Branzoli, F., Amato, M. L., Asllani, I., Colasanti, A., et al. (2022). Diffusion-weighted MR spectroscopy (DW-MRS) is sensitive to LPS-induced changes in human glial morphometry: a preliminary study. Brain Behav. Immun. 99, 256–265. doi: 10.1016/j.bbi.2021.10.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Deigendesch, N., Sironi, L., Kutza, M., Wischnewski, S., Fuchs, V., Hench, J., et al. (2020). Correlates of critical illness-related encephalopathy predominate postmortem COVID-19 neuropathology. Acta Neuropathol. (Berl.) 140, 583–586. doi: 10.1007/s00401-020-02213-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Delgado-Gonzalez, P., Gonzalez-Villarreal, C. A., Roacho-Perez, J. A., Quiroz-Reyes, A. G., Islas, J. F., Delgado-Gallegos, J. L., et al. (2021). Inflammatory effect on the gastrointestinal system associated with COVID-19. World J. Gastroenterol. 27, 4160–4171. doi: 10.3748/wjg.v27.i26.4160

PubMed Abstract | CrossRef Full Text | Google Scholar

Deng, X., Hackbart, M., Mettelman, R. C., O’Brien, A., Mielech, A. M., Yi, G., et al. (2017). Coronavirus nonstructural protein 15 mediates evasion of dsRNA sensors and limits apoptosis in macrophages. Proc. Natl. Acad. Sci. 114, E4251–E4260. doi: 10.1073/pnas.1618310114

PubMed Abstract | CrossRef Full Text | Google Scholar

Dobrovolskaia, M. A., and Vogel, S. N. (2002). Toll receptors, CD14, and macrophage activation and deactivation by LPS. Microbes Infect. 4, 903–914. doi: 10.1016/S1286-4579(02)01613-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Dodd, A. B., Lu, H., Wertz, C. J., Ling, J. M., Shaff, N. A., Wasserott, B. C., et al. (2020). Persistent alterations in cerebrovascular reactivity in response to hypercapnia following pediatric mild traumatic brain injury. J. Cereb. Blood Flow Metab. Off. J. Int. Soc. Cereb. Blood Flow Metab. 40, 2491–2504. doi: 10.1177/0271678X19896883

PubMed Abstract | CrossRef Full Text | Google Scholar

Eckburg, P. B., Bik, E. M., Bernstein, C. N., Purdom, E., Dethlefsen, L., Sargent, M., et al. (2005). Diversity of the human intestinal microbial Flora. Science 308, 1635–1638. doi: 10.1126/science.1110591

PubMed Abstract | CrossRef Full Text | Google Scholar

Elyaspour, Z., Zibaeenezhad, M. J., Razmkhah, M., and Razeghian-Jahromi, I. (2021). Is it all about endothelial dysfunction and thrombosis formation? The secret of COVID-19. Clin. Appl. Thromb. 27:10760296211042940. doi: 10.1177/10760296211042940

PubMed Abstract | CrossRef Full Text | Google Scholar

Eskandarian Boroujeni, M., Sekrecka, A., Antonczyk, A., Hassani, S., Sekrecki, M., Nowicka, H., et al. (2022). Dysregulated interferon response and immune Hyperactivation in severe COVID-19: targeting STATs as a novel therapeutic strategy. Front. Immunol. 13. doi: 10.3389/fimmu.2022.888897

PubMed Abstract | CrossRef Full Text | Google Scholar

Fabbri, V. P., Foschini, M. P., Lazzarotto, T., Gabrielli, L., Cenacchi, G., Gallo, C., et al. (2020). Brain ischemic injury in COVID-19-infected patients: a series of 10 post-mortem cases. Brain Pathol. 31, 205–210. doi: 10.1111/bpa.12901

CrossRef Full Text | Google Scholar

Fasano, A. (2011). Zonulin and its regulation of intestinal barrier function: the biological door to inflammation, autoimmunity, and cancer. Physiol. Rev. 91, 151–175. doi: 10.1152/physrev.00003.2008

PubMed Abstract | CrossRef Full Text | Google Scholar

Fasano, A. (2012). Zonulin, regulation of tight junctions, and autoimmune diseases. Ann. N. Y. Acad. Sci. 1258, 25–33. doi: 10.1111/j.1749-6632.2012.06538.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Fasano, A., Not, T., Wang, W., Uzzau, S., Berti, I., Tommasini, A., et al. (2000). Zonulin, a newly discovered modulator of intestinal permeability, and its expression in coeliac disease. Lancet (North American ed) 355, 1518–1519. doi: 10.1016/S0140-6736(00)02169-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Fierstra, J., van Niftrik, C., Warnock, G., Wegener, S., Piccirelli, M., Pangalu, A., et al. (2018). Staging hemodynamic failure with blood oxygen-level–dependent functional magnetic resonance imaging cerebrovascular reactivity. Stroke 49, 621–629. doi: 10.1161/STROKEAHA.117.020010

PubMed Abstract | CrossRef Full Text | Google Scholar

Fitsiori, A., Pugin, D., Thieffry, C., Lalive, P., and Vargas, M. I. (2020). COVID-19 is associated with an unusual pattern of brain microbleeds in critically ill patients. J. Neuroimaging 30, 593–597. doi: 10.1111/jon.12755

PubMed Abstract | CrossRef Full Text | Google Scholar

Fridman, S., Bres Bullrich, M., Jimenez-Ruiz, A., Costantini, P., Shah, P., Just, C., et al. (2020). Stroke risk, phenotypes, and death in COVID-19: systematic review and newly reported cases. Neurology 95, e3373–e3385. doi: 10.1212/WNL.0000000000010851

PubMed Abstract | CrossRef Full Text | Google Scholar

Fukui, H. (2016). Increased intestinal permeability and decreased barrier function: does it really influence the risk of inflammation? Inflamm. Intest. Dis. 1, 135–145. doi: 10.1159/000447252

