Next Article in Journal
Interactions between the Astrocytic Volume-Regulated Anion Channel and Aquaporin 4 in Hyposmotic Regulation of Vasopressin Neuronal Activity in the Supraoptic Nucleus
Next Article in Special Issue
New Phage-Derived Antibacterial Enzyme PolaR Targeting Rothia spp.
Previous Article in Journal
Efficacy and Safety of Biological Therapies and JAK Inhibitors in Older Patients with Inflammatory Bowel Disease
Previous Article in Special Issue
Accessing the In Vivo Efficiency of Clinically Isolated Phages against Uropathogenic and Invasive Biofilm-Forming Escherichia coli Strains for Phage Therapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Phage Interactions with the Nervous System in Health and Disease

by
Adam Jędrusiak
1,
Wojciech Fortuna
2,3,
Joanna Majewska
1,
Andrzej Górski
1,3,4 and
Ewa Jończyk-Matysiak
1,*
1
Bacteriophage Laboratory, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland
2
Department of Neurosurgery, Wroclaw Medical University, Borowska 213, 54-427 Wroclaw, Poland
3
Phage Therapy Unit, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland
4
Infant Jesus Hospital, The Medical University of Warsaw, 02-006 Warsaw, Poland
*
Author to whom correspondence should be addressed.
Cells 2023, 12(13), 1720; https://doi.org/10.3390/cells12131720
Submission received: 8 May 2023 / Revised: 20 June 2023 / Accepted: 23 June 2023 / Published: 26 June 2023
(This article belongs to the Special Issue Bacteriophages and Their Enzymes as Antibacterial Agents)

Abstract

:
The central nervous system manages all of our activities (e.g., direct thinking and decision-making processes). It receives information from the environment and responds to environmental stimuli. Bacterial viruses (bacteriophages, phages) are the most numerous structures occurring in the biosphere and are also found in the human organism. Therefore, understanding how phages may influence this system is of great importance and is the purpose of this review. We have focused on the effect of natural bacteriophages in the central nervous system, linking them to those present in the gut microbiota, creating the gut-brain axis network, as well as their interdependence. Importantly, based on the current knowledge in the field of phage application (e.g., intranasal) in the treatment of bacterial diseases associated with the brain and nervous system, bacteriophages may have significant therapeutic potential. Moreover, it was indicated that bacteriophages may influence cognitive processing. In addition, phages (via phage display technology) appear promising as a targeted therapeutic tool in the treatment of, among other things, brain cancers. The information collected and reviewed in this work indicates that phages and their impact on the nervous system is a fascinating and, so far, underexplored field. Therefore, the aim of this review is not only to summarize currently available information on the association of phages with the nervous system, but also to stimulate future studies that could pave the way for novel therapeutic approaches potentially useful in treating bacterial and non-bacterial neural diseases.

1. Introduction

Numerous papers have suggested that gut bacteria play a critical role in shaping our health [1,2,3]. It has been demonstrated that a healthy gut microbiota is essential for proper development, immune response, homeostasis, and even learning processes [4,5,6,7,8,9]. Besides the bacterial component of the microbiota, other crucial microorganisms are viruses—more specifically, bacteriophages (phages), which are bacterial viruses, estimated to outnumber bacteria in the gut by one order of magnitude (1015 phages and 1014 bacteria) [10,11,12]. Interestingly, it is probable that 31 billion phage particles pass across the human gut epithelial cell layers each day [13]. The influence of bacteriophages on human health and their role in shaping the overall homeostasis have yet to be fully understood, but it is acknowledged that phages may have a huge impact on the composition of gut microbiota and the function it serves [14,15,16].
Bacteriophages are viruses that invade bacteria. Not only can they eradicate these bacteria, but they also play a significant role in bacterial evolution. This is due to the bacteriophages’ ability to integrate their genetic material with the bacterial genome and facilitate the transfer of genes between bacteria. Phages are ubiquitous, found in both the environment and living organisms [17,18], and may be isolated from different sources, ranging from aquatic samples to soil, or from human specimens [19,20]. Interestingly, they occupy nearly every ecological niche within our bodies, predominantly inhabiting areas such as the urinary tract, lungs, gut, mouth, and skin [21,22], and the composition of each body niche phageome varies significantly [21]. Among different human phageomes, those inhabiting the gut and mouth are probably the most thoroughly characterized because these sites are easily accessible and phages occur there in high concentrations [19,23,24,25].
The central nervous system (CNS) is composed of the brain (cerebrum and cerebellum), spinal cord, and optic nerves, as well as the membranes covering them, and can be infected by different pathogens, regardless of difficulties in penetrating the blood–brain barrier (BBB) [26]. The system is well shielded by the BBB, the major site of a blood–CNS exchange. In humans, it is a neurovascular unit composed of brain microvascular endothelial cells (BMECs), pericytes, astrocytic endfeet, microglia, and neurons. All these cells are connected to each other by tight junctions or adherens junctions, making it extremely difficult for any molecule to penetrate [27]. Surprisingly, recent studies have shown that bacteriophages may also be present in the central nervous system. This is in line with a previous report of bacteriophages’ ability to penetrate the BBB, which opens up a new research area, expanding their therapeutic potential in treating CNS-related bacterial infections, but also in brain cancers and other brain-related diseases (including the study of phages as carriers transporting different drugs through the BBB) [28,29,30,31,32,33,34,35,36].
In this review, we focused on the occurrence of natural bacteriophages in the nervous system, linking their presence in the intestine with the influence they may have on the nervous system. Additionally, we also focused on the current knowledge of phage application and its potential in the treatment of diseases associated with the brain and nervous system (which have presented serious difficulties for treatment).

2. Phage Distribution in the Human Body

2.1. Composition of the Gut Phageome

The intestinal microbiota has been shown to play a major role in physiological processes in host organisms and its dysregulation has been linked to the development of many diseases, such as irritable bowel syndrome (IBS), colorectal cancer, obesity, inflammatory bowel disease (IBD), Clostridium difficile infection, or neurological disorders [37,38,39,40,41]. The bacterial community, which is a primary component, plays a crucial role in mammalian gut physiology. It facilitates metabolic functions, protects against pathogens, and modulates the immune response [7,8,42]. The human gut is also a place where fungi and viruses—especially bacteriophages—are observed [19,23,24,25]. Their role is to shape the structure of the bacterial community by specific lysis (disintegration or decomposition of the cell wall structure) of particular bacteria, but also by modulating the activity of the immune system, mediating the anti-inflammatory response, as well as playing a protective role against invasion of pathogens [14,16,43,44].
Bacteriophages follow one of two main life cycles: lytic and lysogenic. In the lytic cycle, the complete destruction of bacterial cells is observed. These bacteriophages (lytic, virulent) use host cell machinery to replicate and assemble new phage virions. On the other hand, the lysogenic cycle is favored by temperate phages, which base their life cycles on the ability to integrate their genetic material with the host genome and switch to the latent stage in the form of so-called prophages. Such phages can also enter the lytic cycle in response to certain conditions, lyse the cell, and release progeny virions into the environment. It is reported that the lysogenic cycle is favored by phages inhabiting human and animal bodies [45,46]. By comparison, bacteriophages in aquatic ecosystems prefer the classic “kill-the-winner” dynamic [46,47]. The reason for this is still unclear, but Knowles et al. hypothesized that the polarity between the ecosystem of the human body and aquatic ecosystems is based on the observation that temperate phages become increasingly important in diverse ecosystems, especially those of high microbial densities [48]. These phages can modulate gene expression in bacteria and alter their phenotype through transposition, induction, and horizontal gene transfer (HGT). Temperate phages may also transfer virulence or antibiotic-resistance genes between bacterial cells. The vast repertoire of genes can shape bacterial diversity and function; thus, bacteriophages contribute to bacterial balance and homeostasis. Shifts in the composition of the virome (mainly phageome) affect human health indirectly because some diseases are suggested to be linked to alterations in the bacterial community [49,50,51,52,53,54,55,56,57]. Moreover, phages, including temperate ones, may also modulate eukaryotic cell functions and human immune response by inducing, among others, phagocytosis, cytokine production, and leukocytes activation, as well as the polarization of macrophages [58].
Current research suggests that the composition of the gut phageome is highly individualistic, as it consists mostly of long-term colonizing temperate phages [59], but it has also been shown to change throughout the human lifespan. It adapts to our microbiota and changes by affecting not only bacterial hosts but also the human organism by modulating immune response [59,60,61,62,63]. After birth, phage content and diversity are extremely low. However, this changes during the early stages of life, when the human phageome is highly dynamic. Initially, it is dominated by dsDNA tailed bacteriophages, representing sipho-, podo-, and myovirus morphotypes, previously classified (before major changes to bacteriophage taxonomy, reflecting genomic relationships rather than morphology-based classification) as members of—now abolished—Siphoviridae, Podoviridae, and Myoviridae, respectively. Phages belonging to the Microviridae family (ssDNA) were also detected. During infancy, viral and bacterial composition undergo dynamic modifications. Interestingly, the greatest richness and diversity of gut bacteriophages is observed within the first four days of life, and subsequently decreases over time, reaching stability in adulthood [64]. Additionally, it can be disturbed by different factors, such as diet, lifestyle changes, or diseases [65,66,67]. The gastrointestinal tract as well as other human body surfaces are lined with a mucosal layer, utilized by microbes to inhabit and to communicate with the host [68]. Bacteriophages populate mucosal surfaces by binding to mucin glycoproteins through immunoglobulin-like spikes present on their capsid in a process referred to as bacteriophage adherence to mucin (BAM). This phenomenon is believed to most likely play two significant roles: protection from pathogenic bacteria for the human host and providing lysogens with an environment to develop a bacterial symbiotic relationship that benefits the human host [62,65,66].
Reyes et al. (2010) performed a metagenomic analysis of human gut viromes and demonstrated that the fecal virome is highly diverse, while interpersonal viral diversity remains relatively low [46]. Moreover, up to 80% of virome reads generated in that study did not match any known sequences in public databases, which highlights the difficulty in identifying bacteriophage communities and the limitations of these estimates. Interestingly, Dutilh et al. (2014) applied a cross-assembly approach for unknown virome sequence data and identified a ~97 kb circular genome of a novel dsDNA phage named “CrAssphage” [67,68]. It is highly abundant and ubiquitous in all human fecal samples amplified using Bacteroides intestinalis [69]. Manrique et al. (2016) proposed the term “healthy gut phageome”, which consists of core and common phages present in healthy individuals that are likely globally distributed [61]. According to the authors, it is generally dominated by temperate tailed phages (formerly grouped in the—now abolished—order Caudovirales, currently replaced on the higher taxonomic level with the class Caudoviricetes [70,71]) and lytic Microviridae bacteriophages [72,73], while in phage dysbiosis the gut phageome has an increased number of lytic phages and/or activated prophages [74]. The change in phageome composition may stimulate the development of life-threatening diseases or progression into a more severe state of diseases, such as periodontal disease, Parkinson’s disease, type 2 diabetes, cancer, or gastrointestinal disease [74,75,76,77,78,79]. Interestingly, it has been observed that there is a significant decrease in core bacteriophages in patients with gastrointestinal diseases such as Crohn’s disease or ulcerative colitis [61]. At the same time, the occurrence of these diseases was correlated with the expansion of pathogenic Proteobacteria (e.g., Escherichia coli, Fusobacterium) and reduced number of protective bacteria (e.g., Faecalibacterium prausnitzii, Rumininococci) [80,81,82]. An increasing number of research articles suggest that alterations in the gut phageome may be associated with diet, inflammatory bowel disease, malnutrition, or obesity, but it is important to highlight that it is still unknown whether they are simply a consequence of changes in the gut microbiota or they are directly implicated in these pathological states [63,83,84,85].

2.2. The Gut Microbiota–Brain Axis Network and Interdependence

There is evidence that gut microbiota can also modulate functions of the nervous system and impact the maintenance of its homeostasis [86,87,88,89,90,91]. The link directly presenting this dependence is the vagus nerve, which maintains physical bidirectional communication between the CNS, intestinal wall, and enteric nervous system (ENS) [92], consisting of neurons and glial cells, distributed throughout the gastrointestinal tract and responsible for controlling coordinated smooth muscle contractile activity and other gut functions [93]. It has been suggested that the development and function of the ENS are to some extent mediated and modulated by gut microbiota [92]. Communication between them involves both direct and indirect pathways. The direct pathway includes altered intestinal permeability and relies on the release of signaling molecules into the gut lumen from immune and enterochromaffin cells. As a result, motor, sensory, and secretory modalities of the gastrointestinal tract increase [94,95,96]. Besides the ENS, those signaling systems consist of the endocrine-immune system, the hypothalamus–pituitary–adrenal (HPA) axis, and both the sympathetic and parasympathetic arms of the autonomic nervous system (ANS) [94,95,97]. Chemical signaling also occurs through a direct and indirect pathway. A well-reported direct pathway includes short-chain fatty acids (SCFAs), which are lipids produced in the fermentation of dietary fiber by microorganisms. They were proven to take part in regulating neuroplasticity, epigenetics, gene expression, and even immune response in CNS [98]. Other examples of chemicals produced by gut microbiota include hormones, such as corticotrophin-releasing hormone (CRH), serotonin, dopamine, and other neuropeptides [5,99,100,101]. It is also worth mentioning that it was demonstrated that increased uptake of fructose or exposure to SCFAs, such as acetic acid, increases the titer of phages specific to Lactobacillus reuteri and members of the genus Lactococcus [102]. The indirect impact includes modulating the neuroendocrine system [103]. An example of such regulation may include the production of glucagon-like peptide-1 (GLP-1), which was reported to increase appetite. Gut microbiota can also induce the production of neurotransmitters and regulate their concentration, which makes them mediators of classical signaling in the nervous system [104,105,106,107]. Moreover, some intestinal bacteria may also synthesize neurotransmitters—e.g., Bacteroides, Parabacteroides, and Escherichia spp. produce γ-aminobutyric acid (GABA) [104], which is the main inhibitory neurotransmitter in the human cortex [108]. As stated above, the gut phageome contains mostly CrAssphages specific to Bacteroides, thus it provides an extra layer of regulation in the production of these neurotransmitters. Research has also confirmed that the gut microbiota may suppress certain gut–brain signaling pathways through microbial metabolites. As for the immune response, the gut microbiota plays a crucial role not only in the proper modulation and functioning of the peripheral immune system but also in the maturation, development, and activation of microglia, which is essential for the innate immune response of brain cells [109,110]. The disruption of the bottom-up (gut-brain) motif can have a direct, substantial effect on the host’s mental state, potentially leading to depression, anxiety, and symptoms of bipolar disorder. It may also play a role in the pathogenesis, behavioral disturbance, and progression of neurodegenerative disorders, such as multiple sclerosis, Alzheimer’s disease, and Parkinson’s disease [19,111,112,113,114], as well as amyotrophic lateral sclerosis (ALS) [115] and epilepsy [116].
On the other hand, the top-down (brain–gut) motif is based on the alteration of the gut by disturbed brain homeostasis. Scientists have pointed out several immunological and metabolic markers that are altered during neuroinflammation and infection [117]. It was suggested that if the gut–brain axis exists, then alterations in the CNS should be reflected in urine. Theoretically, during chronic CNS bacterial infection, the gut microbiome would be altered, thus leading to a perturbed metabolism both in the CNS and the gut, eventually releasing modified levels of metabolites or even new metabolites into the bloodstream. These metabolites would be filtered by the kidneys and, consequently, should be present in the urine. To confirm this hypothesis, the levels of three immunological biomarkers—interferon-gamma (IFNγ), vascular endothelial growth factor (VEGF), and myeloperoxidase (MPO)—were examined as their levels are altered during Mycobacterium tuberculosis meningitis [118,119]. The studies demonstrated elevated levels of all three biomarkers, indicating that the alteration in the gut–brain axis is indeed detectable in the urine [120,121,122].

