An Expert Report on the Intrabacterial Localization of SARS-CoV-2 in the Human Gut Microbiome
This hypothesis remains speculative and unverified. The observed phenomena could be better explained by artifacts or non-infectious associations.
Executive Summary
The COVID-19 pandemic has spurred an unprecedented volume of research into the multifaceted interactions between SARS-CoV-2 and its human host. A significant body of this work has focused on the gastrointestinal (GI) tract, establishing it as a key site for viral replication, immune modulation, and persistent pathology. Within this context, the question of whether SARS-CoV-2 has been found inside bacteria of the human gut microbiome probes the frontier of our understanding. This report provides an exhaustive analysis of this query, synthesizing available evidence to deliver a nuanced and definitive answer.
The current scientific landscape reveals a complex picture. There is overwhelming evidence for a profound, albeit indirect, relationship between SARS-CoV-2 and the gut microbiota. The virus induces significant gut dysbiosis, which correlates with disease severity and may contribute to post-acute sequelae. The GI tract serves as a durable reservoir for viral RNA, with shedding in feces often long outlasting respiratory clearance. Furthermore, gut bacteria can indirectly modulate host susceptibility to the virus by metabolically altering viral receptors on human cells.
However, the specific claim of SARS-CoV-2 being found inside bacteria, adopting a "bacteriophage-like" behavior, originates exclusively from a series of studies by a single, interconnected research group. These researchers present multiple lines of evidence—including electron microscopy, in vitro culture experiments showing viral replication, isotope labeling demonstrating de novo viral protein synthesis in bacterial cultures, and plaque assays on bacterial lawns—to support their extraordinary hypothesis.
A critical deconstruction of this evidence reveals that while the findings are provocative, they are not yet conclusive. Each line of evidence is susceptible to alternative interpretations, the most significant of which is the potential for unacknowledged contamination of the experimental systems with replication-competent human cellular components. Furthermore, the claim faces profound biological plausibility challenges. The canonical entry mechanism of SARS-CoV-2 is exquisitely adapted to eukaryotic host cells and is fundamentally incompatible with the structure of a prokaryotic bacterial cell wall. The virus lacks the known genetic and structural machinery to overcome this barrier.
Crucially, this "bacteriophage-like" hypothesis has not been independently replicated or corroborated by the wider scientific community. A comprehensive review of the literature reveals a conspicuous absence of this claim in studies from other laboratories, and it is not mentioned in clinical or public health guidelines. In science, particularly in the face of a revolutionary claim, independent verification is the bedrock of acceptance.
Therefore, the conclusion of this report is that while one research group has indeed reported finding SARS-CoV-2 inside gut bacteria, this remains an unverified and highly controversial hypothesis. The observations, though intriguing, are more parsimoniously explained by non-infectious viral-bacterial association or experimental artifacts. Until rigorous, independent replication that definitively rules out alternative explanations is forthcoming, the concept of SARS-CoV-2 infecting gut bacteria should be regarded as a provocative but unsubstantiated outlier, not an established scientific fact.
I. The Gut Microbiome as a Dynamic Theater for SARS-CoV-2 Interaction
Before addressing the specific and controversial claim of intrabacterial viral presence, it is essential to establish the well-documented, complex, and multifaceted relationship between SARS-CoV-2 and the gut microbiome. The GI tract is not a passive bystander in COVID-19; rather, it is an active theater where the virus, the host immune system, and a vast community of microbes engage in a dynamic interplay that significantly influences disease progression, severity, and long-term outcomes. Understanding this broader context is critical for appreciating why the question of direct bacterial infection is both scientifically relevant and significant.
A. The Gut-Lung Axis: Systemic Effects of a Respiratory Virus
While SARS-CoV-2 is primarily a respiratory pathogen, its effects ripple throughout the body, a phenomenon well-explained by the concept of the "gut-lung axis." This axis describes a bidirectional communication network linking the microbial ecosystems of the gastrointestinal and respiratory tracts, primarily mediated by the immune system and microbial metabolites.1 The health and composition of the gut microbiota play a crucial role in calibrating local and systemic immune responses. A balanced gut microbiome helps maintain immune homeostasis, ensuring that the body can mount a robust defense against pathogens without tipping into excessive, damaging inflammation.4
During a respiratory viral infection like COVID-19, this axis becomes critically important. Gut microbiota and their metabolites, such as short-chain fatty acids (SCFAs), can modulate the activity of immune cells far beyond the gut, influencing the host's antiviral response in the lungs.1 Conversely, inflammation originating in the lungs can be transmitted systemically, disrupting the delicate balance of the gut environment and altering the microbial community.1 This crosstalk means that the state of the gut microbiome prior to infection can influence susceptibility and severity, and the infection itself can, in turn, inflict lasting damage on the gut ecosystem. This established biological linkage underscores that events in the gut are not merely localized side effects but are integral to the overall pathophysiology of COVID-19.2
B. SARS-CoV-2-Induced Gut Dysbiosis: A Profile of Altered Communities
One of the most consistent findings in COVID-19 research is that the infection profoundly alters the composition and function of the gut microbiome, a condition known as dysbiosis.8 Numerous studies, employing metagenomic sequencing of fecal samples from patients, have painted a clear picture of this microbial imbalance. The characteristic signature of COVID-19-associated dysbiosis involves a significant reduction in the abundance of beneficial, commensal bacteria, particularly those known for producing anti-inflammatory compounds.12 Key among these depleted microbes are members of the Ruminococcaceae and Lachnospiraceae families, including potent butyrate-producers like Faecalibacterium prausnitzii and Eubacterium rectale, as well as various species of Bifidobacterium.4
Concurrently, this depletion of beneficial microbes is accompanied by an enrichment of opportunistic pathogens. Bacteria such as Enterococcus, Streptococcus, Coprobacillus, and various members of the Enterobacteriaceae family (which includes Escherichia coli) are frequently found in higher abundance in the guts of COVID-19 patients.4 This shift creates a more pro-inflammatory gut environment. The magnitude of this dysbiosis has been shown to correlate directly with disease severity; patients with more severe COVID-19 tend to exhibit more pronounced alterations in their gut microbiota.7 This suggests that the microbiome's composition could serve as a valuable prognostic indicator, helping to identify individuals at higher risk for poor outcomes.4
Furthermore, this dysbiosis is not a transient phenomenon that resolves upon viral clearance. Follow-up studies have demonstrated that these microbial imbalances can persist for weeks or even months after recovery from the acute phase of the illness.10 This long-term disruption is increasingly implicated as a potential contributing factor to the constellation of persistent symptoms known as Post-Acute Sequelae of COVID-19 (PASC), or "Long COVID," particularly the gastrointestinal and fatigue-related symptoms.13
C. The Gastrointestinal Tract as a Reservoir for Viral Replication and Persistence
The GI tract's significant role in COVID-19 is not limited to indirect effects on immunity and microbial composition; it is also a direct target of SARS-CoV-2 infection. The biological basis for this is the high expression of the virus's primary entry receptor, angiotensin-converting enzyme 2 (ACE2), on the surface of intestinal epithelial cells (enterocytes).4 This makes the gut a major extrapulmonary site for viral invasion and replication.
