Next Article in Journal
Rehabilitation of Frail Older Adults after Hip Fracture Surgery: Predictors for the Length of Geriatric Rehabilitation Stay at a Skilled Nursing Home
Previous Article in Journal
Unveiling the Role of Metal Ion Concentration versus Immune Sensitization in Orthodontic Patients—A Long-Term Prospective Evaluation
Previous Article in Special Issue
Andexanet Alfa versus Four-Factor Prothrombin Complex Concentrate for the Reversal of Factor Xa (FXa) Inhibitor-Associated Intracranial Hemorrhage: A Systematic Review of Retrospective Studies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Probiotics in Traumatic Brain Injury: New Insights into Mechanisms and Future Perspectives

by
Diamantoula Pagkou
1,*,
Evangelos Kogias
1,
Nikolaos Foroglou
1 and
Katerina Kotzampassi
2
1
Department of Neurosurgery, AHEPA University Hospital, Aristotle University of Thessaloniki, Kiriakidi 1, 54636 Thessaloniki, Greece
2
Department of Surgery, Aristotle University of Thessaloniki, Kiriakidi 1, 54636 Thessaloniki, Greece
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2024, 13(15), 4546; https://doi.org/10.3390/jcm13154546 (registering DOI)
Submission received: 12 June 2024 / Revised: 19 July 2024 / Accepted: 30 July 2024 / Published: 3 August 2024
(This article belongs to the Special Issue Clinical Treatment for Intracerebral Hemorrhage)

Abstract

:
Traumatic brain injury (TBI) is a serious global public health issue, recognized as a chronic and progressive disease that can affect multiple organs, including the gastrointestinal (GI) tract. Research shows that there is a specific link between the GI tract and the central nervous system, termed the gut–brain axis, which consists of bidirectional exchange between these two. Several preclinical and clinical studies have demonstrated intestinal barrier dysfunction, intestinal inflammation and gut dysbiosis in patients with TBI. It is proven that probiotics can modulate the inflammatory process and modify gut microbiota. Numerous animal studies and human clinical trials have proven the effectiveness of selected bacterial strains as an adjuvant treatment in reducing inflammation, infection rates and time spent in intensive care of hospitalized patients suffering from brain injury. Thus, this review summarizes the current evidence regarding the beneficial effects of probiotic administration in patients suffering from TBI-related complications. This review will help identify novel therapeutic strategies in the future as probiotics have an extensive history of apparently safe use.

1. Introduction

Traumatic brain injury (TBI) occurs when an external mechanical force, most commonly after traffic accident, fall, gun shot or sport injury, acutely disrupts the normal function of the brain [1,2]. TBI represents an important public health issue due to its contribution to transient or permanent disability, decreased lifespan and trauma-related death. The global incidence of all-severity, all-cause TBI is estimated at 939 cases per 100.000 people; thus. 69 million people suffer a TBI every year worldwide [3]. The incidence of TBI globally is rising, mainly owing to injuries associated with the increased use of motor vehicles, particularly in middle-income and low-income countries [4]. Moreover, the recent literature shows that more than 1.1% of the US population lives with TBI, and more than 40% of those with moderate-to-severe injuries have long-term disabilities with healthcare cost estimates ranging from USD 56 to USD 221 billion annually [5].
Patients with severe TBI usually undergo prehospital intubation and the majority of them require tracheostomy during intensive care unit (ICU) stays. These patients often become dependent on mechanical ventilation that increases the ICU length of stay and the risk of severe complications such as mechanical-ventilation-associated pneumonia as well as respiratory and renal failure and sepsis [6,7]. Moreover, recent data strongly suggest that stress-induced hyperglycemia is common among patients with moderate–severe TBI and is associated with high mortality rates [8,9,10]. Additionally, brain-injured individuals in the ICU are subjected to nutritional changes and long-term antibiotic therapy; thus, they have a greater risk of suffering from gastrointestinal (GI) symptoms such as enteral feeding intolerance, gut dysmotility, diarrhea and reflux. Patients suffering from TBI are more likely to develop dysbiosis, a pathological imbalance of gut microbial species and reduction in gut biodiversity [11,12].
The human gut microbiota (GM) refers to a vast number of microorganisms, such as bacteria, fungi, archaea and viruses, which colonize the gastrointestinal tract (GI), and interact with one another and with the human host [13]. On the other hand, the gut microbiome is the term given to describe the collective genomes of these microorganisms [14]. GM mainly includes six phyla, namely, Bacteroidetes, Firmicutes, Proteobacteria, Verrucomicrobia, Actinobacteria, and Fusobacteria [15]. The composition of the GM is crucial for maintaining health and has been linked with vital functions such as protection against pathogens, immune regulation, metabolism and vitamin production. There is growing evidence that changes in the GM’s composition, called dysbiosis, are correlated with neurological and neurocognitive disorders [16].
The relationship between the GM and the brain has been a subject of numerous studies for decades. The bidirectional linkage between the GM and the central nervous system (CNS), termed the “gut microbiome–brain axis” (GMBA), facilitates gut motility, intestinal mucosal integrity, and immunologic homeostasis. TBI results in disruption of the GM starting within hours following injury and is associated with chronic gut dysfunction [17]. Taking into consideration that gut dysbiosis in TBI patients is one of the leading causes of organ failure and multiple system complications, targeting the GM and remodeling the balance of gut microbial community should be considered an important therapeutic goal during hospitalization [18]. In the last decade, many therapeutic strategies focused on restoring the GM have been designed. These include early enteral nutrition, administration of probiotics, appropriate use of antibiotics and fecal microbiota transplantation [19,20].
Recent research supports the option that probiotic administration beginning early after trauma could beneficially affect the GM through the maintenance of intestinal barrier integrity and immunomodulation activity, resulting in lower TBI-related complication rates [21,22]. Several animal and human studies focused on alterations in the GM’s composition following TBI demonstrated the potential role of probiotic consumption in attenuating gut dysbiosis [23]. The purpose of this review is to summarize the current evidence supporting the beneficial effects of probiotic administration in preclinical and clinical studies of TBI-induced GM dysbiosis, and whether the modification of GM could influence the clinical outcome of TBI.