2.3. Naturally Occurring Bacteriophages in the Central Nervous System

Bacteriophages inhabit various sites of the human body. It has been suggested that there is phage translocation from the intestinal tract to other parts of the body [123,124]. It has already been confirmed that they can enter through the epithelial cell barrier by transcytosis [13,125]. This evidence may provide a mechanism by which gut bacteriophages may access previously unknown body sites, such as the nervous system. Interestingly, phages have also been shown to penetrate neural endothelial cells [13]. Figure 1 presents the possible interactions of bacteriophages with the nervous system.
Surprisingly, in 2019, Ghose et al. identified viral particles in the cerebrospinal fluid (CSF) of healthy individuals (which at that time had been commonly considered sterile and unreachable) [28]. Examination of the CSF by epifluorescence microscopy demonstrated virus-like particles (VLPs) at an average of 104 per milliliter, yet other non-sterile human specimens such as saliva contained much more VLPs (106 VLPs/mL). Interestingly, attempts to amplify the V3-4 region of 16S rRNA in order to assess the CSF samples for the presence of any bacterial host were unsuccessful, suggesting that the CSF specimens were bacteria-free. Examination of sequencing reads from the CSF against currently available viral databases revealed that the reads had ~70% homology to known viral sequences, yet further characterization of the putative taxonomic composition determined that most of the contigs (~93%) could not be assigned to any known family, and of those that could be assigned, the vast majority represented tailed bacteriophages. These results suggest that the CSF and probably other parts of the nervous system may be populated by bacteriophages. However, it is still unknown whether these viruses serve a functional role as a non-host-derived immunity against potential bacterial invaders or if they simply entered the CSF with no apparent function [13,28,123,126,127]. Moreover, the effect that phages applied therapeutically may have on the functions of the nervous system has not been described so far.
As phages were confirmed to be naturally present in the nervous system, a new research field emerged to verify whether phages could be used in the treatment of bacterial diseases of the nervous system, such as meningococcal encephalitis, or—as modified bacteriophage—provide a safe carrier to be used in the context of treating cancers such as glioblastoma.

3. Bacteriophages and Cognitive Processing

To study the interplay between the gut phageome, the gut bacteriome, and executive function, which is one of the six key domains of cognition, Mayneris-Perxachs et al. performed fecal shotgun metagenomics and metabolomics analyses in four human cohorts [128]. They also tested mice that had received fecal microbiota transplantations and Drosophila melanogaster after bacteriophage supplementation. All human participants and animals underwent cognitive tests. As a result of these studies, it was observed that the level of tailed bacteriophages previously classified in the—now abolished—order of Caudovirales (now replaced with the class Caudoviricetes [70,71]) was negatively correlated with trial-making test B time (TMTB) (a neuropsychological test that involves visual scanning and working memory and examines the executive functions of the brain), while the level of Microviridae was positively correlated. In TMTB, participants had to connect numbers and letters in an alternating progressive sequence, which means 1 to A, A to 2, 2 to B, and so on, with precision and time being the main priorities in this test. Higher completion times indicate worse executive functions [129,130]. Sex was also identified as a significant factor when it comes to gut microbiota composition [131]. It was observed that women had lower scores in TMTB, which also correlated with higher Caudovirales levels. Surprisingly, no associations were observed in men. Mayneris-Perxachs et al. also performed the backward digit span test, which measures working memory to assess the participant’s ability to hold information in short-term memory and manipulate that information to produce results [132]. In this case, a higher score indicates better executive performance. This time no association was observed in women. The genomic analysis confirmed that most of the Caudovirales were uncultured and uncharacterized, and from those that could be identified, mostly phages infecting Lactococcus spp., Enterobacteriaceae, Firmicutes (e.g., Eubacterium rectale), or Bacteroidetes (Prevotella copri) were detected. These specific Caudovirales phages belong to the three described families, Siphoviridae, Demerecviridae, and Drexlerviridae, which comprised the former Siphoviridae family. As for the new genome-based Siphoviridae family, a negative association between this family and TMTB results was found. On the contrary, Siphoviridae were associated with better performance in the Stroop test (which measures the ability to inhibit cognitive interference that occurs when the processing of a specific stimulus feature impedes the simultaneous processing of a second stimulus) [133] and both short- and long-term memory [128]. Microviridae phages were associated with significant impairment in executive function in both women and men. Bacteriophages from this family were negatively correlated with Lactobacillus, Streptococcus, and Enterococcus, while they were positively linked to Bacteroides and Prevotella species. Conversely, the Caudovirales were positively associated with lactic acid bacteria. These viruses were supposedly responsible for better executive functions and information processing speed, but also for long- and short-term verbal memory and general cognition, but surprisingly, these correlations were only observed in men [128]. Researchers also found a connection between the specific level of Caudovirales and consumption of dietary products and a fat diet containing medium-chain fatty acids, which is in line with findings that supplementation of medium-chain fatty acids improved synaptic plasticity and cognitive function, both in humans and in mice [134,135]. The studies confirmed that a high sugar diet and high uptake of fructose or exposure to SCFAs increases levels of prophages, which can affect bacterial population by two possible mechanisms: firstly, bacteria are killed as a result of prophage induction and entering the lytic cycle, and secondly, the resulting phage progeny can then kill closely related species of bacteria, thus modulating the bacterial community and levels of metabolites they produce [102,136,137].

4. Possible Application of Bacteriophages in the Treatment of CNS-Related Diseases

4.1. CNS Infections

Bacteria, viruses, fungi, and parasites have different strategies for invading the CNS. Among bacteria infecting the CNS, the most common are Mycobacterium spp., Brucella spp., Listeria monocytogenes, Neisseria meningitidis, Streptococcus pneumoniae, Haemophilus influenzae, Borrelia burgdorferi, Staphylococcus aureus, and E. coli [138,139,140]. The spectrum of bacterial infections is wide, ranging from brain abscesses and septicemia to meningoencephalitis, and they can result from hematogenous spread from another site of the patient’s body, contiguous spread from the upper airways, injury, or surgery. Some bacteria, such as M. tuberculosis, can enter the CNS via lymphatic nodes [141]. For newborns, the most threatening bacteria are Streptococcus agalactiae and E. coli, followed by S. pneumoniae and N. meningitidis in the first weeks of life [142]. As an effect of bacterial infection, a specific cascade of events is initiated. In particular, events in the brain significantly modulate the feedback effect on the gut microbiota [19,143,144].
There are three main strategies for neurotropic pathogens to cross the BBB: (i) transcellular migration; (ii) paracellular migration; or (iii) a Trojan horse mechanism [145]. The mechanism of the transcellular entrance is based on the pathogen’s ability to bind to BMECs. The pathogen is then endocytosed, transported within the vacuole through BMECs, and finally released to the brain tissue. In the paracellular mechanism, the pathogen disrupts tight junctions and/or induces the production of pro-inflammatory cytokines, e.g., TNFα, IL-6, or IL-1β. Bacterial infection induces an inflammatory response via glial mediators, which are crucial in the communication between the host’s immune system and the brain, but also between the gut microbiota and the brain [146,147]. In the Trojan horse mechanism, pathogens infect phagocytes and—hidden inside them—cross the BBB and infect the CNS. Several bacteria, including E. coli, group B streptococci, S. pneumoniae, N. meningitidis, L. monocytogenes, and M. tuberculosis, cross the BBB transcellularly, but L. monocytogenes and M. tuberculosis can also cross the BBB via the Trojan horse mechanism [148,149].
Viruses can infect the CNS either by directly traversing the BBB through one of the three routes described above or by taking nonhematogenous routes. Figure 2 presents possible routes of penetrating the BBB and entering the CNS by bacteriophages. This can be achieved by retrograde axonal transport from peripheral nerves to the CNS, the nasal olfactory epithelium, and neurons. These viruses can stimulate the production of pro-inflammatory cytokines just like bacteria, but they were also reported to stimulate the production of metalloproteases in BMECs, astrocytes, and pericytes. As an effect of the activity of these enzymes, tight junctions can be destabilized by initiating the RhoA kinase pathway and enhancing the permeability of BMEC monolayers in vitro [150,151].

4.2. Possibility of Phage Therapy in the Treatment of CNS-Related Bacterial Infections

With dramatically increasing antibiotic resistance, alternative therapeutic strategies are critically needed. Phage therapy (PT) has been considered a vital part of the solution [152,153]. Bacteriophages offer several advantages, including (i) high specificity, allowing selective targeting of pathogenic bacterial strains with no negative impact on the natural microbiota; (ii) self-replication at the side of infection; (iii) the ability to kill antibiotic-resistant bacterial strains; (iv) a lower risk of spreading resistance mechanisms; (v) coevolution with the bacterial host; (vi) safety even for immunocompromised patients; and (vii) little to no side effects [73]. Moreover, the majority of the antibiotics approved over the past decades represent already known classes of antimicrobials, with only a fraction having novel chemical structures [154].
The very first report on using bacteriophages in the treatment of brain-related bacterial diseases dates back to 1943 when Dubos et al. administrated phages specific to Shigella dysenteriae intraperitoneally (ip) in a mouse model [155]. What is more, the applied phages were detected in the brains of the animals to which they were administered. After phage application increased, phage titer was observed in infected mice. Interestingly, out of eight mice treated with high bacteriophage titer (109 PFU/animal), six animals survived, whereas out of eight pup rats treated with lower phage titer (105 PFU/animal) only two survived. The next phage breakthrough came in 1982 when Smith and Huggins conducted an experiment using bacteriophages in the treatment of E. coli producing colicin V, which causes septicemia and meningitis [156]. They demonstrated that Phage R was highly active in vitro, which was reflected in vivo (when 3 × 104 phage particles given intramuscularly or 3 × 103 given intravenously were sufficient) to cure mice administered a potentially lethal intramuscular dose of E. coli. The authors also demonstrated that although phage R-resistant mutants are relatively common in laboratory cultures, they were only found in a few phage-treated mice, and even then they were confined to the inoculation site and outnumbered by phage-sensitive cells. The efficacy of PT vs. antibiotic treatment was compared and showed that one dose of phage R was at least equivalent to multiple doses of streptomycin and more effective than multiple doses of tetracycline, ampicillin, chloramphenicol, or trimethoprim plus sulphafurazole in treating mice infected intramuscularly or intracerebrally with E. coli. The therapeutic success of PT was due to high in vivo activity and the failure of phage-resistant mutants to proliferate during treatment [156]. In 1998, Barrow et al. investigated E. coli causing septicemia and meningitis-like infection and newly isolated phage R (procured from sewage water) selective for K1 antigen-possessing bacterial strains [157]. This antigen is a capsular polysaccharide, associated with a highly virulent nature, and is often found in pathogenic E. coli [158]. When chickens were intramuscularly infected with 106 CFU or intracranially inoculated with 103 CFU of E. coli and simultaneously intramuscularly treated with 106 or 108 PFU of phage R (in the case intramuscular or intracranial bacterial administration, respectively), no morbidity or mortality of the animals was observed [157]. The authors administered lower doses of bacteriophage preparation into the chickens and found that in the case of the intramuscular bacterial inoculation, the application of 102 PFU caused some protection against bacteria, but the effect was not statistically significant. However, with the intracranial bacterial administration, doses lower than 108 PFU were not successful. Interestingly, the authors also observed that newly hatched chickens could acquire some protection against pathogens with lower doses of bacteriophages (106 PFU) compared to older chickens (108 PFU). They also tested the effectiveness of PT on colostrum-deprived calves infected orally and demonstrated that in calves treated intramuscularly with phage R, bacterial infection was controlled, yet the infection could not be prevented altogether. Administration of PT delayed the onset of signs of E. coli bacteremia.
Poulliot et al. demonstrated that isolated lytic bacteriophages can neutralize E. coli strain S242, which causes sepsis and one of the most severe E. coli infections, fatal neonatal meningitis, with a mortality rate of up to 25% [30]. The mortality rate may increase as the infection itself may worsen if the pathogen acquires multi-drug resistance (MDR), and E. coli strains expressing acquired extended-spectrum beta-lactamases (ESBL), such as CTX-M-type enzymes [159], turned out to be especially lethal. The major one is CTX-M1, which is an enzyme with hydrolytic activity against cefotaxime. The EC200PP bacteriophage (isolated from environmental sewage samples in France) was characterized in vivo and ex vivo with regard to stability and pharmacokinetic properties in pup rats. The EC200PP was stable for at least 24 h in adult and pup rats’ serum, while in human serum phage titer decreased by 2 to 3 logs in the first 2 h of incubation and remained stable after that. As stated in the article, serum possesses phage-inhibitory activity, and this activity is not specific to EC200PP, but rather to all phages. When phage preparation was administrated, most of the phage virions were preferably located in the spleen and kidneys, while low titers were observed in urine and the CNS. Because E. coli S242 strain may induce sepsis and meningitis, both models were investigated. For the sepsis model, pup rats were injected with 104 CFU of S242 bacteria ip and then administered 108 PFU/mL of EC200PP subcutaneously 7 h or 24 h after the induction of infection. Interestingly, a 7 h post-infection intervention resulted in 100% recovery, while phage administration 24 h after infection resulted in only 50%. For meningitis models, two scenarios were investigated: with a lower dose of the S242 bacteria administered intrathecally, and with a higher inoculum of bacteria, corresponding to that normally encountered in human meningitis. The first case was based on administering 200 CFU per rat to cistera magna, which resulted in a titer of 106 CFU/mL in the CSF after 24 h. Pup rats were then treated with 108 PFU of EC200PP per pup rat administered ip 1h post-infection, which contributed to a 100% survival rate up to day 5. This also contributed to nondetectable bacterial levels in the CSF, and EC200PP titer of 4.5 +/− 0.2 log PFU/mL 24 h post-infection. Based on the highly successful initial results, a second scenario was then investigated, with a higher concentration of S242 bacteria (2 × 106 CFU) administered intrathecally, relevant to that observed in human meningitis, and then 108 PFU of EC200PP 1h post-infection. As a result, all rats survived to day 5 of the experiment. After 24 h, the CSF samples were positive for bacteria, but no signs of bacteremia were observed, and on day 5, in three out of five rats, the CSF was sterile with no signs of infection. However, when phage treatment was delayed to 2 or 3 h post-infection, the survival rate decreased dramatically [30].
Bacteriophage K1F specific to E. coli strain EV36 also expressing the K1 capsule antigen was used in an in vitro model of bacterial neonatal meningitis in human cerebral microvascular endothelial cells (hCMEC), which are a part of the BBB [160,161,162,163]. The studies revealed that the K1F phage was internalized by phagocytosis upon contact with hCMEC cells, and then degraded through a constitutive pathway—via lysosomal degradation along with PAMP-LC3-dependent phagocytosis, which suggests that phages are recognized by Toll-Like Receptor 3 (TLR3) on mammalian cells. Importantly, the K1F phage did not induce an inflammatory response. The authors also reported temporarily decreased barrier resistance of hCMEC cells, which could facilitate the transition of immune cells across the endothelial vessel in vitro. Importantly, K1F can infect the EV36 strain intracellularly within hCMEC cells without inducing inflammation [29].
Apart from whole phage virions, bacteriophage endolysins were also recently documented to demonstrate great potential for treating severe cases of infections with gram-negative and gram-positive bacteria, such as E. coli, Salmonella spp., Pseudomonas aeruginosa, Pseudomonas putida, Shigella boydii, Shigella flexneri, Vibrio fischeri, Vibrio vulnificus, and many more [164,165,166,167,168]. Phage endolysins are subdivided into five families depending on the peptidoglycan disruption mechanism: glucosaminidases, lysozymes or muramidases, lytic transglycosylases, endopeptidases, and amidases. [169,170,171]. Gram-positive bacteria are more prone to endolysins, which are able to disrupt the peptidoglycan layer. Because gram-negative bacteria possess an outer membrane, shielding the peptidoglycan layer, the activity of endolysins towards gram-negative pathogens may be restricted [172] as the low permeability of the outer membrane makes the exogenous application of endolysins challenging [173] and some endolysins require additional strategies to permeate the outer membrane, such as endogenous lysis mediated by holins, pinholins, and bacterial Sec translocases or exogenous lysis [174]. Their medical potential is highlighted by the fact that, unlike for bacteriophages, there are no reports of bacterial resistance to endolysins acquired using classic mechanisms: by mutations, receptor modification, passive adaptation, restriction modification, CRISPR-Cas, and pseudolysogeny [175]. The most suitable and diverse endolysin described to date is Cpl-1 derived from S. pneumoniae bacteriophage Cp-1 [176]. Usually, bacteriophages are highly selective for a specific strain of bacteria; however, phage endolysins can display a broader activity against different and multiple hosts [166] and they can be easily engineered to enhance their lytic potential [177]. An increasing number of papers confirm their medical potential [178,179,180,181,182], but the results of clinical trials on phages have yet to prove the formal efficacy of PT.

