Abstract
Post-Acute Infection Syndrome (PAIS) is a relatively new medical terminology that represents prolonged sequelae symptoms after acute infection by numerous pathogenic agents. Imposing a substantial public health burden worldwide, PASC (post-acute sequelae of COVID-19 infection) and ME/CFS (myalgic encephalomyelitis/chronic fatigue syndrome) are two of the most recognized and prevalent PAIS conditions. The presences of prior infections and similar symptom profiles in PAIS reflect a plausible common etiopathogenesis. The human microbiome is known to play an essential role in health and disease. In this review, we reviewed and summarized available research on oral and gut microbiota alterations in patients with different infections or PAIS conditions. We discussed key theories about the associations between microbiome dysbiosis and PAIS disease development, aiming to explore the mechanistic roles and potential functions the microbiome may have in the process. Additionally, we discuss the areas of knowledge gaps and propose the potential clinical applications of the microbiome for prevention and treatment of PAIS conditions.
Keywords: Post-acute infection syndromes, Microbiome, PASC, ME/CFS
1. Introduction
As the COVID-19 pandemic wears on, increasing attention has been given to post-acute sequelae of COVID-19 infection (PASC), which is commonly referred to as long-COVID. PASC has imposed a significant burden on global public health, affecting a conservative estimate of 65 million people worldwide [1]. Patients with PASC show persistent symptoms that last for three months following COVID-19 infection that cannot be explained by alternative medical conditions [2]. However, such post-acute sequelae are not exclusive to SARS-CoV-2 infection, as prolonged sequelae symptoms have been documented to be associated with numerous infections. As tail-ends of infections, chronic sequelae caused by the inability to recover from acute infectious diseases are collectively called post-acute infection syndromes (PAIS). Still, PAIS is primarily unexplained in the medical community, featuring non-specific medical manifestations, lack of objective diagnostics, and mysterious mechanisms [3]. A long list of infectious agents, covering various bacterial, viral, and parasite species, is known to trigger the development of PAIS in a proportion of exposed individuals [4]. The majority of other PAIS show symptoms within 2–6 months of the initial infection, with one exception being that post-polio syndrome patients may have signs and symptoms that appear decades after initial polio infection [5]. Besides PASC, myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is another major syndrome that overlaps with the concept of PAIS [6]. ME/CFS is a complex disease characterized by unexplained severe fatigue, with approximately 75% of cases reporting symptoms following an earlier infection. Throughout history, ME/CFS has also been named as post-polio syndrome and post-viral syndrome since patients develop chronic symptoms typically following infections with Polio virus, Epstein Barr virus, Influenza virus, or an array of other pathogens [7].
Currently, our understanding of PAIS is still in a nascent stage. Medical care providers are facing heavy challenges in the recognition, diagnosis, treatment, and prevention of PAIS, largely due to the lack of systematic research that examines the association between acute infections and chronic impairments. While condition-specific symptoms are often aligned with the pathogen tropism and the underlying pathogenesis of related diseases, PAIS patients generally present with a broad spectrum of symptoms in varying degrees of clinical manifestation [8], [9], [10]. Their conditions, however, are shared or characterized by a core set of symptoms, including disproportionate levels of fatigue, exertion intolerance, debilitating flu-like symptoms, chronic or recurrent joint pain, neurocognitive and sensory impairment, and myalgia/arthralgia [3]. Given the similarity of clinical presentations, a unified pathophysiology is needed to facilitate proper understanding and management of PAIS patients. Several mechanistic theories have been speculated to explain PAIS: 1) chronic immune system stimulation by persistent infection or unviable pathogen residues; 2) inflammation and coagulopathy caused by pathogen-induced autoimmunity; 3) irreversible organ injury causing characteristic symptoms in some PAIS conditions; and 4) dysregulation of the microbiota-gut-brain axis may cause chronic disease. It is worth noting that these theories are not mutually exclusive and may coexist or present with varying degrees of impact in different PAIS conditions [11].
The human microbiome generally refers to all microorganisms inhabiting the human body, encompassing their metabolic products and immediate environmental contexts as well. Recent advancements in high-throughput technology have enabled expeditious profiling and quantification of the human microbiota and their interactions with the environment, utilizing metagenomic, transcriptomic, and metabolomic approaches. It is known that human initial exposure to the microbial world occurs during vaginal birth, whereby neonates receive microbial species from the maternal birth canal, skin contact, and breastfeeding, playing a crucial and enduring role in developing the initial microbiome in neonates [12]. Furthermore, the human microbiome undergoes dynamic transformations throughout life, encapsulating diverse factors such as diet, environment, and lifestyle. The holistic microbiome functions as an additional organ system that plays essential roles in human health and development. The intestinal tract solely harbors a complex ecosystem comprising bacteria, fungi, parasites, and viruses, collectively forming the intestinal microbiome, which amounts to an estimated 100 trillion cells (10 times greater than the number of human cells) [13]. To preserve intestinal homeostasis, gut microbes are sequestered from the mucosal immune system by a protective mucous barrier that covers the intestinal epithelium [14]. It necessitates the intestinal immune system to distinguish the vast numbers of harmless or beneficial intestinal microbes from potential pathogens. The mucosal layer serves as the frontline of defense against infections while also providing a foundation for bacterial colonization by supplying a carbon source for the bacterial community [15]. The characteristics and mechanisms of how microbiota affects host susceptibility to infectious agents have already been well described [16], [17]. Either gut dysbiosis or alterations in the gut microbiome may lead to the overgrowth of opportunistic bacterial pathogens. For instance, enteric viruses can alter the diversity and composition of the gut microbiome, resulting in gut dysbiosis and promoting opportunistic bacterial pathogens [18], [19], [20]. Similarly, bacterial infections have been observed to alter microbiome composition that Porphyromonas gingivalis causes the ecological dysbiosis in the oral cavity, consequently leading to periodontitis [21]. Numerous microorganisms from commensal human flora are also capable of transforming into opportunistic pathogens under conditions of microbiota dysregulation and immunosuppression. Staphylococcus aureus is a prevalent member of the normal nasal microbiome in 30% of human population, but it can alter its gene expression to promote a range of ailments, including skin infections and even life-threatening meningitis and endocarditis in severe cases [22]. In a mouse study, gut microbiota of antibiotic treated mice had a significant increase in opportunistic pathogens, which demonstrated the effects of antibiotic therapy on induced dysbiosis and then increased level of opportunistic pathogens [23]. Conversely, gut microbes are essential for restoring the immune system during the recovery from infection in both animal and human research [24]. It has been suggested that a persistent dysbiosis of the gut microbiota lasting for more than a month after disease subsidence maybe linked to the emergence of PASC [25]. Furthermore, common PAIS symptoms (such as fatigue, joint pain, sleep disturbance, diarrhea, anxiety, and headache) are long associated with microbiome dysbiosis, also suggesting that post-acute sequelae may arise from such dysbiosis after acute infection [26].