Compelling evidence supports the active replication of SARS-CoV-2 within the gut. Studies have detected not just viral RNA, but also viral subgenomic RNA (sgRNA) in stool samples from infected individuals.17 The presence of sgRNA is a reliable marker of active viral replication, as these molecules are produced during the transcription of the viral genome and are not typically packaged into new virions. This finding confirms that the gut is more than just a passive conduit for swallowed virus from the respiratory tract; it is a genuine site of viral propagation.17
This active gut infection leads to one of the hallmark features of COVID-19's gastrointestinal involvement: prolonged viral shedding in feces. A substantial proportion of patients continue to test positive for viral RNA in their stool for weeks or even months after their respiratory samples (e.g., nasopharyngeal swabs) have become negative.11 This persistent presence of viral genetic material in the gut long after the resolution of respiratory symptoms creates a significant scientific puzzle. It raises fundamental questions about the nature of this persistence: Is it due to a low-level, smoldering infection of human enterocytes, or could the virus be harbored in another component of the complex gut ecosystem? This very puzzle provides the scientific motivation for investigating more unconventional hypotheses, including the potential for viral interaction with the gut's bacterial residents.
D. Indirect Modulation of Viral Infectivity by Gut Bacteria
The relationship between SARS-CoV-2 and the gut microbiome is bidirectional. Just as the virus affects the bacteria, the bacteria can, in turn, influence the virus. This modulation occurs through several indirect, yet powerful, mechanisms that can alter host susceptibility to infection.
One key mechanism is metabolic modulation. Certain beneficial gut bacteria, particularly fiber-fermenting species from the Clostridia and Bacteroides genera, produce short-chain fatty acids (SCFAs) like butyrate, propionate, and acetate as metabolic byproducts.6 These molecules are not merely waste; they are potent signaling molecules with systemic effects. Preclinical studies in animal models have shown that SCFAs can protect against SARS-CoV-2 infection through multiple pathways. They can downregulate the expression of the ACE2 receptor in both the intestines and the airways, effectively reducing the number of "doors" the virus can use to enter host cells. Additionally, SCFAs have been shown to enhance the host's adaptive immune response, promoting the production of more effective antibodies against the virus.6
A second, more direct mechanism involves the "grooming" of the host cell surface by bacterial enzymes. SARS-CoV-2 uses heparan sulfate (HS), a glycan found abundantly on the surface of human cells, as a co-receptor to facilitate its initial attachment before binding to ACE2.22 Seminal research has demonstrated that certain commensal gut bacteria, most notably Bacteroides species, produce enzymes called heparan lyases. These enzymes can cleave and degrade HS on the surface of adjacent human epithelial cells.22 By "grooming" the cell surface in this manner, these bacteria effectively remove one of the virus's key attachment points. In vitro experiments have confirmed that treating human lung cells with supernatants from Bacteroides cultures, or with purified bacterial heparan lyases, significantly reduces the binding of the SARS-CoV-2 spike protein and subsequent infection by the authentic virus.22
These mechanisms illustrate a sophisticated, albeit indirect, form of antiviral defense conferred by the gut microbiome. The virus's ability to cause dysbiosis, which often involves the depletion of these very Bacteroides and SCFA-producing bacteria, suggests a potential pathogenic strategy. By disrupting the gut ecosystem, SARS-CoV-2 may be dismantling the very microbial defenses that would otherwise limit its ability to infect the host, creating a self-reinforcing cycle of pathology that exacerbates the disease.
II. A Paradigm-Shifting Claim: Intrabacterial Localization of SARS-CoV-2
While the complex, indirect interactions between SARS-CoV-2 and the gut microbiome are well-established, a far more radical hypothesis has been proposed that directly addresses the user's query. This claim posits that SARS-CoV-2, a virus known to exclusively infect eukaryotic cells, can also enter and potentially replicate within prokaryotic cells—specifically, the bacteria of the human gut microbiome. This "bacteriophage-like" hypothesis represents a profound departure from canonical virology and, if substantiated, would necessitate a fundamental rewriting of our understanding of viral host range and pathogenesis.
A. Introduction to the "Bacteriophage-Like" Hypothesis
The core of the claim is that SARS-CoV-2 may exhibit behaviors analogous to those of a bacteriophage, a type of virus that specifically infects bacteria.24 This assertion challenges one of the most fundamental tenets of virology: the principle of host specificity. Viruses have evolved highly specialized molecular machinery to recognize and infect a limited range of host cells. The tools an animal virus like SARS-CoV-2 uses to enter a human cell are radically different from the tools a bacteriophage uses to breach the rigid cell wall of a bacterium.26 The proposition that a single virus could possess the ability to productively infect hosts across different domains of life—from Eukaryota to Bacteria—is extraordinary and requires an exceptionally high burden of proof.