4.3. Phages and Autoimmune Diseases of the Nervous System

Recent data from our and other groups suggest that phages possess immunomodulating activities that may have the potential to control aberrant immune responses, including autoimmune reactions [15,183]. Autoimmune disorders of the nervous system (e.g., inflammatory demyelinating diseases, myasthenia gravis, etc.) pose a significant medical challenge as their treatment is not always satisfactory while side effects of currently available therapy may be serious and sometimes life-threatening [184,185]. Therefore, in those clinical settings one could envisage the potential therapeutic action of selected phages with proven anti-inflammatory and immunosuppressive activity and a virtual lack of side effects [186].

5. Potential of Phage Application in Targeted Therapy for Brain Cancers

Glioblastoma—the most abundant and highly aggressive primary brain tumor in adults—is characterized with a rising incidence rate, currently ranging from 0.59 to 5 per 100,000 persons internationally [187], with a median life expectancy of approximately 14–16 months [188], and only 5% of all patients surviving to 5 years post diagnosis [189]. In terms of risk factors, glioblastoma is poorly understood. Due to the low frequency of the tumor worldwide, it was suggested that the brain demonstrates a higher degree of protection from genotoxic stress than other organs [190]. Because brain cells are protected from mutagens by the BBB, it was postulated that brain tumors, especially glioblastoma, arise from a small pool of adult neuronal stem and progenitor cells (NSPC), which are located in the subventricular zone, subcortical white matter, but also in the dentate gyrus of the hippocampus [191,192,193]. These cells retain the ability to enter mitosis and play a crucial role in learning and memory [194]. Of all these cells, glioma-initiating cells or glioma stem-like cells are the most important; they can self-renew, differentiate, and form secondary tumors in xenotransplantation [195]. The progeny of glioma-initiating cells mostly resembles astrocytes, but some cells were also noted to have features of endothelial cells or pericytes [196,197,198]. In most cases of glioblastoma, tumors are located in the frontal, temporal, and parietal lobes, and rarely in the occipital-lobe, cerebellar, brainstem, and spinal cord [189]. The current standard of care involves surgery, followed by radiotherapy in combination with chemotherapy [195]). In recent years, molecular targeted therapy was suggested as an alternative treatment option in glioblastoma, and this topic was comprehensively reviewed by Le Rhun et al. (2019). Possible targets are molecules involved in oncogenic signaling via tyrosine receptor kinases, cell cycle control, and susceptibility to apoptosis. Repurposing of drugs conventionally used in other diseases, e.g., metformin or the anti-epileptic drug valproic acid, is also considered.
Even though many different drugs that potentially target glioblastoma have been investigated, none of them have entered clinical practice, possibly because most glioblastomas are far from a single-pathway driven disease [195]. As reported, glioma stem-like cells are resistant to radiotherapy and chemotherapy for yet undefined reasons [199,200,201,202,203,204,205]. Especially in the case of brain tumors, the therapeutic effect is difficult to achieve because of the presence of the BBB, which impedes the penetration of drug particles. Moreover, the glioma stem cells (GSCs) turned out to be capable of self-renewal and resistant to standard treatment [202]. This provides an unmet need to create innovative solutions that would allow difficulties in the delivery, pharmacokinetics, and therapeutic use of currently available drugs to be overcome, but also the development of new vectors and novel methods of administration, diagnosis, and monitoring of treatment responses. Bacteriophages may have the potential to meet these conditions.
Filamentous, rod-shaped phages such as f1, fd, and most importantly M13 are widely used in a variety of biochemical and biomedical applications. Recent studies suggest that these phages can greatly impact the development of new therapeutic strategies due to their ability to self-assemble into nanoscale structures [203]. Moreover, rod-shaped nanoparticles possess higher selectivity and specificity as well as targeting abilities to endothelial cells and brain endothelium compared to their spherical counterparts [204]. Filamentous capsids are also robust and monodisperse [205,206]. However, the greatest advantage of these phages comes from their genetic flexibility and easy-to-manipulate structure [206]. One recent breakthrough was the use of bacteriophage nanoparticles in neuronal regeneration and the specific development of biomimetic scaffolds for tissue engineering purposes. Therefore, they may have enormous potential in creating bioactive constructs as a scaffold for repairing and regenerating injured tissues in the nervous system.
The phage display method was developed in order to obtain recombinant, exogenous peptides or proteins exposed on the surface of phage virions [207]. This allows for the preparation of a diverse library of phages presenting peptides/proteins that can be screened to select clones with the highest affinity to the target molecule. Nowadays, phage display technology is commonly used for drug development because it allows for an easy and effective screening of large pools of peptides or antibody variable domains. The development of the method paved the way for a wider use of bacteriophages in nanotechnology and biomedical studies [208]. The phage display technique is a promising tool that may be useful in the treatment of glioma. Targeted therapy, cytotoxic effect, and neuroimaging is achievable [209].
Even though there are alternatives and similar platforms, such as yeast display or ribosome display technology, phage display is still considered the best option due to its high diversity and easy handling [210,211,212,213,214].
As stated earlier, the BBB prevents roughly 98% of potential drugs, pathogens, and molecules from entering the CNS. However, bacteriophages modified with the phage display technique, presenting specific receptors on the surface of their capsids, can penetrate the BBB using the Trojan horse mechanism [215,216]. Moreover, the endocytosis-based mechanism was also confirmed for bacteriophages (Figure 2B) [13,123,217]. The E.coli bacteriophage PK1A2 was demonstrated to bind to polysialic acid on the surface of human neuroblastoma cells, thus sharing structural similarity with the E.coli phage receptor [218,219]. Not only did the phages bind to eucaryotic cells, but they were also internalized into the cells through endocytosis and largely retained infectivity for 24 h [219]. Anand et al. developed a bifunctional viral nanocontainer based on the Salmonella-specific bacteriophage P22 [31]. Ziconotide, an analgesic peptide drug derived from a marine snail, was incorporated into the nanocontainer, while a cell-penetrating peptide HIV-tat was displayed on the exterior surface of the phage capsid. Such a modified P22 was successfully transferred into several cell-based models of rat brain microvascular endothelial cells as well as human brain microvascular endothelial cells. Hence, these findings open another alternative route for drug administration [31]. The F88 phage was also reported to easily penetrate the BBB after intranasal administration. One of the most advantageous features of filamentous bacteriophages is their shape and structure. The linear structure of f88 makes it a favorable nanocarrier to penetrate the BBB. After the intranasal administration of f88, phage particles were detected in the olfactory bulb and hippocampus regions of mice brains [220].
A novel approach using a convection-enhanced delivery method (CED) was used by Ksendzovsky et al., in which a high-pressure gradient of bulk flow is generated as a delivery tool for a desired preparation [221]. The tip of an infusion catheter delivers therapeutics directly through the interstitial spaces of the CNS. By applying this, we eliminate the requirement to deliver high, unwieldy therapeutic concentrations, which enables us to achieve therapeutic concentration of the drug in the brain parenchyma [222]. The results from Ksendzovsky et al.’s experiments indicate that CED is a promising method of phage delivery to the brain due to the fact that the M13 bacteriophage was spotted in the white and grey matter of mice brains.
The development of CNS-related diseases is much more challenging than that of non-CNS diseases due to the presence of the BBB, but also the brain–blood tumor barrier (BBTB). Although the structure itself differs from the BBB, both the BBB and BBTB drastically limit the efficient transport of most currently available drugs, mainly due to their molecular size, which surpasses the threshold limiting the molecules that can readily penetrate BBTS and BBB [223,224]. Additional challenges in CNS drug development include the high costs associated with drugs capable of penetrating the BBB and a potentially increased risk of side effects. Compared to other drugs, those targeting the CNS require higher concentrations due to the indirect route to the CNS via the liver and kidneys. These organs tend to remove a significant portion of these molecules from the bloodstream. One approach to improve drug delivery is through receptor-mediated transcytosis, a method that targets drugs to specific receptors expressed in the BBB. To date, two such monoclonal antibody-based constructs—with either a transferrin receptor or insulin receptor—have successfully completed phase II clinical trials [225,226].
During neurological disorders, the expression of different unique receptors that home in on the CNS can be used to improve drug delivery, and such peptides can be selected with phage display technology [227]. To list a few examples, Eriste et al. identified a novel peptide gHo that effectively binds glioma cells [228] and, when covalently conjugated to the cell-penetrating peptide pVEC and doxorubicin, showed promise in the mouse subcutaneous U87 tumor model, although no effect was demonstrated in the intracranial glioma model [228,229,230]. Chen et al. applied phage display to identify an M1 peptide that localized to the brain and was demonstrated to penetrate the BBB/BBTB [231]. Furthermore, conjugation of paclitaxel to the RGD-modified M1 peptide not only allowed for efficient translocation across the BBB, but also greatly improved—typically low—PTX solubility. Importantly, it was the first vector targeting glioma with the ability to travel through the BBB and BBTS. Kim et al. used in vitro and in vivo phage display screens to select glioblastoma targeting peptides and identified a Cadherin 2-binding peptide with the ability to selectively target GSCs over differentiated glioma and non-neoplastic brain cells [232]. Moreover, in a mouse model of intracranially xenografted glioblastoma, the peptide—administered intravenously—specifically targeted intracranial tumors. Phage display technology has also been used to develop nanobodies targeting CA9 [233] and ABCC3 [234], expression of which was detected to correspond with the worse overall survival of patients with glioblastoma. Another approach could involve combining the use of phage display for identification of tumor-targeting peptides and CAR-T therapy for novel modifications of classic CAR-T, which seems to have potential in the development of novel cytotoxic therapies for glioblastoma, as demonstrated by Potez et al. [202].
Importantly, the potential of phage display is not limited to being a useful platform in the drug discovery process—modified phage particles can also be directly applied as targeted delivery platforms, especially because natural phages generally lack native tropism to eukaryotic cells. Przystal et al. report constructing a dual targeting hybrid vector, containing the genome of human adeno-associated virus (AAV) incorporated into the engineered particles of bacteriophage M13 [32]. The vector was intended for intravenous administration, and its dual tumor targeting system relied on, firstly, targeting αvβ3 integrin receptor by phage-expressed ligand RGD4C, and secondly, tumor-specific and temozolomide-induced gene expression from a Grp78 promotor, acting as suicide gene therapy. The authors demonstrated the potential of this approach in vivo, as the combination of temozolomide and the hybrid vector suppressed the growth of orthotopic glioblastoma in mice.
Many aspects of phage interactions with the nervous system still remain unexplained, i.e., phage binding to the tumor cells, their delivery, and phage titer after its administration (pharmacokinetics). Moreover, it is yet unknown whether they are sufficient to treat tumors in human.
While all these examples focus primarily on the development of novel anti-cancer therapies, it is important to note that the peptides identified with the phage display technology or modified phage particles can also be used for the development of new strategies for the diagnosis and imaging of tumor cells.

6. Intranasal Administration

A different approach to the problem of limited penetration through the BBB was suggested by Wan et al., who focused on bypassing the BBB altogether by choosing a different administration route and simultaneously using a peptide-homing tag, which would theoretically allow issues with the migration of drug molecules to the CNS to be overcome [235]. Intranasal application was proposed as a possible efficient alternative, and the M13 phage display platform was used to screen a peptide library for promising carriers that could then be intranasally administered to rats. Clone7 was selected for further examination based on the highest translocation efficiency—it demonstrated an approximately 50-fold higher translocation efficiency compared to the control phage. The 11 amino acids long sequence of the peptide from Clone7 was identified to be as follows: ACTTPHAWLCG. When administered intranasally to Wistar rats, Clone7 was detectable in the brain in a dose-dependent manner after 15 min, reaching maximum after 45 min, and a higher phage titer was detected in the brain than in the liver, spleen, and blood of the examined rats, indicating that Clone7 was able to bypass the BBB and enter the brain directly. Most of the Clone7 phage particles, as well as the peptide itself, were found along the olfactory nerve. Two possible direct pathways may exist to transport administrated substances from olfactory mucosa into the CNS: the olfactory nerve pathway and the olfactory epithelial pathway (Figure 2A) [236]. The results described above indicate that phages displaying the homing peptide entered the CNS through the olfactory nerve pathway [235]. Interestingly, when administered alone and not on a phage particle, the selected peptide—although clearly observed in the olfactory nerve—could not be detected in the brain in the given time frame of up to 8 h. To date, it has only been observed in the case of a single peptide; however, if this was a more general characteristic of nose-to-brain homing peptides, it would further enforce the application of phage particles as nanocarriers for targeted drug delivery to the brain following intranasal administration. Therefore, this discovery opens a possible new route of drug administration in the treatment of bacterial and viral infections of the CNS and also supports the potential of phage-based solutions in the treatment of brain cancers.
There are also studies of phages with two large, different exogenous peptides that were successfully created in a so-called dual phage display system. The first one, alkaline phosphatase, or green fluorescent protein (GFP), was located on the head, and the other, anti-CEA scFv, on the tail of the lambda phage [237]. A similar approach was used by Rajaram et al., who also created a dual-display system [238] in which a biomarker and a short peptide containing the streptavidin-binding motif Histidine-Proline-Glutamine (HPQ) were displayed on the opposite ends of the M13 bacteriophage capsid. This additional second peptide with a high affinity to streptavidin was introduced to allow for sample (phage) purification on streptavidin-conjugated magnetic beads to increase assay sensitivity and decrease background signal. Finally, Fagète et al. reported the selection of bispecific antibodies, which they described as a new frontier in antibody therapy, that also used a dual-phage system, allowing for the co-selection of pairs of antibodies based on the co-engagement of their respective targets [239]. The authors demonstrated that the use of two complementary leucine zipper domains, which later heterodimerize with a high affinity, allowed for the assembly of two antibody fragments—one displayed on the surface of the phage and the other present in the bacterial cell as a soluble component.
Carrera et al. (2004) prepared phage displaying scFv GNC 92H2, which has an affinity to cocaine receptors. Intranasal administration of GNC 92H2-pVIII (1.0 × 1014) resulted in detection of the modified phage in the brain of rats and, as expected, the displayed protein has the ability to block the psychoactive effects of cocaine [240].
All these approaches enhance the detection or specificity of targeting and may be useful in creating bacteriophages for biomedical purposes, both diagnostic and therapeutic.

7. Conclusions

While PT has recently received greatly increased attention and its prospects appear promising, the potential application of that therapy in treating diseases of the nervous system has rarely been mentioned. Nevertheless, there are some data in the literature and preliminary studies that strongly suggest that the CNS and peripheral nervous system could indeed be targeted by that therapy. Thus, PT could be used as a weapon against bacterial infections of the CNS. Furthermore, recently described immunomodulating and anti-inflammatory effects of phages could pave the way towards their use in treating autoimmune diseases of the nervous system. In addition, phage engineering could allow the use of such modified phages in the fight against some neoplasms of the CNS. It is therefore likely that classical PT as well as “repurposed” PT could provide more efficient means of treatment of both bacterial and non-bacterial diseases of the nervous system.