In summary, the state-of-art techniques used in microbiome research have increased our comprehension of the crucial, albeit underappreciated, roles played by gut microorganisms in both the pathogenesis and prognosis of PAIS. In this perspective, we summarize the latest findings on microbiota alterations in PAIS patients with different microbial sources. We also examine the potential mechanisms of immune interactions linking microbiome dysbiosis to disease development. Lastly, we highlight areas of knowledge gaps and explore the potential clinical applications of the microbiome for diagnosis, prevention, and treatment of PAIS conditions (Fig. 1).
Fig. 1.
Overview of the microbiome in PAIS.
2. Methods
To review and present the full picture of the interaction between microbiome and PAISs, we carried out an exhaustive search in PubMed (U.S. National Library of Medicine) publication database for relevant studies published prior to April 2023. The following keywords were used alone or in combination with a similar search strategy described elsewhere: “microbiome”, “microflora”, “microbiota”, “microorganism”, “dysbiosis”, and all individual PAIS conditions listed in the review by Choutka et. al. [3], Recent publications were preferred, but no limiting period was imposed in our screening. Furthermore, books, general newspapers, and Institutional Websites were reviewed for possible integration.
3. Microbiome dysbiosis
3.1. Microbiome dysbiosis in patients with acute infections
The host immune system works in partnership with microbiota to achieve homeostasis and resilience. When infection occurs, the exogenous pathogens disrupt host’s microbiome, and the impact can be pathogen-specific (Fig. 2). Recent studies of COVID-19 infection in varied populations consistently showed decreased oral and gut microbial diversity, reduced beneficial commensal bacteria, and elevated levels of opportunistic bacterial pathogens [27], [28], [29]. Specifically, the abundance of butyrate producing bacteria such as Faecalibacterium prausnitzii, Clostridium butyricum, Clostridium leptum, and Eubacterium rectale decreased, while the abundance of Enterobacteriaceae and Enterococcus increased [29]. Furthermore, studies of bronchoalveolar lavage fluid in COVID-19 patients also showed a decreased diversity in its microbiome, along with an increase in pathogenic bacteria from the oral and upper respiratory tracts [30]. Besides COVID-19 infection, elevated levels of Enterococcus faecium, Clostridium ramosum, Coprobacillus, Lachnospiraceae, and Eubacterium ventriosum were found in patients with community-acquired pneumonia (CAP) [29]. A cross-sectional study of COVID-19 patients, H1N1 patients, and matched healthy controls found that Blautia, Bifidobacterium, Faecalibacterium, and Eubacterium were decreased in influenza patients compared to healthy controls. Additionally, the microbiome in COVID-19 patients showed a higher relative abundance of the phyla Actinobacteria and Firmicutes compared to those with H1N1 infection [31]. Intriguingly, different subtypes of influenza viruses may have differing impacts on the gut microbiota [32]. The biological interpretation of the differences is still unknown, but it can be crucial knowledge towards deciphering the association between infection and the microbiome. Additionally, consistent changes in the gut microbiota have been reported in patients with chronic infectious diseases like HIV [33], HBV [34], and TB [35], which also lead to a reduction in microbial diversity and depletion of certain key bacterial species. Notably, specific changes observed in the microbiome may just reflect the complexity of conducting research, as variations in sample size, recruitment criteria, subject population, medical treatment, and methods for microbial classification can lead to varied conclusions. Similar observations were found in animal research as well [36]. A longitudinal study in mice found that infection with influenza H1N1 virus altered the composition and diversity of the lower respiratory microbiome, resulting in an increase in the relative abundance of respiratory pathogenic streptococci and staphylococci. This dysbiosis did not return to normal during the recovery phase [37]. In another mice study, the overall gut microbiota composition after infection with either Respiratory Syncytial Virus (RSV) or Influenza A Virus appeared to be similar, which contradicts the findings of the aforementioned human studies [38], suggesting that the underlying mechanism is common to both infections, not a pathogen-specific immune effect.
Fig. 2.
The major taxa alterations in the gut bacteriome of acute infections, chronic infections and PAIS patients. Comparing to healthy controls, the major altered bacterial taxa identified in COVID-19 [27], [28], [29], H1N1 [31], H7N9 [32], CAP (Community-Acquired Pneumonia) [29], Dengue [39], RSV (Respiratory Syncytial Virus) [38], HIV (Human Immunodeficiency Virus) [33], HBV (Hepatitis-B Virus) [34], TB (Tuberculosis) [35], PASC (Post-acute Sequelae of COVID-19) [25], [40], [41], [42], ME/CFS (Myalgic Encephalomyelitis/Chronic Fatigue Syndrome) [43], [44], [45] and PTLDS (Post-Treatment Lyme Disease Syndrome) [46] patients are listed in the figure. All the studies mentioned primarily focused on the gut bacteriome, except for the Dengue infection study, which was centered on the blood bacteriome
3.2. Microbiome dysbiosis in patients with PAIS conditions
Under most circumstances, the infection-triggered microbial disturbance is anticipated to restore over time in convalescent individuals [47], [39]. A longitudinal study investigating the oral and gut microbiomes of COVID-19 patients over a year after infection revealed a gradual recovery [40]. This recovery was accompanied by an increase in microbiome diversity, enhanced prevalence of butyrate producing microbes and Bifidobacterium, and a reduction in the presence of lipopolysaccharide (LPS) producing microbes. Despite this, persistent microbial dysbiosis was found in a proportion of patients with sequelae after acute infections. Given the urgency prompted by COVID-19 pandemic, the microbiome disturbance in PASC is an emerging medical condition that deserves greater attention. Together with PASC, the knowledge previously acquired from ME/CFS research is invaluable for advancing research efforts in PAIS [6].
Several studies have investigated the differences in gut microbiota composition between individuals who fully recovered from COVID-19 and those who developed post-acute sequelae of SARS-CoV-2 infection (PASC). The findings revealed discernible microbial differences at the time of diagnosis [25], three months later [41], six months later [42], and the discrepancy became more pronounced after one year [40]. The microbial composition of gut samples collected at the admission showed potential for predicting PASC outcomes, indicating a connection between the microbiome and the recovery process. Unlike convalescents, patients who developed PASC failed to recover their microbiota richness to normal levels. In contrast to healthy individuals, patients with PASC had reduced bacterial diversity and abundance, characterized by elevated Ruminococcus gnavus, Bacteroides vulgatus, and depleted Faecalibacterium prausnitzii. The incidence of PASC was negatively correlated with the abundance of various commensal bacteria, which positively impact host immunity, including Collinsella aerofaciens, Faecalibacterium prausnitzii, Eubacterium rectale, and Blautia obeum. It is still too early to conclude, but different symptoms in PASC patients seemly represent unique gut microbial features [25]. In addition, oral and nasal microorganisms exhibited immunomodulatory effects among PASC patients, which appeared to be correlated with both the duration of COVID-19 symptoms and PASC development [48].