PubMed Abstract | CrossRef Full Text | Google Scholar

Gajda, A. M., and Storch, J. (2015). Enterocyte fatty acid-binding proteins (FABPs): different functions of liver and intestinal FABPs in the intestine. Prostaglandins Leukot. Essent. Fatty Acids 93, 9–16. doi: 10.1016/j.plefa.2014.10.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Galani, I.-E., Rovina, N., Lampropoulou, V., Triantafyllia, V., Manioudaki, M., Pavlos, E., et al. (2021). Untuned antiviral immunity in COVID-19 revealed by temporal type I/III interferon patterns and flu comparison. Nat. Immunol. 22, 32–40. doi: 10.1038/s41590-020-00840-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Ganda Mall, J.-P., Östlund-Lagerström, L., Lindqvist, C. M., Algilani, S., Rasoal, D., Repsilber, D., et al. (2018). Are self-reported gastrointestinal symptoms among older adults associated with increased intestinal permeability and psychological distress? BMC Geriatr. 18:75. doi: 10.1186/s12877-018-0767-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Gareau, M. G., and Barrett, K. E. (2023). Role of the microbiota-gut-brain axis in postacute COVID syndrome. Am. J. Physiol. - Gastrointest. Liver Physiol. 324, G322–G328. doi: 10.1152/ajpgi.00293.2022

PubMed Abstract | CrossRef Full Text | Google Scholar

Genovese, G., Marjańska, M., Auerbach, E. J., Cherif, L. Y., Ronen, I., Lehéricy, S., et al. (2021). In vivo diffusion-weighted MRS using semi-LASER in the human brain at 3 T: methodological aspects and clinical feasibility. NMR Biomed. 34:e4206. doi: 10.1002/nbm.4206

PubMed Abstract | CrossRef Full Text | Google Scholar

Giannos, P., and Prokopidis, K. (2022). Gut dysbiosis and long COVID-19: feeling gutted. J. Med. Virol. 94, 2917–2918. doi: 10.1002/jmv.27684

PubMed Abstract | CrossRef Full Text | Google Scholar

Gonzalez-Santana, A., and Diaz Heijtz, R. (2020). Bacterial peptidoglycans from microbiota in neurodevelopment and behavior. Trends Mol. Med. 26, 729–743. doi: 10.1016/j.molmed.2020.05.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Grasselli, G., Scaravilli, V., Mangioni, D., Scudeller, L., Alagna, L., Bartoletti, M., et al. (2021). Hospital-acquired infections in critically ill patients with COVID-19. Chest 160, 454–465. doi: 10.1016/j.chest.2021.04.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Gubernatorova, E. O., Gorshkova, E. A., Polinova, A. I., and Drutskaya, M. S. (2020). IL-6: relevance for immunopathology of SARS-CoV-2. Cytokine Growth Factor Rev. 53, 13–24. doi: 10.1016/j.cytogfr.2020.05.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Guedj, E., Campion, J. Y., Dudouet, P., Kaphan, E., Bregeon, F., Tissot-Dupont, H., et al. (2021). 18F-FDG brain PET hypometabolism in patients with long COVID. Eur. J. Nucl. Med. Mol. Imaging 48, 2823–2833. doi: 10.1007/s00259-021-05215-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Guo, C., Che, X., Briese, T., Ranjan, A., Allicock, O., Yates, R. A., et al. (2023). Deficient butyrate-producing capacity in the gut microbiome is associated with bacterial network disturbances and fatigue symptoms in ME/CFS. Cell Host Microbe 31, 288–304.e8. doi: 10.1016/j.chom.2023.01.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Gutiérrez-Castrellón, P., Gandara-Martí, T., Abreu Y Abreu, A. T., Nieto-Rufino, C. D., López-Orduña, E., Jiménez-Escobar, I., et al. (2022). Probiotic improves symptomatic and viral clearance in Covid19 outpatients: a randomized, quadruple-blinded, placebo-controlled trial. Gut Microbes 14:2018899. doi: 10.1080/19490976.2021.2018899

PubMed Abstract | CrossRef Full Text | Google Scholar

Hackbart, M., Deng, X., and Baker, S. C. (2020). Coronavirus endoribonuclease targets viral polyuridine sequences to evade activating host sensors. Proc. Natl. Acad. Sci. 117, 8094–8103. doi: 10.1073/pnas.1921485117

PubMed Abstract | CrossRef Full Text | Google Scholar

Haffke, M., Freitag, H., Rudolf, G., Seifert, M., Doehner, W., Scherbakov, N., et al. (2022). Endothelial dysfunction and altered endothelial biomarkers in patients with post-COVID-19 syndrome and chronic fatigue syndrome (ME/CFS). J. Transl. Med. 20:138. doi: 10.1186/s12967-022-03346-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Hamming, I., Timens, W., Bulthuis, M. L. C., Lely, A. T., Navis, G. J., and van Goor, H. (2004). Tissue distribution of ACE2 protein, the functional receptor for SARS coronavirus. A first step in understanding SARS pathogenesis. J. Pathol. 203, 631–637. doi: 10.1002/path.1570

PubMed Abstract | CrossRef Full Text | Google Scholar

Han, C., Duan, C., Zhang, S., Spiegel, B., Shi, H., Wang, W., et al. (2020). Digestive symptoms in COVID-19 patients with mild disease severity: clinical presentation, stool viral RNA testing, and outcomes. Am. J. Gastroenterol. 115, 916–923. doi: 10.14309/ajg.0000000000000664

PubMed Abstract | CrossRef Full Text | Google Scholar

Han, Q., Zheng, B., Daines, L., and Sheikh, A. (2022). Long-term sequelae of COVID-19: a systematic review and Meta-analysis of one-year follow-up studies on post-COVID symptoms. Pathogens 11:269. doi: 10.3390/pathogens11020269

PubMed Abstract | CrossRef Full Text | Google Scholar

Harapan, B. N., and Yoo, H. J. (2021). Neurological symptoms, manifestations, and complications associated with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and coronavirus disease 19 (COVID-19). J. Neurol. 268, 3059–3071. doi: 10.1007/s00415-021-10406-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Hartmann, L., Taras, D., and Kamlage, B. (2000). A new technique to determine hydrogen excreted by gnotobiotic rats. Lab. Anim. 34, 162–170. doi: 10.1258/002367700780457617

PubMed Abstract | CrossRef Full Text | Google Scholar

Heine, J., Schwichtenberg, K., Hartung, T. J., Rekers, S., Chien, C., Boesl, F., et al. (2023). Structural brain changes in patients with post-COVID fatigue: a prospective observational study. eClinicalMedicine 58:101874. doi: 10.1016/j.eclinm.2023.101874

PubMed Abstract | CrossRef Full Text | Google Scholar

Heithoff, B. P., George, K. K., Phares, A. N., Zuidhoek, I. A., Munoz-Ballester, C., and Robel, S. (2021). Astrocytes are necessary for blood–brain barrier maintenance in the adult mouse brain. Glia 69, 436–472. doi: 10.1002/glia.23908