The proponents of this hypothesis suggest that this novel interaction could explain several puzzling aspects of COVID-19, including the prolonged presence of viral RNA in feces and the significant alterations observed in the gut microbiota of infected individuals. They propose that bacteria could act as a previously unrecognized reservoir for the virus, fundamentally changing the models of COVID-19 transmission, persistence, and evolution.25
B. Overview of the Primary Research and Key Assertions
This paradigm-shifting claim originates from a series of publications, including a case report and several experimental papers, authored by a specific, interconnected research group led by investigators including Carlo Brogna, Mauro Petrillo, and Marina Piscopo.19 Their argument is not based on a single observation but on the convergence of findings from multiple, distinct experimental approaches. The key assertions put forth in their work can be summarized as follows:
Fecal Diagnostics in a Clinical Case: In a 2022 case report, the researchers described a family cluster where a 21-month-old child presented with gastrointestinal and other symptoms of COVID-19. While the child and all asymptomatic family members tested negative via rapid antigen nasopharyngeal swabs, highly sensitive Luminex-based RT-PCR tests detected SARS-CoV-2 RNA in the stool samples of the entire family. This finding was presented as evidence for the diagnostic utility of fecal testing and as the initial observation that prompted further investigation into the virus's behavior in the gut.19
In Vitro Viral Replication in Fecal Cultures: In subsequent experiments, fecal material from a SARS-CoV-2-positive patient was inoculated into a bacterial growth medium. Over a 30-day incubation period, the researchers observed a sustained increase in the concentration of viral RNA, which they interpreted as evidence of active viral replication occurring within the in vitro bacterial culture.28
Visualization of Intrabacterial Virus: To determine the physical location of the virus, the group performed transmission electron microscopy (TEM) on the cultured fecal bacteria. Their published images purport to show virus-like particles, approximately 25–100 nm in diameter, both on the surface of and, crucially, inside the cytoplasm of bacteria. The identity of these particles as SARS-CoV-2 was supported by immunogold labeling, where antibodies against the viral nucleocapsid protein, tagged with gold nanoparticles, appeared to co-localize with these intrabacterial structures.19
Demonstration of De Novo Viral Protein Synthesis: To address whether the increasing RNA was a result of true replication (which requires translation of viral proteins), the researchers conducted an isotope labeling experiment. They grew the fecal bacterial culture in a medium containing a heavy isotope of nitrogen ($^{15}$N). Using mass spectrometry, they reported the detection of newly synthesized viral spike protein that had incorporated the $^{15}$N label. They presented this as definitive proof of de novo synthesis of viral proteins by the machinery present in the bacterial culture.24
Bacteriophage-Like Activity in Plaque Assays: In what is perhaps their most striking claim, the group performed an experiment analogous to a classical bacteriophage plaque assay. They took the cell-free supernatant from their SARS-CoV-2-positive fecal cultures and applied it to lawns of pure-cultured gut bacteria. They reported the formation of clear, circular zones of clearing—resembling lytic plaques—on lawns of two specific species: Faecalibacterium prausnitzii and Dorea formicigenerans. This was interpreted as evidence that a lytic agent in the supernatant, presumed to be SARS-CoV-2, was infecting and killing these specific bacteria.31
The strength of the argument, as presented by its authors, lies in this multi-pronged approach. The hypothesis is constructed from a chain of reasoning that links clinical observation (fecal shedding) to in vitro findings (RNA increase), then to a proposed mechanism (protein synthesis and bacterial lysis), and finally to visual confirmation (electron microscopy). This convergence of disparate lines of evidence makes the claim more substantive than a single, isolated finding would be, and therefore it necessitates an equally rigorous and multifaceted critique.
III. Deconstruction of the Evidentiary Basis for Intrabacterial Viral Presence
While the "bacteriophage-like" hypothesis is supported by a portfolio of experiments, the extraordinary nature of the claim demands a meticulous and critical examination of its evidentiary foundation. Each piece of evidence, from imaging to molecular biology, must be scrutinized for its robustness, specificity, and susceptibility to alternative interpretations that align more closely with established biological principles. This section moves from presenting the claim to rigorously analyzing the evidence, highlighting strengths, weaknesses, and plausible alternative explanations.
A. Analysis of Imaging Data: A Critical Review of TEM and Immunofluorescence Evidence
The most direct evidence presented for the intrabacterial localization of SARS-CoV-2 comes from electron microscopy. The researchers published Transmission Electron Microscope (TEM) images showing what they identify as virus-like particles within the cytoplasm of bacteria from their fecal cultures. These findings were supported by immunogold labeling, where gold nanoparticles conjugated to an anti-SARS-CoV-2 nucleocapsid antibody appeared to co-localize with these intracellular structures, and by immunofluorescence microscopy showing viral protein signals overlapping with bacterial signals.29 While visually compelling, this evidence is subject to several critical challenges.
First, the issue of morphological specificity is paramount. The human gut is a complex environment teeming with a vast and diverse virome, including billions of bacteriophages per gram of content. Many of these native bacteriophages are morphologically similar in size (typically 20-200 nm) and shape to coronaviruses. Without definitive structural features that are unique to SARS-CoV-2 and visible at that resolution, identifying a particle as SARS-CoV-2 based on morphology alone is fraught with uncertainty. The observed particles could very well be endogenous bacteriophages that naturally infect the gut bacteria under study.
Second, the reliability of immunolabeling techniques hinges entirely on the specificity of the antibody and the prevention of artifacts. Non-specific binding of antibodies to cellular components is a common challenge in microscopy. In the context of a complex sample like a fecal culture, which contains a myriad of proteins and cellular debris, the potential for the anti-nucleocapsid antibody to cross-react with bacterial proteins or to bind non-specifically to structures within damaged or dying cells is a significant concern. The evidence provided does not include the rigorous controls—such as using isotype control antibodies or testing on known negative bacteria—that would be necessary to rule out such artifacts.
Finally, while co-localization demonstrates proximity, it does not definitively prove causation or an intracellular location. An immunofluorescence signal showing an overlap between a bacterial marker and a viral marker proves only that the two are in the same focal plane. Similarly, in TEM, a virion that is strongly adhered to the outside of a bacterial cell could, depending on the angle of the ultrathin section, appear to be inside the cell. The images do not unequivocally distinguish between true intracellular localization and strong surface adhesion or engulfment within a bacterial biofilm matrix.
B. Evaluation of Molecular and Microbiological Data
The molecular and microbiological experiments form the functional backbone of the hypothesis, suggesting not just presence but active replication. However, these too warrant critical evaluation. The following table summarizes the key experimental evidence and the authors' interpretations, which will be analyzed in detail below.
The observation of increasing viral RNA load in the fecal culture is a cornerstone of the replication claim.28 While suggestive, this finding does not prove that replication occurred within bacteria. The initial fecal inoculum is a complex slurry containing not only bacteria but also sloughed human epithelial cells, immune cells, and potentially other microbes like phagocytic protozoa. The nutrient broth used for bacterial culture could plausibly sustain some of these contaminating eukaryotic cells or their replication-competent debris long enough for SARS-CoV-2 to replicate within them. The viral replication could be occurring in this small, undetected eukaryotic fraction, with the resulting virions being measured in the total culture. Without stringent controls to ensure the absolute purity of the bacterial culture from any eukaryotic components—such as pre-filtering the inoculum to remove larger cells and testing for human-specific genetic markers throughout the experiment—this alternative explanation cannot be dismissed.
The nitrogen isotope labeling experiment is arguably the most compelling piece of evidence for de novo synthesis within the experimental system.24 It convincingly shows that new viral proteins are being made. However, it is subject to the exact same critique as the RNA detection: it demonstrates that synthesis is happening in the culture, but it does not prove it is happening by the bacterial translational machinery. If contaminating human cells are the site of replication, they would also incorporate the $^{15}$N (Nitrogen-15) from the medium into the new viral proteins they produce. The experiment's conclusion rests on the unproven assumption that the culture is purely bacterial.
Finally, the "plaque assay" experiment is highly evocative but methodologically ambiguous.31 A true virological plaque assay demonstrates that a single, purified viral particle can initiate a spreading infection that results in a zone of lysis on a uniform lawn of host cells. The experiment described used a crude, unpurified supernatant from a complex fecal culture. This supernatant could contain a multitude of agents capable of inhibiting bacterial growth or causing lysis, including native bacteriophages specific to F. prausnitzii or Dorea, bacterial toxins, or metabolic byproducts. To attribute the observed clearing zones solely to SARS-CoV-2 without first isolating and purifying the causative agent from those zones and demonstrating that the purified agent can form new plaques is a significant interpretive leap. The burden of proof would require a rigorous plaque purification protocol, which was not reported.