Studies on gut dysbiosis in ME/CFS patients have identified a clear distinction among different cohorts [43], [44], [45]. Considering various confounding factors, multi-omics analysis on a cohort consisting of 106 patients with ME/CFS and 91 healthy controls has revealed notable differences in the diversity and abundance of the gut microbiome [43]. The abundances of Faecalibacterium prausnitzii and Eubacterium rectale were found to be reduced in individuals with ME/CFS, which were also confirmed with qPCR experiments. The qPCR results also revealed an intriguing finding that a higher fecal total bacterial load in ME/CFS patients comparing to controls. A negative association was observed between the abundance of Faecalibacterium prausnitzii and the severity of fatigue experienced by these individuals. Similar to PASC, dysbiosis of the oral microbiome was observed in ME/CFS individuals, which was characterized by an increased abundance of Leptotrichia, Prevotella, and Fusobacterium species [49]. A study on Post-Treatment Lyme Disease Syndrome (PTLDS) patients also suggested a unique microbiome signature, characterized by Blautia, Staphylococcus aureus, and Roseburia, which enabled accurate classification of over 80% of cases examined with machine learning methods [46]. Specifically, the signature is characterized by an increase in Blautia, a decrease in Bacteroides, along with several other alterations (Fig. 2).
Various fungi, viruses, and archaea coexist with bacteria, and their presences and potential functions cannot be neglected. Patients with long COVID-19 exhibited higher levels of fungal translocation from the gut or lung epithelium, which stimulated cytokine production when compared to patients without PASC or healthy controls [50]. This fungal-related coinfection has been linked to elevated mortality rates in COVID-19 patients, with approximately 54.6% case fatality rate associated with secondary fungal infections [51]. Evidence of active viral and fungal infections in individuals with ME/CFS who experience prolonged and unexplained symptoms was also indicated by previous studies [52], although no significant ME/CFS-specific differences were identified in our recent research utilizing unbiased high-throughput sequencing, capture sequencing, and both multiplex and targeted PCR assays [53].
4. Possible mechanisms
Our current understanding of how infection leads to microbiome dysbiosis is inadequate. To our best knowledge, it is only investigated once that the gut microbiome alteration was linked to immune-mediated inappetence by a mouse study [38]. It remains unexplained for why some individuals would fail to recover from acute infections with a continuing microbiome dysbiosis, as aforementioned in PAIS, and the underlying mechanism appears tightly related to the microbiome dysbiosis in acute infection and PAIS. In this regard, we hypothesize potential mechanisms of PAIS pathogenesis related to the dysregulation of the microbiome generated from the initial infection and ensuing immune responses.
Pathogens in either persistent or latent state can directly cause long-term pathology. Many viruses have been found to either regulate or closely correlate with genes involved in pro-inflammatory and anti-apoptotic pathways, thereby resulting in chronic neuroinflammation [54]. Even though their function is not yet apparent, Ebola viral particles were detected in semen samples several years after the initial infection [55], and Zika viruses were observed to persist for months in both cerebrospinal fluid and lymph nodes of rhesus monkeys following recovery [56]. In another study, influenza viruses were found to regulate the long-term expression of genes related to inflammation, neurons, and glia in mice up to a month after the initial infection [57]. Moreover, ample evidence of residual pathogens was noticed in known PAIS [3]. The prolonged presence of SARS-CoV-2 in the gastrointestinal tract of COVID-19 patients directly contributes to the degradation of the intestinal mucosal barrier through chain protein activity, implying a possible mechanism for long-term ecological dysregulation of the gut [58], [59]. Similarly, Enterovirus protein and RNA were found in patients years after ME/CFS diagnoses in nearly thirty independent studies, suggesting at least a subgroup of ME/CFS is caused by enterovirus infection [60]. It is notable that numerous studies have indicated the virus could exist in bacterial organisms and then can be introduced into human cells [61], [62]. Such interactions could lead to a distinct and potent form of viral persistence, which may partially account for the observed gut dysbiosis in patients with PAIS.
Apart from the original infectious agents, mounting evidence shows that coinfection or secondary infection can worsen or prolong symptoms. For instance, SARS-CoV-2 infection often coincides with the reactivation of latent viruses or opportunistic bacterial pathogens, including Epstein Barr Virus (EBV) [63], Cytomegalovirus [64], various types of Herpes virus [65], and LPS-producing bacteria [66]. The resurrection of latent pathogens can cause profound damage to health [67]. HSV-2 reactivation can cause memory CD4 + T cells to temporarily open the blood-brain barrier, which may impair neural tissues' ability to deal with inflammatory cytokines and leukocytes, ultimately exacerbating neurological symptoms, and leading to PASC [68]. On the other hand, acyclovir is known as a medicine to treat HSV or EBV infections by blocking viral replication. A randomized controlled trial found that additional acyclovir treatment reduced the risk of death in patients with severe COVID-19 [69].
The gut-brain axis (GBA) is a bi-directional communication network between the enteric and central neural systems, influencing immune pathways with inflammatory signaling [70]. Given that over 70% of both microorganisms and immune cells reside in the intestinal tract, it is tempting to speculate on the critical role microbiome plays in modulating GBA pathway in PAIS [71]. A range of pathogens (Coronaviruses, HIV, Zika virus, Enteroviruses, Rotaviruses, Influenza virus, and others) are known to cause GBA disturbance by affecting gut bacteriome, virome and mycobiome, which further regulate the brain activity. Driven by the alteration of microbiome, GBS-associated key modulators, including short-chain fatty acids, 5-hydroxytryptamine, cytokine, cholecystokinin, and LPS have been found altered in different PASC and ME/CFS studies [72]. Furthermore, as one of the potential outcomes, fatigue appears to be linked to neuroinflammation, which is a main clinical manifestation in the PAIS [73]. In a mouse study, the torpor nucleus within the hypothalamus was found to be associated with fatigue, suggesting the activation of GBA in the brain may stimulate relevant fatigue nucleus [74]. The pathogen can either directly cause inflammation in the brain or initially trigger systemic inflammation through gut microbiome, then leading to inflammation and ultimately resulting in elevated inflammatory responses, immune dysregulation, and neurological symptoms in both PASC and ME/CFS [75]. Many gut bacterial species altered in the PAIS, such as Faecalibacterium prausnitzii, Eubacterium rectale, and Bacteroides thetaiotaomicron, were known to have immunomodulatory properties. For instance, Faecalibacterium prausnitzii was inversely correlated with severity of symptoms in ME/CFS [43]. In addition, elevated levels of Prevotella, Veillonella and other gram-negative bacteria were observed in the oral cavity of COVID-19 patients, of whom many developed into PASC later. The altered pattern in oral microbiome is also observed in ME/CFS patients, suggesting the mediated systematic inflammation in PAIS by releasing LPS through the oral-brain axis [49].