PubMed Abstract | CrossRef Full Text | Google Scholar

Huertas, A., Montani, D., Savale, L., Pichon, J., Tu, L., Parent, F., et al. (2020). Endothelial cell dysfunction: a major player in SARS-CoV-2 infection (COVID-19)? Eur. Respir. J. 56:2001634. doi: 10.1183/13993003.01634-2020

PubMed Abstract | CrossRef Full Text | Google Scholar

Hugon, J., Queneau, M., Sanchez Ortiz, M., Msika, E. F., Farid, K., and Paquet, C. (2022). Cognitive decline and brainstem hypometabolism in long COVID: a case series. Brain Behav. 12:e2513. doi: 10.1002/brb3.2513

PubMed Abstract | CrossRef Full Text | Google Scholar

Hwang, L., Low, K., Khoshini, R., Melmed, G., Sahakian, A., Makhani, M., et al. (2010). Evaluating breath methane as a diagnostic test for constipation-predominant IBS. Dig. Dis. Sci. 55, 398–403. doi: 10.1007/s10620-009-0778-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Johnson, K. D., Harris, C., Cain, J. K., Hummer, C., Goyal, H., and Perisetti, A. (2020). Pulmonary and extra-pulmonary clinical manifestations of COVID-19. Front. Med. 7:526. doi: 10.3389/fmed.2020.00526

PubMed Abstract | CrossRef Full Text | Google Scholar

Jung, R. E., and Haier, R. J. (2007). The Parieto-frontal integration theory (P-FIT) of intelligence: converging neuroimaging evidence. Behav. Brain Sci. 30, 135–154; discussion 154-187. doi: 10.1017/S0140525X07001185

PubMed Abstract | CrossRef Full Text | Google Scholar

Kamasak, B., Ulcay, T., Nisari, M., Gorgulu, O., Akca, V., Alpaslan, M., et al. (2023). Effects of COVID-19 on brain and cerebellum: a voxel based morphometrical analysis. Bratisl. Lek. Listy. 124, 442–448. doi: 10.4149/BLL_2023_068

PubMed Abstract | CrossRef Full Text | Google Scholar

Kanda, T., Nakatomi, Y., Ishikawa, H., Hitomi, M., Matsubara, Y., Ono, T., et al. (1992). Intestinal fatty acid-binding protein as a sensitive marker of intestinal ischemia. Dig. Dis. Sci. 37, 1362–1367. doi: 10.1007/BF01296004

PubMed Abstract | CrossRef Full Text | Google Scholar

Keating, J. A., Shaughnessy, C., Baubie, K., Kates, A. E., Putman-Buehler, N., Watson, L., et al. (2019). Characterising the gut microbiome in veterans with gulf war illness: a protocol for a longitudinal, prospective cohort study. BMJ Open 9:e031114. doi: 10.1136/bmjopen-2019-031114

PubMed Abstract | CrossRef Full Text | Google Scholar

Keshavarz, P., Rafiee, F., Kavandi, H., Goudarzi, S., Heidari, F., and Gholamrezanezhad, A. (2021). Ischemic gastrointestinal complications of COVID-19: a systematic review on imaging presentation. Clin. Imaging 73, 86–95. doi: 10.1016/j.clinimag.2020.11.054

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, Y.-K., and Shin, C. (2018). The microbiota-gut-brain Axis in neuropsychiatric disorders: Patho-physiological mechanisms and novel treatments. Curr. Neuropharmacol. 16, 559–573. doi: 10.2174/1570159X15666170915141036

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, Y.-M., and Shin, E.-C. (2021). Type I and III interferon responses in SARS-CoV-2 infection. Exp. Mol. Med. 53, 750–760. doi: 10.1038/s12276-021-00592-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, Y. S., and Täuber, M. G. (1996). Neurotoxicity of glia activated by gram-positive bacterial products depends on nitric oxide production. Infect. Immun. 64, 3148–3153. doi: 10.1128/iai.64.8.3148-3153.1996

PubMed Abstract | CrossRef Full Text | Google Scholar

Kiwango, F., Mremi, A., Masenga, A., and Akrabi, H. (2021). Intestinal ischemia in a COVID-19 patient: case report from northern Tanzania. J. Surg. Case Rep. 2021. doi: 10.1093/jscr/rjaa537

PubMed Abstract | CrossRef Full Text | Google Scholar

Kopel, J., Perisetti, A., Gajendran, M., Boregowda, U., and Goyal, H. (2020). Clinical insights into the gastrointestinal manifestations of COVID-19. Dig. Dis. Sci. 1–8. doi: 10.1007/s10620-020-06362-8

CrossRef Full Text | Google Scholar

Lamers, M. M., Beumer, J., van der Vaart, J., Knoops, K., Puschhof, J., Breugem, T. I., et al. (2020). SARS-CoV-2 productively infects human gut enterocytes. Science 369, 50–54. doi: 10.1126/science.abc1669

PubMed Abstract | CrossRef Full Text | Google Scholar

Lammers, K. M., Lu, R., Brownley, J., Lu, B., Gerard, C., Thomas, K., et al. (2008). Gliadin induces an increase in intestinal permeability and zonulin release by binding to the chemokine receptor CXCR3. Gastroenterology 135, 194–204.e3. doi: 10.1053/j.gastro.2008.03.023

PubMed Abstract | CrossRef Full Text | Google Scholar

Levitt, M. D. (1971). Volume and composition of human intestinal gas determined by means of an intestinal washout technic. N. Engl. J. Med. 284, 1394–1398. doi: 10.1056/NEJM197106242842502

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, J., Richards, E. M., Handberg, E. M., Pepine, C. J., and Raizada, M. K. (2021). Butyrate regulates COVID-19-relevant genes in gut epithelial organoids from normotensive rats. Hypertens. Dallas Tex 1979, e13–e16. doi: 10.1161/HYPERTENSIONAHA.120.16647

CrossRef Full Text | Google Scholar

Liu, Q., Mak, J. W. Y., Su, Q., Yeoh, Y. K., Lui, G. C. Y., Ng, S. S. S., et al. (2022). Gut microbiota dynamics in a prospective cohort of patients with post-acute COVID-19 syndrome. Gut 71, 544–552. doi: 10.1136/gutjnl-2021-325989

PubMed Abstract | CrossRef Full Text | Google Scholar

Lu, H., Liu, P., Yezhuvath, U., Cheng, Y., Marshall, O., and Ge, Y. (2014). MRI mapping of cerebrovascular reactivity via gas inhalation challenges. J. Vis. Exp. JoVE. doi: 10.3791/52306-v