8. Future Perspective

The current state of the research in the treatment of bacterial infections in the CNS indicates that this area is vastly unexplored. While some foundational research has been conducted, there is much potential for broadening the search for phages specific to bacterial strains that commonly infect the CNS. Although partial ground research has been carried out, it will be highly beneficial if a wide search for phages specific to bacteria commonly infecting the CNS is initiated. After that, the next crucial step would be to determine phage pharmacokinetics, host range, stability, and safety. Then, universal and easily monitored models should be created to study penetration through the BBB of natural and modified bacteriophages, as well as phage-based nanocarriers and their effectiveness in delivering drugs to the targeted tissues and cells. One more highly important direction is testing larger groups of subjects with the treatment regimens that show promising results. One future possibility is bacteriophage-based modifications of the composition of gut microbiota to improve the efficiency of cognitive processes Another field altogether is cancer research, where bacteriophages might fit perfectly due to their natural properties and opportunities provided by the phage display technique—yet their potential is still underexploited. The only issue is if the phage constructs would effectively penetrate the BBB and how—if at all—the immune system would be activated by these particles. Excitingly, recent publications in 2022 have linked bacteriophages with CRISPR-like antiviral systems, a groundbreaking development [241,242,243,244,245,246]. Bacteriophage antiviral systems belong to all six known types of CRISPR-Cas systems and occur as divergent and hypercompact antiviral systems. Some of these systems lack key components, which suggests alternative functional roles or host complementation [246]. There are even reports of using the CRISPR-Cas system to genetically modify bacteriophages, thus giving them new mechanisms of action [247], which is an interesting perspective in the context of therapeutic phage applications.

Author Contributions

Conceptualization, A.J. and E.J.-M.; writing parts of the manuscript, A.J., W.F., J.M. and E.J.-M.; writing—review and editing, A.J., W.F., J.M., E.J.-M. and A.G.; figure preparation, A.J.; support and conceptual advice at all stages of manuscript preparation, A.G. and E.J.-M. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by statutory funds from the Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, and funds from the Lower Silesian Foundation for the Development of Healthcare “Dolfroz”.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

A. Górski is the co-inventor of patents owned by the L. Hirszfeld Institute covering phage preparations. The other authors declared that the research was conducted without any commercial or financial relationships that could be construed.