Specific mechanistic pathway may underlie individual PAIS conditions. It has been hypothesized that antibiotics trigger ME/CFS through D-lactate toxicity, though no improvement in fatigue has been seen in recent studies by targeting the D-lactic acid producing bacteria with antibiotics or probiotics [52]. It is also notable that antibiotics may exert profound influence on the disease progression in both ME/CFS and PASC [1], [54]. In COVID-19, ACE2 acts as a primary receptor for the virus to enter human cells, but it is also vital in regulating the renin-angiotensin-aldosterone system and controlling the composition of the intestinal microbiome to regulate immune responses. Alterations in the ACE2 balance have been linked to unfavorable outcomes for COVID-19 patients, ultimately leading to PASC [76], [77]. It is also reported that Bacteroides thetaiotaomicron can affect PASC by regulating both ACE2 expression and anti-inflammatory activity [78]. A recent study has shown that SARS-CoV-2 can replicate in human gut bacteria before attaching to human cells [79], which demonstrated the possibility that SARS-CoV-2 can act as a bacteriophage infecting human microbiota. Additionally, differences in the prevalence and clinical severity of PASC and ME/CFS were observed between sexes. Among patients diagnosed with ME/CFS, the prevalence can be 4 times higher in female than in male, and male patients were also observed to have milder symptoms and a better quality of life compared to female patients [80]. Also, female gender was recognized as a key factor associated with PASC diagnosis [81]. It may indicate a potential role for sex hormones and chromosomes, particularly estrogens and testosterones, in PAIS development [82], [83]. Since females consistently exhibit increased inflammatory responses compared to males after puberty, the interplay between sex hormones, microbiome, and PASC pathogenesis cannot be ignored. Future studies focusing on PASC among genders may assist in improving understanding of the mechanism [81]. Lastly, it is worth noting that the proposed hypotheses above may exhibit overlapping and interconnected mechanisms, and pathways in the etiopathogenesis of PAIS unrelated to the microbiome are not discussed in this review.
5. Potential therapeutic interventions targeting microbiome
When seemingly disparate findings from different PAIS are interpreted through the lens of these microbiome-based paradigms, a cohesive picture of the disease progression emerges. The pathogenesis of PAIS was triggered by varied pathogenic infections originating from pathogen-induced malfunction, which led to microbiome dysbiosis that varies depending on the patient's individual disease history and surroundings. Interventions to promote the human immune response may aid in the reversal of the inflammatory disease process in PAIS patients. Potential therapeutic approaches including prebiotics, probiotics, fecal transplants, antibiotic, bacteriophages therapy, and microbiome-derived metabolites may similarly modify host flora and decrease virus-induced intestinal inflammation, therefore, to prevent or alleviate PAIS development.
When consumed, probiotics are live microorganisms and prebiotics are typically high-fiber foods for human microflora to maintain or improve healthy microflora. The effects of probiotics/prebiotics therapy have been evaluated in several clinical and experimental human studies. A recent study demonstrated that treating patients with Lactobacillus probiotics and inulin can relieve acute and long-term symptoms among individuals with COVID-19 [84]. Another randomized controlled trial indicated probiotic intake had a positive effect on PASC by improving a variety of symptoms [85]. Prebiotics/probiotics can modulate the gut microbiome to help manage gastrointestinal symptoms. Increasing dietary fiber intake can cause structural alterations in gut microbiota, mitigating both gastrointestinal and non-gastrointestinal symptoms such as anxiety and palpitations in patients with PASC [86]. A case report showed a two-month treatment of high-fiber formula helped alleviate gastrointestinal symptoms by modulating the gut microbiota in one PASC patient [87]. There have been numerous studies involving treatment for ME/CFS as well [88]. However, it is still inconclusive to consider probiotics as a treatment for gastrointestinal symptoms in ME/CFS patients due to the overall poor study quality [89]. Nowadays, the effects of prebiotics/probiotics therapy have been assessed in ongoing clinical trials for both ME/CFS and PASC (NCT04741841, NCT04950803, and NCT05080244).
Fecal microbiota transplantation (FMT) is another therapeutic method using gut microorganisms from donated feces to treat the gut microbiome dysbiosis associated with pathological health conditions. It has achieved impressive therapeutic effects for recurrent Clostridioides difficile infection in 90% of cases [90]. In terms of treating ME/CFS, fecal transplantation has also been discussed. There are several preliminary indications that the microbiome might be an important player in the disease, especially its neurological symptoms [91]. Two controlled studies in double-blind randomized trials are presently underway (NCT04158427 and NCT03691987) to assess the effectiveness of FMT in treating ME/CFS. Meanwhile, there is no completed trial to evaluate the clinical efficacy of FMT treatment in post-COVID-19 neuropsychiatric conditions yet (NCT05556733).
Due to the possibility of COVID-19 leading to bacterial coinfection, antibiotics were frequently prescribed to patients. Antibiotic usage can eliminate bacterial pathogens while also affecting the normal symbiotic microbiota, and excessive use can lead to increased bacterial resistance and dysbiosis of the microbiota. Although numerous studies have suggested antibiotic usage is not associated with disease development in both PACS and ME/CFS [52], [81], more studies are warranted to investigate the role of antibiotics in the PAISs, and antibiotic usage should be taken as a potential covariable in the analysis. Meanwhile, the issues of antibiotics encourage the search for alternative methods of limiting bacterial growth and restoring the normal balance of the microbiota in the human body. Bacteriophages are promising candidates as potential regulators of the microbiota, facing the increasing problem of bacterial resistance to antibiotics [92]. Bacteriophage cocktail for oral administration targeting multiple species of bacteria was used for the rehabilitation of PASC patients and showed significant improvements in saturation and respiratory rate as well as a decrease in inflammatory markers [93].
Furthermore, metformin is usually prescribed for diabetes to control blood sugar levels, partly by altering the gut microbiome. A recent study revealed that the administration of metformin induced a 42% reduction in COVID-19 sequelae relative to controls [94]. Increased Escherichia coli abundance due to metformin supplementation, as well as decreased Intestinibacter bartlettii abundance and increased SCFA production in humans, may be responsible for metformin-mediated health effects, specifically with regard to gut microbiota modifications [95]. It has been suggested that by activating AMP-activated protein kinase, metformin causes ACE2 to be phosphorylated, leading to a conformational change that might prevent SARS-CoV-2 from binding to the receptor. In vitro, metformin increases the mRNA expression of ACE2 and TMPRSS2 in human hepatocytes [96]. Lastly, the level of ACE2 activity is modulated by the local microbiome. The success of metformin also sheds light on other potential therapies for COVID-19 bowel physiology and PASC.