2. The Gut Microbiota–Brain Axis (GΜBA)

In recent years, there has been a growing interest in the bidirectional communication between the brain and GM [24]. The GMBA refers to a complex network, linking the central nervous system and the GM, modulated by neurons, neurotransmitters, immune system molecules and hormones. Numerous studies have shown the influence of the GM on cognitive capabilities such as learning and memory, psychiatric and neurodevelopmental disorders and neurodegenerative diseases [25].
The top–down pathway between the brain and GM involves the autonomic and the enteric nervous system, as well as the hypothalamic–pituitary–adrenal (HPA) axis. The autonomic and the enteric nervous system regulate important gut functions such as intestinal homeostasis, motility and secretion of gastric acid and bile, intestinal barrier permeability, and mucosal immune response, thus leading to microbial diversity [26]. Stress-induced HPA axis activation leads to the production of stress hormones such as catecholamines and glucocorticoids, which increase intestinal permeability and alter the GM’s composition, resulting in bacterial inflammatory cytokine production, including interleukin-1 (IL-1), interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) [27].
Recent studies have provided insights into the possible pathways that connect the GM to the brain. In fact, recent evidence widely suggests that the GM modulates brain development, function and behavior [28]. The GM affects the host through neural, immune, neuroendocrine and metabolic pathways. The key communication routes between the enteric microbiota and the brain are the vagal nerve, tryptophan metabolites, and microbial metabolite products, such as short-chain fatty acids (SCFAs) and lipopolysaccharide (LPS) [29]. SCFAs are among the most important class of gut microbiota bio-products. SCFAs are produced in the colon mainly by bacterial fermentation of dietary fibers and resistant starch. Butyrate, acetate and propionate are the most abundant SCFAs in the human body. SCFAs improve gut health through a number of local effects, ranging from the maintenance of intestinal barrier integrity to mucus production, and protection against inflammation. Furthermore, SCFAs are able to cross the blood–brain barrier and have been shown to regulate microglia homoeostasis, which is required for proper brain development and brain tissue homoeostasis [30]. Another decisive bacterial metabolite is LPS, which is principally derived from the cell walls of Gram-negative enterobacteria. LPS can enter into the systemic circulation via intestinal epithelial tight junction’s defects and cross the BBB, causing neuroinflammation [31]. Moreover, microbial-induced metabolism of dietary tryptophan, particularly by certain species of the Lactobacillus genus, results in the production of neuroprotective and neurotoxic metabolites [32]. Nowadays, it is clear that certain bacteria are strain-specifically able to produce different essential neurotransmitters and specific neuromodulators. Neurotransmitters, such as gamma-aminobutyric acid (GABA), serotonin, catecholamines and acetylcholine, are produced by bacteria belonging to Lactobacillus, Bifidobacteria, Enterococcus, and Streptococcus species. These neurotransmitters induce epithelial cells to release molecules that, in turn, have the ability to modulate neural signaling within the enteric nervous system and subsequently control brain function and behavior. In the GI epithelium, enterochromaffin cells are responsible for producing 90% of the body’s serotonin [33].

3. Effects of TBI on Gut Microbiome

TBI consist of two phases: the primary injury and the secondary one. The primary injury takes place within seconds to minutes following trauma and involves direct damage to neurons as a result of the initial local impact. It can result in intracranial hemorrhage, brain edema, acute brain ischemia and, in severe cases, diffuse axonal injury. The secondary injury follows after hours or weeks in response to the primary event, and induces a complex cascade of ischemic, inflammatory, immunological and cytotoxic processes, leading to blood–brain barrier disruption, and thus to secondary brain damage [34].
TBI is increasingly recognized not only as an acute condition but also as a chronic disease with long-term devastating sequelae outside the nervous system. Gut dysbiosis, intestinal barrier dysfunction and chronic enteric inflammation are parallel changes that happen after TBI, which could ulteriorly aggravate the injury profile. Dysbiosis is related to the imbalance between bacteria that normally inhabit the intestine and virulent bacteria, resulting in beneficial bacteria decrease, pathogenic bacteria overgrowth and overall bacterial diversity loss. The phenomenon of dysbiosis could absolutely cause misinterpretation of gut signals in the brain and alter the everyday physiological functions between gut and CNS [31]. Although dysbiosis has been thoroughly studied in the pathogenesis of GI neurodegenerative and metabolic disorders, the investigation of TBI-induced gut dysbiosis in preclinical and clinical models remains very limited and only recently has it been explored [35].
Intestinal barrier dysfunction is often considered a major outcome among TBI patients that further contributes to long-term complications [36,37]. Experimental TBI models in mice demonstrated decreased villous length, crypt depth, and surface area, epithelial cell death, and loss of tight-junction proteins such as zonulin-1 (ZO-1) and occludin creating a “leaky gut” [38,39]. In a Drosophila melanogaster model of closed-head brain injury, disruption of the intestinal barrier and GI permeability was observed, mediated by adrenergic signaling [37]. Damaged intestinal epithelium leads to severe consequences such as malnutrition, electrolyte disorders, bacterial translocation, systemic inflammatory response, sepsis, and multiple organ failure, which further increase mortality and prolongs the length of hospital stay. Common GI symptoms following TBI include feeding intolerance, dyspepsia and chronic diarrhea, causing additional morbidity [40].
According to numerous studies on TBI, there is an increasing evidence that acute and chronic inflammatory events contribute to secondary damage and long-term neuronal disorders. The neuroinflammatory response following TBI is the result of complex interactions between components from both the CNS and periphery and has been shown to be involved in sepsis, systemic inflammatory response syndrome (SIRS) and, consequently, multiple organ failure syndrome [41]. In the gut, TBI induces an increased enteric glial cells activation as an adaptation to chronic brain-derived stimulus mechanism, serving to promote mucosal homeostasis and attenuate the inflammation in response to intestinal barrier disruption [42,43,44]. Attenuating the detrimental aspects of the inflammatory response is a promising strategy to ameliorate the secondary injury.

3.1. Effects of TBI on the Gut Microbiome in Animal Studies

Animal studies have shed light on the potential interaction mechanisms between microbiota and their host organisms. In recent decades, numerous animal experimental models have been developed to mimic various aspects of human TBI. Mice and rats are the most commonly used animal models for studying TBI. Controlled cortical impact injury, fluid percussion injury, weight-drop TBI, and repeated mild TBI models are among the most used in animal TBI experimental studies [45]. The controlled cortical impact model uses an electromagnetic device to produce focal brain contusion injury. The fluid percussion injury model creates a rapid fluid pulse injection, through a craniotomy, directly to the intact dural surface, creating a diffuse deformation of the brain. In the weight-drop model, brain injury is caused by a free-failing weight which directly impacts the animal exposed brain [46].
Houlden et al. [47] identified a positive correlation between gut dysbiosis and injury severity in a TBI mice model 72 h after a closed-head impact. This study demonstrated a significant decrease in the levels of Prevotellaceace (Firmicutes phylum) and an increase in the levels of Peptococcaceae (Bacteroidetes), correlated with the extent of the injury. In another study, Waligora-Dupriet et al. [48] found significant differences in GM after head injury induction in 12 male rats in comparison to controls: a reduced abundance of the beneficial anaerobic groups within the Bacteroidetes, Actinobacteria and Firmicutes phyla and increases in facultative aerobes of the Enterobacteriaceae family. Treangen et al. [49] demonstrated in mice a rapid significant decrease in Lactobacillus gasseri, Ruminococcus flavefaciens, and Eubacterium ventriosum and a significant increase in Eubacterium sulci, and Marvinbryantia formatexigens as early as 24 h post-TBI.
In a controlled cortical impact rodent model of moderate TBI in rodents, Nicholson et al. [50] observed changes in the gut microbiota that favor the pathogenic Gram-negative species of Bacteroidetes and Proteobacteria over the beneficial from the Firmicutes phylum as early as 2 h post-injury. These alterations remained for more than a week, causing an increase in gut permeability. The same finding of long duration of GM disruption was also documented in a recent study, where fecal samples were studied both at the 7th and the 28th post-TBI days. It is of interest to note that authors referred to a decrease in the population of Akkermansia municiphila, a beneficial bacterium belonging to Verrucomicrobia [51,52]. However, in a similar experimental mice TBI model, increased levels of Akkermansia municiphila were detected at 1, 2, and 3 days after brain injury, indicating that alterations in the abundance of Akkermansia municiphila may be divided into two phases [53].
Finally, a significant decrease in beneficial butyrate producing bacteria—Agathobacter, Faecalibacterium, and Eubacterium—and an increase in species belonging to Prevotellaceae and Rikenellaceae families were also found more recently by others [54], while a systematic review performed in 2020 also concluded that there was an increase in Proteobacteria and Firmicutes bacterial populations in all six studies assessed [55].