PubMed Abstract | CrossRef Full Text | Google Scholar

Lu, R., Zhao, X., Li, J., Niu, P., Yang, B., Wu, H., et al. (2020). Genomic characterisation and epidemiology of 2019 novel coronavirus: implications for virus origins and receptor binding. Lancet 395, 565–574. doi: 10.1016/S0140-6736(20)30251-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Lukiw, W. J., Pogue, A., and Hill, J. M. (2022). SARS-CoV-2 infectivity and neurological targets in the brain. Cell. Mol. Neurobiol. 42, 217–224. doi: 10.1007/s10571-020-00947-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Matschke, J., Lütgehetmann, M., Hagel, C., Sperhake, J. P., Schröder, A. S., Edler, C., et al. (2020). Neuropathology of patients with COVID-19 in Germany: a post-mortem case series. Lancet Neurol. 19, 919–929. doi: 10.1016/S1474-4422(20)30308-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Mavrikaki, M., Lee, J. D., Solomon, I. H., and Slack, F. J. (2022). Severe COVID-19 is associated with molecular signatures of aging in the human brain. Nat. Aging 2, 1130–1137. doi: 10.1038/s43587-022-00321-w

PubMed Abstract | CrossRef Full Text | Google Scholar

Mayr, F. B., Yende, S., and Angus, D. C. (2014). Epidemiology of severe sepsis. Virulence 5, 4–11. doi: 10.4161/viru.27372

PubMed Abstract | CrossRef Full Text | Google Scholar

Meinhardt, J., Radke, J., Dittmayer, C., Franz, J., Thomas, C., Mothes, R., et al. (2021). Olfactory transmucosal SARS-CoV-2 invasion as a port of central nervous system entry in individuals with COVID-19. Nat. Neurosci. 24, 168–175. doi: 10.1038/s41593-020-00758-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Merad, M., and Martin, J. C. (2020). Pathological inflammation in patients with COVID-19: a key role for monocytes and macrophages. Nat. Rev. Immunol. 20, 355–362. doi: 10.1038/s41577-020-0331-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Meringer, H., and Mehandru, S. (2022). Gastrointestinal post-acute COVID-19 syndrome. Nat. Rev. Gastroenterol. Hepatol. 19, 345–346. doi: 10.1038/s41575-022-00611-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Metz-Zumaran, C., Kee, C., Doldan, P., Guo, C., Stanifer, M. L., and Boulant, S. (2022). Increased sensitivity of SARS-CoV-2 to type III interferon in human intestinal epithelial cells. J. Virol. 96, e01705–e01721. doi: 10.1128/jvi.01705-21

CrossRef Full Text | Google Scholar

Montagnana, M., Danese, E., and Lippi, G. (2018). Biochemical markers of acute intestinal ischemia: possibilities and limitations. Ann. Transl. Med. 6:341. doi: 10.21037/atm.2018.07.22

PubMed Abstract | CrossRef Full Text | Google Scholar

Mordstein, M., Neugebauer, E., Ditt, V., Jessen, B., Rieger, T., Falcone, V., et al. (2010). Lambda interferon renders epithelial cells of the respiratory and gastrointestinal tracts resistant to viral infections. J. Virol. 84, 5670–5677. doi: 10.1128/JVI.00272-10

PubMed Abstract | CrossRef Full Text | Google Scholar

Moser, G., Fournier, C., and Peter, J. (2018). Intestinal microbiome-gut-brain axis and irritable bowel syndrome. Wien. Med. Wochenschr. 1946, 62–66. doi: 10.1007/s10354-017-0592-0

CrossRef Full Text | Google Scholar

Mueller, A. L., McNamara, M. S., and Sinclair, D. A. (2020). Why does COVID-19 disproportionately affect older people? Aging 12, 9959–9981. doi: 10.18632/aging.103344

PubMed Abstract | CrossRef Full Text | Google Scholar

Myhre, A. E., Aasen, A. O., Thiemermann, C., and Wang, J. E. (2006). Peptidoglycan-an endotoxin in its own right? Shock 25, 227–235. doi: 10.1097/01.shk.0000191378.55274.37

PubMed Abstract | CrossRef Full Text | Google Scholar

Nakajima, M., Arimatsu, K., Kato, T., Matsuda, Y., Minagawa, T., Takahashi, N., et al. (2015). Oral Administration of P. gingivalis induces Dysbiosis of gut microbiota and impaired barrier function leading to dissemination of Enterobacteria to the liver. PLoS One 10:e0134234. doi: 10.1371/journal.pone.0134234

PubMed Abstract | CrossRef Full Text | Google Scholar

Nicosia, R. F., Ligresti, G., Caporarello, N., Akilesh, S., and Ribatti, D. (2021). COVID-19 vasculopathy: mounting evidence for an indirect mechanism of endothelial injury. Am. J. Pathol. 191, 1374–1384. doi: 10.1016/j.ajpath.2021.05.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Nori, P., Cowman, K., Chen, V., Bartash, R., Szymczak, W., Madaline, T., et al. (2021). Bacterial and fungal coinfections in COVID-19 patients hospitalized during the new York City pandemic surge. Infect. Control Hosp. Epidemiol. 42, 84–88. doi: 10.1017/ice.2020.368

PubMed Abstract | CrossRef Full Text | Google Scholar

Norsa, L., Valle, C., Morotti, D., Bonaffini, P. A., Indriolo, A., and Sonzogni, A. (2020). Intestinal ischemia in the COVID-19 era. Dig. Liver Dis. 52, 1090–1091. doi: 10.1016/j.dld.2020.05.030

PubMed Abstract | CrossRef Full Text | Google Scholar

Pacheco, P., Vieira-de-Abreu, A., Gomes, R. N., Barbosa-Lima, G., Wermelinger, L. B., Maya-Monteiro, C. M., et al. (2007). Monocyte chemoattractant Protein-1/CC chemokine ligand 2 controls microtubule-driven biogenesis and leukotriene B4-synthesizing function of macrophage lipid bodies elicited by innate immune response. J. Immunol. 179, 8500–8508. doi: 10.4049/jimmunol.179.12.8500

PubMed Abstract | CrossRef Full Text | Google Scholar

Pan, Y.-Y., Wang, L.-C., Yang, F., and Yu, M. (2023). Interferon-lambda: new role in intestinal symptoms of COVID-19. World J. Gastroenterol. 29, 1942–1954. doi: 10.3748/wjg.v29.i13.1942