C. Methodological Considerations and Alternative Interpretations
Synthesizing these critiques, a central alternative hypothesis emerges: the observations reported by the research group may result from the close and persistent association of SARS-CoV-2 with bacteria and other components of the gut milieu, rather than a true, productive intracellular infection of bacteria. This overarching interpretation can encompass the various findings without invoking a biologically unprecedented cross-domain infection.
Several specific scenarios could explain the data:
Strong Surface Adhesion: SARS-CoV-2 virions, released from infected human gut cells, could bind tenaciously to the surfaces of nearby bacteria. Bacteria are known to form biofilms, and their surfaces are decorated with exopolysaccharides and proteins that could serve as non-specific attachment sites for viruses. Virions trapped in this biofilm matrix or adhered to individual cells would be carried along with the bacteria during sample preparation, leading to co-localization in microscopy and the presence of viral RNA in bacterial pellets.
Contamination with Eukaryotic Cellular Debris: The "bacterial cultures" initiated with raw fecal samples are almost certain to contain replication-competent fragments of infected human intestinal cells. These fragments, or even intact cells, could be the true source of the observed increase in viral RNA and the de novo synthesis of viral proteins. The bacterial growth may simply provide a supportive environment for these eukaryotic remnants to continue producing virus for a period.
Phagocytosis by Gut Protozoa: The human gut harbors various species of phagocytic protozoa (e.g., amoebas) that feed on bacteria. It is conceivable that these eukaryotic microbes could be susceptible to SARS-CoV-2 infection. If an infected protozoan also ingests bacteria, subsequent analysis could find both viral components and intact bacteria within the same organism, creating a "Trojan horse" scenario that mimics intrabacterial localization.
The fundamental weakness across the entire body of evidence for the "bacteriophage-like" hypothesis is the lack of rigorous controls to definitively exclude these more plausible alternative explanations. The claim is extraordinary, and as such, it requires extraordinary evidence that systematically rules out every conventional biological possibility. As it stands, the evidence is intriguing and warrants further investigation, but it does not yet meet this high burden of proof.
IV. Assessing Biological Plausibility: A Virological and Microbiological Perspective
Beyond the critique of the specific experimental evidence, the "bacteriophage-like" hypothesis must be evaluated against the fundamental principles of virology and microbiology. A key question is whether such a cross-domain infection is mechanistically plausible. This requires a detailed comparison of the highly specialized entry pathway of SARS-CoV-2 with the formidable and fundamentally different defensive barrier of a bacterial cell.
A. The Canonical Eukaryotic Entry Pathway of SARS-CoV-2
The process by which SARS-CoV-2 enters human cells is a well-characterized, multi-step molecular ballet, exquisitely evolved for a eukaryotic host.32 The entire process is mediated by the viral Spike (S) protein.
Attachment: The first step is the binding of the S protein's receptor-binding domain (RBD) to its primary receptor on the host cell surface, angiotensin-converting enzyme 2 (ACE2). This interaction is often stabilized by an initial, lower-affinity binding to co-receptors like heparan sulfate (HS).16
Proteolytic Priming: For the virus to become fusion-competent, the S protein must be cleaved by specific host proteases. This cleavage occurs at what is known as the S2' site. If the host cell expresses the protease TMPRSS2 on its surface, this cleavage happens at the plasma membrane. If TMPRSS2 is absent, the virus is taken up into an endosome, where acidic conditions activate other proteases, such as cathepsins, to perform the cleavage.32
Membrane Fusion: Once cleaved, the S protein undergoes a dramatic conformational change, exposing a fusion peptide that inserts into the host cell membrane. This action pulls the viral envelope and the host membrane together, causing them to fuse and creating a pore through which the viral RNA genome is released into the host cell's cytoplasm.32
This entire pathway is a cascade of highly specific protein-protein and protein-glycan interactions, dependent on the precise molecular landscape of a mammalian cell surface and the availability of specific host enzymes.
B. The Prokaryotic Barrier: The Structure and Composition of the Bacterial Cell Envelope
The surface of a bacterium is fundamentally different from that of a human cell, presenting a formidable barrier to a virus not specifically evolved to breach it. The bacterial cell envelope's primary structural component is the cell wall, which is composed of peptidoglycan—a rigid, mesh-like polymer of sugars and amino acids that provides structural integrity and protection.38
Gram-positive bacteria, the group to which the proposed hosts Faecalibacterium and Dorea belong, have a particularly thick layer of peptidoglycan external to their single plasma membrane. Gram-negative bacteria have a thinner peptidoglycan layer sandwiched between an inner plasma membrane and a protective outer membrane composed of lipopolysaccharides (LPS).
Crucially, bacteria lack the key components required for the canonical SARS-CoV-2 entry pathway. Their genomes do not encode for ACE2, TMPRSS2, or cathepsins.32 Their surfaces are decorated with molecules like teichoic acids (in Gram-positives) or LPS (in Gram-negatives), not the specific protein and glycan receptors used by SARS-CoV-2. Therefore, the virus's highly specialized "key" (the Spike protein) has no corresponding "lock" on the bacterial surface.
C. A Tale of Two Hosts: Contrasting Animal Viruses and Bacteriophages
The immense biological gulf between infecting a human cell and a bacterial cell is best illustrated by a direct comparison of their respective viruses. The following table starkly contrasts the key features of SARS-CoV-2 with those of a typical bacteriophage.
This comparison highlights the "mechanistic chasm" that the "bacteriophage-like" hypothesis must overcome. SARS-CoV-2 relies on hijacking the fluid dynamics of a eukaryotic membrane and exploiting specific host enzymes.37 In contrast, a bacteriophage uses a mechanical approach, often employing a syringe-like tail structure to physically puncture the rigid bacterial cell wall and inject its genetic material directly inside.27 For SARS-CoV-2 to infect a bacterium, it would need to abandon its evolved strategy and spontaneously develop a new one for which it possesses no known genetic blueprint. This is not merely unlikely; it runs contrary to our fundamental understanding of viral evolution and host adaptation.
D. Examining the Proposed Bacterial Hosts: Faecalibacterium prausnitzii and Dorea formicigenerans
The specific bacteria identified in the plaque assay experiments, F. prausnitzii and D. formicigenerans, are well-studied members of the healthy human gut microbiome.31 Both are Gram-positive, strictly anaerobic bacteria belonging to the phylum Firmicutes (Clostridium cluster IV and the family Lachnospiraceae, respectively).41 They are renowned for their beneficial, anti-inflammatory properties, largely due to their prolific production of the SCFA butyrate.45
A review of their known biology reveals nothing to suggest a unique susceptibility to a eukaryotic virus. Their genomes have been sequenced, and they contain no genes with significant homology to ACE2 or other known SARS-CoV-2 entry factors.43 In fact, genomic analyses have shown that some strains of these bacteria harbor prophages—the remnants of integrated bacteriophages.45 This indicates that they are conventional hosts for prokaryotic viruses, not eukaryotic ones.
