Acessibilidade / Reportar erro

The brain-gut-microbiota axis in the treatment of neurologic and psychiatric disorders

O eixo microbiota-intestino-cérebro no tratamento de desordens neurológicas e psiquiátricas

Abstract

The human gut microbiota is a complex ecosystem made of trillions of microorganisms. The composition can be affected by diet, metabolism, age, geography, stress, seasons, temperature, sleep, and medications. The increasing evidence about the existence of a close and bi-directional correlation between the gut microbiota and the brain indicates that intestinal imbalance may play a vital role in the development, function, and disorders of the central nervous system. The mechanisms of interaction between the gut-microbiota on neuronal activity are widely discussed. Several potential pathways are involved with the brain-gut-microbiota axis, including the vagus nerve, endocrine, immune, and biochemical pathways. Gut dysbiosis has been linked to neurological disorders in different ways that involve activation of the hypothalamic-pituitary-adrenal axis, imbalance in neurotransmitter release, systemic inflammation, and increase in the permeability of the intestinal and the blood-brain barrier. Mental and neurological diseases have become more prevalent during the coronavirus disease 2019pandemic and are an essential issue in public health globally. Understanding the importance of diagnosing, preventing, and treating dysbiosis is critical because gut microbial imbalance is a significant risk factor for these disorders. This review summarizes evidence demonstrating the influence of gut dysbiosis on mental and neurological disorders.

Keywords
Gastrointestinal Microbiome; Dysbiosis; Central Nervous System; Therapeutics

Resumo

A microbiota intestinal humana é um ecossistema complexo feito de trilhões de microrganismos, cuja composição pode ser afetada pela dieta, pelo metabolismo, pela idade, geografia, pelo estresse, pelas estações do ano, pela temperatura, pelo sono e por medicamentos. A crescente evidência sobre a existência de uma correlação estreita e bidirecional entre a microbiota intestinal e o cérebro indica que o desequilíbrio intestinal pode desempenhar um papel vital no desenvolvimento, na função e nos distúrbios do sistema nervoso central. Os mecanismos de interação entre a microbiota intestinal e a atividade neuronal são amplamente discutidos. Várias vias potenciais estão envolvidas com o eixo microbiota-intestino-cérebro, incluindo o nervo vago e as vias endócrinas, imunes e bioquímicas. A disbiose intestinal tem sido associada a distúrbios neurológicos de diferentes maneiras que envolvem a ativação do eixo hipotálamo-hipófise-adrenal, o desequilíbrio na liberação de neurotransmissores, a inflamação sistêmica e o aumento da permeabilidade das barreiras intestinal e hematoencefálica. As doenças mentais e neurológicas tornaram-se mais prevalentes durante a pandemia de coronavirus disease 2019 e são uma questão global essencial na saúde pública. Compreender a importância de diagnosticar, prevenir e tratar a disbiose é fundamental porque o desequilíbrio microbiano intestinal é um fator de risco significativo para esses distúrbios. Esta revisão resume as evidências que demonstram a influência da disbiose intestinal em distúrbios mentais e neurológicos.

Palavras-chave
Microbioma Gastrointestinal; Disbiose; Sistema Nervoso Central; Terapêutica

INTRODUCTION

There are 100 billion neurons in the human brain and 500 million in our gut. The gut is connected to our central nervous system (CNS) and influences brain function through nerves, blood circulation, and lymphatic ways. Another essential component of the interaction between gut and brain is the gut microbiota. This microbiota is a complex ecosystem configured by trillions of microorganisms. The gut microbiota is affected by diet, metabolism, age, geography, stress, seasons, temperature, sleep, medications, and others. A correct balance in the composition of the gut microbiota is vital for mental and physical health, and for the prevention and management of several diseases. The interaction between these numerous structures in the brain and gastrointestinal tract involving the microbiota encompasses the concept of the brain-gut microbiota axis (BGMA), a target of increasing clinical interest. The relationship between the higher prevalence of neurological, psychiatric, and sleep disorders and changes in gut microbiota is being explored to unravel the mechanisms of action of this relationship so that treatment strategies can be developed.11 Eckburg PB, Bik EM, Bernstein CN, et al. Diversity of the human intestinal microbial flora. Science 2005;308(5728):1635–1638

In 2007, the Human Microbiome Project began to unravel the abundance, diversity, and function of genes present in the microbiota and their interference in health and disease.22 Integrative HMPRNCIntegrative HMP (iHMP) Research Network Consortium. The Integrative Human Microbiome Project. Nature 2019;569(7758):641–648 Approximately 15 years of studies have passed, and the knowledge acquired in this area is still insufficient to treat neurological and mental disorders effectively. We know that lifestyle habits such as increased consumption of processed foods and decreased consumption of complex carbohydrates, fruits and vegetables alter the composition of the intestinal microbiota, leading to a condition called dysbiosis. Gut dysbiosis is an imbalance of the intestinal microbiome, which involves the loss of commensal species and increase in the pathogenic microorganism. This microbiome disharmony can alter nutrient availability, trigger chronic inflammation, weaken the human immune system, and begin with many types of diseases. Mental and neurological diseases have become more prevalent during the coronavirus disease 2019 (COVID-19) pandemic and are an essential issue in public health globally. Considering that dysbiosis is a significant risk factor for these disorders, understanding the importance of diagnosing, preventing, and treating dysbiosis is critical. This review summarizes evidence demonstrating the influence of gut dysbiosis on mental and neurological pathophysiology. We also explore dysbiosis diagnosis and the potential of the gut microbiota as a therapeutic target for brain disorders.33 Toor D,Wsson MK, Kumar P, et al. Dysbiosis Disrupts Gut Immune Homeostasis and Promotes Gastric Diseases. Int J Mol Sci 2019;20 (10):20

METHODS

A literature review was performed to identify relevant studies on the role of the microbiome in neurological disorders, including brain, psychiatric, and sleep disorders, by searching the electronic databases of PubMed, ScienceDirect, and Scopus.

To provide a broader perspective on this issue, we reviewed the literature published between 2000 and 2022 in English. Two authors (M. F. N. and F. M. S. C.) conducted the study selection process independently. They identified articles by title, abstract, and full text. Any disagreements were discussed and resolved.