6. Future directions of research on the microbiome of PAIS
Consisting of PASC, ME/CFS, and other under-recognized post-acute infection sequelae, PAIS conditions represent vital global public health concerns. The overlap in symptoms, signs, and clinical features suggests a unified pathological mechanism. The associations among the gut microbiome, acute infection, and PAIS appear obvious, while current research has not yet established causality. With the various constellations of symptoms and severity observed in PAIS patients, multiple mechanisms may be involved. To solve the mystery and advance the field, well-designed longitudinal animal and human studies that consider physical activity levels, antibiotic intake, vaccination status, and other potential confounding factors are crucial to systematically investigate the association between microbiome and the development of PAIS. Despite the complexity, the field is expanding with substantial opportunities, and current knowledge presents a unique opportunity to apply microbiome-relevant applications in prevention, diagnosis, and treatment of PAIS.
Author contributions
JL and CG contributed to conception of the review. CG, BY and JW wrote the manuscript. CG and BY contributed equally and share the first authorship. All authors read and approved the final manuscript.
Funding
This work was supported by the National Key Research and Development Program of China (2018YFE0208000).
Declaration of Competing Interest
The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
References
- 1.Davis H.E., McCorkell L., Vogel J.M., Topol E.J. Long COVID: major findings, mechanisms and recommendations. Nat Rev Microbiol. 2023;21:133–146. doi: 10.1038/s41579-022-00846-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Soriano J.B., Murthy S., Marshall J.C., Relan P., Diaz J.V. WHO clinical case definition working group on post-COVID-19 condition. A clinical case definition of post-COVID-19 condition by a Delphi consensus. Lancet Infect Dis. 2022;22:e102–e107. doi: 10.1016/S1473-3099(21)00703-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Choutka J., Jansari V., Hornig M., Iwasaki A. Unexplained post-acute infection syndromes. Nat Med. 2022;28:911–923. doi: 10.1038/s41591-022-01810-6. [DOI] [PubMed] [Google Scholar]
- 4.Owens B. How “long covid” is shedding light on postviral syndromes. BMJ. 2022;378:o2188. doi: 10.1136/bmj.o2188. [DOI] [PubMed] [Google Scholar]
- 5.Jubelt B., Agre J.C. Characteristics and management of postpolio syndrome. JAMA. 2000;284(4):412. doi: 10.1001/jama.284.4.412. [DOI] [PubMed] [Google Scholar]
- 6.Komaroff A.L., Lipkin W.I. Insights from myalgic encephalomyelitis/chronic fatigue syndrome may help unravel the pathogenesis of postacute COVID-19 syndrome. Trends Mol Med. 2021;27:895–906. doi: 10.1016/j.molmed.2021.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.O’Boyle S., Nacul L., Nacul F.E., Mudie K., Kingdon C.C., Cliff J.M., et al. A natural history of disease framework for improving the prevention, management, and research on post-viral fatigue syndrome and other forms of myalgic encephalomyelitis/chronic fatigue syndrome. Front Med. 2021;8 doi: 10.3389/fmed.2021.688159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Rodríguez-Morales A.J., Cardona-Ospina J.A., Fernanda Urbano-Garzón S., Sebastian Hurtado-Zapata J. Prevalence of post-chikungunya infection chronic inflammatory arthritis: a systematic review and meta-analysis. Arthritis Care Res (Hoboken) 2016;68:1849–1858. doi: 10.1002/acr.22900. [DOI] [PubMed] [Google Scholar]
- 9.Litleskare S., Rortveit G., Eide G.E., Hanevik K., Langeland N., Wensaas K.-A. Prevalence of irritable bowel syndrome and chronic fatigue 10 years after giardia infection. Clin Gastroenterol Hepatol. 2018;16:1064–1072.e4. doi: 10.1016/j.cgh.2018.01.022. [DOI] [PubMed] [Google Scholar]
- 10.Shantha J.G., Crozier I., Yeh S. An update on ocular complications of Ebola virus disease. Curr Opin Ophthalmol. 2017;28:600–606. doi: 10.1097/ICU.0000000000000426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Sherif Z.A., Gomez C.R., Connors T.J., Henrich T.J., Reeves W.B. RECOVER Mechanistic Pathway Task Force. Pathogenic mechanisms of post-acute sequelae of SARS-CoV-2 infection (PASC) Elife. 2023;12 doi: 10.7554/eLife.86002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Tamburini S., Shen N., Wu H.C., Clemente J.C. The microbiome in early life: implications for health outcomes. Nat Med. 2016;22:713–722. doi: 10.1038/nm.4142. [DOI] [PubMed] [Google Scholar]
- 13.Sender R., Fuchs S., Milo R. Revised estimates for the number of human and bacteria cells in the body. PLoS Biol. 2016;14 doi: 10.1371/journal.pbio.1002533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Soderholm A.T., Pedicord V.A. Intestinal epithelial cells: at the interface of the microbiota and mucosal immunity. Immunology. 2019;158:267–280. doi: 10.1111/imm.13117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Clemente J.C., Manasson J., Scher J.U. The role of the gut microbiome in systemic inflammatory disease. BMJ. 2018;360:j5145. doi: 10.1136/bmj.j5145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Libertucci J., Young V.B. The role of the microbiota in infectious diseases. Nat Microbiol. 2019;4:35–45. doi: 10.1038/s41564-018-0278-4. [DOI] [PubMed] [Google Scholar]
- 17.Karst S.M. The influence of commensal bacteria on infection with enteric viruses. Nat Rev Microbiol. 2016;14:197–204. doi: 10.1038/nrmicro.2015.25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Cortez V., Margolis E., Schultz-Cherry S. Astrovirus and the microbiome. Curr Opin Virol. 2019;37:10–15. doi: 10.1016/j.coviro.2019.05.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Kim A.H., Hogarty M.P., Harris V.C., Baldridge M.T. The complex interactions between rotavirus and the gut microbiota. Front Cell Infect Microbiol. 2020;10 doi: 10.3389/fcimb.2020.586751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Lockhart A., Mucida D., Parsa R. Immunity to enteric viruses. Immunity. 2022;55:800–818. doi: 10.1016/j.immuni.2022.04.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Lamont R.J., Fitzsimonds Z.R., Wang H., Gao S. Role of Porphyromonas gingivalis in oral and orodigestive squamous cell carcinoma. Periodontol 2000. 2022;89:154–165. doi: 10.1111/prd.12425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Jenul C., Horswill A.R. Regulation of staphylococcus aureus virulence. Microbiol Spectr. 2019;7 doi: 10.1128/microbiolspec.GPP3-0031-2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Raplee I., Walker L., Xu L., Surathu A., Chockalingam A., Stewart S., et al. Emergence of nosocomial associated opportunistic pathogens in the gut microbiome after antibiotic treatment. Antimicrob Resist Infect Control. 2021;10 doi: 10.1186/s13756-021-00903-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Zheng D., Liwinski T., Elinav E. Interaction between microbiota and immunity in health and disease. Cell Res. 2020;30:492–506. doi: 10.1038/s41422-020-0332-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Liu Q., Mak J.W.Y., Su Q., Yeoh Y.K., Lui G.C.-Y., Ng S.S.S., et al. Gut microbiota dynamics in a prospective cohort of patients with post-acute COVID-19 syndrome. Gut. 2022;71:544–552. doi: 10.1136/gutjnl-2021-325989. [DOI] [PubMed] [Google Scholar]