References

  1. Lau, H.C.H. Gut Microbiome in Health and Gastrointestinal Cancer. In Microbiome in Gastrointestinal Cancer; Springer Nature: Singapore, 2023; pp. 5–21. [Google Scholar]
  2. Park, W. Gut microbiomes and their metabolites shape human and animal health. J. Microbiol. 2018, 56, 151–153. [Google Scholar] [CrossRef] [Green Version]
  3. Xu, Z.; Knight, R. Dietary effects on human gut microbiome diversity. BJN 2015, 113, S1–S5. [Google Scholar] [CrossRef] [Green Version]
  4. Everard, A.; Cani, P.D. Diabetes, obesity and gut microbiota. Best Pract. Res. Clin. Gastroenterol. 2013, 27, 73–83. [Google Scholar] [CrossRef] [Green Version]
  5. Dinan, T.G.; Cryan, J.F. Brain–gut–microbiota axis-mood, metabolism and behaviour. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 69–70. [Google Scholar] [CrossRef]
  6. Dinan, T.G.; Cryan, J.F. Gut instincts: Microbiota as a key regulator of brain development, ageing and neurodegeneration. J. Physiol. 2017, 595, 489–503. [Google Scholar] [CrossRef] [Green Version]
  7. Belkaid, Y.; Hand, T.W. Role of the Microbiota in Immunity and Inflammation. Cell 2014, 157, 121–141. [Google Scholar] [CrossRef] [Green Version]
  8. Sokol, H.; Pigneur, B.; Watterlot, L.; Lakhdari, O.; Bermúdez-Humarán, L.G.; Gratadoux, J.J.; Blugeon, S.; Bridonneau, C.; Furet, J.P.; Corthier, G.; et al. Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc. Natl. Acad. Sci. USA 2008, 105, 16731–16736. [Google Scholar] [CrossRef] [Green Version]
  9. Sonnenburg, J.L.; Xu, J.; Leip, D.D.; Chen, C.H.; Westover, B.P.; Weatherford, J.; Buhler, J.D.; Gordon, J.I. Glycan Foraging in Vivo by an Intestine-Adapted Bacterial Symbiont. Science 2005, 307, 1955–1959. [Google Scholar] [CrossRef] [Green Version]
  10. Dalmasso, M.; Hill, C.; Ross, R.P. Exploiting gut bacteriophages for human health. Trends Microbiol. 2014, 22, 399–405. [Google Scholar] [CrossRef]
  11. Carding, S.R.; Davis, N.; Hoyles, L. Review article: The human intestinal virome in health and disease. Aliment. Pharmacol. Ther. 2017, 46, 800–815. [Google Scholar] [CrossRef]
  12. Qin, J.; Li, R.; Raes, J.; Arumugam, M.; Burgdorf, K.S.; Manichanh, C.; Nielsen, T.; Pons, N.; Levenez, F.; Yamada, T.; et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature 2010, 464, 59–65. [Google Scholar] [CrossRef] [Green Version]
  13. Nguyen, S.; Baker, K.; Padman, B.S.; Patwa, R.; Dunstan, R.A.; Weston, T.A.; Schlosser, K.; Bailey, B.; Lithgow, T.; Lazarou, M.; et al. Bacteriophage Transcytosis Provides a Mechanism To Cross Epithelial Cell Layers. mBio 2017, 8, 01874–17. [Google Scholar] [CrossRef] [Green Version]
  14. Ogilvie, L.A.; Jones, B.V. The human gut virome: A multifaceted majority. Front. Microbiol. 2015, 6, e01874-17. [Google Scholar] [CrossRef] [Green Version]
  15. Górski, A.; Dąbrowska, K.; Międzybrodzki, R.; Weber-Dąbrowska, B.; Łusiak-Szelachowska, M.; Jończyk-Matysiak, E.; Borysowski, J. Phages and immunomodulation. Future Microbiol. 2017, 12, 905–914. [Google Scholar] [CrossRef] [Green Version]
  16. Gogokhia, L.; Round, J.L. Immune–bacteriophage interactions in inflammatory bowel diseases. Curr. Opin. Virol. 2021, 49, 30–35. [Google Scholar] [CrossRef]
  17. Hendrix, R.W. Evolution: The long evolutionary reach of viruses. Curr. Biol. 1999, 9, 914–917. [Google Scholar] [CrossRef] [Green Version]
  18. Letarov, A.; Kulikov, E. The bacteriophages in human- and animal body-associated microbial communities. J. Appl. Microbiol. 2009, 107, 1–13. [Google Scholar] [CrossRef]
  19. Clokie, M.R.J.; Millard, A.D.; Letarov, V.A.; Heaphy, S. Phages in nature. Bacteriophage 2011, 1, 31–45. [Google Scholar] [CrossRef] [Green Version]
  20. Weber-Dąbrowska, B.; Jończyk-Matysiak, E.; Żaczek, M.; Łobocka, M.; Łusiak-Szelachowska, M.; Górski, A. Bacteriophage Procurement for Therapeutic Purposes. Front. Microbiol. 2016, 7, 1177. [Google Scholar] [CrossRef] [Green Version]
  21. Barr, J.J. A bacteriophages journey through the human body. Immunol. Rev. 2017, 279, 106–122. [Google Scholar] [CrossRef]
  22. Garretto, A.; Miller-Ensminger, T.; Wolfe, A.J.; Putonti, C. Bacteriophages of the lower urinary tract. Nat. Rev. Urol. 2019, 16, 422–432. [Google Scholar] [CrossRef]
  23. Sender, R.; Fuchs, S.; Milo, R. Are We Really Vastly Outnumbered? Revisiting the Ratio of Bacterial to Host Cells in Humans. Cell 2016, 164, 337–340. [Google Scholar] [CrossRef] [Green Version]
  24. Kim, M.S.; Park, E.J.; Roh, S.W.; Bae, J.W. Diversity and Abundance of Single-Stranded DNA Viruses in Human Feces. Appl. Environ. Microbiol. 2011, 77, 8062–8070. [Google Scholar] [CrossRef] [Green Version]
  25. Reyes, A.; Wu, M.; McNulty, N.P.; Rohwer, F.L.; Gordon, J.I. Gnotobiotic mouse model of phage–bacterial host dynamics in the human gut. Proc. Natl. Acad. Sci. USA 2013, 110, 20236–20241. [Google Scholar] [CrossRef] [Green Version]
  26. Archibald, L.K.; Quisling, R.G. Central Nervous System Infections. In Textbook of Neurointensive Care; Springer: London, UK, 2013; pp. 427–517. [Google Scholar]
  27. Dickens, D.; Radisch, S.; Pirmohamed, M. Drug Transporters at the Blood-Brain Barrier. In Drug Transporters: Volume 1: Role and Importance in ADME and Drug Development; Royal Society of Chemistry: London, UK, 2016; pp. 151–183. [Google Scholar]
  28. Ghose, C.; Ly, M.; Schwanemann, L.K.; Shin, J.H.; Atab, K.; Barr, J.J.; Little, M.; Schoolet, R.T.; Chopyk, J.; Pride, D.T. The Virome of Cerebrospinal Fluid: Viruses Where We Once Thought There Were None. Front. Microbiol. 2019, 10, 2061. [Google Scholar] [CrossRef]
  29. Møller-Olsen, C.; Ross, T.; Leppard, K.N.; Foisor, V.; Smith, C.; Grammatopoulos, D.K.; Sagona, A.P. Bacteriophage K1F targets Escherichia coli K1 in cerebral endothelial cells and influences the barrier function. Sci. Rep. 2020, 10, 1–17. [Google Scholar] [CrossRef]
  30. Pouillot, F.; Chomton, M.; Blois, H.; Courroux, C.; Noelig, J.; Bidet, P.; Bingen, E.; Bonacorsi, S. Efficacy of Bacteriophage Therapy in Experimental Sepsis and Meningitis Caused by a Clone O25b:H4-ST131 Escherichia coli Strain Producing CTX-M-15. Antimicrob. Agents Chemother. 2012, 56, 3568–3575. [Google Scholar] [CrossRef] [Green Version]
  31. Anand, P.; O’Neil, A.; Lin, E.; Douglas, T.; Holford, M. Tailored delivery of analgesic ziconotide across a blood brain barrier model using viral nanocontainers. Sci. Rep. 2015, 5, srep12497. [Google Scholar] [CrossRef] [Green Version]
  32. Przystal, J.M.; Waramit, S.; Pranjol, M.Z.I.; Yan, W.; Chu, G.; Chongchai, A.; Samarth, G.; Olaciregui, N.G.; Tabatabai, G.; Carcaboso, A.M.; et al. Efficacy of systemic temozolomide-activated phage-targeted gene therapy in human glioblastoma. EMBO Mol. Med. 2019, 11, e8492. [Google Scholar] [CrossRef]
  33. Staquicini, F.I.; Ozawa, M.G.; Moya, C.A.; Driessen, W.H.P.; Barbu, E.M.; Nishimori, H.; Soghomonyan, S.; Flores, L.G., 2nd; Liang, X.; Paolillo, V.; et al. Systemic combinatorial peptide selection yields a non-canonical iron-mimicry mechanism for targeting tumors in a mouse model of human glioblastoma. J. Clin. Investig. 2011, 121, 161–173. [Google Scholar] [CrossRef]
  34. Zhang, X.; Zhang, X.; Gao, H.; Qing, G. Phage display derived peptides for Alzheimer’s disease therapy and diagnosis. Theranostics 2022, 12, 2041–2062. [Google Scholar] [CrossRef]
  35. Frenkel, D.; Katz, O.; Solomon, B. Immunization against Alzheimer’s β-amyloid plaques via EFRH phage administration. Proc. Natl. Acad. Sci. USA 2000, 97, 11455–11459. [Google Scholar] [CrossRef] [Green Version]
  36. Messing, J. Phage M13 for the treatment of Alzheimer and Parkinson disease. Gene 2016, 583, 85–89. [Google Scholar] [CrossRef]
  37. Chey, W.D.; Kurlander, J.; Eswaran, S. Irritable Bowel Syndrome. JAMA 2015, 313, 949. [Google Scholar] [CrossRef]
  38. Sharon, G.; Sampson, T.R.; Geschwind, D.H.; Mazmanian, S.K. The Central Nervous System and the Gut Microbiome. Cell 2016, 167, 915–932. [Google Scholar] [CrossRef] [Green Version]
  39. Halfvarson, J.; Brislawn, C.J.; Lamendella, R.; Vázquez-Baeza, Y.; Walters, W.A.; Bramer, L.M.; D’Amato, M.; Bonfiglio, F.; McDonald, D.; Gonzalez, A.; et al. Dynamics of the human gut microbiome in inflammatory bowel disease. Nat. Microbiol. 2017, 2, 1–15. [Google Scholar] [CrossRef] [Green Version]
  40. Liu, R.; Zhang, C.; Shi, Y.; Zhang, F.; Li, L.; Wang, X.; Ling, Y.; Fu, H.; Dong, W.; Shen, J.; et al. Dysbiosis of Gut Microbiota Associated with Clinical Parameters in Polycystic Ovary Syndrome. Front. Microbiol. 2017, 8, 324. [Google Scholar] [CrossRef]
  41. Wirbel, J.; Pyl, P.T.; Kartal, E.; Zych, K.; Kashani, A.; Milanese, A.; Fleck, J.S.; Voigt, A.Y.; Palleja, A.; Ponnudurai, R.; et al. Meta-analysis of fecal metagenomes reveals global microbial signatures that are specific for colorectal cancer. Nat. Med. 2019, 25, 679–689. [Google Scholar] [CrossRef] [Green Version]
  42. Bäckhed, F.; Ley, R.E.; Sonnenburg, J.L.; Peterson, D.A.; Gordon, J.I. Host-Bacterial Mutualism in the Human Intestine. Science 2005, 307, 1915–1920. [Google Scholar] [CrossRef] [Green Version]
  43. Łusiak-Szelachowska, M.; Weber-Dąbrowska, B.; Jończyk-Matysiak, E.; Wojciechowska, R.; Górski, A. Bacteriophages in the gastrointestinal tract and their implications. Gut. Pathog. 2017, 9, 1–5. [Google Scholar] [CrossRef] [Green Version]
  44. Górski, A.; Borysowski, J.; Miȩdzybrodzki, R. Bacteriophage Interactions with Epithelial Cells: Therapeutic Implications. Front. Microbiol. 2021, 11, 631161. [Google Scholar] [CrossRef]
  45. Hobbs, Z.; Abedon, S.T. Diversity of phage infection types and associated terminology: The problem with ‘Lytic or lysogenic’. FEMS Microbiol. Lett. 2016, 363, fnw047. [Google Scholar] [CrossRef] [Green Version]
  46. Reyes, A.; Haynes, M.; Hanson, N.; Angly, F.E.; Heath, A.C.; Rohwer, F.; Gordon, J.I. Viruses in the faecal microbiota of monozygotic twins and their mothers. Nature 2010, 466, 334–338. [Google Scholar] [CrossRef] [Green Version]
  47. Minot, S.; Bryson, A.; Chehoud, C.; Wu, G.D.; Lewis, J.D.; Bushman, F.D. Rapid evolution of the human gut virome. Proc. Natl. Acad. Sci. USA 2013, 110, 12450–12455. [Google Scholar] [CrossRef] [Green Version]
  48. Knowles, B.; Silveira, C.B.; Bailey, B.A.; Barott, K.; Cantu, V.A.; Cobián-Güemes, A.G.; Coutinho, F.H.; Dinsdale, E.A.; Felts, B.; Furby, K.A.; et al. Lytic to temperate switching of viral communities. Nature 2016, 531, 466–470. [Google Scholar] [CrossRef]
  49. Keen, E.C.; Dantas, G. Close Encounters of Three Kinds: Bacteriophages, Commensal Bacteria, and Host Immunity. Trends Microbiol. 2018, 26, 943–954. [Google Scholar] [CrossRef]
  50. O’Brien, A.D.; Newland, J.W.; Miller, S.F.; Holmes, R.K.; Smith, H.W.; Formal, S.B. Shiga-Like Toxin-Converting Phages from Escherichia coli Strains That Cause Hemorrhagic Colitis or Infantile Diarrhea. Science 1984, 226, 694–696. [Google Scholar] [CrossRef]
  51. Allué-Guardia, A.; García-Aljaro, C.; Muniesa, M. Bacteriophage-Encoding Cytolethal Distending Toxin Type V Gene Induced from Nonclinical Escherichia coli Isolates. Infect. Immun. 2011, 79, 3262–3272. [Google Scholar] [CrossRef] [Green Version]
  52. Penadés, J.R.; Chen, J.; Quiles-Puchalt, N.; Carpena, N.; Novick, R.P. Bacteriophage-mediated spread of bacterial virulence genes. Curr. Opin. Microbiol. 2015, 23, 171–178. [Google Scholar] [CrossRef]
  53. Colomer-Lluch, M.; Jofre, J.; Muniesa, M. Antibiotic Resistance Genes in the Bacteriophage DNA Fraction of Environmental Samples. PLoS ONE 2011, 6, e17549. [Google Scholar] [CrossRef] [Green Version]
  54. Haaber, J.; Leisner, J.J.; Cohn, M.T.; Catalan-Moreno, A.; Nielsen, J.B.; Westh, H.; Penadés, J.R.; Ingmer, H. Bacterial viruses enable their host to acquire antibiotic resistance genes from neighbouring cells. Nat. Commun. 2016, 7, 13333. [Google Scholar] [CrossRef] [Green Version]
  55. Muniesa, M.; García, A.; Miró, E.; Mirelis, B.; Prats, G.; Jofre, J.; Navarro, F. Bacteriophages and Diffusion of β-lactamase Genes. Emerg. Infect. Dis. 2004, 10, 1134–1137. [Google Scholar] [CrossRef]
  56. Ross, J.; Topp, E. Abundance of Antibiotic Resistance Genes in Bacteriophage following Soil Fertilization with Dairy Manure or Municipal Biosolids, and Evidence for Potential Transduction. Appl. Environ. Microbiol. 2015, 81, 7905–7913. [Google Scholar] [CrossRef] [Green Version]
  57. Colavecchio, A.; Cadieux, B.; Lo, A.; Goodridge, L.D. Bacteriophages Contribute to the Spread of Antibiotic Resistance Genes among Foodborne Pathogens of the Enterobacteriaceae Family—A Review. Front. Microbiol. 2017, 8, 1108. [Google Scholar] [CrossRef] [Green Version]
  58. Cieślik, M.; Bagińska, N.; Jończyk-Matysiak, E.; Węgrzyn, A.; Węgrzyn, G.; Górski, A. Temperate Bacteriophages—The Powerful Indirect Modulators of Eukaryotic Cells and Immune Functions. Viruses 2021, 13, 1013. [Google Scholar] [CrossRef]
  59. Basso, J.T.R.; Ankrah, N.Y.D.; Tuttle, M.J.; Grossman, A.S.; Sandaa, R.A.; Buchan, A. Genetically similar temperate phages form coalitions with their shared host that lead to niche-specific fitness effects. ISME J. 2020, 14, 1688–1700. [Google Scholar] [CrossRef] [Green Version]
  60. Obeng, N.; Pratama, A.A.; van Elsas, J.D. The Significance of Mutualistic Phages for Bacterial Ecology and Evolution. Trends Microbiol. 2016, 24, 440–449. [Google Scholar] [CrossRef]
  61. Manrique, P.; Bolduc, B.; Walk, S.T.; van der Oost, J.; de Vos, W.M.; Young, M.J. Healthy human gut phageome. Proc. Natl. Acad. Sci. USA 2016, 113, 10400–10405. [Google Scholar] [CrossRef] [Green Version]
  62. Barr, J.J.; Auro, R.; Furlan, M.; Whiteson, K.L.; Erb, M.L.; Pogliano, J.; Stotland, A.; Wolkowicz, R.; Cutting, A.S.; Doran, K.S.; et al. Bacteriophage adhering to mucus provide a non–host-derived immunity. Proc. Natl. Acad. Sci. USA 2013, 110, 10771–10776. [Google Scholar] [CrossRef] [Green Version]
  63. Norman, J.M.; Handley, S.A.; Baldridge, M.T.; Droit, L.; Liu, C.Y.; Keller, B.C.; Kambal, A.; Monaco, C.L.; Zhao, G.; Fleshner, P.; et al. Disease-Specific Alterations in the Enteric Virome in Inflammatory Bowel Disease. Cell 2015, 160, 447–460. [Google Scholar] [CrossRef] [Green Version]
  64. Lawrence, D.; Baldridge, M.T.; Handley, S.A. Phages and Human Health: More Than Idle Hitchhikers. Viruses 2019, 11, 587. [Google Scholar] [CrossRef] [Green Version]
  65. Barr, J.J.; Auro, R.; Sam-Soon, N.; Kassegne, S.; Peters, G.; Bonilla, N.; Jeremy, J.; Hatay, M.; Mourtada, S.; Bailey, B.; et al. Subdiffusive motion of bacteriophage in mucosal surfaces increases the frequency of bacterial encounters. Proc. Natl. Acad. Sci. USA 2015, 112, 13675–13680. [Google Scholar] [CrossRef] [Green Version]
  66. Barr, J.J.; Youle, M.; Rohwer, F. Innate and acquired bacteriophage-mediated immunity. Bacteriophage 2013, 3, e25857. [Google Scholar] [CrossRef] [Green Version]
  67. Dutilh, B.E.; Cassman, N.; McNair, K.; Sanchez, S.E.; Silva, G.G.Z.; Boling, L.; Barr, J.J.; Speth, D.R.; Seguritan, V.; Aziz, R.K.; et al. A highly abundant bacteriophage discovered in the unknown sequences of human faecal metagenomes. Nat. Commun. 2014, 5, 4498. [Google Scholar] [CrossRef] [Green Version]