Pathways involved with brain-gut microbiota axis

The discussion about putative mechanisms of the indirect and direct impact of the microbiota on neuronal activity is widespread. There are several potential pathways involved with BGMA. First, the neural way is the fastest communication channel between the brain, gut, and microbiota. Moreover, microbiota can affect the CNS's function bidirectionally via the vagus nerve. An article published in 2011 showed that the vagus nerve plays a significant role in the gastrointestinal tract and CNS. The mechanism involved depends on alterations in brain gamma-aminobutyric acid (GABA)mRNA. Also, the neurochemical and behavioral effects of nonpathogenic bacteria treatment depend on the vagus, considering vagotomized mice do not show alterations.44 Bravo JA, Forsythe P, Chew MV, et al. Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc Natl Acad Sci U S A 2011;108(38):16050–16055 In addition, human studies involving nonpathogenic bacteria administration confirmed the potential translatability of such findings. How gut bacteria can use the vagus nerve to communicate with the CNS remains to be clarified.55 Allen AP, Hutch W, Borre YE, et al. Bifidobacterium longum 1714 as a translational psychobiotic:modulation of stress, electrophysiology and neurocognition in healthy volunteers. Transl Psychiatry 2016;6(11):e939

Secondly, the endocrine pathway (e.g., circulation of neuropeptides or hormones) interacts with these distant structures gut and brain. Any neuroactive substances (precursors and neurotransmitters) in the gut can affect the neuronal activity in the brain. Many neurotransmitter precursors from the gut can cross through the blood-brain barrier (BBB), influencing various neurological electrophysical and biochemical interactions. Third, the immune pathway is influenced by the gut microbiota as components of the microbiota interact with cells and their receptors in the gut mucosa all the time, stimulating homeostatic control of immune responses, which are important for the symbiotic relationship between the microbiota and the host. However, some inflammatory triggers, such as gut pathogenic bacteria or exposure to a toxic substance, can activate immune cells, which may modulate neuronal activity via the release of neurotransmitters and cytokines.66 Houser MC, Tansey MG. The gut-brain axis: is intestinal inflammation a silent driver of Parkinson’s disease pathogenesis? NPJ Parkinsons Dis 2017;3:3

Finally, various microbiota bacteria can metabolize tryptophan, limiting its use as a precursor of indole, melatonin, and serotonin. Low levels of serotonin have been linked to mental and neurological disorders. Besides these biochemical pathways, gut microbiota can produce metabolites such as short-chain fatty acid (SCFA) and synthesize other neurotransmitters. Short-chain fatty acid can cross the BBB contributing to the GABA metabolic cycle in the hypothalamus. Interestingly, 11C- and 13C-labeled acetate from the gut acts in the hypothalamus, stimulating tricarboxylic acid, glutamate-glutamine, and GABA transcellular cycles. Also, microbiota produces enzymes that can facilitate the synthesis of neurotransmitters or their precursors, such as glutamate, GABA, dopamine, and serotonin.77 Frost G, Sleeth ML, Sahuri-Arisoylu M, et al. The short-chain fatty acid acetate reduces appetite via a central homeostatic mechanism. Nat Commun 2014;5:3611

Usually, these neurotransmitters do not penetrate the BBB, but capillary endothelial cells can actively transport many neurotransmitter precursors into the brain. The concentration of many of them, such as tyrosine and tryptophan, contribute to neurotransmitter-producing. Dysbiosis can disrupt these pathways inducing mental and neurological diseases (Figure 1).77 Frost G, Sleeth ML, Sahuri-Arisoylu M, et al. The short-chain fatty acid acetate reduces appetite via a central homeostatic mechanism. Nat Commun 2014;5:3611

Figure 1
The brain-gut-microbiota axis. Abbreviations: CCK, cholecystokinin; ENS, enteric nervous system; FMT, fecal microbiota transplantation; GLP1, glucagon-like peptide 1; HPA, hypothalamic–pituitary–adrenal; LBPs, live biotherapeutic products; PYY, peptide YY; SCFA, short-chain fatty acids.

Gut dysbiosis diagnosis

Feces samples are usually collected for the human gut microbiome analysis. However, fecal samples are just a proxy of the human intestinal microbiota, and researchers comprehend that might be an essential distinction between the microbial composition of feces and intestinal mucosa. Another way to assess gut microbiota is by endoscopic biopsy, which can investigate distinct areas of the gastrointestinal (GI) tract. However, biopsies are invasive methods to determine the microbiome; thus, it is not appropriate to investigate healthy individuals.88 Tang Q, Jin G, Wang G, et al. Current Sampling Methods for Gut Microbiota: A Call for More Precise Devices. Front Cell Infect Microbiol 2020;10:151

Other possible sampling methods for collecting gut microbiota are luminal brush, laser capture microdissection, catheter aspiration, intelligent capsule, surgery, ileostomy, and fluorescence in situ hybridization (FISH). Nevertheless, they are invasive, inappropriate for healthy individuals, and some are unsuitable for most studies and mainly used for commercial analysis.88 Tang Q, Jin G, Wang G, et al. Current Sampling Methods for Gut Microbiota: A Call for More Precise Devices. Front Cell Infect Microbiol 2020;10:151

After sample collection, it is necessary to apply an established methodology to extract the microbial DNA, to assess and quantify the gut microbiota composition. There are some offered methods to determine and quantify dysbiosis. A recent study has classified the dysbiosis indexes into five categories according to the methodology: Large-scale bacterial markers; relevant taxon-based plans; neighborhood classification; Random Forest prediction, and combined α and β diversity.99 Wei S, BahlMI, Baunwall SMD, Hvas CL, Licht TR. Determining Gut Microbial Dysbiosis: a Review of Applied Indexes for Assessment of Intestinal Microbiota Imbalances. Appl Environ Microbiol 2021;87(11):87

Although diagnosing gut dysbiosis is essential to elucidate and treat numerous diseases, including neurological disorders (such as Alzheimer, Parkinson, multiple sclerosis, mood disorders, and others), for many reasons, it is still a challenge for physicians to assess intestinal microbiota imbalance in their clinical practices. Faulty sampling methods are part of the adversity for converting scientific studies into medical interventions. Moreover, the microbiome is highly complex, resulting in datasets that demand refined statistical analysis, which cannot afford commercial purposes. Besides, the diagnosis methods currently available are costly.88 Tang Q, Jin G, Wang G, et al. Current Sampling Methods for Gut Microbiota: A Call for More Precise Devices. Front Cell Infect Microbiol 2020;10:151