PubMed Abstract | CrossRef Full Text | Google Scholar

Peiris, S., Mesa, H., Aysola, A., Manivel, J., Toledo, J., Borges-Sa, M., et al. (2021). Pathological findings in organs and tissues of patients with COVID-19: a systematic review. PLoS One 16:e0250708. doi: 10.1371/journal.pone.0250708

PubMed Abstract | CrossRef Full Text | Google Scholar

Pekny, M., and Nilsson, M. (2005). Astrocyte activation and reactive gliosis. Glia 50, 427–434. doi: 10.1002/glia.20207

PubMed Abstract | CrossRef Full Text | Google Scholar

Pelsers, M. M. A. L., Namiot, Z., Kisielewski, W., Namiot, A., Januszkiewicz, M., Hermens, W. T., et al. (2003). Intestinal-type and liver-type fatty acid-binding protein in the intestine. Tissue distribution and clinical utility. Clin. Biochem. 36, 529–535. doi: 10.1016/S0009-9120(03)00096-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Peluso, M. J., Deveau, T. -M., Munter, S. E., Ryder, D., Buck, A., Beck-Engeser, G., et al. (2023). Chronic viral coinfections differentially affect the likelihood of developing long COVID. J. Clin. Invest. 133:e163669. doi: 10.1172/JCI163669

PubMed Abstract | CrossRef Full Text | Google Scholar

Philippens, I. H. C. H. M., Böszörményi, K. P., Wubben, J. A. M., Fagrouch, Z. C., van Driel, N., Mayenburg, A. Q., et al. (2022). Brain inflammation and intracellular α-Synuclein aggregates in macaques after SARS-CoV-2 infection. Viruses 14:776. doi: 10.3390/v14040776

PubMed Abstract | CrossRef Full Text | Google Scholar

Pimentel, M., Saad, R. J., Long, M. D., and Rao, S. S. C. (2020). ACG clinical guideline: small intestinal bacterial overgrowth. Off. J. Am. Coll. Gastroenterol. ACG 115, 165–178. doi: 10.14309/ajg.0000000000000501

PubMed Abstract | CrossRef Full Text | Google Scholar

Pott, J., Mahlakõiv, T., Mordstein, M., Duerr, C. U., Michiels, T., Stockinger, S., et al. (2011). IFN-lambda determines the intestinal epithelial antiviral host defense. Proc. Natl. Acad. Sci. U. S. A. 108, 7944–7949. doi: 10.1073/pnas.1100552108

PubMed Abstract | CrossRef Full Text | Google Scholar

Pott, J., and Stockinger, S. (2017). Type I and III interferon in the gut: tight balance between host protection and immunopathology. Front. Immunol. 8:258. doi: 10.3389/fimmu.2017.00258

PubMed Abstract | CrossRef Full Text | Google Scholar

Prasad, R., Patton, M. J., Floyd, J. L., Vieira, C. P., Fortmann, S., DuPont, M., et al. (2021). Plasma microbiome in COVID-19 subjects: an indicator of gut barrier defects and dysbiosis. bioRxiv 2021.04.06.438634. doi: 10.1101/2021.04.06.438634

PubMed Abstract | CrossRef Full Text | Google Scholar

Proal, A. D., and VanElzakker, M. B. (2021). Long COVID or post-acute sequelae of COVID-19 (PASC): an overview of biological Factors that may contribute to persistent symptoms. Front. Microbiol. 12. doi: 10.3389/fmicb.2021.698169

PubMed Abstract | CrossRef Full Text | Google Scholar

Pulverer, J. E., Rand, U., Lienenklaus, S., Kugel, D., Ziętara, N., Kochs, G., et al. (2010). Temporal and spatial resolution of type I and III interferon responses in vivo. J. Virol. 84, 8626–8638. doi: 10.1128/JVI.00303-10

PubMed Abstract | CrossRef Full Text | Google Scholar

Qin, Y., Wu, J., Chen, T., Li, J., Zhang, G., Wu, D., et al. (2021). Long-term microstructure and cerebral blood flow changes in patients recovered from COVID-19 without neurological manifestations. J. Clin. Invest. 131:e147329. doi: 10.1172/JCI147329

CrossRef Full Text | Google Scholar

Queiroz, M. A. F., Neves, P. F. M., Lima, S. S., Lopes, J. C., Torres, M. K. S., Vallinoto, I. M. V. C., et al. (2022). Cytokine profiles associated with acute COVID-19 and Long COVID-19 syndrome. Front. Cell. Infect. Microbiol. 12:922422. doi: 10.3389/fcimb.2022.922422

PubMed Abstract | CrossRef Full Text | Google Scholar

Raetz, C. R. H., and Whitfield, C. (2002). Lipopolysaccharide Endotoxins. Annu. Rev. Biochem. 71, 635–700. doi: 10.1146/annurev.biochem.71.110601.135414

PubMed Abstract | CrossRef Full Text | Google Scholar

Raman, B., Bluemke, D. A., Lüscher, T. F., and Neubauer, S. (2022). Long COVID: post-acute sequelae of COVID-19 with a cardiovascular focus. Eur. Heart J. 43, 1157–1172. doi: 10.1093/eurheartj/ehac031

PubMed Abstract | CrossRef Full Text | Google Scholar

Ranjbar, M., Rahimi, A., Baghernejadan, Z., Ghorbani, A., and Khorramdelazad, H. (2022). Role of CCL2/CCR2 axis in the pathogenesis of COVID-19 and possible treatments: all options on the table. Int. Immunopharmacol. 113:109325. doi: 10.1016/j.intimp.2022.109325

PubMed Abstract | CrossRef Full Text | Google Scholar

Reiken, S., Sittenfeld, L., Dridi, H., Liu, Y., Liu, X., and Marks, A. R. (2022). Alzheimer’s-like signaling in brains of COVID-19 patients. Alzheimers Dement (N Y) 18, 955–965. doi: 10.1002/alz.12558

PubMed Abstract | CrossRef Full Text | Google Scholar

Reinold, J., Farahpour, F., Fehring, C., Dolff, S., Konik, M., Korth, J., et al. (2021). A pro-inflammatory gut microbiome characterizes SARS-CoV-2 infected patients and a reduction in the connectivity of an anti-inflammatory bacterial network associates with severe COVID-19. Front. Cell. Infect. Microbiol. 11. doi: 10.3389/fcimb.2021.747816