3.2. Effects of TBI on the Gut Microbiome in Human Studies

To date, only four clinical studies have addressed the issue of the potential impact of brain injury on the GM. In 101 patients with moderate-to-severe TBI, feces received by means of rectal swabs revealed colonization by the Proteobacteria phylum, with Enterobacteriaceae forming the largest group [56].
Moderate to severe TBI patients were recently examined by Pyles et al. [57]. The researchers collected fecal samples at the time of the injury as well as 5 years post-TBI. Interestingly, significant alterations in the fecal microbiota were detected 5 years after brain injury. Corynebacterium, Alistipes and Pseudomonas genus were estimated with higher concentrations compared to control profiles.
Brenner et al. [58], in an effort to document the longevity of GM changes, analyzed fecal samples from veterans with a history of brain trauma 28 years before, but no difference was found in alpha and beta diversity between those having experienced TBI of different severities in relation to the control group, namely, veterans without TBI history. On the contrary, Urban et al. [59] found a significant reduction in the Prevotella and Bacteroides species and an increase in the Ruminococcaceae family in permanent care residents who suffered from chronic brain trauma consequences in relation to healthy controls, although there is no information regarding whether they were also residents of the same facilities or not.

4. Probiotics and Their Effects on TBI-Related Gut Dysbiosis and Inflammation

According to the Food and Agriculture Organization of the United Nations and the World Health Organization (FAO/WHO), probiotics are defined as “live microorganisms which, when administered in adequate amounts, confer a health benefit to the host [60,61]. Probiotics are currently in the spotlight of research given the growing evidence with regard to their potential effects to prevent and/or restore gut microbial dysbiosis [62,63,64,65], thus improving patients outcomes [66]. It has been demonstrated that TBI patients are at high risk of surgical and nosocomial infections resulting from prolonged hospitalization [67]. A post hoc analysis by Tzikos et al. in multiple-trauma patients with concomitant head injury showed that a probiotic mixture significantly reduced the incidence of surgical site infection [68]. Since growing evidence suggests the ability of probiotics to manipulate the GM, we explore the potential role of probiotics as an effective therapeutic strategy to ameliorate TBI-induced pathology and symptoms. The potential role of probiotics in the treatment of TBI systemic effects has been investigated in several experimental and clinical studies [69].

4.1. Animal Studies

In preclinical TBI models, the impact of probiotics on restoring GM and modifying immune system and inflammatory responses is well established. These models demonstrated the efficacy of the administration of several different species of probiotic bacteria, including lactobacilli and butyrate-producing gut bacteria on animal head injury models.
In recent decades, numerous animal experimental models in mice and rats have been developed as an approximation of the actual pathophysiological events of human TBI [70]. Controlled cortical impact injury, fluid percussion injury, weight-drop TBI, and repeated mild TBI models are among the most commonly used in animal TBI experimental studies [45]. The controlled cortical impact injury model uses an electromagnetic device to produce focal brain contusion. In the weight-drop model, brain injury is caused by a free-falling weight which directly impacts the exposed brain of the animal. The fluid percussion injury model takes advantage of a rapid fluid pulse injection, through a craniotomy, directly to the intact dural surface, causing a diffuse deformation of the brain [46].
Li et al. [71] evaluated the effect of Clostridium butyricum in a mouse model of TBI induced by weight-drop impact head injury. The mice were intragastrically given C. butyricum (109 CFU/mL) or placebo once daily for 14 consecutive days prior to the onset of TBI, and for another 14 days thereafter. C. butyricum was found to significantly ameliorate neurological dysfunction, brain edema, neurodegeneration, intestinal barrier permeability and blood–brain barrier impairment in parallel with an increase in the glucagon-like peptide (GLP)-1 levels in the colon in relation to the control treatment.
In a mice weight-drop model of TBI induction, a single strain of Lactobacillus acidophilus at 2 × 1010 CFU/mL was given on the day of injury and for 1, 3, and 7 days thereafter. Probiotic treatment was found to significantly improve the neurological manifestation of the injury: intestinal barrier permeability, neuroinflammation, brain edema, neuronal loss, and sensorimotor deficits in relation to the placebo treatment group [72]. Furthermore, L. acidophilus demonstrated a decreased abundance of the family Pseudomonadaceae and an increase in Prevotellaceae, while controls exhibited a progressive, injury-severity-dependent increase in Alpha-proteobacteria, and specifically of the Porphyromonadaceae and Pseudomonadaceae families [47,50].
Recently, Cui et al. [73] randomized 40 male adult rats surgically subjected to a standardized brain trauma to receive either combination of a brain protein plus Bifidobacterium lactis or placebo. After 7 days of treatment, a decreased secondary inflammatory response and blood–brain barrier repair were prominent in B. lactis treated rats, in parallel with an increase in the immunoregulatory regulatory T cells (Tregs), in addition to a significant enhancement in the cerebral expression of ZO-1 and occludin proteins related to the strengthening of the blood–brain barrier and a decrease in the secondary inflammatory response of brain tissue.
Wild-type mice were subjected to controlled cortical impact injury and subsequently received daily, for the next 3 days, Akkermansia muciniphila [10 9 CFU] in 200 μL sterile phosphate-buffered saline. At the 7th post-injury day, A. muciniphila-treated mice presented with reduced intestinal mucosal injury and a better neurofunctional score in comparison to the control treated at days 3, 7, and 14 [74].
Based on the above, probiotic administration seems to alleviate TBI-induced gut dysbiosis in standardized animal models, mimicking different mechanisms of brain injury. Animal models, despite their advantages, still have limitations that need to be addressed. Human TBI is a complex disease, very difficult to be accurately reflected by animal models. Consequently, findings from experiments in animals cannot fully translate to humans. Thus, it is extremely important to further develop and increasingly use larger species with brains that are more anatomically and functionally closer to man. In addition, more research into the effect of age, sex and animal species or probiotic strain on the outcome of TBI is necessary. Larger as well as well-designed animal studies are required to extrapolate these preclinical findings to humans.