Thus, prospective studies are still necessary to improve from a descriptive phase of research, to better elucidate and uncomplicate dysbiosis assessment and, other than that, conclude about types of dysbiosis (e.g., loss of beneficial bacteria, overgrowth of pathogenic bacteria, reduction of bacterial diversity) and if each type has equally or distinct weights to trigger or aggravate different categories of diseases.1010 Brüssow H. Problems with the concept of gut microbiota dysbiosis. Microb Biotechnol 2020;13(02):423–434

Neurological disorders

Alzheimer’s disease

Alzheimer’s disease (AD) is a degenerative and progressive cognitive impairment disease, with no treatment available to interrupt the degenerative process and revive degenerated brain cells, and it is associated with the malfunction of several neurotransmitters, such as acetylcholine, dopamine, GABA, serotonin, glutamate, and norepinephrine. The concentration of these neurotransmitters depends on bacterial taxa such as E. coli, Bacteroides, Eubacterium, and Bifidobacterium.1111 Horiuchi Y, Kimura R, Kato N, et al. Evolutional study on acetylcholine expression. Life Sci 2003;72(15):1745–1756

Studies have reported dysbiosis in patients with AD. There is an imbalance in gut microbiota (rises of Bacteroides, Escherichia/Shigella, and Ruminococcus and reduction of Dialister and E. rectale, Bifidobacterium) influencing the progress of AD due to dysregulation of the synthesis of neurotransmitters and the precursors of them.1212 Cattaneo A, Cattane N, Galluzzi S, et al; INDIA-FBP Group. Association of brain amyloidosis with pro-inflammatory gut bacterial taxa and peripheral inflammation markers in cognitively impaired elderly. Neurobiol Aging 2017;49:60–68 A recent study characterized an abundance of several bacteria taxa in mild AD patients (Anaerostipes, Mitsuokella, Prevotella, Bosea, Fusobacterium, Anaerotruncus, Clostridium, and Coprobacillus), and there was an abundance of an emerging probiotic (Akkermansia) and a traditional probiotic (Bifidobacteria). The authors defend this pattern of the fecal microbiome in mild AD patients.1313 Chen Y, Xu J, Chen Y. Regulation of Neurotransmitters by the Gut Microbiota and Effects on Cognition in Neurological Disorders. Nutrients 2021;13(06):13

The treatment of AD patients is related to a pharmacological treatment with acetylcholinesterase inhibitors (tacrine, donepezil, rivastigmine, and galantamine), N-methyl-D-aspartate (NMDA) receptor antagonist (memantine), and the recent aducanumab.1414 Cummings J, Aisen P, Apostolova LG, Atri A, Salloway S,Weiner M. Aducanumab: Appropriate Use Recommendations. J Prev Alzheimers Dis 2021;8(04):398–410

However, there is a growing interest in dietary interventions to treat AD patients with fermented foods and drug development. The diagnosis of this gut dysbiosis is not a reality in real life yet. However, potential fermented food products have been studied with safe long-term eating to control the disease outcome and minimize commercial drug side effects and toxicity. Recently, a systematic review on the impact of probiotics in animal AD studies demonstrated that although the effect of probiotic administration on Aβ remains ambiguous, probiotics such as B. longum (NK46), C. butyricum, and the mixture SLAB51 have exhibited promising effects to relieve AD symptoms1515 de Rijke TJ, Doting MHE, van Hemert S, et al. A Systematic Review on the Effects of Different Types of Probiotics in Animal Alzheimer’s Disease Studies. Front Psychiatry 2022;13:879491

Parkinson’s disease

Parkinson’s disease (PD) is a neurodegenerative disorder with dysregulation of the dopamine system due to aggregation of insoluble α-synuclein (µ-syn) protein in vulnerable populations of neurons and glia. Gut dysbiosis has also been observed in PD patients (increase of Akkermansia, Catabacter, Lactobacillus, Bifidobacterium, Bifidobacteriaceae, Ruminococcaceae, Verrucomicrobiaceae, Christensenellaceae, and decrease of Roseburia, Faecalibacterium, Lachnospiraceae ND3007 group, Prevotellaceae, Blautia, Coprococcus, and Lachnospira).1313 Chen Y, Xu J, Chen Y. Regulation of Neurotransmitters by the Gut Microbiota and Effects on Cognition in Neurological Disorders. Nutrients 2021;13(06):13

The other factors associated with gut microbiota were constipation severity, physical activity, and pharmacological therapies associated with β diversity. They concluded that the gut microbiota combined with macronutrient intake could predict the development of PD. Recent reports on the gut microbiome in PD showed no differences in the fungi abundance, decreased total virus richness, and significantly increased Lactobacillaceae abundance, consistent with the overrepresentation of Lactobacillales.1616 Cirstea MS, Sundvick K, Golz E, et al. The Gut Mycobiome in Parkinson’s Disease. J Parkinsons Dis 2021;11(01):153–158

The pathological process suggested for PD is active retrograde transport of ∝-syn from the enteric nervous system (ENS) to the CNS by the dorsal motor nucleus of the vagus nerve (DMNV). An extracellular amyloid protein named “curli” (secreted by Escherichia coli) can be implicated, inducing neuronal deposition of a-syn in the gut and promoting neuroinflammation. [80] Experimental animal models with gut dysbiosis show that gut microbiota is related to motor impairments and gastrointestinal (GI) dysfunction. The microbial molecules, via the gut–brain, impact microglia activation, neuroinflammation, and ∝-syn aggregation.1717 Sampson TR, Debelius JW, Thron T, et al. Gut Microbiota Regulate Motor Deficits and Neuroinflammation in a Model of Parkinson’s Disease. Cell 2016;167(06):1469–1480.e12