- 26.Simpson C.A., Diaz-Arteche C., Eliby D., Schwartz O.S., Simmons J.G., Cowan C.S.M. The gut microbiota in anxiety and depression - A systematic review. Clin Psychol Rev. 2021;83 doi: 10.1016/j.cpr.2020.101943. [DOI] [PubMed] [Google Scholar]
- 27.Wu Y., Cheng X., Jiang G., Tang H., Ming S., Tang L., et al. Altered oral and gut microbiota and its association with SARS-CoV-2 viral load in COVID-19 patients during hospitalization. NPJ Biofilms Micro. 2021;7 doi: 10.1038/s41522-021-00232-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Cheng X., Zhang Y., Li Y., Wu Q., Wu J., Park S.K., et al. Meta-analysis of 16S rRNA microbial data identified alterations of the gut microbiota in COVID-19 patients during the acute and recovery phases. BMC Microbiol. 2022;22(1):13. doi: 10.1186/s12866-022-02686-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Zuo T., Zhang F., Lui G.C.Y., Yeoh Y.K., Li A.Y.L., Zhan H., et al. Alterations in gut microbiota of patients with COVID-19 during time of hospitalization. Gastroenterology. 2020;159:944–955.e8. doi: 10.1053/j.gastro.2020.05.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Candel S., Tyrkalska S.D., Álvarez-Santacruz C., Mulero V. The nasopharyngeal microbiome in COVID-19. Emerg Microbes Infect. 2023;12 doi: 10.1080/22221751.2023.2165970. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Gu S., Chen Y., Wu Z., Chen Y., Gao H., Lv L., et al. Alterations of the gut microbiota in patients with coronavirus disease 2019 or H1N1 influenza. Clin Infect Dis. 2020;71:2669–2678. doi: 10.1093/cid/ciaa709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Woodall C.A., McGeoch L.J., Hay A.D., Hammond A. Respiratory tract infections and gut microbiome modifications: a systematic review. PLoS One. 2022;17 doi: 10.1371/journal.pone.0262057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Zhou J., Zhang Y., Cui P., Luo L., Chen H., Liang B., et al. Gut microbiome changes associated with HIV infection and sexual orientation. Front Cell Infect Microbiol. 2020;10 doi: 10.3389/fcimb.2020.00434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Milosevic I., Russo E., Vujovic A., Barac A., Stevanovic O., Gitto S., et al. Microbiota and viral hepatitis: state of the art of a complex matter. World J Gastroenterol. 2021;27:5488–5501. doi: 10.3748/wjg.v27.i33.5488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Liu Y., Wang J., Wu C. Microbiota and tuberculosis: a potential role of probiotics, and postbiotics. Front Nutr. 2021;8:1–10. doi: 10.3389/fnut.2021.626254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Sencio V., Barthelemy A., Tavares L.P., Machado M.G., Soulard D., Cuinat C., et al. Gut dysbiosis during influenza contributes to pulmonary pneumococcal superinfection through altered short-chain fatty acid production. Cell Rep. 2020;30 doi: 10.1016/j.celrep.2020.02.013. 2934-2947.e6. [DOI] [PubMed] [Google Scholar]
- 37.Yildiz S., Mazel-Sanchez B., Kandasamy M., Manicassamy B., Schmolke M. Influenza A virus infection impacts systemic microbiota dynamics and causes quantitative enteric dysbiosis. Microbiome. 2018;6 doi: 10.1186/s40168-017-0386-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Groves H.T., Higham S.L., Moffatt M.F., Cox M.J., Tregoning J.S. Respiratory viral infection alters the gut microbiota by inducing inappetence. MBio. 2020;11 doi: 10.1128/mBio.03236-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Chancharoenthana W., Kamolratanakul S., Ariyanon W., Thanachartwet V., Phumratanaprapin W., Wilairatana P., et al. Abnormal blood bacteriome, gut dysbiosis, and progression to severe dengue disease. Front Cell Infect Microbiol. 2022;12:1–13. doi: 10.3389/fcimb.2022.890817. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Cui G.-Y., Rao B.-C., Zeng Z.-H., Wang X.-M., Ren T., Wang H.-Y., et al. Characterization of oral and gut microbiome and plasma metabolomics in COVID-19 patients after 1-year follow-up. Mil Med Res. 2022;9 doi: 10.1186/s40779-022-00387-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Zhou Y., Zhang J., Zhang D., Ma W.-L., Wang X. Linking the gut microbiota to persistent symptoms in survivors of COVID-19 after discharge. J Microbiol. 2021;59:941–948. doi: 10.1007/s12275-021-1206-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Chen Y., Gu S., Chen Y., Lu H., Shi D., Guo J., et al. Six-month follow-up of gut microbiota richness in patients with COVID-19. Gut. 2022;71:222–225. doi: 10.1136/gutjnl-2021-324090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Guo C., Che X., Briese T., Ranjan A., Allicock O., Yates R.A., et al. Deficient butyrate-producing capacity in the gut microbiome is associated with bacterial network disturbances and fatigue symptoms in ME/CFS. Cell Host Microbe. 2023;31:288–304.e8. doi: 10.1016/j.chom.2023.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Xiong R., Gunter C., Fleming E., Vernon S.D., Bateman L., Unutmaz D., et al. Multi-’omics of gut microbiome-host interactions in short- and long-term myalgic encephalomyelitis/chronic fatigue syndrome patients. Cell Host Microbe. 2023;31 doi: 10.1016/j.chom.2023.01.001. 273-287.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Nagy-Szakal D., Williams B.L., Mishra N., Che X., Lee B., Bateman L., et al. Fecal metagenomic profiles in subgroups of patients with myalgic encephalomyelitis/chronic fatigue syndrome. Microbiome. 2017;5 doi: 10.1186/s40168-017-0261-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Morrissette M., Pitt N., González A., Strandwitz P., Caboni M., Rebman A.W., et al. A distinct microbiome signature in posttreatment lyme disease patients. MBio. 2020;11 doi: 10.1128/mBio.02310-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Newsome R.C., Gauthier J., Hernandez M.C., Abraham G.E., Robinson T.O., Williams H.B., et al. The gut microbiome of COVID-19 recovered patients returns to uninfected status in a minority-dominated United States cohort. Gut Microbes. 