  68. Guerin, E.; Shkoporov, A.; Stockdale, S.R.; Clooney, A.G.; Ryan, F.J.; Sutton, T.D.S.; Draper, L.A.; Gonzalez-Tortuero, E.; Ross, R.P.; Hill, C. Biology and Taxonomy of crAss-like Bacteriophages, the Most Abundant Virus in the Human Gut. Cell. Host. Microbe 2018, 24, 653–664. [Google Scholar] [CrossRef] [Green Version]
  69. Shkoporov, A.N.; Khokhlova, E.v.; Fitzgerald, C.B.; Stockdale, S.R.; Draper, L.A.; Ross, R.P.; Hill, C. ΦCrAss001 represents the most abundant bacteriophage family in the human gut and infects Bacteroides intestinalis. Nat. Commun. 2018, 9, 4781. [Google Scholar] [CrossRef] [Green Version]
  70. Zhu, Y.; Shang, J.; Peng, C.; Sun, Y. Phage family classification under Caudoviricetes: A review of current tools using the latest ICTV classification framework. Front. Microbiol. 2022, 13, 1032186. [Google Scholar] [CrossRef]
  71. Turner, D.; Shkoporov, A.N.; Lood, C.; Millard, A.D.; Dutilh, B.E.; Alfenas-Zerbini, P.; van Zyl, L.J.; Aziz, R.K.; Oksanen, H.M.; Poranen, M.M.; et al. Abolishment of morphology-based taxa and change to binomial species names: 2022 taxonomy update of the ICTV bacterial viruses subcommittee. Arch. Virol. 2023, 168, 74. [Google Scholar] [CrossRef]
  72. Shkoporov, A.N.; Clooney, A.G.; Sutton, T.D.S.; Ryan, F.J.; Daly, K.M.; Nolan, J.A.; McDonnell, S.A.; Khokhlova, E.V.; Draper, L.A.; Forde, A.; et al. The Human Gut Virome Is Highly Diverse, Stable, and Individual Specific. Cell Host Microbe 2019, 26, 527–541. [Google Scholar] [CrossRef]
  73. Creasy, A.; Rosario, K.; Leigh, B.; Dishaw, L.; Breitbart, M. Unprecedented Diversity of ssDNA Phages from the Family Microviridae. Detected within the Gut of a Protochordate Model Organism (Ciona robusta). Viruses 2018, 10, 404. [Google Scholar] [CrossRef] [Green Version]
  74. Manrique, P.; Dills, M.; Young, M. The Human Gut Phage Community and Its Implications for Health and Disease. Viruses 2017, 9, 141. [Google Scholar] [CrossRef] [Green Version]
  75. Kowalski, J.; Górska, R.; Cieślik, M.; Górski, A.; Jończyk-Matysiak, E. What Are the Potential Benefits of Using Bacteriophages in Periodontal Therapy? Antibiotics 2022, 11, 446. [Google Scholar] [CrossRef]
  76. Souza, B.M.; Preisser, T.M.; Pereira, V.B.; Zurita-Turk, M.; de Castro, C.P.; da Cunha, V.P.; de Oliveira, R.P.; Gomes-Santos, A.C.; de Faria, A.M.C.; Machado, D.C.C.; et al. Lactococcus lactis carrying the pValac eukaryotic expression vector coding for IL-4 reduces chemically-induced intestinal inflammation by increasing the levels of IL-10-producing regulatory cells. Microb. Cell Factories 2016, 15, 150. [Google Scholar] [CrossRef] [Green Version]
  77. Ma, Y.; You, X.; Mai, G.; Tokuyasu, T.; Liu, C. A human gut phage catalog correlates the gut phageome with type 2 diabetes. Microbiome 2018, 6, 24. [Google Scholar] [CrossRef] [Green Version]
  78. Santiago-Rodriguez, T.M.; Hollister, E.B. Human Virome and Disease: High-Throughput Sequencing for Virus Discovery, Identification of Phage-Bacteria Dysbiosis and Development of Therapeutic Approaches with Emphasis on the Human Gut. Viruses 2019, 11, 656. [Google Scholar] [CrossRef] [Green Version]
  79. Maronek, M.; Link, R.; Ambro, L.; Gardlik, R. Phages and Their Role in Gastrointestinal Disease: Focus on Inflammatory Bowel Disease. Cells 2020, 9, 1013. [Google Scholar] [CrossRef] [Green Version]
  80. Frank, D.N.; st. Amand, A.L.; Feldman, R.A.; Boedeker, E.C.; Harpaz, N.; Pace, N.R. Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proc. Natl. Acad. Sci. USA 2007, 104, 13780–13785. [Google Scholar] [CrossRef] [Green Version]
  81. Hoarau, G.; Mukherjee, P.K.; Gower-Rousseau, C.; Hager, C.; Chandra, J.; Retuerto, M.A.; Neut, C.; Vermeire, S.; Clemente, J.; Colombel, J.F.; et al. Bacteriome and Mycobiome Interactions Underscore Microbial Dysbiosis in Familial Crohn’s Disease. mBio 2016, 7, e01250-16. [Google Scholar] [CrossRef] [Green Version]
  82. Lloyd-Price, J.; Arze, C.; Ananthakrishnan, A.N.; Schirmer, M.; Avila-Pacheco, J.; Poon, T.W.; Andrews, E.; Ajami, N.K.; Bonham, K.S.; Brislawn, C.J.; et al. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature 2019, 569, 655–662. [Google Scholar] [CrossRef]
  83. Reyes, A.; Blanton, L.v.; Cao, S.; Zhao, G.; Manary, M.; Trehan, I.; Smith, M.I.; Wang, D.; Virgin, H.W.; Rohwer, F.; et al. Gut DNA viromes of Malawian twins discordant for severe acute malnutrition. Proc. Natl. Acad. Sci. USA 2015, 112, 11941–11946. [Google Scholar] [CrossRef] [Green Version]
  84. Jung, M.J.; Lee, J.; Shin, N.R.; Kim, M.S.; Hyun, D.W.; Yun, J.H.; Kim, P.S.; Whon, T.W.; Bae, J.W. Chronic Repression of mTOR Complex 2 Induces Changes in the Gut Microbiota of Diet-induced Obese Mice. Sci. Rep. 2016, 6, 30887. [Google Scholar] [CrossRef] [Green Version]
  85. Zou, S.; Fang, L.; Lee, M.H. Dysbiosis of gut microbiota in promoting the development of colorectal cancer. Gastroenterol. Rep. 2018, 6, gox031. [Google Scholar] [CrossRef] [Green Version]
  86. Cryan, J.F.; O’Riordan, K.J.; Cowan, C.S.M.; Sandhu, K.v.; Bastiaanssen, T.F.S.; Boehme, M.; Codagnone, M.G.; Cussotto, S.; Fulling, C.; Golubeva, A.V.; et al. The Microbiota-Gut-Brain Axis. Physiol. Rev. 2019, 99, 1877–2013. [Google Scholar] [CrossRef]
  87. Erny, D.; Hrabě de Angelis, A.L.; Jaitin, D.; Wieghofer, P.; Staszewski, O.; David, E.; Keren-Shaul, H.; Mahlakoiv, T.; Jakobshagen, K.; Buch, T.; et al. Host microbiota constantly control maturation and function of microglia in the CNS. Nat. Neurosci. 2015, 18, 965–977. [Google Scholar] [CrossRef]
  88. Clarke, G.; Grenham, S.; Scully, P.; Fitzgerald, P.; Moloney, R.D.; Shanahan, F.; Dinan, T.G.; Cryan, T.G. The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner. Mol. Psychiatry 2013, 18, 666–673. [Google Scholar] [CrossRef] [Green Version]
  89. Lyte, M. Vagal Pathways for Microbiome-Brain-Gut Axis Communication. In Microbial Endocrinology and the Microbiota-Gut-Brain Axis. Advances in Experimental Medicine and Biology; Springer: New York, NY, USA, 2014; Volume 817, pp. 3–24. [Google Scholar]
  90. Sharon, G.; Cruz, N.J.; Kang, D.W.; Gandal, M.J.; Wang, B.; Kim, Y.M.; Erika, M.; Zink, E.M.; Casey, C.P.; Taylor, B.C.; et al. Human Gut Microbiota from Autism Spectrum Disorder Promote Behavioral Symptoms in Mice. Cell 2019, 177, 1600–1618. [Google Scholar] [CrossRef] [Green Version]
  91. Martin, C.R.; Osadchiy, V.; Kalani, A.; Mayer, E.A. The Brain-Gut-Microbiome Axis. Cell. Mol. Gastroenterol. Hepatol. 2018, 6, 133–148. [Google Scholar] [CrossRef] [Green Version]
  92. Yoo, B.B.; Mazmanian, S.K. The Enteric Network: Interactions between the Immune and Nervous Systems of the Gut. Immunity 2017, 46, 910–926. [Google Scholar] [CrossRef] [Green Version]
  93. Sasselli, V.; Pachnis, V.; Burns, A.J. The enteric nervous system. Dev. Biol. 2012, 366, 64–73. [Google Scholar] [CrossRef] [Green Version]
  94. Rhee, S.H.; Pothoulakis, C.; Mayer, E.A. Principles and clinical implications of the brain–gut–enteric microbiota axis. Nat. Rev. Gastroenterol. Hepatol. 2009, 6, 306–314. [Google Scholar] [CrossRef] [Green Version]
  95. Grenham, S.; Clarke, G.; Cryan, J.F.; Dinan, T.G. Brain-Gut-Microbe Communication in Health and Disease. Front. Physiol. 2011, 2, 94. [Google Scholar] [CrossRef] [Green Version]
  96. Eisenstein, M. Microbiome: Bacterial broadband. Nature 2016, 533, S104–S106. [Google Scholar] [CrossRef]
  97. Cong, X.; Henderson, W.A.; Graf, J.; McGrath, J.M. Early Life Experience and Gut Microbiome. Adv. Neonatal Care 2015, 15, 314–323. [Google Scholar] [CrossRef] [Green Version]
  98. Dalile, B.; van Oudenhove, L.; Vervliet, B.; Verbeke, K. The role of short-chain fatty acids in microbiota–gut–brain communication. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 461–478. [Google Scholar] [CrossRef]
  99. Lyte, M. Microbial Endocrinology in the Microbiome-Gut-Brain Axis: How Bacterial Production and Utilization of Neurochemicals Influence Behavior. PLoS Pathog. 2013, 9, e1003726. [Google Scholar] [CrossRef]
  100. Holzer, P.; Farzi, A. Neuropeptides and the Microbiota-Gut-Brain Axis. Advances in Experimental Medicine and Biology; Springer: New York, NY, USA, 2014; Volume 817, pp. 195–219. [Google Scholar]
  101. Wong, M.L.; Inserra, A.; Lewis, M.D.; Mastronardi, C.A.; Leong, L.; Choo, J.; Kentish, S.; Xie, P.; Morrison, M.; Wesselingh, S.L.; et al. Inflammasome signaling affects anxiety- and depressive-like behavior and gut microbiome composition. Mol. Psychiatry 2016, 21, 797–805. [Google Scholar] [CrossRef] [Green Version]
  102. Oh, J.H.; Alexander, L.M.; Pan, M.; Schueler, K.L.; Keller, M.P.; Attie, A.D.; Walter, J.; van Pijkeren, J.P. Dietary Fructose and Microbiota-Derived Short-Chain Fatty Acids Promote Bacteriophage Production in the Gut Symbiont Lactobacillus reuteri. Cell. Host. Microbe 2019, 25, 273–284. [Google Scholar] [CrossRef] [Green Version]
  103. Sudo, N.; Chida, Y.; Aiba, Y.; Sonoda, J.; Oyama, N.; Yu, X.N.; Kubo, C.; Kogaet, Y. Postnatal microbial colonization programs the hypothalamic-pituitary-adrenal system for stress response in mice. J. Physiol. 2004, 558, 263–275. [Google Scholar] [CrossRef]
  104. Strandwitz, P.; Kim, K.H.; Terekhova, D.; Liu, J.K.; Sharma, A.; Levering, J.; McDonald, D.; Dietrich, D.; Ramadhar, T.R.; Lekbua, A.; et al. GABA-modulating bacteria of the human gut microbiota. Nat. Microbiol. 2018, 4, 396–403. [Google Scholar] [CrossRef]
  105. Barrett, R.; Zhang, X.; Koon, H.W.; Vu, M.; Chang, J.Y.; Yeager, N.; Nguyen, M.A.; Michelsen, K.S.; Berel, D.; Pothoulakis, C.; et al. Constitutive TL1A Expression under Colitogenic Conditions Modulates the Severity and Location of Gut Mucosal Inflammation and Induces Fibrostenosis. Am. J. Pathol. 2012, 180, 636–649. [Google Scholar] [CrossRef] [Green Version]
  106. Yano, J.M.; Yu, K.; Donaldson, G.P.; Shastri, G.G.; Ann, P.; Ma, L.; Nagler, C.R.; Mazmanian, S.K.; Ismagilov, R.F.; Hsiao, E.Y. Indigenous Bacteria from the Gut Microbiota Regulate Host Serotonin Biosynthesis. Cell 2015, 161, 264–276. [Google Scholar] [CrossRef] [Green Version]
  107. Poutahidis, T.; Cappelle, K.; Levkovich, T.; Lee, C.W.; Doulberis, M.; Ge, Z.; Fox, J.G.; Horwitz, B.H.; Erdmanet, S.E. Pathogenic Intestinal Bacteria Enhance Prostate Cancer Development via Systemic Activation of Immune Cells in Mice. PLoS ONE 2013, 8, e73933. [Google Scholar] [CrossRef] [Green Version]
  108. Boonstra, E.; de Kleijn, R.; Colzato, L.S.; Alkemade, A.; Forstmann, B.U.; Nieuwenhuis, S. Neurotransmitters as food supplements: The effects of GABA on brain and behavior. Front. Psychol. 2015, 6, 1520. [Google Scholar] [CrossRef] [Green Version]
  109. Zheng, D.; Li, R.; An, J.; Xie, T.; Han, Z.; Xu, R.; Fang, Y.; Zhang, X.Z. Prebiotics-Encapsulated Probiotic Spores Regulate Gut Microbiota and Suppress Colon Cancer. Adv. Mater. 2020, 32, e2004529. [Google Scholar] [CrossRef]
  110. Abdel-Haq, R.; Schlachetzki, J.C.M.; Glass, C.K.; Mazmanian, S.K. Microbiome–microglia connections via the gut–brain axis. J. Exp. Med. 2019, 216, 41–59. [Google Scholar] [CrossRef] [Green Version]
  111. Mayer, E.A.; Knight, R.; Mazmanian, S.K.; Cryan, J.F.; Tillisch, K. Gut Microbes and the Brain: Paradigm Shift in Neuroscience. J. Neurosci. 2014, 34, 15490–15496. [Google Scholar] [CrossRef] [Green Version]
  112. Powell, N.; Walker, M.M.; Talley, N.J. The mucosal immune system: Master regulator of bidirectional gut–brain communications. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 143–159. [Google Scholar] [CrossRef]
  113. Cekanaviciute, E.; Yoo, B.B.; Runia, T.F.; Debelius, J.W.; Singh, S.; Nelson, C.A.; Kanner, R.; Bencosme, Y.; Lee, Y.K.; Hauser, S.L.; et al. Gut bacteria from multiple sclerosis patients modulate human T cells and exacerbate symptoms in mouse models. Proc. Natl. Acad. Sci. USA 2017, 114, 10713–10718. [Google Scholar] [CrossRef] [Green Version]
  114. Ambrosini, Y.M.; Borcherding, D.; Kanthasamy, A.; Kim, H.J.; Willette, A.A.; Jergens, A.; Allenspach, K.; Mochel, J.P. The Gut-Brain Axis in Neurodegenerative Diseases and Relevance of the Canine Model: A Review. Front. Aging Neurosci. 2019, 11, 130. [Google Scholar] [CrossRef]
  115. Hong, D.; Zhang, C.; Wu, W.; Lu, X.; Zhang, L. Modulation of the gut-brain axis via the gut microbiota: A new era in treatment of amyotrophic lateral sclerosis. Front. Neurol. 2023, 14, 1133546. [Google Scholar] [CrossRef]
  116. Darch, H.; McCafferty, C.P. Gut microbiome effects on neuronal excitability & activity: Implications for epilepsy. Neurobiol. Dis. 2022, 165, 105629. [Google Scholar]
  117. Isaiah, S.; Loots, D.T.; Solomons, R.; van der Kuip, M.; Tutu Van Furth, A.M.; Mason, S. Overview of brain-to-gut axis exposed to chronic CNS bacterial infection(s) and a predictive urinary metabolic profile of a brain infected by Mycobacterium tuberculosis. Front. Neurosci. 2020, 14, 296. [Google Scholar] [CrossRef] [Green Version]
  118. An, M.; Gao, Y. Urinary Biomarkers of Brain Diseases. Genom. Proteom. Bioinform. 2015, 13, 345–354. [Google Scholar] [CrossRef] [Green Version]
  119. Emwas, A.H.; Luchinat, C.; Turano, P.; Tenori, L.; Roy, R.; Salek, R.M.; Ryan, D.; Merzaban, J.S.; Kaddurah-Daouk, R.; Zeri, A.C.; et al. Standardizing the experimental conditions for using urine in NMR-based metabolomic studies with a particular focus on diagnostic studies: A review. Metabolomics 2015, 11, 872–894. [Google Scholar] [CrossRef] [Green Version]
  120. Matsuyama, W.; Hashiguchi, T.; Umehara, F.; Matsuura, E.; Kawabata, M.; Arimura, K.; Maruyama, I.; Osame, M. Expression of vascular endothelial growth factor in tuberculous meningitis. J. Neurol. Sci. 2001, 186, 75–79. [Google Scholar] [CrossRef]
  121. de Visser, V.; Sotgiu, G.; Lange, C.; Aabye, M.G.; Bakker, M.; Bartalesi, F.; Brat, K.; Chee, C.B.E.; Dheda, K.; Dominguez, J.; et al. False-negative interferon-γ release assay results in active tuberculosis: A TBNET study. Eur. Respir. J. 2015, 45, 279–283. [Google Scholar] [CrossRef] [Green Version]
  122. Mansour, A.M.; Frenck, R.W.; Darville, T.; Nakhla, I.A.; Wierzba, T.F.; Sultan, Y.; Bassiouny, M.I.; McCarthy, K.; Jacobs, R.F. Relationship between Intracranial Granulomas and Cerebrospinal Fluid Levels of Gamma Interferon and Interleukin-10 in Patients with Tuberculous Meningitis. Clin. Vaccine Immunol. 2005, 12, 363–365. [Google Scholar] [CrossRef] [Green Version]
  123. Górski, A.; Weber-Dabrowska, B. The potential role of endogenous bacteriophages in controlling invading pathogens. CMLS 2005, 62, 511–519. [Google Scholar] [CrossRef]
  124. Górski, A.; Wazna, E.; Weber-Dabrowska, B.; Dabrowska, K.; Switała-Jeleń, K.; Miedzybrodzki, R. Bacteriophage translocation. FEMS Immunol. Med. Microbiol. 2006, 46, 313–319. [Google Scholar] [CrossRef]
  125. Miernikiewicz, P.; Dąbrowska, K. Endocytosis of Bacteriophages. Curr. Opin. Virol. 2022, 52, 229–235. [Google Scholar] [CrossRef]
  126. Duerkop, B.A.; Hooper, L.V. Resident viruses and their interactions with the immune system. Nat. Immunol. 2013, 14, 654–659. [Google Scholar] [CrossRef]
  127. Rees, J.C.; Barr, J.R. Detection of methicillin-resistant Staphylococcus aureus using phage amplification combined with matrix-assisted laser desorption/ionization mass spectrometry. Anal. Bioanal. Chem. 2017, 409, 1379–1386. [Google Scholar] [CrossRef] [Green Version]