This leads to a significant paradox. The very bacteria proposed as replication factories for SARS-CoV-2 are the same species consistently reported to be depleted in the gut of COVID-19 patients.4 If these bacteria were indeed a major reservoir for viral replication, one might expect a more complex population dynamic, perhaps with the virus driving the selection of resistant strains. The observed simple depletion of these beneficial species is far more parsimoniously explained by the indirect effects of the host's systemic inflammatory response and the resulting gut dysbiosis, which creates an environment less hospitable to these strictly anaerobic, beneficial commensals. The "infection" hypothesis does not neatly align with the observed population-level data from numerous independent studies.
V. The Specter of Horizontal Gene Transfer and Chimeric RNA
While a direct, productive infection of bacteria by SARS-CoV-2 faces insurmountable biological plausibility hurdles, this does not mean that viral and bacterial genetic material cannot interact in the gut. The gut microbiome is a dynamic environment characterized by frequent genetic exchange. Exploring these more plausible, albeit still complex, phenomena may offer alternative explanations for the molecular findings that underpin the "bacteriophage-like" hypothesis.
A. Horizontal Gene Transfer (HGT) in the Gut Microbiome
The mammalian gut is a recognized hotspot for Horizontal Gene Transfer (HGT), the process by which organisms exchange genetic material outside of traditional parent-to-offspring inheritance.49 This is a primary driver of microbial evolution, allowing bacteria to rapidly acquire new traits. The main mechanisms of HGT are:
Conjugation: The transfer of plasmids (small, circular DNA molecules) through direct cell-to-cell contact.
Transformation: The uptake of "naked" DNA from the environment, often released from lysed cells.
Transduction: The transfer of genetic material from one bacterium to another via a bacteriophage.51
These processes are rampant in the dense, competitive environment of the gut, facilitating the spread of genes for antibiotic resistance, metabolism of novel substrates, and virulence.49 This establishes a clear precedent for genetic material—including viral DNA from bacteriophages—moving between different microbial cells within the gut ecosystem.
B. The Potential for Viral-Bacterial Recombination Events
Given the high concentration of both SARS-CoV-2 RNA (from lysed human cells) and bacteria in the gut of an infected individual, the potential for interaction exists. While a full infection cycle is implausible, other forms of genetic interaction could occur.
One intriguing study has provided bioinformatic evidence for such an interaction. The authors suggest that the SARS-CoV-2 RNA-dependent RNA polymerase (RdRp), the enzyme the virus uses to replicate its own genome, might be capable of binding to and replicating bacterial messenger RNAs (mRNAs) that it encounters. This template-switching event would result in the production of viral-bacterial "chimeric RNAs"—single RNA molecules containing fused sequences from both the virus and a bacterium.53 This would create a physical linkage between viral and bacterial genetic information without requiring the virus to actually enter and take over the bacterium.
Furthermore, research on SARS-CoV-2 in human cells has shown that viral RNA can be reverse-transcribed into DNA and integrated into the host human genome. This process is thought to be mediated by the machinery of endogenous retrotransposons like LINE1.54 While bacteria lack LINE1 elements and this specific mechanism is therefore not applicable, it demonstrates the general capacity of the virus's genetic information to be converted to DNA and become integrated into host DNA under certain conditions. It is theoretically possible, though entirely unproven, that fragments of viral RNA could be taken up by bacteria and integrated into plasmids or the bacterial chromosome via the bacteria's own recombination machinery.
C. Distinguishing True Infection from Genetic Artifacts
These alternative genetic mechanisms provide a more biologically conservative framework for explaining the molecular findings of the Brogna et al. group. If bacteria in the gut can take up free-floating viral RNA from the environment (released from the millions of lysed human enterocytes), or if chimeric viral-bacterial RNAs are formed, then highly sensitive molecular tests like RT-PCR performed on a bacterial pellet would inevitably yield a positive result.
This scenario would create the appearance of an infection. One would find viral genetic sequences physically associated with bacteria, but this association would be non-productive and non-infectious. It would be a genetic artifact of proximity and the known "stickiness" of nucleic acids, rather than evidence of a true biological takeover.
According to the principle of Occam's Razor, which posits that the simplest explanation is most likely the correct one, these alternative hypotheses are more parsimonious. They build upon established principles of microbial genetics (HGT, promiscuity of polymerases) rather than requiring the complete overthrow of fundamental tenets of virology (cross-domain infection). Therefore, until the "bacteriophage-like" hypothesis can be supported by evidence that rigorously excludes these more plausible genetic interactions, it is more likely that the molecular data reflects a non-infectious association rather than a true replicative cycle.
VI. Scientific Consensus and the Path Forward: The Need for Independent Corroboration
In science, the journey of a novel finding from a preliminary observation to an accepted fact is a rigorous process, with independent replication by the broader scientific community serving as the most critical gatekeeper. This is especially true for extraordinary claims that challenge established paradigms. An assessment of the "bacteriophage-like" hypothesis is therefore incomplete without evaluating its reception and status within the global research community.
A. Current Status of the Hypothesis in Peer-Reviewed Literature
A comprehensive analysis of the available scientific literature reveals that the claim of SARS-CoV-2 infecting gut bacteria is, at present, an isolated one. The hypothesis originates from and is exclusively promoted by a series of papers from a single, interconnected group of researchers.19
Citation analysis of these key papers indicates that they have not been widely integrated into the work of other laboratories. The number of independent citations is extremely low, suggesting that other researchers in the field are not currently building upon these findings.57 More telling is the conspicuous absence of this claim in the vast body of other research on COVID-19 and the gut microbiome. Dozens of review articles and original research papers from independent groups worldwide have explored the gut-lung axis, dysbiosis, and viral persistence in the gut, yet none of them mention, discuss, or report any evidence of SARS-CoV-2 acting as a bacteriophage.4 Furthermore, major clinical and public health guidelines from organizations like the WHO and IDSA, which synthesize the totality of evidence on viral transmission and pathology, make no mention of a bacterial reservoir or a fecal-bacterial transmission route.60
This silence from the wider community is not merely a lack of data; in the context of a global pandemic that has generated an unprecedented firehose of research, it is a powerful data point in itself. The scientific community has intensely scrutinized the gut's role in COVID-19. A revolutionary finding with such profound implications for pathogenesis, diagnostics, and transmission would normally trigger a flurry of activity from other labs seeking to replicate, validate, and extend the work. The fact that the claim remains confined to its originators strongly suggests that it has either not been successfully replicated or is not considered a sufficiently credible or well-supported hypothesis by the broader community to warrant the significant investment of time and resources required for replication.