4.2. Human Studies

Several human clinical studies conducted to date have shown a reduction in unfavorable outcomes among TBI patients which received probiotics during hospitalization. These studies demonstrated that probiotic supplementation in patients with brain injury could effectively normalize the altered GM and decrease TBI-related complications such as lung, urinary tract and intracranial infections. There is also evidence that probiotics could prevent TBI-related GI symptoms such as diarrhea, vomiting, abdominal distension, constipation, stress ulcer and reflux. Finally, it was found that probiotic administration reduced time spent in the ICU and mechanical ventilation duration, as well as life-threatening conditions such as sepsis and multiple organ failure [75,76,77].
A randomized controlled trial (RCT) evaluated the effect of probiotics’ administration plus early enteral nutrition in 23 severe-TBI patients in the ICU. Eleven of them were randomly assigned to the probiotic group—240 mL of fermented milk with the probiotic strain Lactobacillus johnsonii plus 30 g of glutamine daily—and 12 received only a polymeric diet. The results demonstrated that the probiotic group had a significantly lower incidence of infections, shorter length of ICU stay and fewer days on mechanical ventilation than the control group [78].
In another single-blind RCT, 52 severe-TBI patients were randomized to receive seven sachets of a three-probiotics mixture composed of Bifidobacterium longum [0.5 × 108 cfu], Lactobacillus bulgaricus [0.5 × 107], and 0.5 × 107 Streptococcus thermophilus [0.5 × 107] or placebo. Probiotic-treated patients were found to have a lower incidence of nosocomial infections, shorter ICU stay and reduction in IL-4 and IL-10 levels [79]. Similar to previous studies, severe-TBI patients who received Lactobacillus bulgaricus plus enteral nutrition within the first 48 h of hospitalization experienced a shorter length of ICU stay and fewer days on mechanical ventilation, as well as lower nosocomial infection rates in relation to those on the placebo treatment [80].
Seventy-six severe-TBI patients were also randomized into probiotics—Bifidobacterium longum, Lactobacillus bulgaricus, and Enterococcus faecalis—or control. The probiotic-treated group demonstrated significantly reduced serum levels of IL-6, IL-10, and tumor necrosis factor TNF-α, along with reduced hospital stays and lower rates of respiratory infection. Moreover, GCS at 15 days was significantly lower in the probiotic group compared to the control group [81]. The same probiotic regime when administered in 68 severe-TBI patients led to a significant reduction in inflammatory response, immune function and TBI-related complications compared to a group of 68 similar patients treated with placebo [82].
A recent small-scale pilot RCT tested the efficacy of Lactobacillus Reuteri versus placebo in 31 U.S. war veterans with a history of mild TBI and (PTSD) symptoms. Probiotic supplementation for 8 weeks was associated with a significant decrease in serum CRP levels, suggesting that CRP may be a useful biomarker to monitor treatment effect in future studies, although the scope of this study did not include the detection of treatment-related changes in clinical PTSD symptoms [83].
In a more recent, phase IIa, randomized placebo-controlled trial, the same author discussed an experimental design to assess the effects of daily oral administration of Lactobacillus rhamnosus (1010 CFU) in 118 US military Veterans with TBI suffering from symptoms related to PTSD. Participants were equally but randomly assigned to either a probiotic or a placebo group, wherein each participant consumed one LGG or placebo capsule daily, for 8 ± 2 weeks. Given the chronic inflammation state among TBI patients and the anti-inflammatory effects of Lactobacillus rhamnosus, the primary outcome measure of this study is the plasma concentration of high-sensitivity CRP [84].
A meta-analysis of four studies (out of 8.781 articles identified) including 325 patients with severe TBI and post-traumatic stress disorder (PTSD) who received prebiotics/probiotics during hospitalization was carried out [85]. Two RCTs included participants who received probiotic treatment, which have previously been described in detail [78,79]. The retrospective cohort study by Painter and colleagues was not discussed here as they used an immune-enhancing nutrition formula containing prebiotics [86]. Finally, the fourth study by Gocan et al. [87], who examined the effects of a fermented soy formulation on veterans with PTSD suffering from depression and anxiety, was not included in this review. Although the findings suggested some promise about the beneficial effects of probiotics in TBI patients, the research in this area remains very limited with significant heterogeneity among the studies in terms of supplements administered, dosing strategies, timing, as well as the duration of administration. Given the widespread use of probiotics and their potential therapeutic effects, there is a need for larger prospective trials to explore the efficacy of specific probiotic supplements for patients with TBI.
In 2019, Yi et al. [88] conducted a meta-analysis of 18 RCTSs, which included 1016 patents between 2010 and 2016. The authors concluded that early enteral nutrition supplemented with probiotics in patients with severe head injury resulted in a significant reduction in infections, mortality rate and ICU length of stay. Similarly, another meta-analysis which included 39 studies found that probiotic supplementation in enteral nutrition resulted in a decreased risk of mortality, infection and gastrointestinal complications in patients suffering from a severe head injury [89]. These studies cover both Chinese and English language articles, resulting in a large number of articles included.
The published research on the use of probiotics in the acute care of TBI has mostly shown favorable findings with respect to decreased infections and improved outcomes. However, a recent systematic review and meta-analysis which reviewed seven RCTs indicated that the administration of probiotics in TBI patients had no significant effects on CRP, IL-6 and the length of staying in ICU. Thus, there was evidence of considerable data heterogeneity; a subsequent subgroup analysis was performed, which showed beneficial results. As a result, more carefully designed RCTs are needed to investigate the effect of probiotics on inflammatory biomarkers and ICU length of stay in TBI patients [90].
Overall, it appears that there is evidence of a positive effect from probiotic use on TBI outcomes. Probiotic treatment in the clinical context seems to be most beneficial in reducing infection, morbidity and mortality rates and decreasing inflammation. However, no studies exist examining the neurological or cognitive outcomes of probiotic supplementation on TBI patients. Additionally, probiotics are widely available for use in preclinical and clinical settings, they are relatively inexpensive, and have been generally demonstrated to be safe for human consumption. However, there is still a relative sparsity of human clinical studies on this subject, and a greater amount of research is needed to be able to more accurately determine the safety and efficacy of probiotic treatment among TBI patients before recommendations for clinical use. Furthermore, the vast majority of microorganisms colonizing the gut have remained unidentified until recently as they are mostly facultative anaerobe bacteria and, consequently, difficult to cultivate. With the development of human genome sequencing and techniques, newly beneficial bacterial strains have been isolated, cultured, and researched, defined as next-generation probiotics (NGPs) [91]. Among these, Akkermansia muciniphila, Faecalibacterium prausnitzii, Eubacterium hallii Bacteroides uniformis, Bacteroides coprocola, Parabacteroides distasonis, Parabacteroides goldsteinii, Hafnia alvei, Odoribacter laneus and Christensenella minuta have demonstrated efficacy in improving gut barrier integrity, exerting anti-inflammatory effects, modulating intestinal flora, and ameliorating intestinal dysbiosis in patients affected by neurodegenerative diseases [92]. Although there are many studies confirming the beneficial effects of traditional probiotics on TBI, repeated preclinical experiments on mice, clinical human trials and complex screening processes are needed to go beyond investigating the exact benefits of NGPs on TBI-patients.

5. Conclusions

As TBI constitutes a major public health issue worldwide, more preventive and therapeutic approaches against TBI and its related complications are urgently needed. Growing clinical evidence reveals that targeting the GMΒA is of great importance in the treatment of TBI patients and there is increasing evidence about the beneficial properties of probiotics among these patients. In this review of the literature, we summarize preclinical and clinical studies that demonstrate the potential importance of gut dysbiosis in the pathogenesis and progression of TBI complications. We also highlighted the significance of modulating the composition of gut microbiome by probiotics as an important and promising therapeutic strategy to promote gut eubiosis and immune function and modulate inflammatory responses.
However, there are still gaps in the literature that need to be addressed. To confirm the positive effects of probiotics on TBI, and to determine the optimal strain and dose with clinical efficacy, as well as timing and duration of administration, more well-designed randomized controlled clinical trials should be conducted. Further research on new-generation probiotics regarding their effects on TBI patients should be verified in animal experiments and human trials, which would lead to the development of novel therapeutic approaches to improve the intestinal barrier, alleviate symptoms, and allow patients suffering from TBI to recover quickly from disease.