Enterococcus and Bacillus are bacteria capable of synthesizing dopamine in gut microbiota. The intraduodenal infusion of levodopa-carbidopa intestinal gel (LCIG) is related to an increased relative abundance of the Enterobacteriaceae family, as well as to lower absorption of L-dopa in PD patients infected by Helicobacter pylori. Indeed, there is a bidirectional approach between medications and gut microbiota in PD patients.1818 Lorente-Picón M, Laguna A. New Avenues for Parkinson’s Disease Therapeutics: Disease-Modifying Strategies Based on the Gut Microbiota. Biomolecules 2021;11(03):11

Authors have demonstrated the influence of E. faecalis in the activity of levodopa decarboxylation with potential clinical response influence. However, a recent study showed that Levodopa was not associated with changes in microbiota composition and concluded that more extensive and longer-term studies must be done.1919 PalaciosN, Hannoun A, Flahive J, et al. Effect of Levodopa Initiation on the Gut Microbiota in Parkinson’s Disease. Front Neurol 2021; 12:574529

Several clinical trials with fecal microbiota transplantation from healthy controls to patients with PD have demonstrated significant effects on attenuating the motor symptoms, meanwhile improving non-motor symptoms such as the quality of sleep and life and relieving anxiety and depression constipation symptoms in PD.2020 Xue LJ, Yang XZ, Tong Q, et al. Fecal microbiota transplantation therapy for Parkinson’s disease: A preliminary study. Medicine (Baltimore) 2020;99(35):e22035

Multiple sclerosis

Multiple sclerosis (MS) is a prevalent neurological disease, which is more likely to occur in Europe, North America, and Australasia. The risk factors involved in the pathology are highly complex and combine genetic variants and environmental factors. Evidence supports that the gut microbiota composition of patients with MS suffers alteration, as several studies observed that the intestinal microbiome profile of these patients differs from healthy controls.2121 Jangi S, Gandhi R, Cox LM, et al. Alterations of the human gut microbiome in multiple sclerosis. Nat Commun 2016; 7:12015

The modification of the bacterial population detected in the gut of patients with MS seems to lead to a proinflammatory state. At the same time, dysbiosis increases the BBB permeability. It results in CNS inflammation and neurodegeneration, which might culminate in the development of MS.2222 Adamczyk-Sowa M, Medrek A, Madej P, Michlicka W, Dobrakowski P. Does the Gut Microbiota Influence Immunity and Inflammation in Multiple Sclerosis Pathophysiology? J Immunol Res 2017;2017:7904821

An overview of clinical trials that evaluated the gut microbiota composition of patients with MS observed that, compared with healthy controls, these patients had increased concentrations of Pedobacteria, Flavobacterium, Pseudomonas, Mycoplana, Acinetobacter, and Eggerthella, Dorea, Blautia, Streptococcus, and Akkermansia. In contrast, their gut microbiota presents lower amounts of Prevotella, Bacteroides, Parabacteroides, Haemophilus, Sutterela, Adlercreutzia, Croprobacillus, Lactobacillus, Clostridium, Anaerostipes, and Faecalibacterium, showing that dysbiosis is a fact in patients with MS. The authors suggest that the dysbiosis treatment seems to reduce inflammation and reactivate the immune system by regulating the T lymphocytes effect in humans.2323 Schepici G, Silvestro S, Bramanti P, Mazzon E. The Gut Microbiota in Multiple Sclerosis: An Overview of Clinical Trials. Cell Transplant 2019;28(12):1507–1527

Epilepsy

The gut microbiota of patients with epilepsy has also been studied. As shown in other neurological disorders, it seems to be altered compared with healthy controls, which increases inflammation and may influence drug-resistant seizures.2424 Dahlin M, Prast-Nielsen S. The gut microbiome and epilepsy. EBioMedicine 2019;44:741–746

In a recent longitudinal study in drug-naïve children with epilepsy, researchers observed that the intestinal microbiome of these patients had an increased abundance of Akkermansia ssp. and Proteobacteria and a decreased number of Faecalibacterium ssp, suggesting that dysbiosis and taxonomy may happen in the gut of patients with epilepsy. However, clinical trials evaluating the gut microbiota of patients with epilepsy are still scarce, and further studies must be performed to confirm these findings (Table 1).2525 Ceccarani C, Viganò I, Ottaviano E, et al. Is Gut Microbiota a Key Player in Epilepsy Onset? A Longitudinal Study in Drug-Naive Children. Front Cell Infect Microbiol 2021;11:749509

Table 1
Summary of studies assessing the response to probiotic administration for treating brain and psychologic disorders

Gut microbiota and sleep disorders

Obstructive sleep apnea

Obstructive sleep apnea (OSA) is a pathology that occurs due to a collapse of the upper airway during sleep, leading to a decrease in or interruption of ventilation. Consequently, the affected individual has hypoxia, hypercapnia (due to reduced airflow during airway obstruction), and an increase in the number of awakenings during sleep, usually associated with events, to restore breathing.2626 International Classification of Sleep Disorders – Third edition (ICSD-3). 3rd ed. American Academy of Sleep Medicine; 2014 Obstructive sleep apnea is related to type 2 diabetes, arterial hypertension, stroke, cardiac arrhythmias, coronary diseases, and cognitive alterations.2727 Veasey SC, Rosen IM. Obstructive Sleep Apnea in Adults. N Engl J Med 2019;380(15):1442–1449, 2828 Badran M, Mashaqi S, Gozal D. The gut microbiome as a target for adjuvant therapy in obstructive sleep apnea. Expert Opin Ther Targets 2020;24(12):1263–1282

The intermittent hypoxia that occurs in OSA can cause an increase in anaerobic organisms in the gut. This dysbiosis increases tissue permeability, inflammation, and oxidative stress, besides being linked to cardiometabolic, neurobehavioral, and GI changes often found in individuals with OSA. An increase in Firmicutes/Bacteroidetes and a decrease in Actinobacteria/Proteobacteria was observed in individuals who snore, indicating it may influence the gut microbiota. In children with OSA, there is a decrease in the diversity of the intestinal microbiota with an increase in inflammation in the intestinal barrier due to intermittent hypoxia.2929 Valentini F, Evangelisti M, Arpinelli M, et al. Gut microbiota composition in childrenwith obstructive sleep apnoea syndrome: a pilot study. Sleep Med 2020;76:140–147