B. The Critical Role of Independent Replication in Validating Extraordinary Claims
The scientific method is a self-correcting process. A foundational pillar of this process is that for a finding to be considered valid, it must be reproducible. Another researcher, following the same methods, should be able to achieve the same results. This process of independent verification is what separates a one-off anomaly or a potential artifact from a robust scientific discovery.
The adage, often attributed to Carl Sagan, that "extraordinary claims require extraordinary evidence" is central here. The claim that a human respiratory virus can infect bacteria is undeniably extraordinary. The corollary to this rule is that this extraordinary evidence must itself be independently verifiable. Until the findings from the Brogna et al. group are replicated by other, unaffiliated research teams, the hypothesis remains precisely that: a hypothesis, not an established fact.
C. Defining the Research Required for Verification or Falsification
To move the "bacteriophage-like" hypothesis from a provocative outlier to either a proven phenomenon or a disproven artifact, a clear and rigorous experimental path is needed. Any attempt at replication would need to address the key methodological weaknesses of the original studies head-on. The required experiments would include:
Rigorous Purity Controls: The in-vitro culture experiments must be repeated with unimpeachable controls to ensure the absolute absence of any contaminating eukaryotic cells or replication-competent cellular debris. This would involve techniques like passing the fecal inoculum through filters with a pore size small enough to block eukaryotic cells (e.g., 0.45 µm) while allowing bacteria to pass, followed by continuous monitoring of the culture for human-specific DNA and RNA markers (e.g., via PCR for human housekeeping genes) to confirm purity.
Purification of the Causative Agent: The plaque assay experiment must be advanced beyond using a crude supernatant. If true plaques are formed, the putative lytic agent must be isolated from a single, well-defined plaque. This agent would then need to be purified using standard virological techniques, such as cesium chloride density gradient centrifugation. The purified agent, confirmed to be SARS-CoV-2 via sequencing and microscopy, must then be shown to form new plaques on a fresh bacterial lawn. This would definitively link the lytic activity to the virus itself, rather than to some other component of the original supernatant.
Detailed Genetic and Functional Analysis: If a purified, bacteria-replicating SARS-CoV-2 could be isolated, it would be a treasure trove for scientific discovery. Its genome would need to be sequenced and compared to the parental virus to identify any mutations that might enable this cross-domain jump. Furthermore, analyzing its proteins could reveal whether it is using the bacterial translational machinery (e.g., prokaryotic ribosomes), which might leave specific biochemical signatures.
Only through such a rigorous and transparent process of independent research can the scientific community confidently accept or reject this paradigm-challenging claim.
VII. Implications and Future Research Directions
While the "bacteriophage-like" hypothesis remains unsubstantiated, it is a valuable scientific exercise to consider its implications if it were to be proven true. The potential consequences would be far-reaching, transforming our understanding of viral pathogenesis, public health, and even fundamental biology. Contemplating these possibilities underscores why the question is so important, regardless of the current answer.
A. Potential Consequences for Viral Pathogenesis and Persistence
If gut bacteria could indeed serve as a host for SARS-CoV-2, it would introduce a completely new dimension to COVID-19 pathogenesis.
A Vast Bacterial Reservoir: The human gut contains trillions of bacteria. If even a small fraction of these could be infected, they would constitute a vast and durable reservoir for the virus. This could readily explain the phenomenon of prolonged fecal shedding, as the virus would have a continuous source of replication independent of the host's immune clearance of infected human cells. This reservoir could also potentially serve as a source for disease relapse, where the virus re-emerges from the gut to cause a new systemic infection without any external re-exposure.25
A Factory for Novel Variants: Evolution is driven by replication in new environments. A virus replicating within a prokaryotic host would be subject to entirely different evolutionary pressures than in a human host. It would be using different replication machinery (bacterial polymerases and ribosomes) and interacting with different cellular components. This could serve as a powerful engine for generating novel mutations and, potentially, entirely new viral variants with unpredictable characteristics, such as altered transmissibility, virulence, or antigenicity.31 This has even been speculatively linked to the origin of highly mutated variants like Omicron.
B. Implications for Diagnostics, Therapeutics, and Public Health Surveillance
The confirmation of a bacterial reservoir would have immediate and profound practical implications.
Diagnostics: It would strongly validate the use of fecal testing as a primary and perhaps more reliable long-term diagnostic tool than respiratory swabs, particularly for monitoring persistent infection or in cases with atypical presentations.19
Therapeutics: It would open the door to entirely new therapeutic strategies. For instance, if specific bacterial species are the hosts, one could envision using targeted antibiotics to eliminate that part of the viral reservoir, a possibility hinted at in the original studies.28 Conversely, probiotic therapies could be developed using beneficial bacteria that could outcompete the susceptible host bacteria for resources and space in the gut niche.
Public Health Surveillance: Our understanding of transmission would need to be re-evaluated. A long-term bacterial reservoir would increase the potential duration of an individual's infectiousness, potentially impacting quarantine guidelines. It would also complicate the interpretation of wastewater surveillance data, as the presence of viral RNA in sewage could reflect shedding from this bacterial reservoir long after acute community transmission has subsided.31
C. Broader Questions on Virus-Microbe Interactions Across Biological Domains
Perhaps the most profound implication would be for the field of biology itself. The confirmation that a complex animal virus can productively infect a bacterium would shatter the long-held paradigm of a rigid barrier between the viromes of different biological domains. It would suggest that the rules of host specificity are more fluid than previously imagined and would launch an entirely new field of "cross-domain virology." Researchers would be compelled to ask whether this phenomenon is unique to SARS-CoV-2 or if other human viruses might also have unappreciated interactions with our vast microbiome. It would force a fundamental re-evaluation of the intricate and still mysterious relationships between viruses, microbes, and their multicellular hosts.25
VIII. Concluding Analysis: An Unverified but Provocative Hypothesis
In synthesizing the totality of the available evidence, a definitive and nuanced answer to the query—"Have there been any cases of SARS-CoV-2 being found inside bacteria of the human gut microbiome?"—emerges. The answer is a qualified "yes, but." Yes, a single research group has published a series of reports claiming to have observed this phenomenon. However, this affirmative is immediately followed by a cascade of critical caveats that place the claim firmly in the category of an unverified and highly controversial hypothesis.
The evidence presented in support of this "bacteriophage-like" behavior, while multifaceted and intriguing, does not withstand rigorous scrutiny. The imaging data is ambiguous, the molecular and microbiological findings are vulnerable to a central, unaddressed methodological flaw—the potential for contamination with replication-competent eukaryotic material—and the conclusions are not supported by the broader, population-level data on gut dysbiosis in COVID-19.