Author Contributions

The authors contributed as follows: Conceptualization, K.K., E.K. and N.F.; Writing—Original Draft Preparation, D.P. and K.K.; Writing—Review and Editing, K.K, E.K., N.F. and D.P.; Supervision, K.K., E.K. and N.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Blennow, K.; Brody, D.L.; Kochanek, P.M.; Levin, H.; McKee, A.; Ribbers, G.M.; Yaffe, K.; Zetterberg, H. Traumatic brain in-juries. Nat. Rev. Dis. Primers 2016, 2, 16084. [Google Scholar] [CrossRef] [PubMed]
  2. Galgano, M.; Toshkezi, G.; Qiu, X.; Russell, T.; Chin, L.; Zhao, L.-R. Traumatic brain injury: Current treatment strategies and future endeavors. Cell Transplant. 2017, 26, 1118–1130. [Google Scholar] [CrossRef] [PubMed]
  3. Dewan, M.C.; Rattani, A.; Gupta, S.; Baticulon, R.E.; Hung, Y.C.; Punchak, M.; Agrawal, A.; Adeleye, A.O.; Shrime, M.G.; Rubiano, A.M.; et al. Estimating the global incidence of traumatic brain injury. J. Neurosurg. 2018, 130, 1039–1408. [Google Scholar] [CrossRef]
  4. Global Burden of Disease Collaborators. Global, regional, and national burden of neurological disorders, 1990–2016: A systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 2019, 18, 459–480. [Google Scholar] [CrossRef] [PubMed]
  5. Capizzi, A.; Woo, J.; Verduzco-Gutierrez, M. Traumatic brain injury: An overview of epidemiology, pathophysiology, and medical management. Med. Clin. 2020, 104, 213–238. [Google Scholar]
  6. Faez, D.C.; Kosour, C.; Figueiredo, L.C.; Tonella, R.M.; Pinto, V.G.S.; Dragosavac, D.; Falcao, A.L.E. Weaning from mechanical ventilation in patients with severe head trauma: A proposol of tracheostomy antecipation. J. Neurol. Res. 2016, 6, 35–40. [Google Scholar] [CrossRef]
  7. Corral, L.; Javierre, C.F.; Ventura, J.L.; Marcos, P.; Herrero, J.I.; Manez, R. Impact of non-neurological complications in severe traumatic brain injury outcome. Crit. Care 2012, 16, R44. [Google Scholar] [CrossRef] [PubMed]
  8. Prisco, L.; Iscra, F.; Ganau, M.; Berlot, G. Early predictive factors on mortality in head injured patients: A retrospective analysis of 112 traumatic brain injured patients. J. Neurosurg. Sci. 2012, 56, 131–136. [Google Scholar]
  9. Nwafor, D.; Goeckeritz, J.; Hasanpour, Z.; Davidson, C.; Lucke-Wold, B. Nutritional Support Following Traumatic Brain Injury: A Comprehensive Review. Explor. Res. Hypothesis Med. 2022, 8, 236–247. [Google Scholar] [CrossRef]
  10. Rau, C.-S.; Wu, S.-C.; Chen, Y.-C.; Chien, P.-C.; Hsieh, H.-Y.; Kuo, P.-J.; Hsieh, C.-H. Stress-Induced Hyperglycemia, but Not Diabetic Hyperglycemia, Is Associated with Higher Mortality in Patients with Isolated Moderate and Severe Traumatic Brain Injury: Analysis of a Propensity Score-Matched Population. Int. J. Environ. Res. Public Health 2017, 14, 1340. [Google Scholar] [CrossRef]
  11. Szychowiak, P.; Villageois-Tran, K.; Patrier, J.; Timsit, J.F.; Ruppé, É. The role of the microbiota in the management of intensive care patients. Ann. Intensive Care 2022, 12, 3. [Google Scholar] [CrossRef]
  12. Yuan, B.; Lu, X.-J.; Wu, Q. Gut Microbiota and Acute Central Nervous System Injury: A New Target for Therapeutic Intervention. Front. Immunol. 2021, 12, 800796. [Google Scholar] [CrossRef]
  13. Cresci, G.A.M.; Izzo, K. Gut microbiome. In Adult Short Bowel Syndrome, 1st ed.; Corrigan, M.L., Roberts, K., Steiger, E., Eds.; Academic Press: London, UK, 2019; Chapter 4; pp. 45–54. [Google Scholar]
  14. Hou, K.; Wu, Z.-X.; Chen, X.-Y.; Wang, J.-Q.; Zhang, D.; Xiao, C.; Zhu, D.; Koya, J.B.; Wei, L.; Li, J.; et al. Microbiota in Health and Diseases. Signal Transduct. Target. Ther. 2022, 7, 135. [Google Scholar] [CrossRef]
  15. Thursby, E.; Juge, N. Introduction to the human gut microbiota. Biochem. J. 2017, 474, 1823–1836. [Google Scholar] [CrossRef]
  16. Krakovski, M.A.; Arora, N.; Jain, S.; Glover, J.; Dombrowski, K.; Hernandez, B.; Yadav, H.; Sarma, A.K. Diet-microbiome-gut-brain nexus in acute and chronic brain injury. Front. Neurosci. 2022, 16, 1002266. [Google Scholar] [CrossRef]
  17. Marshall, W.A.; Adams, L.M.; Weaver, J.L. The Brain–Gut Axis in Traumatic Brain Injury: Implications for Nutrition Support. Curr. Surg. Rep. 2022, 10, 172–179. [Google Scholar] [CrossRef]
  18. Corriero, A.; Gadaleta, R.M.; Puntillo, F.; Inchingolo, F.; Moschetta, A.; Brienza, N. The Central Role of the Gut in Intensive Care. Critical Care 2022, 26, 379. [Google Scholar] [CrossRef] [PubMed]
  19. Gagliardi, A.; Totino, V.; Cacciotti, F.; Iebba, V.; Neroni, B.; Bonfiglio, G.; Trancassini, M.; Passariello, C.; Pantanella, F.; Schippa, S. Rebuilding the gut microbiota ecosystem. Int. J. Environ. Res. Public Health 2018, 15, 1679. [Google Scholar] [CrossRef] [PubMed]
  20. Zhu, C.S.; Grandhi, R.; Patterson, T.T.; Nicholson, S.E. A review of traumatic brain injury and the gut microbiome: Insights into novel mechanisms of secondary brain injury and promising targets for neuroprotection. Brain Sci. 2018, 8, 113. [Google Scholar] [CrossRef]
  21. Aghakhani, N. Relationship between Mild Traumatic Brain Injury and the Gut Microbiome: A Scoping Review. J. Neurosci. Res. 2022, 100, 827–834. [Google Scholar] [CrossRef]
  22. Martínez-Guardado, I.; Arboleya, S.; Grijota, F.J.; Kaliszewska, A.; Gueimonde, M.; Arias, N. The Therapeutic Role of Exercise and Probiotics in Stressful Brain Conditions. Int. J. Mol. Sci. 2022, 23, 3610. [Google Scholar] [CrossRef] [PubMed]
  23. Munley, J.A.; Kirkpatrick, S.L.; Gillies, G.S.; Bible, L.E.; Efron, P.A.; Nagpal, R.; Mohr, A.M. The Intestinal Microbiome after Traumatic Injury. Microorganisms 2023, 11, 1990. [Google Scholar] [CrossRef] [PubMed]
  24. George, A.K.; Behera, J.; Homme, R.P.; Tyagi, N.; Tyagi, S.C.; Singh, M. Rebuilding Microbiome for Mitigating Traumatic Brain Injury: Importance of Restructuring the Gut-Microbiome-Brain Axis. Mol. Neurobiol. 2021, 58, 3614–3627. [Google Scholar] [CrossRef] [PubMed]