Studies that evaluated the intestinal microbiota of individuals with OSA observed intestinal dysbiosis in these patients.3030 Durgan DJ. Obstructive Sleep Apnea-Induced Hypertension: Role of the Gut Microbiota. Curr Hypertens Rep 2017;19(04):35

Experimental studies show that this dysbiosis may improve with the use of pre and probiotics.2626 International Classification of Sleep Disorders – Third edition (ICSD-3). 3rd ed. American Academy of Sleep Medicine; 2014, 2828 Badran M, Mashaqi S, Gozal D. The gut microbiome as a target for adjuvant therapy in obstructive sleep apnea. Expert Opin Ther Targets 2020;24(12):1263–1282 Prebiotics and probiotics with weight loss increase the quality of life and decrease liver fat, visceral fat, abdominal circumference, interleukin 6 (IL-6), other proinflammatory cytokines, and cortisol.2828 Badran M, Mashaqi S, Gozal D. The gut microbiome as a target for adjuvant therapy in obstructive sleep apnea. Expert Opin Ther Targets 2020;24(12):1263–1282

Sleep deprivation

Sleep deprivation occurs when an individual sleeps less time than is necessary for rest and health maintenance. It can cause or lead to several pathologies, such as cardiovascular diseases, atherosclerosis, coronary diseases, alterations in the autonomic nervous system, immunological alterations, metabolic, respiratory, and GI disorders, cognitive, mood, and attention and memory alterations, and stroke. It is also related to increased appetite, insulin resistance, and increased obesity.3131 Liew SC, Aung T. Sleep deprivation and its association with diseases- a review. Sleep Med 2021;77:192–204

Sleep deprivation or sleep fragmentation are stressors capable of altering the intestinal microbiota, leading to dysbiosis, and increasing intestinal permeability, consequently increasing inflammation.3232 Reynolds AC, Paterson JL, Ferguson SA, Stanley D, Wright KP Jr, Dawson D. The shift work and health research agenda: Considering changes in gut microbiota as a pathway linking shift work, sleep loss and circadian misalignment, and metabolic disease. Sleep Med Rev 2017;34:3–9 After acute sleep deprivation, a decrease in gut microbiota diversity was found.2828 Badran M, Mashaqi S, Gozal D. The gut microbiome as a target for adjuvant therapy in obstructive sleep apnea. Expert Opin Ther Targets 2020;24(12):1263–1282, 3333 Wang Z, Chen WH, Li SX, et al. Gut microbiota modulates the inflammatory response and cognitive impairment induced by sleep deprivation. Mol Psychiatry 2021;26(11):6277–6292 In experimental models, sleep deprivation has been observed to cause oxidative stress and intestinal cell damage.3434 Karl JP, Hatch AM, Arcidiacono SM, et al. Effects of Psychological, Environmental and Physical Stressors on the Gut Microbiota. Front Microbiol 2018;9:2013

Microbiome diversity was correlated with sleep efficiency and wakefulness after sleep onset. We found positive correlations between total microbiome diversity and IL-6, a cytokine previously observed for its effects on sleep.3535 Smith RP, Easson C, Lyle SM, et al. Gut microbiome diversity is associated with sleep physiology in humans. PLoS One 2019;14 (10):e0222394

Studies have shown an increase in the abundance of Firmicutes related to sleep efficiency and a reduction in the quantity of Bacteroidetes in the intestinal microbiota in sleep-deprived individuals. This change is associated with obese individuals and could explain why sleep-deprived individuals tend to gain weight.3535 Smith RP, Easson C, Lyle SM, et al. Gut microbiome diversity is associated with sleep physiology in humans. PLoS One 2019;14 (10):e0222394

Another sleep disorder that is associated with decreased sleep time is insomnia. Reduced bacterial diversity could be related to acute and chronic insomnia. Patients with the condition showed a decrease in anaerobic microorganisms. There is an alteration in the microbiome structure of patients with chronic insomnia, and there may be a decrease in butyrate-producing microorganisms and a consequent increase in inflammatory markers. There was also an increase in the genus Blautia and a reduction in Faecalibacterium, which could be associated with inflammatory changes and neuropsychological diseases, including diabetes and GI disorders.3636 Li Y, Zhang B, Zhou Y, et al. Gut Microbiota Changes and Their Relationship with Inflammation in Patients with Acute and Chronic Insomnia. Nat Sci Sleep 2020;12:895–905 It is also observed that sleep-deprived patients may have decreased colon and stool melatonin levels.3737 Park YS, Kim SH, Park JW, et al. Melatonin in the colon modulates intestinal microbiota in response to stress and sleep deprivation. Intest Res 2020;18(03):325–336

Circadian rhythm and gut microbiota

The human body follows the circadian rhythm for around 24 hours. The sleep-wake period's circadian rhythm follows endogenous patterns independent of external and exogenous factors such as daylight. The interruption in the circadian rhythm leads to a proinflammatory state and increases the risk of cardiovascular diseases, diabetes, and metabolic syndrome, among others.3838 Hernández-García J, Navas-Carrillo D, Orenes-Piñero E. Alterations of circadian rhythms and their impact on obesity, metabolic syndrome and cardiovascular diseases. Crit Rev Food Sci Nutr 2020;60(06):1038–1047

The circadian rhythm influences the intestinal bacteria population, fluctuating during the day.3939 Voigt RM, Forsyth CB, Green SJ, Engen PA, Keshavarzian A. Circadian Rhythm and the Gut Microbiome. Int Rev Neurobiol 2016;131:193–205 The feeding schedule is an important marker off- and impact on- the rhythm, and when the former changes, so does the latter. With the interruption of the circadian rhythm, dysbiosis occurs in the intestinal microbiota, and researchers could detect changes in cell permeability and inflammatory and immune responses.4040 Parekh PJ, Oldfield EC IV, Johnson DA. The Effects of Sleep on the Commensal Microbiota: Eyes Wide Open? J Clin Gastroenterol 2018;52(03):204–209

It was observed that, in experimental models, the interruption of the circadian rhythm of animals alters functions, such as the rhythmicity of the intestinal microbiota, and causes changes in their genetics. Thus, there is a change in the composition of this microbiota. With dysbiosis, there is a change in intestinal permeability and increased inflammation, with possible harmful health effects. The observed effects are increased glucose intolerance and weight gain.3434 Karl JP, Hatch AM, Arcidiacono SM, et al. Effects of Psychological, Environmental and Physical Stressors on the Gut Microbiota. Front Microbiol 2018;9:2013