  128. Mayneris-Perxachs, J.; Castells-Nobau, A.; Arnoriaga-Rodríguez, M.; Garre-Olmo, J.; Puig, J.; Ramos, R.; Martínez-Hernández, F.; Burokas, A.; Coll, C.; Moreno-Navarrete, J.M.; et al. Caudovirales bacteriophages are associated with improved executive function and memory in flies, mice, and humans. Cell. Host. Microbe 2022, 30, 340–356.e8. [Google Scholar] [CrossRef]
  129. McMorris, T. History of Research into the Acute Exercise–Cognition Interaction. In Exercise-Cognition Interaction; Academic Press Elsevier: London, UK; Oxford, UK; San Diego, CA, USA; Waltham, MA, USA, 2016; pp. 1–28. [Google Scholar]
  130. Ciolek, C.H.; Lee, S.Y. Cognitive Issues in the Older Adults. In Guccione’s Geriatric Physical Therapy; Mosby: Maryland Heights, MI, USA, 2020; pp. 425–452. ISBN 9780323609128. [Google Scholar]
  131. Mayneris-Perxachs, J.; Arnoriaga-Rodríguez, M.; Luque-Córdoba, D.; Priego-Capote, F.; Pérez-Brocal, V.; Moya, A.; Burokas, A.; Maldonado, R.; Fernández-Real, J.M. Gut microbiota steroid sexual dimorphism and its impact on gonadal steroids: Influences of obesity and menopausal status. Microbiome 2020, 8, 136. [Google Scholar] [CrossRef]
  132. Wahlstrom, D.; Lawrence, G.; Weiss, L.G.; Donald, H.; Saklofske, D.H. Practical Issues in the WISC-V Administration and Scoring. In WISC-V Assessment and Interpretation: Scientist-Practitioner Perspectives; Elsevier Academic Press: Amsterdam, The Netherlands, 2016; pp. 25–62. [Google Scholar]
  133. Scarpina, F.; Tagini, S. The Stroop Color and Word Test. Front. Psychol. 2017, 8, 557. [Google Scholar] [CrossRef] [Green Version]
  134. Page, K.A.; Williamson, A.; Yu, N.; McNay, E.C.; Dzuira, J.; McCrimmon, R.J.; Sherwin, R.S. Medium-Chain Fatty Acids Improve Cognitive Function in Intensively Treated Type 1 Diabetic Patients and Support In Vitro Synaptic Transmission During Acute Hypoglycemia. Diabetes 2009, 58, 1237–1244. [Google Scholar] [CrossRef] [Green Version]
  135. Wang, D.; Mitchell, E.S. Cognition and Synaptic-Plasticity Related Changes in Aged Rats Supplemented with 8- and 10-Carbon Medium Chain Triglycerides. PLoS ONE 2016, 11, e0160159. [Google Scholar] [CrossRef] [Green Version]
  136. Howe, A.; Ringus, D.L.; Williams, R.J.; Choo, Z.N.; Greenwald, S.M.; Owens, S.M.; Coleman, M.L.; Meyer, F.; Chang, E.B. Divergent responses of viral and bacterial communities in the gut microbiome to dietary disturbances in mice. ISME J. 2016, 10, 1217–1227. [Google Scholar] [CrossRef] [Green Version]
  137. Duerkop, B.A.; Clements, C.v.; Rollins, D.; Rodrigues, J.L.M.; Hooper, L.v. A composite bacteriophage alters colonization by an intestinal commensal bacterium. Proc. Natl. Acad. Sci. USA 2012, 109, 17621–17626. [Google Scholar]
  138. Drevets, D.A.; Leenen, P.J.M.; Greenfield, R.A. Invasion of the Central Nervous System by Intracellular Bacteria. Clin. Microbiol. Rev. 2004, 17, 323–347. [Google Scholar] [CrossRef] [Green Version]
  139. John, C.C.; Carabin, H.; Montano, S.M.; Bangirana, P.; Zunt, J.R.; Peterson, P.K. Global research priorities for infections that affect the nervous system. Nature 2015, 527, S178–S186. [Google Scholar] [CrossRef]
  140. Robertson, F.C.; Lepard, J.R.; Mekary, R.A.; Davis, M.C.; Yunusa, I.; Gormley, W.B.; Baticulon, R.E.; Mahmud, M.R.; Misra, B.K.; Rattani, A.; et al. Epidemiology of central nervous system infectious diseases: A meta-analysis and systematic review with implications for neurosurgeons worldwide. J. Neurosurg. 2019, 130, 1107–1126. [Google Scholar] [CrossRef] [Green Version]
  141. Slane, V.H.; Unakal, C.G. Tuberculous Meningitis. [Updated 2022 Nov 18]. In StatPearls [Internet]; StatPearls Publishing: Treasure Island, FL, USA, 2023. Available online: https://www.ncbi.nlm.nih.gov/books/NBK541015/ (accessed on 10 February 2023).
  142. van de Beek, D.; Cabellos, C.; Dzupova, O.; Esposito, S.; Klein, M.; Kloek, A.T.; Leib, S.L.; Mourvillier, B.; Ostergaard, C.; Pagliano, P.; et al. ESCMID guideline: Diagnosis and treatment of acute bacterial meningitis. Clin. Microbiol. Infect. 2016, 22, S37–S62. [Google Scholar] [CrossRef] [Green Version]
  143. Dando, S.J.; Mackay-Sim, A.; Norton, R.; Currie, B.J.; st. John, J.A.; Ekberg, J.A.K.; Batzloff, M.; Ulett, G.C.; Beacham, I.R. Pathogens Penetrating the Central Nervous System: Infection Pathways and the Cellular and Molecular Mechanisms of Invasion. Clin. Microbiol. Rev. 2014, 27, 691–726. [Google Scholar] [CrossRef] [Green Version]
  144. Bauer, K.C.; Huus, K.E.; Finlay, B.B. Microbes and the mind: Emerging hallmarks of the gut microbiota-brain axis. Cell. Microbiol. 2016, 18, 632–644. [Google Scholar] [CrossRef] [Green Version]
  145. Le Govic, Y.; Demey, B.; Cassereau, J.; Bahn, Y.S.; Papon, N. Pathogens infecting the central nervous system. PLoS Pathog. 2022, 18, e1010234. [Google Scholar] [CrossRef]
  146. DiSabato, D.J.; Quan, N.; Godbout, J.P. Neuroinflammation: The devil is in the details. J. Neurochem. 2016, 139, 136–153. [Google Scholar] [CrossRef] [Green Version]
  147. Geyer, S.; Jacobs, M.; Hsu, N.J. Immunity Against Bacterial Infection of the Central Nervous System: An Astrocyte Perspective. Front. Mol. Neurosci. 2019, 12, 57. [Google Scholar] [CrossRef] [Green Version]
  148. Kim, K.S. Mechanisms of microbial traversal of the blood–brain barrier. Nat. Rev. Microbiol. 2008, 6, 625–634. [Google Scholar] [CrossRef] [Green Version]
  149. van Sorge, N.M.; Doran, K.S. Defense at the border: The blood–brain barrier versus bacterial foreigners. Future. Microbiol. 2012, 7, 383–394. [Google Scholar] [CrossRef] [Green Version]
  150. Miner, J.J.; Diamond, M.S. Mechanisms of restriction of viral neuroinvasion at the blood–brain barrier. Curr. Opin. Immunol. 2016, 38, 18–23. [Google Scholar] [CrossRef]
  151. Koyuncu, O.O.; Hogue, I.B.; Enquist, L.W. Virus Infections in the Nervous System. Cell. Host. Microbe 2013, 13, 379–393. [Google Scholar] [CrossRef] [Green Version]
  152. Kuehn, M.; Bridget, M. CDC Establishes Global Networks to Combat Antimicrobial Resistance. JAMA 2022, 327, 315. [Google Scholar] [CrossRef]
  153. Yu, P.; Liao, H.; Wang, N.; Goel, R.K. Harness the power of bacteriophages to combat the global antimicrobial resistance Front. Microbiol. Sec. Phage Biol. 2023, 14. [Google Scholar]
  154. Miethke, M.; Pieroni, M.; Weber, T.; Brönstrup, M.; Hammann, P.; Halby, L.; Arimondo, P.B.; Glaser, P.; Aigle, B.; Bode, H.B.; et al. Towards the sustainable discovery and development of new antibiotics. Nat. Rev. Chem. 2021, 5, 726–749. [Google Scholar] [CrossRef]
  155. Dubos, R.J.; Straus, J.H.; Pierce, C. the multiplication of bacteriophage in vivo and its protective effect against an experimental infection with Shigella dysenteriae. J. Exp. Med. 1943, 78, 161–168. [Google Scholar] [CrossRef]
  156. Smith, H.W.; Huggins, M.B. Successful Treatment of Experimental Escherichia coli Infections in Mice Using Phage: Its General Superiority over Antibiotics. Microbiology 1982, 128, 307–318. [Google Scholar] [CrossRef] [Green Version]
  157. Barrow, P.; Lovell, M.; Berchieri, A. Use of Lytic Bacteriophage for Control of Experimental Escherichia coli Septicemia and Meningitis in Chickens and Calves. Clin. Diagn. Lab. Immunol. 1998, 5, 294–298. [Google Scholar] [CrossRef] [Green Version]
  158. Proquot, M.; Jamal, L.N.; Plouzeau-Jayle, C.; Michaud, A.; Broutin, L.; Burucoa, C.; Cremniter, J.; Pichon, M. K1 Antigen Is Associated with Different AST Profile in Escherichia coli: A One-Month-Long Pilot Study. Microorganisms 2021, 9, 1884. [Google Scholar] [CrossRef]
  159. Gaschignard, J.; Levy, C.; Romain, O.; Cohen, R.; Bingen, E.; Aujard, Y.; Boileau, P. Neonatal Bacterial Meningitis. Pediatr. Infect. Dis. J. 2011, 30, 212–217. [Google Scholar] [CrossRef]
  160. Barichello, T.; Fagundes, G.D.; Generoso, J.S.; Elias, S.G.; Simões, L.R.; Teixeira, A.L. Pathophysiology of neonatal acute bacterial meningitis. J. Med. Microbiol. 2013, 62, 1781–1789. [Google Scholar] [CrossRef] [Green Version]
  161. Hoffman, O.; Weber, J.R. Review: Pathophysiology and treatment of bacterial meningitis. Ther. Adv. Neurol. Disord. 2009, 2, 401–412. [Google Scholar] [CrossRef] [Green Version]
  162. Bingen, E.; Picard, B.; Brahimi, N.; Mathy, S.; Desjardins, P.; Elion, J.; Denamur, E. Phylogenetic Analysis of Escherichia coli Strains Causing Neonatal Meningitis Suggests Horizontal Gene Transfer from a Predominant Pool of Highly Virulent B2 Group Strains. J. Infect. Dis. 1998, 177, 642–650. [Google Scholar] [CrossRef] [Green Version]
  163. Scholl, D.; Adhya, S.; Merril, C. Escherichia coli K1’s Capsule Is a Barrier to Bacteriophage T7. Appl. Environ. Microbiol. 2005, 71, 4872–4874. [Google Scholar] [CrossRef] [Green Version]
  164. Guo, M.; Feng, C.; Ren, J.; Zhuang, X.; Zhang, Y.; Zhu, Y.; Dong, K.; He, P.; Guo, X.; Qin, J. A Novel Antimicrobial Endolysin, LysPA26, against Pseudomonas aeruginosa. Front. Microbiol. 2017, 8, 293. [Google Scholar] [CrossRef] [Green Version]
  165. Lim, J.A.; Shin, H.; Heu, S.; Ryu, S. Exogenous Lytic Activity of SPN9CC Endolysin Against Gram-Negative Bacteria. J. Microbiol. Biotechnol. 2014, 24, 803–811. [Google Scholar] [CrossRef]
  166. Kim, S.; Lee, D.W.; Jin, J.S.; Kim, J. Antimicrobial activity of LysSS, a novel phage endolysin, against Acinetobacter baumannii and Pseudomonas aeruginosa. J. Glob. Antimicrob. Resist. 2020, 22, 32–39. [Google Scholar] [CrossRef]
  167. Antonova, N.; Vasina, D.; Rubalsky, E.; Fursov, M.; Savinova, A.; Grigoriev, I.; Usachev, E.V.; Shevlyagina, N.V.; Zhukhovitsky, V.G.; Balabanyan, V.U.; et al. Modulation of Endolysin LysECD7 Bactericidal Activity by Different Peptide Tag Fusion. Biomolecules 2020, 10, 440. [Google Scholar] [CrossRef] [Green Version]
  168. Antonova, N.; Vasina, D.; Lendel, A.; Usachev, E.; Makarov, V.; Gintsburg, A.; Tkachuk, A.P.; Gushchin, V.A. Broad Bactericidal Activity of the Myoviridae Bacteriophage Lysins LysAm24, LysECD7, and LysSi3 against Gram-Negative ESKAPE Pathogens. Viruses 2019, 11, 284. [Google Scholar] [CrossRef] [Green Version]
  169. Gontijo, M.T.P.; Vidigal, P.M.P.; Lopez, M.E.S.; Brocchi, M. Bacteriophages that infect Gram-negative bacteria as source of signal-arrest-release motif lysins. Res. Microbiol. 2021, 172, 103794. [Google Scholar] [CrossRef]
  170. Ghose, C.; Euler, C.W. Gram-Negative Bacterial Lysins. Antibiotics 2020, 9, 74. [Google Scholar] [CrossRef] [Green Version]
  171. Love, M.J.; Abeysekera, G.S.; Muscroft-Taylor, A.C.; Billington, C.; Dobson, R.C.J. On the catalytic mechanism of bacteriophage endolysins: Opportunities for engineering. Biochim. Et Biophys. Acta (BBA)-Proteins Proteom. 2020, 1868, 140302. [Google Scholar] [CrossRef]
  172. Murray, E.; Draper, L.A.; Ross, R.P.; Hill, C. The Advantages and Challenges of Using Endolysins in a Clinical Setting. Viruses 2021, 13, 680. [Google Scholar] [CrossRef]
  173. Oliveira, H.; Vilas Boas, D.; Mesnage, S.; Kluskens, L.D.; Lavigne, R.; Sillankorva, S.; Secundo, F.; Azeredo, J. Structural and Enzymatic Characterization of ABgp46, a Novel Phage Endolysin with Broad Anti-Gram-Negative Bacterial Activity. Front. Microbiol. 2016, 7, 208. [Google Scholar] [CrossRef] [Green Version]
  174. Gontijo, M.T.P.; Jorge, G.P.; Brocchi, M. Current Status of Endolysin-Based Treatments against Gram-Negative Bacteria. Antibiotics 2021, 10, 1143. [Google Scholar] [CrossRef]
  175. Mondal, S.I.; Draper, L.A.; Ross, R.P.; Hill, C. Bacteriophage endolysins as a potential weapon to combat Clostridioides difficile infection. Gut Microbes 2020, 12, 1813533. [Google Scholar] [CrossRef]
  176. Harhala, M.; Nelson, D.C.; Miernikiewicz, P.; Heselpoth, R.D.; Brzezicka, B.; Majewska, J.; Linden, S.B.; Shang, X.; Szymczak, A.; Lecion, D.; et al. Safety Studies of Pneumococcal Endolysins Cpl-1 and Pal. Viruses 2018, 10, 638. [Google Scholar] [CrossRef] [Green Version]
  177. Gerstmans, H.; Grimon, D.; Gutiérrez, D.; Lood, C.; Rodríguez, A.; van Noort, V.; Lammertyn, J.; Lavigne, R.R.; Briers, Y. A Versatile-driven platform for rapid hit-to-lead development of engineered lysins. Sci. Adv. 2020, 6, eaaz1136. [Google Scholar] [CrossRef]
  178. Grandgirard, D.; Loeffler, J.M.; Fischetti, V.A.; Leib, S.L. Phage Lytic Enzyme Cpl-1 for Antibacterial Therapy in Experimental Pneumococcal Meningitis. J. Infect. Dis. 2008, 197, 1519–1522. [Google Scholar] [CrossRef] [Green Version]
  179. Entenza, J.M.; Loeffler, J.M.; Grandgirard, D.; Fischetti, V.A.; Moreillon, P. Therapeutic Effects of Bacteriophage Cpl-1 Lysin against Streptococcus pneumoniae Endocarditis in Rats. Antimicrob. Agents Chemother. 2005, 49, 4789–4792. [Google Scholar] [CrossRef] [Green Version]
  180. Witzenrath, M.; Schmeck, B.; Doehn, J.M.; Tschernig, T.; Zahlten, J.; Loeffler, J.M.; Zemlin, M.; Müller, H.; Gutbier, B.; Schütte, H.; et al. Systemic use of the endolysin Cpl-1 rescues mice with fatal pneumococcal pneumonia*. Crit. Care Med. 2009, 37, 642–649. [Google Scholar] [CrossRef]
  181. Jado, I. Phage lytic enzymes as therapy for antibiotic-resistant Streptococcus pneumoniae infection in a murine sepsis model. J. Antimicrob. Chemother. 2003, 52, 967–973. [Google Scholar] [CrossRef] [Green Version]
  182. Loeffler, J.M.; Djurkovic, S.; Fischetti, V.A. Phage Lytic Enzyme Cpl-1 as a Novel Antimicrobial for Pneumococcal Bacteremia. Infect. Immun. 2003, 71, 6199–6204. [Google Scholar] [CrossRef] [Green Version]
  183. Van Belleghem, J.D.; Clement, F.; Merabishvili, M.; Lavigne, R.; Vaneechoutte, M. Pro- and anti-inflammatory responses of peripheral blood mononuclear cells induced by Staphylococcus aureus and Pseudomonas aeruginosa phages. Sci. Rep. 2017, 7, 8004. [Google Scholar] [CrossRef] [Green Version]
  184. Saccà, F.; Pane, C.; Espinosa, P.E.; Sormani, M.P.; Signori, A. Efficacy of innovative therapies in myasthenia gravis: A systematic review, meta-analysis and network meta-analysis. Eur. J. Neurol. 2023. Epub ahead of print. [Google Scholar] [CrossRef]
  185. Coutinho Costa, V.G.; Araújo, S.E.S.; Alves-Leon, S.V.; Gomes, F.C.A. Central nervous system demyelinating diseases: Glial cells at the hub of pathology. Front. Immunol. 2023, 14, 1135540. [Google Scholar] [CrossRef]
  186. Górski, A.; Międzybrodzki, R.; Jończyk-Matysiak, E.; Żaczek, M.; Borysowski, J. Phage-specific diverse effects of bacterial viruses on the immune system. Future Microbiol. 2019, 14, 1171–1174. [Google Scholar] [CrossRef] [Green Version]
  187. Grech, N.; Dalli, T.; Mizzi, S.; Meilak, L.; Calleja, N.; Zrinzo, A. Rising Incidence of Glioblastoma Multiforme in a Well-Defined Population. Cureus 2020, 12, e8195. [Google Scholar] [CrossRef]
  188. Inada, M.; Tanimoto, T.; Murata, Y.; Kaneda, Y.; Fujieda, T. Letter to the editor: Caregiver burden by treatment and clinical characteristics of patients with glioblastoma. Support. Care Cancer 2023, 3, 1–2. [Google Scholar] [CrossRef]
  189. Lai, R.K.; Chen, Y.; Guan, X.; Nousome, D.; Sharma, C.; Canoll, P.; Jeffrey Bruce, J.; Sloan, A.E.; Cortes, E.; Vonsattel, J.P.; et al. Genome-Wide Methylation Analyses in Glioblastoma Multiforme. PLoS ONE 2014, 9, e89376. [Google Scholar] [CrossRef]