  25. Maiuolo, J.; Gliozzi, M.; Musolino, V.; Carresi, C.; Scarano, F.; Nucera, S.; Scicchitano, M.; Oppedisano, F.; Bosco, F.; Ruga, S.; et al. The Contribution of Gut Microbiota-Brain Axis in the Development of Brain Disorders. Front. Neurosci. 2021, 15, 616883. [Google Scholar] [CrossRef] [PubMed]
  26. 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] [PubMed]
  27. Freimer, D.; Yang, T.T.; Ho, T.C.; Tymofiyeva, O.; Leung, C. The gut microbiota, HPA axis, and brain in adolescent-onset depression: Probiotics as a novel treatment. Brain Behav. Immun.-Health 2022, 26, 100541. [Google Scholar] [CrossRef] [PubMed]
  28. Liu, L.; Huh, J.R.; Shah, K. Microbiota and the gut-brain-axis: Implications for new therapeutic design in the CNS. eBioMedicine 2022, 77, 103908. [Google Scholar] [CrossRef] [PubMed]
  29. Morais, L.H.; Schreiber, H.L.; Mazmanian, S.K. The gut microbiota–brain axis in behaviour and brain disorders. Nat. Rev. Microbiol. 2021, 19, 241–255. [Google Scholar] [CrossRef] [PubMed]
  30. Chakrabarti, A.; Geurts, L.; Hoyles, L.; Iozzo, P.; Kraneveld, A.D.; La Fata, G.; Miani, M.; Patterson, E.; Pot, B.; Shortt, C.; et al. The microbiota–gut–brain axis: Pathways to better brain health. Perspectives on what we know, what we need to investigate and how to put knowledge into practice. Cell. Mol. Life Sci. 2022, 79, 80. [Google Scholar] [CrossRef] [PubMed]
  31. Raval, U.; Harary, J.; Zeng, E.; Pasinetti, G.M. The dichotomous role of the gut microbiome in exacerbating and ameliorating neurodegenerative disorders. Expert Rev. Neurother. 2020, 20, 673–686. [Google Scholar] [CrossRef]
  32. Sun, Y.; Wang, S.; Liu, B.; Hu, W.; Zhu, Y. Host-Microbiome Interactions: Tryptophan Metabolism and Aromatic Hydrocarbon Receptors after Traumatic Brain Injury. Int. J. Mol. Sci. 2023, 24, 10820. [Google Scholar] [CrossRef] [PubMed]
  33. Rutsch, A.; Kantsjö, J.B.; Ronchi, F. The Gut-Brain Axis: How Microbiota and Host Inflammasome Influence Brain Physiology and Pathology. Front. Immunol. 2020, 11, 3237. [Google Scholar] [CrossRef]
  34. Ng, S.Y.; Lee, A.Y.W. Traumatic brain injuries: Pathophysiology and potential therapeutic targets. Front. Cell Neurosci. 2019, 13, 528. [Google Scholar] [CrossRef] [PubMed]
  35. Margolis, K.G.; Cryan, J.F.; Mayer, E.A. The Microbiota-Gut-Brain Axis: From Motility to Mood. Gastroenterology 2021, 160, 1486–1501. [Google Scholar] [CrossRef] [PubMed]
  36. Pan, P.; Song, Y.; Du, X.; Bai, L.; Hua, X.; Xiao, Y.; Yu, X. Intestinal barrier dysfunction following traumatic brain injury. Neurol. Sci. 2019, 40, 1105–1110. [Google Scholar] [CrossRef] [PubMed]
  37. Scharenbrock, A.R.; Katzenberger, R.J.; Fischer, M.C.; Ganetzky, B.; Wassarman, D.A. Beta-blockers reduce intestinal permeability and early mortality following traumatic brain injury in Drosophila. MicroPubl. Biol. 2021, 10, 17912. [Google Scholar]
  38. Bansal, V.; Costantini, T.; Kroll, L.; Peterson, C.; Loomis, W.; Eliceiri, B.; Baird, A.; Wolf, P.; Coimbra, R. Traumatic brain injury and intestinal dysfunction: Uncovering the neuro-enteric axis. J. Neurotrauma 2009, 26, 1353–1359. [Google Scholar] [CrossRef] [PubMed]
  39. Ma, E.L.; Smith, A.D.; Desai, N.; Cheung, L.; Hanscom, M.; Stoica, B.A.; Loane, D.J.; Shea-Donohue, T.; Faden, A.I. Bidirectional Brain-Gut Interactions and Chronic Pathological Changes after Traumatic Brain Injury in Mice. Brain Behav. Immun. 2017, 66, 56–69. [Google Scholar] [CrossRef]
  40. Hanscom, M.; Loane, D.J.; Shea-Donohue, T. Brain-Gut Axis Dysfunction in the Pathogenesis of Traumatic Brain Injury. J. Clin. Investig. 2021, 131, e143777. [Google Scholar] [CrossRef]
  41. Faden, A.I.; Barrett, J.P.; Stoica, B.A.; Henry, R.J. Bidirectional Brain-Systemic Interactions and Outcomes after TBI. Trends Neurosci. 2021, 44, 406–418. [Google Scholar] [CrossRef]
  42. McKee, C.A.; Lukens, J.R. Emerging Roles for the Immune System in Traumatic Brain Injury. Front. Immunol. 2016, 7, 556. [Google Scholar] [CrossRef] [PubMed]
  43. Yang, S.H.; Gangidine, M.; Pritts, T.A.; Goodman, M.D.; Lentsch, A.B. Interleukin 6 mediates neuroinflammation and motor coordination deficits after mild traumatic brain injury and brief hypoxia in mice. Shock 2013, 40, 471–475. [Google Scholar] [CrossRef] [PubMed]
  44. Timmerman, K.L.; Amonette, W.E.; Markofski, M.M.; Ansinelli, H.A.; Gleason, E.A.; Rasmussen, B.B.; Mossberg, K.A. Blunted IL-6 and IL-10 response to maximal aerobic exercise in patients with traumatic brain injury. Eur. J. Appl. Physiol. 2015, 115, 111–118. [Google Scholar] [CrossRef] [PubMed]
  45. Xiong, Y.; Mahmood, A.; Chopp, M. Animal models of traumatic brain injury. Nat. Rev. Neurosci. 2013, 14, 128. [Google Scholar] [CrossRef] [PubMed]
  46. Shah, E.J.; Gurdziel, K.; Ruden, D. Mammalian Models of Traumatic Brain Injury and a Place for Drosophila in TBI Research. Front. Neurosci. 2019, 13, 409. [Google Scholar] [CrossRef] [PubMed]
  47. Houlden, A.; Goldrick, M.; Brough, D.; Vizi, E.S.; Lénárt, N.; Martinecz, B.; Roberts, I.S.; Denes, A. Brain injury induces specific changes in the caecal microbiota of mice via altered autonomic activity and mucoprotein production. Brain Behav. Immun. 2016, 57, 10–20. [Google Scholar] [CrossRef] [PubMed]
  48. Waligora-Dupriet, A.J.; Lafleur, S.; Charrueau, C.; Choisy, C.; Cynober, L.; Butel, M.J.; Moinard, C. Head injury profoundly affects gut microbiota homeostasis: Results of a pilot study. Nutrition 2017, 45, 104–107. [Google Scholar] [CrossRef]
  49. Treangen, T.J.; Wagner, J.; Burns, M.P.; Villapol, S. Traumatic brain injury in mice induces acute bacterial dysbiosis within the fecal microbiome. Front. Immunol. 2018, 9, 2757. [Google Scholar] [CrossRef] [PubMed]
  50. Nicholson, S.E.; Burmeister, D.M.; Merrill, D.; Scroggins, S.; Zou, Y.; Lai, Z.; Grandhi, R.; Lewis, A.M.; Newton, L.M.; Eastridge, B.J.; et al. Moderate traumatic brain injury alters the gastrointestinal microbiome in a time-dependent manner. Shock 2018, in press. [Google Scholar] [CrossRef]