More fundamentally, the hypothesis faces a formidable wall of biological implausibility. The known entry and replication strategy of SARS-CoV-2 is exquisitely adapted for a eukaryotic host and is mechanistically incompatible with the fundamentally different structure and molecular makeup of a prokaryotic cell. The virus simply lacks the genetic toolkit required to recognize, breach, and hijack a bacterium. Alternative, more parsimonious explanations, such as non-infectious surface adhesion or the formation of chimeric viral-bacterial RNA, can account for the molecular findings without requiring the dismissal of core virological principles.
The final and most decisive factor is the stark lack of independent corroboration. In a global research environment that has exhaustively studied every facet of COVID-19, the silence from the wider scientific community on this revolutionary claim is deafening. Without independent replication, the claim cannot transition from a provocative hypothesis to an accepted scientific fact.
Therefore, this report concludes that there is currently no credible, independently verified evidence to support the assertion that SARS-CoV-2 can productively infect bacteria of the human gut microbiome. The claim remains an outlier, a scientifically valuable one for its role in pushing the boundaries of our thinking, but an outlier nonetheless. It serves as a powerful reminder of the immense complexity of the gut ecosystem and of the many unanswered questions that persist regarding the long-term, intricate interactions between SARS-CoV-2 and its human host. It is a testament to the fact that even within one of the most intensely studied diseases in human history, there are still frontiers of knowledge to be explored and fundamental assumptions to be challenged.
Acknowledgement
I acknowledge the assistance of Gemini AI in the preparation of the subject research plan, the execution of the research, and the preparation of this report.
Works cited
SARS-CoV-2 and immune-microbiome interactions: Lessons from respiratory viral infections, accessed July 30, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC7891052/
Impact of SARS-CoV2 infection on gut microbiota dysbiosis - OAE Publishing Inc., accessed July 30, 2025, https://www.oaepublish.com/articles/mrr.2023.48
Regulation of SARS-CoV-2 infection by diet-modulated gut microbiota - Frontiers, accessed July 30, 2025, https://www.frontiersin.org/journals/cellular-and-infection-microbiology/articles/10.3389/fcimb.2023.1167827/full
The relationship between gut microbiota and COVID-19 progression: new insights into immunopathogenesis and treatment - Frontiers, accessed July 30, 2025, https://www.frontiersin.org/journals/immunology/articles/10.3389/fimmu.2023.1180336/full
Genetic support of the causal association between gut microbiome and COVID-19: a bidirectional Mendelian randomization study - Frontiers, accessed July 30, 2025, https://www.frontiersin.org/journals/immunology/articles/10.3389/fimmu.2023.1217615/full
Gut microbiota-derived metabolites confer protection against SARS-CoV-2 infection, accessed July 30, 2025, https://pubmed.ncbi.nlm.nih.gov/35915556/
Alteration of the gut microbiota following SARS-CoV-2 infection correlates with disease severity in hamsters - PubMed, accessed July 30, 2025, https://pubmed.ncbi.nlm.nih.gov/34965194/
Microbiome dysbiosis in SARS-CoV-2 infection: implication for pathophysiology and management strategies of COVID-19 - Frontiers, accessed July 30, 2025, https://www.frontiersin.org/journals/cellular-and-infection-microbiology/articles/10.3389/fcimb.2025.1537456/full
The Effect of COVID-19 on Gut Microbiota: Exploring the Complex Interplay and Implications for Human Health - MDPI, accessed July 30, 2025, https://www.mdpi.com/2624-5647/5/3/28
Gut Microbiome Disruption Following SARS-CoV-2: A Review - PubMed, accessed July 30, 2025, https://pubmed.ncbi.nlm.nih.gov/38257958/
Alterations of the gut microbiota in coronavirus disease 2019 and its therapeutic potential, accessed July 30, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9813939/
COVID-19 and the Human Gut Microbiome: An Under-Recognized Association - PMC, accessed July 30, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9535107/
Gut Microbiome Disruption Following SARS-CoV-2: A Review - MDPI, accessed July 30, 2025, https://www.mdpi.com/2076-2607/12/1/131
SARS-CoV-2 infection is associated with intestinal permeability, systemic inflammation, and microbial dysbiosis in hospitalized COVID-19 patients | bioRxiv, accessed July 30, 2025, https://www.biorxiv.org/content/10.1101/2023.12.07.570670v1
Gut Microbiome Signatures During Acute Infection Predict Long COVID - bioRxiv, accessed July 30, 2025, https://www.biorxiv.org/content/10.1101/2024.12.10.626852v1
Mechanisms of infection by SARS-CoV-2, inflammation and potential links with the microbiome - PubMed Central, accessed July 30, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC7876557/
SARS-CoV-2 infection perturbs the gastrointestinal tract and induces modest microbial translocation across the intestinal barrier - PubMed, accessed July 30, 2025, https://pubmed.ncbi.nlm.nih.gov/39264207/
Depicting SARS-CoV-2 faecal viral activity in association with gut microbiota composition in patients with COVID-19, accessed July 30, 2025, https://gut.bmj.com/content/70/2/276
The first report on detecting SARS-CoV-2 inside bacteria of the human gut microbiome: A case series on asymptomatic family members and a child with COVID-19 - PubMed Central, accessed July 30, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11502994/
Gut microbiome and clinical and lifestyle host factors associated with recurrent positive RT-PCR for SARS-CoV-2 - Frontiers, accessed July 30, 2025, https://www.frontiersin.org/journals/cellular-and-infection-microbiology/articles/10.3389/fcimb.2024.1494193/full
Preclinical Study Suggests Gut Bacteria May Influence Viral Entry, Immune Response and Coagulation in COVID-19 - Pediatrics | Weill Cornell Medicine, accessed July 30, 2025, https://pediatrics.weill.cornell.edu/news/preclinical-study-suggests-gut-bacteria-may-influence-viral-entry-immune-response-and
SARS-CoV-2 infectivity can be modulated through bacterial ..., accessed July 30, 2025, https://journals.asm.org/doi/10.1128/mbio.04015-24
SARS-CoV-2 infectivity can be modulated through bacterial grooming of the glycocalyx, accessed July 30, 2025, https://www.researchgate.net/publication/389323424_SARS-CoV-2_infectivity_can_be_modulated_through_bacterial_grooming_of_the_glycocalyx
Could SARS-CoV-2 Have Bacteriophage Behavior or Induce the ..., accessed July 30, 2025, https://www.mdpi.com/2076-393X/10/5/708