Microbiota, other sleep disorders, and related comorbidities

Other sleep disorders that worsen sleep quality could be related to changes in the intestinal microbiota. Even neurological diseases and cognitive alterations are related to sleep and could be related to intestinal alterations. A study showed that changes in sleep and consequently in the microbiota could be a factor that would bring cognitive changes to the elderly.4141 Anderson JR, Carroll I, Azcarate-Peril MA, et al. A preliminary examination of gut microbiota, sleep, and cognitive flexibility in healthy older adults. Sleep Med 2017;38:104–107

The intestinal microbiota of children with autism spectrum disorder and with sleep problems was unbalanced, with an increase in Faecalibacterium and a decrease in Agathobacter. There was a decrease in the production of butyrate and its metabolites, worsening sleep and aggravating other central symptoms in these individuals.4242 Hua X, Zhu J, Yang T, et al. The Gut Microbiota and Associated Metabolites Are Altered in Sleep Disorder of Children With Autism Spectrum Disorders. Front Psychiatry 2020;11:855

No differences were found in patients with type 1 narcolepsy relative to controls, between groups for microbial diversity, population richness, and uniformity.4343 Lecomte A, Barateau L, Pereira P, et al. Gutmicrobiota composition is associated with narcolepsy type 1. Neurol Neuroimmunol Neuroinflamm 2020;7(06):7

In studies with bipolar patients, a relationship was found between Faecalibacterium and Lactobacterium and sleep quality, suggesting that increasing these two types could improve sleep in these patients (Table 1).4444 Wagner-Skacel J, Dalkner N, Moerkl S, et al. Sleep and Microbiome in Psychiatric Diseases. Nutrients 2020;12(08):12

Psychiatric disorders

Depression

Before the COVID-19 pandemic, the global prevalence of depression and anxiety disorders was alarming. The World Health Organization (WHO) estimated that, globally, the number of individuals with depression in 2015 exceeded 300 million.4545 Organization WH. Depression and Other Common Mental Disorders Global Health Estimates.World Health Organization; 2017 Nevertheless, research performed in specific countries showed that the incidence of anxiety and depression has increased significantly throughout the COVID-19 pandemic. According to the scientific brief released by the WHO, the global prevalence of anxiety and depression increased by 25%.4646 Canet-Juric L, Andrés ML, Del Valle M, et al. A Longitudinal Study on the Emotional Impact Cause by the COVID-19 Pandemic Quarantine on General Population. Front Psychol 2020; 11:565688

Although antidepressants, the primary drugs used for depression, are often adequate to treat this disabling mental disorder, it often causes side effects.4747 Bet PM, Hugtenburg JG, Penninx BW, Hoogendijk WJ. Side effects of antidepressants during long-term use in a naturalistic setting. Eur Neuropsychopharmacol 2013;23(11):1443–1451 Since side effects increase with longer-term use, and antidepressants, in most cases, should not be taken for more than 2 years, patients and their physicians usually desire to get off the medication as the depression symptoms get better.4848 Bowers HM, Williams SJ, Geraghty AWA, et al. Helping people discontinue long-term antidepressants: views of health professionals in UK primary care. BMJ Open 2019;9(07):e027837

There is increasing use of pre and probiotics with psychotherapy, such as complementary and alternative therapies for mental disorders.4949 Dinan TG, Cryan JF. Brain-Gut-Microbiota Axis andMental Health. Psychosom Med 2017;79(08):920–926 It is well known that the microbiota-gut-brain axis has a bidirectional communication and microbiota imbalance leads to adverse effects on central nervous system functions that increase the symptoms and mental illness.5050 Foster JA, Baker GB, Dursun SM. The Relationship Between the Gut Microbiome-Immune System-Brain Axis and Major Depressive Disorder. Front Neurol 2021;12:721126 Although the mechanism by which the gut microbiota plays a role in mental health is still controversial, researchers suggest that factors such as the hypothalamic-pituitary-adrenal (HPA) axis and serotonergic, GABAergic, and other neurotransmitters signaling systems are probably the central part of this correlation.5151 Suganya K, Koo BS. Gut-Brain Axis: Role of Gut Microbiota on Neurological Disorders and How Probiotics/Prebiotics Beneficially Modulate Microbial and Immune Pathways to Improve Brain Functions. Int J Mol Sci 2020;21(20):21

The HPA axis is the human central stress response system, and hyperactivity is identified in several patients diagnosed with depression and is activated by gut dysbiosis through specific mechanisms.5252 Misiak B, Łoniewski I, MarliczW, et al. The HPA axis dysregulation in severe mental illness: Can we shift the blame to gut microbiota? Prog Neuropsychopharmacol Biol Psychiatry 2020; 102:109951

Gut dysbiosis can also be related to depression symptoms, impacting tryptophan metabolism and the serotoninergic system. Tryptophan (an essential amino acid) is the exclusive precursor of serotonin, and the human intestinal microbiota has a crucial role in regulating the tryptophan metabolism.5353 Waclawiková B, El Aidy S. Role of Microbiota and Tryptophan Metabolites in the Remote Effect of Intestinal Inflammation on Brain and Depression. Pharmaceuticals (Basel) 2018;11(03):11 Additionally, the GI tract has high levels of 5-hydroxytryptamine (5-HT), and studies suggest that the epithelial enterochromaffin (ECCs) cells are responsible for 90% of 5-HT secretion, while only 10% remains from the enteric nervous system.5454 Kim DY, Camilleri M. Serotonin: a mediator of the brain-gut connection. Am J Gastroenterol 2000;95(10):2698–2709 Bacteria, such as Escherichia, Streptococcus, Enterococcus, and Candida, are some gut microorganisms capable of producing serotonin.5555 Strandwitz P. Neurotransmitter modulation by the gut microbiota. Brain Res 2018;1693(Pt B):128–133 Brain functions can be affected by gut dysbiosis, as intestinal microorganisms influence not only serotonin synthesis/metabolism but also the synthesis of other neurotransmitters.44 Bravo JA, Forsythe P, Chew MV, et al. Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc Natl Acad Sci U S A 2011;108(38):16050–16055