  51. Zheng, Z.; Wang, S.; Wu, C.; Cao, Y.; Gu, Q.; Zhu, Y.; Zhang, W.; Hu, W. Gut Microbiota Dysbiosis after Traumatic Brain Injury Contributes to Persistent Microglial Activation Associated with Upregulated Lyz2 and Shifted Tryptophan Metabolic Phenotype. Nutrients 2022, 14, 3467. [Google Scholar] [CrossRef]
  52. Cani, P.D.; Depommier, C.; Derrien, M.; Everard, A.; de Vos, W.M. Akkermansia muciniphila: Paradigm for next-generation beneficial microorganisms. Nat. Rev. Gastroenterol. Hepatol. 2022, 19, 625–637. [Google Scholar] [CrossRef] [PubMed]
  53. DeSana, A.J.; Estus, S.; Barrett, T.A.; Saatman, K.E. Acute gastrointestinal permeability after traumatic brain injury in mice precedes a bloom in Akkermansia muciniphila supported by intestinal hypoxia. Sci. Rep. 2024, 14, 2990. [Google Scholar] [CrossRef] [PubMed]
  54. Taraskina, A.; Ignatyeva, O.; Lisovaya, D.; Ivanov, M.; Ivanova, L.; Golovicheva, V.; Baydakova, G.; Silachev, D.; Popkov, V.; Ivanets, T.; et al. Effects of Traumatic Brain Injury on the Gut Microbiota Composition and Serum Amino Acid Profile in Rats. Cells 2022, 11, 1409. [Google Scholar] [CrossRef] [PubMed]
  55. Pathare, N.; Sushilkumar, S.; Haley, L.; Jain, S.; Osier, N. The Impact of Traumatic Brain Injury on Microbiome Composition: A Systematic Review. Biol. Res. Nurs. 2020, 22, 495–505. [Google Scholar] [CrossRef] [PubMed]
  56. Mahajan, C.; Khurana, S.; Kapoor, I.; Sokhal, S.; Kumar, S.; Prabhakar, H.; Mathur, P.; Mani, K. Characteristics of Gut Microbiome after Traumatic Brain Injury. J. Neurosurg. Anesthesiol. 2021. online ahead of print. [Google Scholar] [CrossRef] [PubMed]
  57. Pyles, R.B.; Miller, A.L.; Urban, R.J.; Sheffield-Moore, M.; Wright, T.J.; Maxwell, C.A.; Randolph, K.M.; Danesi, C.P.; McGovern, K.A.; Vargas, J.; et al. The altered TBI fecal microbiome is stable and functionally distict. Front. Mol. Neurosci. 2024, 17, 1341808. [Google Scholar] [CrossRef] [PubMed]
  58. Brenner, L.A.; Stamper, C.E.; Hoisington, A.J.; Stearns-Yoder, K.A.; Stanislawksi, M.A.; Brostow, D.P. Microbial Diversity and Community Structures among Those with Moderate to Severe TBI: A United States-Veteran Microbiome Project Study. J. Head Trauma Rehabil. 2020, 35, 332–341. [Google Scholar] [CrossRef] [PubMed]
  59. Urban, R.J.; Pyles, R.B.; Stewart, C.J.; Ajami, N.; Randolph, M.K.; Durham, W.J.; Danesi, C.P.; Dillon, E.L.; Summons, M.J.R.; Singh, C.K.; et al. Altered Fecal Microbiome Years after Traumatic Brain Injury. J. Neurotrauma 2020, 37, 1037–1051. [Google Scholar] [CrossRef] [PubMed]
  60. Hill, C.; Guarner, F.; Reid, G.; Gibson, G.R.; Merenstein, D.J.; Pot, B.; Morelli, L.; Canani, R.B.; Flint, H.J.; Salminen, S.; et al. Expert consensus document. The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat. Rev. Gastroenterol. Hepatol. 2014, 11, 506–514. [Google Scholar]
  61. Salminen, S.; Collado, M.C.; Endo, A.; Hill, C.; Lebeer, S.; Quigley, E.M.M.; Sanders, M.E.; Shamir, R.; Swann, J.R.; Szajewska, H.; et al. The International Scientific Association of Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of postbiotics. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 649–667. [Google Scholar] [CrossRef]
  62. Sanders, M.E.; Merenstein, D.J.; Reid, G.; Gibson, G.R.; Rastall, R.A. Probiotics and prebiotics in intestinal health and disease: From biology to the clinic. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 605–616. [Google Scholar] [CrossRef] [PubMed]
  63. Suez, J.; Zmora, N.; Segal, E.; Elinav, E. The pros, cons, and many unknowns of probiotics. Nat. Med. 2019, 25, 716–729. [Google Scholar] [CrossRef]
  64. Day, R.L.J.; Harper, A.J.; Woods, R.M.; Davies, O.G.; Heaney, L.M. Probiotics: Current landscape and future horizons. Future Sci. OA 2019, 5, FSO391. [Google Scholar] [CrossRef] [PubMed]
  65. Tegegne, B.A.; Kebede, B. Probiotics, their prophylactic and therapeutic applications in human health development: A review of the literature. Heliyon 2022, 8, e09725. [Google Scholar] [CrossRef] [PubMed]
  66. Kotzampassi, K. Why Give My Surgical Patients Probiotics. Nutrients 2022, 14, 4389. [Google Scholar] [CrossRef]
  67. Sharma, R.; Shultz, S.R.; Robinson, M.; Belli, A.; Hibbs, M.L.; O’Brien, T.J.; Semple, B.D. Infections after a traumatic brain injury: The complex interplay between the immune and neurological systems. Brain Behav. Immun. 2019, 79, 63–74. [Google Scholar] [CrossRef]
  68. Tzikos, G.; Tsalkatidou, D.; Stavrou, G.; Thoma, G.; Chorti, A.; Tsilika, M.; Michalopoulos, A.; Papavramidis, T.; Giamarellos-Bourboulis, E.J.; Kotzampassi, K. A Four-Probiotic Regime to Reduce Surgical Site Infections in Multi-Trauma Patients. Nutrients 2022, 14, 2620. [Google Scholar] [CrossRef]
  69. Rice, M.W.; Pandya, J.D.; Shear, D.A. Gut Microbiota as a Therapeutic Target to Ameliorate the Biochemical, Neuroanatomical, and Behavioral Effects of Traumatic Brain Injuries. Front. Neurol. 2019, 10, 875. [Google Scholar] [CrossRef]
  70. Petersen, A.; Soderstrom, M.; Saha, B.; Sharma, P. Animal models of traumatic brain injury: A review of pathophysiology to biomarkers and treatments. Exp. Brain Res. 2021, 239, 2939–2950. [Google Scholar] [CrossRef]
  71. Li, H.; Sun, J.; Du, J.; Wang, F.; Fang, R.; Yu, C.; Xiong, J.; Chen, W.; Lu, Z.; Liu, J. Clostridium butyricum exerts a neuroprotective effect in a mouse model of traumatic brain injury via the gut-brain axis. Neurogastroenterol. Motil. 2018, 30, 13260. [Google Scholar] [CrossRef]
  72. Ma, Y.; Liu, T.; Fu, J.; Fu, S.; Hu, C.; Sun, B.; Fan, X.; Zhu, J. Lactobacillus acidophilus Exerts Neuroprotective Effects in Mice with Traumatic Brain Injury. J. Nutr. 2019, 149, 1543–1552. [Google Scholar] [CrossRef] [PubMed]
  73. Cui, Y.; Xu, L.; Wang, F.; Wang, Z.; Tong, X.; Yan, H. Orally Administered Brain Protein Combined with Probiotics Increases Treg Differentiation to Reduce Secondary Inflammatory Damage Following Craniocerebral Trauma. Front. Immunol. 2022, 13, 928343. [Google Scholar] [CrossRef] [PubMed]