Evidence of SARS-CoV-2 bacteriophage potential in human gut microbiota - JRC Publications Repository, accessed July 30, 2025, https://publications.jrc.ec.europa.eu/repository/handle/JRC125662
Interactions between Bacteriophages and Eukaryotic Cells - PMC, accessed July 30, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC7301238/
What Are Viruses and How Do They Infect Us? | bioMérieux Living Diagnostics Blog, accessed July 30, 2025, https://www.biomerieux.com/corp/en/blog/infectious-diseases/what-are-viruses-and-how-do-they-infect-us--.html
Increase of SARS-CoV-2 RNA load in faecal samples prompts for rethinking of SARS-CoV-2 biology and COVID-19 epidemiology, accessed July 30, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8283343/
The first report on detecting SARS-CoV-2 inside bacteria of the ..., accessed July 30, 2025, https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11502994/
Evidence of SARS-CoV-2 bacteriophage potential in human gut microbiota - PMC, accessed July 30, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC12120431/
Evidence of SARS-CoV-2 bacteriophage potential in human gut microbiota., accessed July 30, 2025, https://f1000research.com/articles/11-292
Structural understanding of SARS-CoV-2 virus entry to host cells - Frontiers, accessed July 30, 2025, https://www.frontiersin.org/journals/molecular-biosciences/articles/10.3389/fmolb.2023.1288686/full
SARS-CoV-2 Spike-Mediated Entry and Its Regulation by Host Innate Immunity - PMC, accessed July 30, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10059852/
COVID-19 - Wikipedia, accessed July 30, 2025, https://en.wikipedia.org/wiki/COVID-19
TMPRSS2 expression dictates the entry route used by SARS‐CoV‐2 to infect host cells, accessed July 30, 2025, https://www.embopress.org/doi/10.15252/embj.2021107821
Glycosylated extracellular mucin domains protect against SARS-CoV-2 infection at the respiratory surface | PLOS Pathogens - Research journals, accessed July 30, 2025, https://journals.plos.org/plospathogens/article?id=10.1371/journal.ppat.1011571
Viral entry - Wikipedia, accessed July 30, 2025, https://en.wikipedia.org/wiki/Viral_entry
Methicillin-Resistant Staphylococcus aureus (MRSA): Resistance, Prevalence, and Coping Strategies - MDPI, accessed July 30, 2025, https://www.mdpi.com/2079-6382/14/8/771
The SARS-CoV-2 Spike protein has a broad tropism for mammalian ACE2 proteins, accessed July 30, 2025, https://journals.plos.org/plosbiology/article?id=10.1371/journal.pbio.3001016
Replication of the coronavirus genome: A paradox among positive-strand RNA viruses - PMC - PubMed Central, accessed July 30, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8994683/
Faecalibacterium - Wikipedia, accessed July 30, 2025, https://en.wikipedia.org/wiki/Faecalibacterium
Faecalibacterium: a bacterial genus with promising human health applications - PMC, accessed July 30, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10410495/
lepA - Elongation factor 4 - Dorea formicigenerans ATCC 27755 | UniProtKB | UniProt, accessed July 30, 2025, https://www.uniprot.org/uniprot/B0G870_9FIRM
The Controversial Role of Human Gut Lachnospiraceae - MDPI, accessed July 30, 2025, https://www.mdpi.com/2076-2607/8/4/573
Full article: Faecalibacterium prausnitzii regulates carbohydrate metabolic functions of the gut microbiome in C57BL/6 mice, accessed July 30, 2025, https://www.tandfonline.com/doi/full/10.1080/19490976.2025.2455503
Butyrate-producing Faecalibacterium prausnitzii suppresses natural killer/T-cell lymphoma by dampening the JAK-STAT pathway | Gut, accessed July 30, 2025, https://gut.bmj.com/content/74/4/557
Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients | PNAS, accessed July 30, 2025, https://www.pnas.org/doi/10.1073/pnas.0804812105
Phage tail tape measure protein domain-containing protein - Dorea formicigenerans | UniProtKB | UniProt, accessed July 30, 2025, https://www.uniprot.org/uniprotkb/A0A3E5GPN2
Gut inflammation can boost horizontal gene transfer between pathogenic and commensal Enterobacteriaceae | PNAS, accessed July 30, 2025, https://www.pnas.org/doi/10.1073/pnas.1113246109
Horizontal gene transfer overrides mutation in Escherichia coli colonizing the mammalian gut | PNAS, accessed July 30, 2025, https://www.pnas.org/doi/10.1073/pnas.1906958116
Enhancing insights into diseases through horizontal gene transfer event detection from gut microbiome - Oxford Academic, accessed July 30, 2025, https://academic.oup.com/nar/article/52/14/e61/7694277
Genomic insights into bacteriophages: a new frontier in AMR detection and phage therapy - Oxford Academic, accessed July 30, 2025, https://academic.oup.com/bfg/article/doi/10.1093/bfgp/elaf011/8215392?rss=1
The human microbiota is a beneficial reservoir for SARS-CoV-2 mutations | mBio, accessed July 30, 2025, https://journals.asm.org/doi/10.1128/mbio.03187-23
pmc.ncbi.nlm.nih.gov, accessed July 30, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8166107/#:~:text=We%20show%20here%20that%20SARS,detected%20in%20patient%2Dderived%20tissues.
Reverse-transcribed SARS-CoV-2 RNA can integrate into the genome of cultured human cells and can be expressed in patient-derived tissues - PubMed, accessed July 30, 2025, https://pubmed.ncbi.nlm.nih.gov/33958444/
SARS-CoV-2 Can Integrate Into the Genome, Cause Positive Tests | Microbiology - Labroots, accessed July 30, 2025, https://www.labroots.com/trending/microbiology/24783/sars-cov-2-integrate-genome-cause-positive-tests
The first report on detecting SARS-CoV-2 inside bacteria of the human gut microbiome: A case series on asymptomatic family members and a child with COVID-19 - Consensus, accessed July 30, 2025, https://consensus.app/papers/the-first-report-on-detecting-sarscov2-inside-bacteria-of-petrillo-piscopo/1aa403b67f1a536e99f45d358a1fed63
The first report on detecting SARS-CoV-2 inside bacteria of the human gut microbiome: A case series on asymptomatic family members and a child with COVID-19 - ResearchGate, accessed July 30, 2025, https://www.researchgate.net/publication/384985758_The_first_report_on_detecting_SARS-CoV-2_inside_bacteria_of_the_human_gut_microbiome_A_case_series_on_asymptomatic_family_members_and_a_child_with_COVID-19
Mild SARS-CoV-2 infection results in long-lasting microbiota instability - PMC, accessed July 30, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10470529/
IDSA Guidelines on the Treatment and Management of Patients with COVID-19, accessed July 30, 2025, https://www.idsociety.org/practice-guideline/covid-19-guideline-treatment-and-management/
Modes of transmission of virus causing COVID-19: implications for IPC precaution recommendations - World Health Organization (WHO), accessed July 30, 2025, https://www.who.int/news-room/commentaries/detail/modes-of-transmission-of-virus-causing-covid-19-implications-for-ipc-precaution-recommendations