PubMed Abstract | CrossRef Full Text | Google Scholar

Rezaie, A., Buresi, M., Lembo, A., Lin, H., McCallum, R., Rao, S., et al. (2017). Hydrogen and methane-based breath testing in gastrointestinal disorders: the north American consensus. Off. J. Am. Coll. Gastroenterol. ACG 112, 775–784. doi: 10.1038/ajg.2017.46

PubMed Abstract | CrossRef Full Text | Google Scholar

Rodriguez-Hernandez, C. J., Sokoloski, K. J., Stocke, K. S., Dukka, H., Jin, S., Metzler, M. A., et al. (2021). Microbiome-mediated incapacitation of interferon lambda production in the oral mucosa. Proc. Natl. Acad. Sci. 118:e2105170118. doi: 10.1073/pnas.2105170118

PubMed Abstract | CrossRef Full Text | Google Scholar

Rogosnitzky, M., and Branch, S. (2016). Gadolinium-based contrast agent toxicity: a review of known and proposed mechanisms. Biometals 29, 365–376. doi: 10.1007/s10534-016-9931-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Ruff, W. E., Greiling, T. M., and Kriegel, M. A. (2020). Host–microbiota interactions in immune-mediated diseases. Nat. Rev. Microbiol. 18, 521–538. doi: 10.1038/s41579-020-0367-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Ryman, S., Vakhtin, A. A., Richardson, S. P., and Lin, H. C. (2023). Microbiome–gut–brain dysfunction in prodromal and symptomatic Lewy body diseases. J. Neurol. 270, 746–758. doi: 10.1007/s00415-022-11461-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Sapone, A., de Magistris, L., Pietzak, M., Clemente, M. G., Tripathi, A., Cucca, F., et al. (2006). Zonulin upregulation is associated with increased gut permeability in subjects with type 1 diabetes and their relatives. Diabetes 55, 1443–1449. doi: 10.2337/db05-1593

PubMed Abstract | CrossRef Full Text | Google Scholar

Sashindranath, M., and Nandurkar, H. H. (2021). Endothelial dysfunction in the brain. Stroke 52, 1895–1904. doi: 10.1161/STROKEAHA.120.032711

PubMed Abstract | CrossRef Full Text | Google Scholar

Schrijver, I. A., van Meurs, M., Melief, M. J., Wim Ang, C., Buljevac, D., Ravid, R., et al. (2001). Bacterial peptidoglycan and immune reactivity in the central nervous system in multiple sclerosis. Brain J. Neurol. 124, 1544–1554. doi: 10.1093/brain/124.8.1544

PubMed Abstract | CrossRef Full Text | Google Scholar

Schubert, J., Tonietto, M., Turkheimer, F., Zanotti-Fregonara, P., and Veronese, M. (2021). Supervised clustering for TSPO PET imaging. Eur. J. Nucl. Med. Mol. Imaging 49, 257–268. doi: 10.1007/s00259-021-05309-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Schultheiß, C., Willscher, E., Paschold, L., Gottschick, C., Klee, B., Henkes, S. S., et al. (2022). The IL-1β, IL-6, and TNF cytokine triad is associated with post-acute sequelae of COVID-19. Cell Rep. Med. 3:100663. doi: 10.1016/j.xcrm.2022.100663

PubMed Abstract | CrossRef Full Text | Google Scholar

Shao, X., Ma, S. J., Casey, M., D’Orazio, L., Ringman, J. M., and Wang, D. J. J. (2019). Mapping water exchange across the blood-brain barrier using 3D diffusion-prepared arterial spin labeled perfusion MRI. Magn. Reson. Med. 81, 3065–3079. doi: 10.1002/mrm.27632

PubMed Abstract | CrossRef Full Text | Google Scholar

Shimizu, T., Tani, T., Endo, Y., Hanasawa, K., Tsuchiya, M., and Kodama, M. (2002). Elevation of plasma peptidoglycan and peripheral blood neutrophil activation during hemorrhagic shock: plasma peptidoglycan reflects bacterial translocation and may affect neutrophil activation. Crit. Care Med. 30, 77–82. doi: 10.1097/00003246-200201000-00012

PubMed Abstract | CrossRef Full Text | Google Scholar

Singh, B., and Kaur, P. (2021). COVID-19 and acute mesenteric ischemia: a review of literature. Hematol. Transfus. Cell Ther. 43, 112–116. doi: 10.1016/j.htct.2020.10.959

PubMed Abstract | CrossRef Full Text | Google Scholar

Siow, I., Lee, K. S., Zhang, J. J. Y., Saffari, S. E., and Ng, A. (2021). Encephalitis as a neurological complication of COVID-19: a systematic review and meta-analysis of incidence, outcomes, and predictors. Eur. J. Neurol. 28, 3491–3502. doi: 10.1111/ene.14913

CrossRef Full Text | Google Scholar

Smith, E. E., and Beaudin, A. E. (2018). New insights into cerebral small vessel disease and vascular cognitive impairment from MRI. Curr. Opin. Neurol. 31, 36–43. doi: 10.1097/WCO.0000000000000513

PubMed Abstract | CrossRef Full Text | Google Scholar

Solomon, T. (2021). Neurological infection with SARS-CoV-2 — the story so far. Nat. Rev. Neurol. 17, 65–66. doi: 10.1038/s41582-020-00453-w

PubMed Abstract | CrossRef Full Text | Google Scholar

Solomon, I. H., Normandin, E., Bhattacharyya, S., Mukerji, S. S., Keller, K., Ali, A. S., et al. (2020). Neuropathological features of Covid-19. N. Engl. J. Med. 383, 989–992. doi: 10.1056/NEJMc2019373

PubMed Abstract | CrossRef Full Text | Google Scholar

Song, E., Zhang, C., Israelow, B., Lu-Culligan, A., Prado, A. V., Skriabine, S., et al. (2021). Neuroinvasion of SARS-CoV-2 in human and mouse brainNeuroinvasion of SARS-CoV-2 in humans and mice. J. Exp. Med. 218. doi: 10.1084/jem.20202135

PubMed Abstract | CrossRef Full Text | Google Scholar

Stevens, B. R., Goel, R., Seungbum, K., Richards, E. M., Holbert, R. C., Pepine, C. J., et al. (2018). Increased human intestinal barrier permeability plasma biomarkers zonulin and FABP2 correlated with plasma LPS and altered gut microbiome in anxiety or depression. Gut 67, 1555.2–1555.1557. doi: 10.1136/gutjnl-2017-314759