Anxiety

Anxiety symptoms are frequent in patients with GI imbalance or GI disorders. For instance, more than 50% of patients diagnosed with irritable bowel syndrome have anxiety or depression as a comorbidity.5656 Whitehead WE, Palsson O, Jones KR. Systematic review of the comorbidity of irritable bowel syndrome with other disorders: what are the causes and implications? Gastroenterology 2002; 122(04):1140–1156

The correlation between anxiety symptoms and the alteration of gut microbiota diversity and complexity has been extensively documented. The factors implicated in this correlation are due to the role of gut microbiota and brain mutual signals, including neurotransmitter systems and immunologic factors. The mechanisms responsible for the close relationship between gut dysbiosis and anxiety are similar to those that correlate with gut microbiota imbalance and depression. Hypothalamic-pituitary-adrenal axis activation is the protagonist in stress and anxiety symptoms by activating and releasing adrenocorticotropic hormone (ACTH), which stimulates the production of glucocorticoids (e.g., cortisol) into the bloodstream.5757 Faravelli C, Lo Sauro C, Godini L, et al. Childhood stressful events, HPA axis and anxiety disorders. World J Psychiatry 2012;2(01): 13–25 The intestinal microbiota imbalance leads to activation of the HPA axis, which provokes mental condition alterations and mental disorders such as anxiety.5858 Huo R, Zeng B, Zeng L, et al. Microbiota Modulate Anxiety-Like Behavior and Endocrine Abnormalities in Hypothalamic-Pituitary-Adrenal Axis. Front Cell Infect Microbiol 2017;7:489

Although the anxiety pathophysiology is not completely clear, it is well known that neurotransmitters, including serotonin, dopamine, and GABA, are linked to anxiety disorders, and intestinal microbiota has been shown to alter all these neurotransmitter modulations in the brain. There is growing evidence indicating that this impairment triggered by dysbiosis can also explain the relationship of gut microbiota with anxiety disorder.5959 O’Mahony SM, Clarke G, Borre YE, Dinan TG, Cryan JF. Serotonin, tryptophan metabolism and the brain-gut-microbiome axis. Behav Brain Res 2015;277:32–48

Besides tryptophan and serotonin modulation, several experimental studies have demonstrated that gut microbiota similarly plays a role in dopamine modulation, one of the most important neurotransmitters involved in anxiety symptoms in distinct brain parts.6060 Huang F, Wu X. Brain Neurotransmitter Modulation by Gut Microbiota in Anxiety and Depression. Front Cell Dev Biol 2021; 9:649103

Autism spectrum disorder (ASD)

Numerous studies have already demonstrated that patients with ASD suffer from several gut-related comorbidities, including diarrhea, constipation, nausea, abdominal pain, vomiting, and reflux. Besides, their microbiota is distinctive from healthy subjects, and gut dysbiosis is much more frequent.6161 Song Y, Liu C, Finegold SM. Real-time PCR quantitation of clostridia in feces of autistic children. Appl Environ Microbiol 2004;70 (11):6459–6465

Studies have been showing that the chances of developing ASD are higher in some situations that trigger dysbiosis in early childhood, including children born by cesarean, children who were not breastfed, those who were treated with antibiotics during early childhood, children born to a mother that used antibiotics during pregnancy, and those whose mother had to be hospitalized due to infection when pregnant.

In a recent meta-analysis, researchers observed that reduced quantities of Streptococcus and Bifidobacterium were associated with ASD diagnosis. In contrast, another meta-analysis found that people with ASD present an intestinal microbiota with increased concentration of Bacteroides and Clostridium and decreased amounts of Bacteroidetes, Firmicutes, and Proteobacteria phyla.6262 Iglesias-Vázquez L, Van Ginkel Riba G, Arija V, Canals J. Composition of Gut Microbiota in Children with Autism Spectrum Disorder: A Systematic Review and Meta-Analysis. Nutrients 2020;12 (03):12

Schizophrenia and bipolar disorder

Similar to what has been observed in ASD, studies suggest that imbalance that might happen in newborns' gut microbiota seems to be associated with an increased risk of developing schizophrenia. Moreover, these patients also usually suffer from GI disorders.6363 Socała K, Doboszewska U, Szopa A, et al. The role of microbiotagut-brain axis in neuropsychiatric and neurological disorders. Pharmacol Res 2021;172:105840

Studies have shown that the infection of the gut microbiota by the protozoa Toxoplasma gondii seems to cause main changes in the intestinal composition, which are associated with an environmental risk factor for the onset of schizophrenia and bipolar disorder (BD).6464 Bhadra R, Cobb DA, Weiss LM, Khan IA. Psychiatric disorders in toxoplasma seropositive patients–the CD8 connection. Schizophr Bull 2013;39(03):485–489 The differences in gut microbiota diversity and quantity of patients with schizophrenia are still unclear as some studies show no difference, while others observed distinct variations. Elevated levels of Lactobacilli appear to be the only alteration that is consistent among studies.6363 Socała K, Doboszewska U, Szopa A, et al. The role of microbiotagut-brain axis in neuropsychiatric and neurological disorders. Pharmacol Res 2021;172:105840

Concerning the intestinal microbiota composition of patients with BD, it is also unclear how discrepant it is compared with healthy volunteers.6565 Kim YK, Shin C. The Microbiota-Gut-Brain Axis in Neuropsychiatric Disorders: Pathophysiological Mechanisms and Novel Treatments. Curr Neuropharmacol 2018;16(05):559–573 Nevertheless, studies show that it seems to be altered, as a lower amount of Faecalibacterium and Ruminococcaceae and higher levels of Actinobacteria and Coriobacteria were found when compared with healthy controls.6666 Painold A, Mörkl S, Kashofer K, et al. A step ahead: Exploring the gut microbiota in inpatients with bipolar disorder during a depressive episode. Bipolar Disord 2019;21(01):40–49 Another observation that implies an association between BD and dysbiosis is that evidence shows that hospitalized patients with acute mania received antimicrobial medications nearly twice as much as control patients, as they have an elevated rate of bacterial infections (Table 1).6767 Yolken R, Adamos M, Katsafanas E, et al. Individuals hospitalized with acute mania have increased exposure to antimicrobial medications. Bipolar Disord 2016;18(05):404–409