2021;13:1–15. doi: 10.1080/19490976.2021.1926840. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Xiang Z., Koo H., Chen Q., Zhou X., Liu Y., Simon-Soro A. Potential implications of SARS-CoV-2 oral infection in the host microbiota. J Oral Microbiol. 2020;13 doi: 10.1080/20002297.2020.1853451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Haran J.P., Bradley E., Zeamer A.L., Cincotta L., Salive M.-C., Dutta P., et al. Inflammation-type dysbiosis of the oral microbiome associates with the duration of COVID-19 symptoms and long COVID. JCI Insight. 2021;6 doi: 10.1172/jci.insight.152346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Giron L.B., Peluso M.J., Ding J., Kenny G., Zilberstein N.F., Koshy J., et al. Markers of fungal translocation are elevated during post-acute sequelae of SARS-CoV-2 and induce NF-κB signaling. JCI Insight. 2022;7 doi: 10.1172/jci.insight.160989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Borman A.M., Fountain H., Guy R., Casale E., Gerver S.M., Elgohari S., et al. Increased mortality in COVID-19 patients with fungal co- and secondary infections admitted to intensive care or high dependency units in NHS hospitals in England. J Infect. 2022;84:579–613. doi: 10.1016/j.jinf.2021.12.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.König R.S., Albrich W.C., Kahlert C.R., Bahr L.S., Löber U., Vernazza P., et al. The gut microbiome in myalgic encephalomyelitis (ME)/chronic fatigue syndrome (CFS) Front Immunol. 2021;12 doi: 10.3389/fimmu.2021.628741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Briese T., Tokarz R., Bateman L., Che X., Guo C., Jain K., et al. A multicenter virome analysis of blood, feces, and saliva in myalgic encephalomyelitis/chronic fatigue syndrome. J Med Virol. 2023;95 doi: 10.1002/jmv.28993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Proal A., Marshall T. Myalgic encephalomyelitis/chronic fatigue syndrome in the era of the human microbiome: persistent pathogens drive chronic symptoms by interfering with host metabolism, gene expression, and immunity. Front Pedia. 2018;6 doi: 10.3389/fped.2018.00373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Thorson A.E., Deen G.F., Bernstein K.T., Liu W.J., Yamba F., Habib N., et al. Persistence of Ebola virus in semen among Ebola virus disease survivors in Sierra Leone: a cohort study of frequency, duration, and risk factors. PLoS Med. 2021;18 doi: 10.1371/journal.pmed.1003273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Aid M., Abbink P., Larocca R.A., Boyd M., Nityanandam R., Nanayakkara O., et al. Zika virus persistence in the central nervous system and lymph nodes of rhesus monkeys. Cell. 2017;169 doi: 10.1016/j.cell.2017.04.008. 610-620.e14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Hosseini S., Wilk E., Michaelsen-Preusse K., Gerhauser I., Baumgärtner W., Geffers R., et al. Long-term neuroinflammation induced by influenza A virus infection and the impact on hippocampal neuron morphology and function. J Neurosci. 2018;38:3060–3080. doi: 10.1523/JNEUROSCI.1740-17.2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Wu Y., Guo C., Tang L., Hong Z., Zhou J., Dong X., et al. Prolonged presence of SARS-CoV-2 viral RNA in faecal samples. Lancet Gastroenterol Hepatol. 2020;5:434–435. doi: 10.1016/S2468-1253(20)30083-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Xu E., Xie Y., Al-Aly Z. Long-term gastrointestinal outcomes of COVID-19. Nat Commun. 2023;14 doi: 10.1038/s41467-023-36223-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Chia J.K.S. The role of enterovirus in chronic fatigue syndrome. J Clin Pathol. 2005;58:1126–1132. doi: 10.1136/jcp.2004.020255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Zajac V., Mego M., Martinický D., Stevurková V., Cierniková S., Ujházy E., et al. Testing of bacteria isolated from HIV/AIDS patients in experimental models. Neuro Endocrinol Lett. 2006;27(Suppl 2):61–64. [PubMed] [Google Scholar]
- 62.Zajac V., Matelova L., Liskova A., Mego M., Holec V., Adamcikova Z., et al. Confirmation of HIV-like sequences in respiratory tract bacteria of Cambodian and Kenyan HIV-positive pediatric patients. Med Sci Monit. 2011;17:CR154–CR158. doi: 10.12659/msm.881449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Gold J.E., Okyay R.A., Licht W.E., Hurley D.J. Investigation of Long COVID prevalence and its relationship to epstein-barr virus reactivation. Pathog (Basel, Switz. 2021;10 doi: 10.3390/pathogens10060763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Gatto I., Biagioni E., Coloretti I., Farinelli C., Avoni C., Caciagli V., et al. Cytomegalovirus blood reactivation in COVID-19 critically ill patients: risk factors and impact on mortality. Intensive Care Med. 2022;48:706–713. doi: 10.1007/s00134-022-06716-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Klein J., Wood J., Jaycox J., Lu P., Dhodapkar R.M., Gehlhausen J.R., et al. Distinguishing features of Long COVID identified through immune profiling. MedRxiv Prepr Serv Heal Sci. 2022 doi: 10.1101/2022.08.09.22278592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Teixeira P.C., Dorneles G.P., Santana Filho P.C., da Silva I.M., Schipper L.L., Postiga I.A.L., et al. Increased LPS levels coexist with systemic inflammation and result in monocyte activation in severe COVID-19 patients. Int Immunopharmacol. 2021;100 doi: 10.1016/j.intimp.2021.108125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Waltz E. Could long COVID be linked to herpes viruses? Early data offer a hint. Nature. 2022 doi: 10.1038/d41586-022-02296-5. [DOI] [PubMed] [Google Scholar]
- 68.Zubchenko S., Kril I., Nadizhko O., Matsyura O., Chopyak V. Herpesvirus infections and post-COVID-19 manifestations: a pilot observational study. Rheuma Int. 2022;42:1523–1530. doi: 10.1007/s00296-022-05146-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.German E.R., Jairath M.K., Caston J. Treatment of long-Haul COVID patients with off-label acyclovir. Cureus. 2023 doi: 10.7759/cureus.37926. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Agirman G., Yu K.B., Hsiao E.Y. Signaling inflammation across the gut-brain axis. Science. 2021;374:1087–1092. doi: 10.1126/science.abi6087. [DOI] [PubMed] [Google Scholar]
- 71.Yoo B.B., Mazmanian S.K. The enteric network: interactions between the immune and nervous systems of the gut. Immunity. 2017;46:910–926. doi: 10.1016/j.immuni.2017.05.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Vakili K., Fathi M., Yaghoobpoor S., Sayehmiri F., Nazerian Y., Nazerian A., et al. 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. 2022;12 doi: 10.3389/fcimb.2022.983089. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Omdal R. The biological basis of chronic fatigue: neuroinflammation and innate immunity. Curr Opin Neurol. 2020;33:391–396. doi: 10.1097/WCO.0000000000000817. [DOI] [PubMed] [Google Scholar]