  74. Chen, Y.; Chen, J.; Wei, H.; Gong, K.; Meng, J.; Long, T.; Guo, J.; Hong, J.; Yang, L.; Qiu, J.; et al. Akkermansia muciniphila-Nlrp3 is involved in the neuroprotection of phosphoglycerate mutase 5 deficiency in traumatic brain injury mice. Front. Immunol. 2023, 14, 1172710. [Google Scholar] [CrossRef] [PubMed]
  75. Willman, J.; Willman, M.; Reddy, R.; Fusco, A.; Sriram, S.; Mehkri, Y.; Charles, J.; Goeckeritz, J.; Lucke-Wold, B. Gut microbiome and neurosurgery: Implications for treatment. Clin. Transl. Discov. 2022, 2, e139. [Google Scholar] [CrossRef]
  76. Danehower, S. Targeting gut dysbiosis as a means to enhance recovery from surgical brain injury. Surg. Neurol. Int. 2021, 12, 210. [Google Scholar] [CrossRef]
  77. Li, C.; Lu, F.; Chen, J.; Ma, J.; Xu, N. Probiotic Supplementation Prevents the Development of Ventilator-Associated Pneumonia for Mechanically Ventilated ICU Patients: A Systematic Review and Network Meta-analysis of Randomized Controlled Trials. Front. Nutr. 2022, 9, 919156. [Google Scholar] [CrossRef] [PubMed]
  78. Falcão de Arruda, I.S.; de Aguilar-Nascimento, J.E. Benefits of Early Enteral Nutrition with Glutamine and Probiotics in Brain Injury Patients. Clin. Sci. 2004, 106, 287–292. [Google Scholar] [CrossRef] [PubMed]
  79. Tan, M.; Zhu, J.-C.; Du, J.; Zhang, L.-M.; Yin, H.-H. Effects of Probiotics on Serum Levels of Th1/Th2 Cytokine and Clinical Outcomes in Severe Traumatic Brain-Injured Patients: A Prospective Randomized Pilot Study. Crit. Care 2011, 15, R290. [Google Scholar] [CrossRef] [PubMed]
  80. Pavelescu, D.; Mirea, L.; Grintescu, I. Could selected probiotics have beneficial effects on clinical outcome of severe traumatic brain injury patients? Crit. Care 2014, 18, P472. [Google Scholar] [CrossRef]
  81. Wan, G.; Wang, L.; Zhang, G.; Zhang, J.; Lu, Y.; Li, J.; Yi, X. Effects of Probiotics Combined with Early Enteral Nutrition on Endothelin-1 and C-Reactive Protein Levels and Prognosis in Patients with Severe Traumatic Brain Injury. J. Int. Med. Res. 2020, 48. [Google Scholar] [CrossRef]
  82. Zhang, T.; Lv, G.G.; Song, Y.; Wang, F. The Effects of Early Enteral Nutrition when Combined with Probiotics in Patient with TBI. Prog. Nutr. 2021, 23, 2021214. [Google Scholar]
  83. Brenner, L.A.; Forster, J.E.; Stearns-Yoder, K.A.; Stamper, C.E.; Hoisington, A.J.; Brostow, D.P.; Mealer, M.; Wortzel, H.S.; Postolache, T.T.; Lowry, C.A. Evaluation of an Immunomodulatory Probiotic Intervention for Veterans with Co-Occurring Mild Traumatic Brain Injury and Posttraumatic Stress Disorder: A Pilot Study. Front. Neurol. 2020, 11, 1015. [Google Scholar] [CrossRef]
  84. Brenner, L.A.; Stearns-Yoder, K.A.; Stamper, C.E. Rationale, design, and methods: A randomized placebo-controlled trial of an immunomodulatory probiotic intervention for Veterans with PTSD. Contemp. Clin. Trials Commun. 2022, 28, 100960. [Google Scholar] [CrossRef] [PubMed]
  85. Brenner, L.A.; Stearns-Yoder, K.A.; Hoffberg, A.S.; Penzenik, M.E.; Starosta, A.J.; Hernández, T.D.; Hadidi, D.A.; Lowry, C.A. Growing Literature but Limited Evidence: A Systematic Review Regarding Prebiotic and Probiotic Interventions for Those with Traumatic Brain Injury and/or Posttraumatic Stress Disorder. Brain Behav. Immun. 2017, 65, 57–67. [Google Scholar] [CrossRef]
  86. Painter, T.J.; Rickerds, J.; Alban, R.F. Immune enhancing nutrition in traumatic brain injury—A preliminary study. Int. J. Surg. 2015, 21, 70–74. [Google Scholar] [CrossRef] [PubMed]
  87. Gocan, A.G.; Bachg, D.; Schindler, A.E.; Rohr, U.D. Balancing steroidal hormone cascade in treatment-resistant veteran soldiers with PTSD using a fermented soy product (FSWW08): A pilot study. Horm. Mol. Biol. Clin. Investig. 2012, 10, 301–314. [Google Scholar] [CrossRef]
  88. Yi, L.J.; Tian, X.; Shi, B.; Pi, Y.P.; Chen, W.Q. Early enteral nutrition supplemented with probiotics improved the clinical outcomes in severe head injury: Some promising findings from Chinese patients. Medicine 2019, 98, 15426. [Google Scholar] [CrossRef]
  89. Du, T.; Jing, X.; Song, S.; Lu, S.; Xu, L.; Tong, X.; Yan, H. Therapeutic Effect of Enteral Nutrition Supplemented with Probiotics in the Treatment of Severe Craniocerebral Injury: A Systematic Review and Meta-Analysis. World Neurosurg. 2020, 139, e553–e571. [Google Scholar] [CrossRef]
  90. Noshadi, N.; Heidari, M.; Naemi Kermanshahi, M.; Zarezadeh, M.; Sanaie, S.; Ebrahimi-Mameghani, M. Effects of Probiotics Supplementation on CRP, IL-6, and Length of ICU Stay in Traumatic Brain Injuries and Multiple Trauma Patients: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Evid. Based Complement. Altern. Med. 2022, 2022, 4674000. [Google Scholar] [CrossRef]
  91. Khan, M.T.; Dwibedi, C.; Sundh, D.; Pradhan, M.; Kraft, J.D.; Caesar, R.; Tremaroli, V.; Lorentzon, M.; Bäckhed, F. Synergy and oxygen adaptation for development of next-generation probiotics. Nature 2023, 620, 381–385. [Google Scholar] [CrossRef]
  92. Chang, C.-J.; Lin, T.-L.; Tsai, Y.-L.; Wu, T.-R.; Lai, W.-F.; Lu, C.-C.; Lai, H.-C. Next generation probiotics in disease amelioration. J. Food Drug Anal. 2019, 27, 615–622. [Google Scholar] [CrossRef] [PubMed]
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

Pagkou, D.; Kogias, E.; Foroglou, N.; Kotzampassi, K. Probiotics in Traumatic Brain Injury: New Insights into Mechanisms and Future Perspectives. J. Clin. Med. 2024, 13, 4546. https://doi.org/10.3390/jcm13154546

AMA Style

Pagkou D, Kogias E, Foroglou N, Kotzampassi K. Probiotics in Traumatic Brain Injury: New Insights into Mechanisms and Future Perspectives. Journal of Clinical Medicine. 2024; 13(15):4546. https://doi.org/10.3390/jcm13154546

Chicago/Turabian Style

Pagkou, Diamantoula, Evangelos Kogias, Nikolaos Foroglou, and Katerina Kotzampassi. 2024. "Probiotics in Traumatic Brain Injury: New Insights into Mechanisms and Future Perspectives" Journal of Clinical Medicine 13, no. 15: 4546. https://doi.org/10.3390/jcm13154546

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