  190. Wirsching, H.G.; Weller, M. Glioblastoma. In Malignant Brain Tumors; Springer International Publishing: Cham, Switzerland, 2017; pp. 265–288. [Google Scholar]
  191. Sanai, N.; Tramontin, A.D.; Quiñones-Hinojosa, A.; Barbaro, N.M.; Gupta, N.; Kunwar, S.; Berger, S.; Alvarez-Buylla, A. Unique astrocyte ribbon in adult human brain contains neural stem cells but lacks chain migration. Nature 2004, 427, 740–744. [Google Scholar] [CrossRef]
  192. Nunes, M.C.; Roy, N.S.; Keyoung, H.M.; Goodman, R.R.; McKhann, G.; Jiang, L.; Kang, J.; Nedergaard, M.; Goldman, S.A. Identification and isolation of multipotential neural progenitor cells from the subcortical white matter of the adult human brain. Nat. Med. 2003, 9, 439–447. [Google Scholar] [CrossRef]
  193. Hellström, N.A.K.; Björk-Eriksson, T.; Blomgren, K.; Kuhn, H.G. Differential Recovery of Neural Stem Cells in the Subventricular Zone and Dentate Gyrus After Ionizing Radiation. Stem Cells 2009, 27, 634–641. [Google Scholar] [CrossRef]
  194. Sanai, N.; Alvarez-Buylla, A.; Berger, M.S. Neural Stem Cells and the Origin of Gliomas. N. Engl. J. Med. 2005, 353, 811–822. [Google Scholar] [CrossRef]
  195. le Rhun, E.; Preusser, M.; Roth, P.; Reardon, D.A.; van den Bent, M.; Wen, P.; Reifenberger, G.; Weller, M. Molecular targeted therapy of glioblastoma. Cancer Treat. Rev. 2019, 80, 101896. [Google Scholar] [CrossRef]
  196. Ricci-Vitiani, L.; Pallini, R.; Biffoni, M.; Todaro, M.; Invernici, G.; Cenci, T.; Maira, G.; Parati, E.A.; Stassi, G.; Larocca, L.M.; et al. Tumour vascularization via endothelial differentiation of glioblastoma stem-like cells. Nature 2010, 468, 824–828. [Google Scholar] [CrossRef]
  197. Wang, R.; Chadalavada, K.; Wilshire, J.; Kowalik, U.; Hovinga, K.E.; Geber, A.; Fligelman, B.; Leversha, M.; Brennan, C.; Tabar, V. Glioblastoma stem-like cells give rise to tumour endothelium. Nature 2010, 468, 829–833. [Google Scholar] [CrossRef]
  198. Cheng, L.; Huang, Z.; Zhou, W.; Wu, Q.; Donnola, S.; Liu, J.K.; Fang, X.; Sloan, A.E.; Mao, Y.; Lathia, J.D.; et al. Glioblastoma Stem Cells Generate Vascular Pericytes to Support Vessel Function and Tumor Growth. Cell 2013, 153, 139–152. [Google Scholar] [CrossRef] [Green Version]
  199. Bao, S.; Wu, Q.; McLendon, R.E.; Hao, Y.; Shi, Q.; Hjelmeland, A.B.; Dewhirst, M.W.; Bigner, D.D.; Rich, J.N. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature 2006, 444, 756–760. [Google Scholar] [CrossRef]
  200. Beier, D.; Röhrl, S.; Pillai, D.R.; Schwarz, S.; Kunz-Schughart, L.A.; Leukel, P.; Proescholdt, M.; Brawanski, A.; Bogdahn, U.; Trampe-Kieslich, A.; et al. Temozolomide Preferentially Depletes Cancer Stem Cells in Glioblastoma. Cancer Res. 2008, 68, 5706–5715. [Google Scholar] [CrossRef] [Green Version]
  201. Chen, J.; Li, Y.; Yu, T.S.; McKay, R.M.; Burns, D.K.; Kernie, S.G.; Parada, L.F. A restricted cell population propagates glioblastoma growth after chemotherapy. Nature 2012, 488, 522–526. [Google Scholar] [CrossRef] [Green Version]
  202. Potez, M.; Snedal, S.; She, C.; Kim, J.; Thorner, K.; Tran, T.H.; Ramello, M.C.; Abate-Daga, D.; Liu, J.K.C. Use of phage display biopanning as a tool to design CAR-T cells against glioma stem cells. Front. Oncol. 2023, 13, 1124272. [Google Scholar] [CrossRef]
  203. Moon, J.S.; Kim, W.G.; Kim, C.; Park, G.T.; Heo, J.; Yoo, S.Y.; Oh, J.W. M13 Bacteriophage-Based Self-Assembly Structures and Their Functional Capabilities. Mini. Rev. Org. Chem. 2015, 12, 271–281. [Google Scholar] [CrossRef] [Green Version]
  204. Kolhar, P.; Anselmo, A.C.; Gupta, V.; Pant, K.; Prabhakarpandian, B.; Ruoslahti, E.; Mitragotri, S. Using shape effects to target antibody-coated nanoparticles to lung and brain endothelium. Proc. Natl. Acad. Sci. USA 2013, 110, 10753–11078. [Google Scholar] [CrossRef] [Green Version]
  205. Yang, S.H.; Chung, W.J.; McFarland, S.; Lee, S.W. Assembly of Bacteriophage into Functional Materials. Chem. Rec. 2013, 13, 43–59. [Google Scholar] [CrossRef]
  206. Bakhshinejad, B.; Karimi, M.; Sadeghizadeh, M. Bacteriophages and medical oncology: Targeted gene therapy of cancer. Med Oncol. 2014, 31, 110. [Google Scholar] [CrossRef]
  207. Smith, G.P. Filamentous Fusion Phage: Novel Expression Vectors That Display Cloned Antigens on the Virion Surface. Science 1985, 228, 1315–1317. [Google Scholar] [CrossRef]
  208. Pierzynowska, K.; Morcinek-Orłowska, J.; Gaffke, L.; Jaroszewicz, W.; Skowron, P.M.; Węgrzyn, G. Applications of the phage display technology in molecular biology, biotechnology and medicine. Crit. Rev. Microbiol. 2023, 4, 2219741. [Google Scholar] [CrossRef]
  209. Petrov, G.; Dymova, M.; Richter, V. Bacteriophage-Mediated Cancer Gene Therapy. Int. J. Mol. Sci. 2022, 23, 14245. [Google Scholar] [CrossRef]
  210. Yan, X.; Xu, Z. Ribosome-display technology: Applications for directed evolution of functional proteins. Drug Discov. Today 2006, 11, 911–916. [Google Scholar] [CrossRef]
  211. Kunamneni, A.; Ogaugwu, C.; Bradfute, S.; Durvasula, R. Ribosome Display Technology: Applications in Disease Diagnosis and Control. Antibodies 2020, 9, 28. [Google Scholar] [CrossRef]
  212. Feldhaus, M.J.; Siegel, R.W. Yeast display of antibody fragments: A discovery and characterization platform. J. Immunol. Methods 2004, 290, 69–80. [Google Scholar] [CrossRef] [PubMed]
  213. Traxlmayr, M.W.; Obinger, C. Directed evolution of proteins for increased stability and expression using yeast display. Arch. Biochem. Biophys. 2012, 526, 174–180. [Google Scholar] [CrossRef]
  214. Kristensen, P.; Ravn, P.; Jensen, K.B.; Jensen, K. Applying phage display technology in aging research. Biogerontology 2000, 1, 67–78. [Google Scholar] [CrossRef]
  215. Majerova, P.; Hanes, J.; Olesova, D.; Sinsky, J.; Pilipcinec, E.; Kovac, A. Novel Blood-Brain Barrier Shuttle Peptides Discovered through the Phage Display Method. Molecules 2020, 25, 874. [Google Scholar] [CrossRef] [Green Version]
  216. Podlacha, M.; Grabowski, Ł.; Kosznik-Kawśnicka, K.; Zdrojewska, K.; Stasiłojć, M.; Węgrzyn, G.; Węgrzyn, A. Interactions of Bacteriophages with Animal and Human Organisms—Safety Issues in the Light of Phage Therapy. Int. J. Mol. Sci. 2021, 22, 8937. [Google Scholar] [CrossRef]
  217. Tian, Y.; Wu, M.; Liu, X.; Liu, Z.; Zhou, Q.; Niu, Z.; Huang, Y. Probing the Endocytic Pathways of the Filamentous Bacteriophage in Live Cells Using Ratiometric pH Fluorescent Indicator. Adv. Healthc. Mater. 2015, 4, 413. [Google Scholar] [CrossRef]
  218. Finne, J.; Leinonen, M.; Mäkelä, P.H. Antigenic similarities between brain components and bacteria causing meningitis. Implications for vaccine development and pathogenesis. Lancet 1983, 322, 355–357. [Google Scholar] [CrossRef]
  219. Lehti, T.A.; Pajunen, M.I.; Skog, M.S.; Finne, J. Internalization of a polysialic acid-binding Escherichia coli bacteriophage into eukaryotic neuroblastoma cells. Nat. Commun. 2017, 8, 1915. [Google Scholar] [CrossRef] [Green Version]
  220. Frenkel, D.; Solomon, B. Filamentous phage as vector-mediated antibody delivery to the brain. Proc. Natl. Acad. Sci. USA 2002, 99, 5675–5679. [Google Scholar] [CrossRef] [Green Version]
  221. Ksendzovsky, A.; Walbridge, S.; Saunders, R.C.; Asthagiri, A.R.; Heiss, J.D.; Lonser, R.R. Convection-enhanced delivery of M13 bacteriophage to the brain. J. Neurosurg. 2012, 117, 197–203. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  222. Mehta, A.; Sonabend, A.M.; Bruce, J.N. Convection-Enhanced Delivery. Neurotherapeutics 2017, 14, 358–371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  223. Pangalos, M.N.; Schechter, L.E.; Hurko, O. Drug development for CNS disorders: Strategies for balancing risk and reducing attrition. Nat. Rev. Drug. Discov. 2007, 6, 521–532. [Google Scholar] [CrossRef] [PubMed]
  224. Wang, Y.; Sheng, J.; Chai, J.; Zhu, C.; Li, X.; Yang, W.; Cui, R.; Ge, T. Filamentous Bacteriophage—A Powerful Carrier for Glioma Therapy. Front. Immunol. 2021, 12, 729336. [Google Scholar] [CrossRef] [PubMed]
  225. Okuyama, T.; Eto, Y.; Sakai, N.; Minami, K.; Yamamoto, T.; Sonoda, H.; Yamaoka, M.; Tachibana, K.; Hirato, T.; Sato, Y. Iduronate-2-Sulfatase with Anti-human Transferrin Receptor Antibody for Neuropathic Mucopolysaccharidosis II: A Phase 1/2 Trial. Mol. Ther. 2019, 27, 456–464. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  226. Giugliani, R.; Giugliani, L.; de Oliveira Poswar, F.; Donis, K.C.; Corte, A.D.; Schmidt, M.; Boado, R.J.; Nestrasil, I.; Nguyen, C.; Chen, S.; et al. Neurocognitive and somatic stabilization in pediatric patients with severe Mucopolysaccharidosis Type I after 52 weeks of intravenous brain-penetrating insulin receptor antibody-iduronidase fusion protein (valanafusp alpha): An open label phase 1-2 trial. Orphanet. J. Rare. Dis. 2018, 13, 110. [Google Scholar] [CrossRef]
  227. Bazan, J.; Całkosiński, I.; Gamian, A. Phage display--a powerful technique for immunotherapy: 1. Introduction and potential of therapeutic applications. Hum. Vaccin. Immunother. 2012, 8, 1817–1828. [Google Scholar] [CrossRef] [Green Version]
  228. Eriste, E.; Kurrikoff, K.; Suhorutšenko, J.; Oskolkov, N.; Copolovici, D.M.; Jones, S.; Laakkonen, P.; Howl, J.; Langel, Ü. Peptide-Based Glioma-Targeted Drug Delivery Vector gHoPe2. Bioconjug. Chem. 2013, 24, 305–313. [Google Scholar] [CrossRef] [PubMed]
  229. Lundin, P.; Johansson, H.; Guterstam, P.; Holm, T.; Hansen, M.; Langel, Ü.; Andaloussi, S. Distinct Uptake Routes of Cell-Penetrating Peptide Conjugates. Bioconjug. Chem. 2008, 19, 2535–2542. [Google Scholar] [CrossRef]
  230. Elmquist, A.; Hansen, M.; Langel, Ü. Structure–activity relationship study of the cell-penetrating peptide pVEC. Biochim. et Biophys. Acta (BBA)-Biomembr. 2006, 1758, 721–729. [Google Scholar] [CrossRef] [Green Version]
  231. Chen, L.; Zeng, D.; Xu, N.; Li, C.; Zhang, W.; Zhu, X.; Gao, Y.; Chen, P.R.; Lin, J. Blood–Brain Barrier- and Blood–Brain Tumor Barrier-Penetrating Peptide-Derived Targeted Therapeutics for Glioma and Malignant Tumor Brain Metastases. ACS Appl. Mater. Interfaces 2019, 11, 41889–41897. [Google Scholar] [CrossRef] [PubMed]
  232. Kim, J.M.; Marine, P.; Chunhua, S.; Ping, H.; Qiulian, W.; Shideng, B.; Rich, J.N.; Liu, J.K.C. CGlioblastoma Stem Cell Targeting Peptide Isolated Through Phage Display Binds Cadherin 2. Stem Cells 2023, sxad045. [Google Scholar] [CrossRef] [PubMed]
  233. Araste, F.; Ebrahimizadeh, W.; Rasooli, I.; Rajabibazl, M.; Mousavi, G.S.L. A novel VHH nanobody against the active site (the CA domain) of tumor-associated, carbonic anhydrase isoform IX and its usefulness for cancer diagnosis. Biotechnol. Lett. 2014, 36, 21–28. [Google Scholar] [CrossRef]
  234. Ruiz-López, E.; Jovčevska, I.; González-Gómez, R.; Tejero, H.; Al-Shahrour, F.; Muyldermans, S.; Schuhmacher, A.J. Nanobodies targeting ABCC3 for immunotargeted applications in glioblastoma. Sci. Rep. 2022, 12, 22581. [Google Scholar] [CrossRef] [PubMed]
  235. Wan, X.M.; Chen, Y.P.; Xu, W.R.; Yang, W.j.; Wen, L.P. Identification of nose-to-brain homing peptide through phage display. Peptides 2009, 30, 343–350. [Google Scholar] [CrossRef]
  236. Mathison, S.; Nagilla, R.; Kompella, U.B. Nasal Route for Direct Delivery of Solutes to the Central Nervous System: Fact or Fiction? J. Drug Target 1998, 5, 415–441. [Google Scholar] [CrossRef]
  237. Pavoni, E.; Vaccaro, P.; D’Alessio, V.; de Santis, R.; Minenkova, O. Simultaneous display of two large proteins on the head and tail of bacteriophage lambda. BMC Biotechnol. 2013, 13, 79. [Google Scholar] [CrossRef] [Green Version]
  238. Rajaram, K.; Vermeeren, V.; Somers, K.; Somers, V.; Michiels, L. Construction of helper plasmid-mediated dual-display phage for autoantibody screening in serum. Appl. Microbiol. Biotechnol. 2014, 98, 6365–6373. [Google Scholar] [CrossRef] [Green Version]
  239. Fagète, S.; Botas-Perez, L.; Rossito-Borlat, I.; Adea, K.; Gueneau, F.; Ravn, U.; Rousseau, F.; Kosco-Vilbois, M.; Fischer, N.; Hartley, O. Dual display: Phage selection driven by co-engagement of two targets by two different antibody fragments. PEDS 2017, 30, 575–582. [Google Scholar] [CrossRef]
  240. Carrera, M.R.; Kaufmann, G.F.; Mee, J.M.; Meijler, M.M.; Koob, G.F.; Janda, K.D. Treating cocaine addiction with viruses. Proc. Natl. Acad. Sci. USA 2004, 101, 10416–10421. [Google Scholar] [CrossRef] [Green Version]
  241. Deng, X.; Yuan, J.; Chen, L.; Chen, H.; Wei, C.; Nielsen, P.H.; Wuertz, S.; Qiu, G. CRISPR-Cas phage defense systems and prophages in Candidatus Accumulibacter. Water Res. 2023, 235, 119906. [Google Scholar] [CrossRef] [PubMed]
  242. Pons, B.J.; van Houte, S.; Westra, E.R.; Chevallereau, A. Ecology and evolution of phages encoding anti-CRISPR proteins. J. Mol. Biol. 2023, 435, 167974. [Google Scholar] [CrossRef] [PubMed]
  243. Marino, N.D. Phage Against the Machine: Discovery and Mechanism of Type V Anti-CRISPRs. J. Mol. Biol. 2023, 435, 168054. [Google Scholar] [CrossRef]
  244. Stanley, S.Y.; Maxwell, K.L. Phage-Encoded Anti-CRISPR Defenses. Annu. Rev. Genet. 2018, 52, 445–464. [Google Scholar] [CrossRef]
  245. Pausch, P.; Al-Shayeb, B.; Bisom-Rapp, E.; Tsuchida, C.A.; Li, Z.; Cress, B.F.; Knott, G.J.; Jacobsen, S.E.; Banfield, J.F.; Doudna, J.A. CRISPR-CasΦ from huge phages is a hypercompact genome editor. Science 2020, 369, 333–337. [Google Scholar] [CrossRef]
  246. Al-Shayeb, B.; Skopintsev, P.; Soczek, K.M.; Stahl, E.C.; Li, Z.; Groover, E.; Smock, D.; Eggers, A.R.; Pausch, P.; Cress, B.F.; et al. Diverse virus-encoded CRISPR-Cas systems include streamlined genome editors. Cell 2022, 185, 4574–4586. [Google Scholar] [CrossRef] [PubMed]
  247. Zhang, X.; Zhang, C.; Liang, C.; Li, B.; Meng, F.; Ai, Y. CRISPR–Cas9 Based Bacteriophage Genome Editing. Microbiol. Spectr. 2022, 10, 1–12. [Google Scholar] [CrossRef]
Figure 1. Possible phages interactions with CNS.
Figure 1. Possible phages interactions with CNS.
Cells 12 01720 g001
Figure 2. Possible routes of entering the CNS by bacteriophages. (A) Intranasal administration; (a) transcellular pathway; (b) intracellular pathway. (B) Entering from circulation through the BBB by either transcellular pathway or by the Trojan horse mechanism.
Figure 2. Possible routes of entering the CNS by bacteriophages. (A) Intranasal administration; (a) transcellular pathway; (b) intracellular pathway. (B) Entering from circulation through the BBB by either transcellular pathway or by the Trojan horse mechanism.
Cells 12 01720 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Jędrusiak, A.; Fortuna, W.; Majewska, J.; Górski, A.; Jończyk-Matysiak, E. Phage Interactions with the Nervous System in Health and Disease. Cells 2023, 12, 1720. https://doi.org/10.3390/cells12131720

AMA Style

Jędrusiak A, Fortuna W, Majewska J, Górski A, Jończyk-Matysiak E. Phage Interactions with the Nervous System in Health and Disease. Cells. 2023; 12(13):1720. https://doi.org/10.3390/cells12131720

Chicago/Turabian Style

Jędrusiak, Adam, Wojciech Fortuna, Joanna Majewska, Andrzej Górski, and Ewa Jończyk-Matysiak. 2023. "Phage Interactions with the Nervous System in Health and Disease" Cells 12, no. 13: 1720. https://doi.org/10.3390/cells12131720

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

Article Metrics

Back to TopTop