PubMed Abstract | CrossRef Full Text | Google Scholar

Stopa, E. G., Tanis, K. Q., Miller, M. C., Nikonova, E. V., Podtelezhnikov, A. A., Finney, E. M., et al. (2018). Comparative transcriptomics of choroid plexus in Alzheimer’s disease, frontotemporal dementia and Huntington’s disease: implications for CSF homeostasis. Fluids Barriers CNS 15:18. doi: 10.1186/s12987-018-0102-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Sur, S., Lin, Z., Li, Y., Yasar, S., Rosenberg, P., Moghekar, A., et al. (2020). Association of cerebrovascular reactivity and Alzheimer pathologic markers with cognitive performance. Neurology 95, e962–e972. doi: 10.1212/WNL.0000000000010133

PubMed Abstract | CrossRef Full Text | Google Scholar

Suzzi, S., Tsitsou-Kampeli, A., and Schwartz, M. (2023). The type I interferon antiviral response in the choroid plexus and the cognitive risk in COVID-19. Nat. Immunol. 24, 220–224. doi: 10.1038/s41590-022-01410-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Swank, Z., Senussi, Y., Manickas-Hill, Z., Yu, X. G., Li, J. Z., Alter, G., et al. (2023). Persistent circulating severe acute respiratory syndrome coronavirus 2 spike is associated with post-acute coronavirus disease 2019 sequelae. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 76, e487–e490. doi: 10.1093/cid/ciac722

PubMed Abstract | CrossRef Full Text | Google Scholar

Tabata, T., Tani, T., Endo, Y., and Hanasawa, K. (2002). Bacterial translocation and peptidoglycan translocation by acute ethanol administration. J. Gastroenterol. 37, 726–731. doi: 10.1007/s005350200118

PubMed Abstract | CrossRef Full Text | Google Scholar

Taruffi, L., Muccioli, L., Mitolo, M., Ferri, L., Descovich, C., Mazzoni, S., et al. (2023). Neurological manifestations of Long COVID: a single-center one-year experience. Neuropsychiatr. Dis. Treat. 19, 311–319. doi: 10.2147/NDT.S387501

PubMed Abstract | CrossRef Full Text | Google Scholar

Teixeira, P. C., Dorneles, G. P., Santana Filho, P. C., da Silva, I. M., Schipper, L. L., Postiga, I. A. L., et al. (2021). Increased LPS levels coexist with systemic inflammation and result in monocyte activation in severe COVID-19 patients. Int. Immunopharmacol. 100:108125. doi: 10.1016/j.intimp.2021.108125

PubMed Abstract | CrossRef Full Text | Google Scholar

Thapaliya, K., Marshall-Gradisnik, S., Barth, M., Eaton-Fitch, N., and Barnden, L. (2023). Brainstem volume changes in myalgic encephalomyelitis/chronic fatigue syndrome and long COVID patients. Front. Neurosci. 17:1125208. doi: 10.3389/fnins.2023.1125208

PubMed Abstract | CrossRef Full Text | Google Scholar

Theobald, S. J., Simonis, A., Georgomanolis, T., Kreer, C., Zehner, M., Eisfeld, H. S., et al. (2021). Long-lived macrophage reprogramming drives spike protein-mediated inflammasome activation in COVID-19. EMBO Mol. Med. 13:e14150. doi: 10.15252/emmm.202114150

PubMed Abstract | CrossRef Full Text | Google Scholar

Theoharides, T. C. (2022). Could SARS-CoV-2 spike protein be responsible for Long-COVID syndrome? Mol. Neurobiol. 59, 1850–1861. doi: 10.1007/s12035-021-02696-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Toor, D., Wsson, M. K., Kumar, P., Karthikeyan, G., Kaushik, N. K., Goel, C., et al. (2019). Dysbiosis disrupts gut immune homeostasis and promotes gastric diseases. Int. J. Mol. Sci. 20:E2432. doi: 10.3390/ijms20102432

CrossRef Full Text | Google Scholar

Triantafyllou, K., Chang, C., and Pimentel, M. (2014). Methanogens, methane and gastrointestinal motility. J. Neurogastroenterol. Motil. 20, 31–40. doi: 10.5056/jnm.2014.20.1.31

PubMed Abstract | CrossRef Full Text | Google Scholar

Tsunooka, N., Maeyama, K., Hamada, Y., Imagawa, H., Takano, S., Watanabe, Y., et al. (2004). Bacterial translocation secondary to small intestinal mucosal ischemia during cardiopulmonary bypass. Measurement by diamine oxidase and peptidoglycan. Eur. J. Cardiothorac. Surg. 25, 275–280. doi: 10.1016/j.ejcts.2003.11.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Urenjak, J., Williams, S. R., Gadian, D. G., and Noble, M. (1993). Proton nuclear magnetic resonance spectroscopy unambiguously identifies different neural cell types. J. Neurosci. 13, 981–989. doi: 10.1523/JNEUROSCI.13-03-00981.1993

PubMed Abstract | CrossRef Full Text | Google Scholar

Vakhtin, A. A., Ryman, S. G., Flores, R. A., and Jung, R. E. (2014). Functional brain networks contributing to the Parieto-frontal integration theory of intelligence. NeuroImage 103, 349–354. doi: 10.1016/j.neuroimage.2014.09.055

PubMed Abstract | CrossRef Full Text | Google Scholar

Vakili, K., Fathi, M., Yaghoobpoor, S., Sayehmiri, F., Nazerian, Y., Nazerian, A., et al. (2022). The contribution of gut-brain axis to development of neurological symptoms in COVID-19 recovered patients: a hypothesis and review of literature. Front. Cell. Infect. Microbiol. 12. doi: 10.3389/fcimb.2022.983089

PubMed Abstract | CrossRef Full Text | Google Scholar

Valles-Colomer, M., Falony, G., Darzi, Y., Tigchelaar, E. F., Wang, J., Tito, R. Y., et al. (2019). The neuroactive potential of the human gut microbiota in quality of life and depression. Nat. Microbiol. 4, 623–632. doi: 10.1038/s41564-018-0337-x

PubMed Abstract | CrossRef Full Text | Google Scholar

van Winkle, J. A., Peterson, S. T., Kennedy, E. A., Wheadon, M. J., Ingle, H., Desai, C., et al. (2022). Homeostatic interferon-lambda response to bacterial microbiota stimulates preemptive antiviral defense within discrete pockets of intestinal epithelium. elife 11:e74072. doi: 10.7554/eLife.74072