- 74.Hrvatin S., Sun S., Wilcox O.F., Yao H., Lavin-Peter A.J., Cicconet M., et al. Neurons that regulate mouse torpor. Nature. 2020;583:115–121. doi: 10.1038/s41586-020-2387-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Tate W., Walker M., Sweetman E., Helliwell A., Peppercorn K., Edgar C., et al. Molecular mechanisms of neuroinflammation in ME/CFS and long COVID to sustain disease and promote relapses. Front Neurol. 2022;13 doi: 10.3389/fneur.2022.877772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Oudit G.Y., Wang K., Viveiros A., Kellner M.J., Penninger J.M. Angiotensin-converting enzyme 2-at the heart of the COVID-19 pandemic. Cell. 2023;186:906–922. doi: 10.1016/j.cell.2023.01.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Yu Z., Yang Z., Wang Y., Zhou F., Li S., Li C., et al. Recent advance of ACE2 and microbiota dysfunction in COVID-19 pathogenesis. Heliyon. 2021;7 doi: 10.1016/j.heliyon.2021.e07548. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Alenazy M.F., Aljohar H.I., Alruwaili A.R., Daghestani M.H., Alonazi M.A., Labban R.S., et al. Gut microbiota dynamics in relation to long-COVID-19 syndrome: role of probiotics to combat psychiatric complications. Metabolites. 2022;12 doi: 10.3390/metabo12100912. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Brogna C., Brogna B., Bisaccia D.R., Lauritano F., Marino G., Montano L., et al. Could SARS-CoV-2 have bacteriophage behavior or induce the activity of other bacteriophages? Vaccines. 2022;10 doi: 10.3390/vaccines10050708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Faro M., Sàez-Francás N., Castro-Marrero J., Aliste L., Fernández de Sevilla T., Alegre J. Gender differences in chronic fatigue syndrome. Reum Clin. 2016;12:72–77. doi: 10.1016/j.reuma.2015.05.007. [DOI] [PubMed] [Google Scholar]
- 81.Bai F., Tomasoni D., Falcinella C., Barbanotti D., Castoldi R., Mulè G., et al. Female gender is associated with long COVID syndrome: a prospective cohort study. Clin Microbiol Infect. 2022;28:611.e9–611.e16. doi: 10.1016/j.cmi.2021.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Thomas N., Gurvich C., Huang K., Gooley P.R., Armstrong C.W. The underlying sex differences in neuroendocrine adaptations relevant to Myalgic Encephalomyelitis Chronic Fatigue Syndrome. Front Neuroendocr. 2022;66 doi: 10.1016/j.yfrne.2022.100995. [DOI] [PubMed] [Google Scholar]
- 83.Subramanian A., Nirantharakumar K., Hughes S., Myles P., Williams T., Gokhale K.M., et al. Symptoms and risk factors for long COVID in non-hospitalized adults. Nat Med. 2022;28:1706–1714. doi: 10.1038/s41591-022-01909-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Thomas R., Aldous J., Forsyth R., Chater A., Williams M. The influence of a blend of probiotic lactobacillus and prebiotic inulin on the duration and severity of symptoms among individuals with covid-19. Infect Dis Diagn Treat. 2021;5 doi: 10.29011/2577-1515.100182. [DOI] [Google Scholar]
- 85.Gutiérrez-Castrellón P., Gandara-Martí T., Abreu Y Abreu A.T., Nieto-Rufino C.D., López-Orduña E., Jiménez-Escobar I., et al. Probiotic improves symptomatic and viral clearance in Covid19 outpatients: a randomized, quadruple-blinded, placebo-controlled trial. Gut Microbes. 2022;14 doi: 10.1080/19490976.2021.2018899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Barrea L., Grant W.B., Frias-Toral E., Vetrani C., Verde L., de Alteriis G., et al. Dietary recommendations for Post-COVID-19 Ssyndrome. Nutrients. 2022;14 doi: 10.3390/nu14061305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Wang Y., Wu G., Zhao L., Wang W. Nutritional modulation of gut microbiota alleviates severe gastrointestinal symptoms in a patient with post-acute COVID-19 syndrome. MBio. 2022;13 doi: 10.1128/mbio.03801-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Varesi A., Deumer U.-S., Ananth S., Ricevuti G. The emerging role of gut microbiota in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS): current evidence and potential therapeutic applications. J Clin Med. 2021;10 doi: 10.3390/jcm10215077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Corbitt M., Campagnolo N., Staines D., Marshall-Gradisnik S. A systematic review of probiotic interventions for gastrointestinal symptoms and irritable bowel syndrome in chronic fatigue syndrome/myalgic encephalomyelitis (CFS/ME) Probiotics Antimicrob Proteins. 2018;10:466–477. doi: 10.1007/s12602-018-9397-8. [DOI] [PubMed] [Google Scholar]
- 90.Mullish B.H., Quraishi M.N., Segal J.P., McCune V.L., Baxter M., Marsden G.L., et al. The use of faecal microbiota transplant as treatment for recurrent or refractory Clostridium difficile infection and other potential indications: joint British Society of Gastroenterology (BSG) and Healthcare Infection Society (HIS) guidelines. Gut. 2018;67:1920–1941. doi: 10.1136/gutjnl-2018-316818. [DOI] [PubMed] [Google Scholar]
- 91.Kenyon J.N., Coe S., Izadi H. A retrospective outcome study of 42 patients with Chronic Fatigue Syndrome, 30 of whom had Irritable Bowel Syndrome. Half were treated with oral approaches, and half were treated with Faecal Microbiome Transplantation. Hum Micro J. 2019;13 doi: 10.1016/j.humic.2019.100061. [DOI] [Google Scholar]
- 92.Gordillo Altamirano F.L., Barr J.J. Phage Therapy in the Postantibiotic Era. Clin Microbiol Rev. 2019;32 doi: 10.1128/CMR.00066-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Zurabov F.M., Chernevskaya E.A., Beloborodova N.V., Zurabov A.Y., Petrova M.V., Yadgarov M.Y., et al. Bacteriophage cocktails in the post-COVID rehabilitation. Viruses. 2022;14 doi: 10.3390/v14122614. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Bramante C.T., Buse J.B., Liebovitz D., Nicklas J., Puskarich M.A., Cohen K., et al. Outpatient treatment of Covid-19 with metformin, ivermectin, and fluvoxamine and the development of Long Covid over 10-month follow-up. MedRxiv Prepr Serv Heal Sci. 2022 doi: 10.1101/2022.12.21.22283753. [DOI] [Google Scholar]
- 95.Mueller N.T., Differding M.K., Zhang M., Maruthur N.M., Juraschek S.P., Miller E.R., et al. Metformin affects gut microbiome composition and function and circulating short-chain fatty acids: a randomized trial. Diabetes Care. 2021;44:1462–1471. doi: 10.2337/dc20-2257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Malhotra A., Hepokoski M., McCowen K.C., Y-J Shyy J. ACE2, metformin, and COVID-19. IScience. 2020;23 doi: 10.1016/j.isci.2020.101425. [DOI] [PMC free article] [PubMed] [Google Scholar]