PubMed Abstract | CrossRef Full Text | Google Scholar

Vargas-Caraveo, A., Sayd, A., Maus, S. R., Caso, J. R., Madrigal, J. L. M., García-Bueno, B., et al. (2017). Lipopolysaccharide enters the rat brain by a lipoprotein-mediated transport mechanism in physiological conditions. Sci. Rep. 7:13113. doi: 10.1038/s41598-017-13302-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Venzon, M., Bernard-Raichon, L., Klein, J., Axelrad, J. E., Hussey, G. A., Sullivan, A. P., et al. (2021). Gut microbiome dysbiosis during COVID-19 is associated with increased risk for bacteremia and microbial translocation. Res. Sq. rs.3.rs-726620. doi: 10.21203/rs.3.rs-726620/v1

PubMed Abstract | CrossRef Full Text | Google Scholar

Villapol, S. (2020). Gastrointestinal symptoms associated with COVID-19: impact on the gut microbiome. Transl. Res. 226, 57–69. doi: 10.1016/j.trsl.2020.08.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Visser, L., et al. (2005). Proinflammatory bacterial peptidoglycan as a cofactor for the development of central nervous system autoimmune disease. J. Immunol. Baltim. Md 1950, 808–816. doi: 10.4049/jimmunol.174.2.808

CrossRef Full Text | Google Scholar

von Weyhern, C. H., Kaufmann, I., Neff, F., and Kremer, M. (2020). Early evidence of pronounced brain involvement in fatal COVID-19 outcomes. Lancet Lond. Engl. 395:e109. doi: 10.1016/S0140-6736(20)31282-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Wirusanti, N. I., Baldridge, M. T., and Harris, V. C. (2022). Microbiota regulation of viral infections through interferon signaling. Trends Microbiol. 30, 778–792. doi: 10.1016/j.tim.2022.01.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Wolf, P. G., Biswas, A., Morales, S. E., Greening, C., and Gaskins, H. R. (2016). H2 metabolism is widespread and diverse among human colonic microbes. Gut Microbes 7, 235–245. doi: 10.1080/19490976.2016.1182288

PubMed Abstract | CrossRef Full Text | Google Scholar

Xie, Y., Xu, E., Bowe, B., and Al-Aly, Z. (2022). Long-term cardiovascular outcomes of COVID-19. Nat. Med. 28, 583–590. doi: 10.1038/s41591-022-01689-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Xiong, R., Gunter, C., Fleming, E., Vernon, S. D., Bateman, L., Unutmaz, D., et al. (2023). Multi-‘omics of gut microbiome-host interactions in short- and long-term myalgic encephalomyelitis/chronic fatigue syndrome patients. Cell Host Microbe 31, 273–287.e5. doi: 10.1016/j.chom.2023.01.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, A. C., Kern, F., Losada, P. M., Agam, M. R., Maat, C. A., Schmartz, G. P., et al. (2021). Dysregulation of brain and choroid plexus cell types in severe COVID-19. Nature 595, 565–571. doi: 10.1038/s41586-021-03710-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Yeoh, Y. K., Zuo, T., Lui, G. C. Y., Zhang, F., Liu, Q., Li, A. Y. L., et al. (2021). Gut microbiota composition reflects disease severity and dysfunctional immune responses in patients with COVID-19. Gut 70, 698–706. doi: 10.1136/gutjnl-2020-323020

PubMed Abstract | CrossRef Full Text | Google Scholar

Yesilkaya, U. H., Sen, M., and Balcioglu, Y. H. (2021). COVID-19-related cognitive dysfunction may be associated with transient disruption in the DLPFC glutamatergic pathway. J. Clin. Neurosci. 87, 153–155. doi: 10.1016/j.jocn.2021.03.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Yong, S. J. (2021). Persistent brainstem dysfunction in Long-COVID: a hypothesis. ACS Chem. Neurosci. 12, 573–580. doi: 10.1021/acschemneuro.0c00793

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, Y., Avery, T., Vakhtin, A. A., Mathersul, D. C., Tranvinh, E., Wintermark, M., et al. (2020a). Brainstem atrophy in gulf war illness. Neurotoxicology 78, 71–79. doi: 10.1016/j.neuro.2020.02.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, Y., Vakhtin, A. A., Dietch, J., Jennings, J. S., Yesavage, J. A., Clark, J. D., et al. (2021). Brainstem damage is associated with poorer sleep quality and increased pain in gulf war illness veterans. Life Sci. 280:119724. doi: 10.1016/j.lfs.2021.119724

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, Y., Vakhtin, A. A., Jennings, J. S., Massaband, P., Wintermark, M., Craig, P. L., et al. (2020b). Diffusion tensor tractography of brainstem fibers and its application in pain. PLoS One 15:e0213952. doi: 10.1371/journal.pone.0213952

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, L., Zhou, L., Bao, L., Liu, J., Zhu, H., and Lv, Q. (2021). SARS-CoV-2 crosses the blood–brain barrier accompanied with basement membrane disruption without tight junctions alteration. Signal Transduct. Target. Ther. 6, 1–12. doi: 10.1038/s41392-021-00719-9

CrossRef Full Text | Google Scholar

Zhou, P., Yang, X. L., Wang, X. G., Hu, B., Zhang, L., Zhang, W., et al. (2020). A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature 579, 270–273. doi: 10.1038/s41586-020-2012-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: COVID-19, post-acute COVID-19 syndrome (PACS), post-acute sequelae of COVID-19 (PASC), gut-brain axis, neuroinflammation, endotoxicity

Citation: Plummer AM, Matos YL, Lin HC, Ryman SG, Birg A, Quinn DK, Parada AN and Vakhtin AA (2023) Gut-brain pathogenesis of post-acute COVID-19 neurocognitive symptoms. Front. Neurosci. 17:1232480. doi: 10.3389/fnins.2023.1232480

Received: 31 May 2023; Accepted: 01 September 2023;
Published: 28 September 2023.

Edited by:

Wakiro Sato, National Center of Neurology and Psychiatry, Japan

Reviewed by:

Hiroaki Masuoka, RIKEN Yokohama, Japan
George D. Vavougios, University of Cyprus, Cyprus

Copyright © 2023 Plummer, Matos, Lin, Ryman, Birg, Quinn, Parada and Vakhtin. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Andrei A. Vakhtin, avakhtin@mrn.org

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