Dysbiosis treatment

The first guidance health professionals must understand and disseminate is the importance of maintaining a delicate gut microbiota balance as the top priority goal of dysbiosis prevention. Thus, factors related to microbiota imbalance that can consequently affect or even lead to the development of neuropathologies should be part of physicians' knowledge and orientation.6868 Parkin K, Christophersen CT, Verhasselt V, Cooper MN,Martino D. Risk Factors for Gut Dysbiosis in Early Life.Microorganisms 2021; 9(10):9

The factors that can negatively impact gut microbiota include delivery methods (cesarean sections); short, or absence of, breastfeeding (inclination for formula-feeding); early introduction of solid food (3 months of age or earlier); antibiotic use during pregnancy; antibiotics use in early life; and poor dietary habits and food choice (poor diets), including high consumption of ultraprocessed food, high rates of antibiotics consumption during life, drug use, smoking tobacco or electronic cigarettes, alcohol consumption, and sedentarism.6868 Parkin K, Christophersen CT, Verhasselt V, Cooper MN,Martino D. Risk Factors for Gut Dysbiosis in Early Life.Microorganisms 2021; 9(10):9

It is essential to highlight that early infancy is a relevant intestinal microbiota colonization period. Thus, raising awareness about gut microbiota during pregnancy and in the earliest years of life (at least until 3 years of age) is necessary to avoid dysbiosis, which is lined to extraintestinal diseases, such as neurological ones.6868 Parkin K, Christophersen CT, Verhasselt V, Cooper MN,Martino D. Risk Factors for Gut Dysbiosis in Early Life.Microorganisms 2021; 9(10):9

Another strategy to treat gut dysbiosis includes dietary modification.6969 Mörkl S, Butler MI, Holl A, Cryan JF, Dinan TG. Probiotics and the Microbiota-Gut-Brain Axis: Focus on Psychiatry. Curr Nutr Rep 2020;9(03):171–182 Dietary methods to treat dysbiosis and keep a healthy gut microbiota composition include limiting processed food as much as possible (Mediterranean diet). Furthermore, diets should incorporate fermented food that is rich in probiotics (e.g., kefir, yogurt with live active cultures, pickled vegetables, fermented milk products, kombucha tea, kimchi, and miso), high-fiber foods (e.g., fresh fruits, whole grains, vegetables, legumes, and beans), spices, and foods that are rich in polyphenols, oleic acids, and omega-3 fatty acids. Patients should avoid the consumption of saturated fatty acids, refined carbohydrates, and high protein and/or sugar intake.

Polyunsaturated fatty acids (PUFAs), especially omega 3, are essential in the human diet. A few studies have described the benefit of omega 3 in patients with PD. Although it is a good and safe profile with ease of administration, more studies with PUFAs, controlling for all possible covariates, must be done.7070 Bousquet M, Calon F, Cicchetti F. Impact of ω-3 fatty acids in Parkinson’s disease. Ageing Res Rev 2011;10(04):453–463

Antibiotics can inhibit or eliminate some microorganisms, causing the vanishing of some and the advent and progression of new bacterial species, with consequent dysbiosis. The knowledge about the relationship between the coexisting species and how they interact with their host is a challenge for new therapeutic approaches to treat dysbiosis.7171 Modi SR, Collins JJ, Relman DA. Antibiotics and the gutmicrobiota. J Clin Invest 2014;124(10):4212–4218 The action of antibiotics, such as ceftriaxone, moxycycline, minocycline, and rifampicin, has been related to gut dysbiosis and neuroprotection. Still, the long-term use due to toxicity and resistant bacterial selection is a risk in clinical practice.7272 Cankaya S, Cankaya B, Kilic U, Kilic E, Yulug B. The therapeutic role of minocycline in Parkinson’s disease. Drugs Context 2019; 8:212553

The probiotic (active microorganisms with health benefits), prebiotic (non-digestible food ingredients that stimulate the activity of particular microorganisms), and symbiotic (combination of probiotics and prebiotics) supplementation is a potential therapeutic intervention for gut dysbiosis, and it is considered safe for most of the population (Table 1).6969 Mörkl S, Butler MI, Holl A, Cryan JF, Dinan TG. Probiotics and the Microbiota-Gut-Brain Axis: Focus on Psychiatry. Curr Nutr Rep 2020;9(03):171–182, 7373 Tait C, Sayuk GS. The Brain-Gut-Microbiotal Axis: A framework for understanding functional GI illness and their therapeutic interventions. Eur J Intern Med 2021;84:1–99090 Reininghaus EZ, Wetzlmair LC, Fellendorf FT, et al. The Impact of Probiotic Supplements on Cognitive Parameters in Euthymic Individuals with Bipolar Disorder: A Pilot Study. Neuropsychobiology 2018;•••:1–8

As shown in Table 1, most of the revised manuscripts were randomized-controlled trials that administrated probiotics in patients diagnosed with brain, psychiatric, or sleep disorders, and most of them showed a high level of evidence (level II) of probiotics to treat gut dysbiosis and alleviate the symptoms of the studied disease, with no side effects or toxicity. Although the results are promising, the therapeutic approaches need to be better understood in relation to the mechanisms involved.

Psychobiotics are a type of probiotics that, when consumed in appropriate quantity, have the potential to improve mental health. Psychobiotics may improve mental health by downregulating the HPA axis, decreasing proinflammatory cytokines levels, attenuating the disruption of the BBB, enhancing the production of neurotransmitters such as serotonin and GABA, and modulating neurotransmitter balancing. These same types of probiotics might be beneficial for patients with sleep disorders, serving to alleviate symptoms by the same mechanisms.9191 Del Toro-Barbosa M, Hurtado-Romero A, Garcia-Amezquita LE, García-Cayuela T. Psychobiotics: Mechanisms of Action, Evaluation Methods and Effectiveness in Applications with Food Products. Nutrients 2020;12(12):12

Live biotherapeutic products (LBPs) are distinguished from probiotic supplements because they have evidence for treating or preventing diseases. They can be the result of engineering by addition, erasing, or adjusting genetic material within the organisms (E. coli, Bacteroides, and lactic acid bacteria).9292 Pedrolli DB, RibeiroNV, Squizato PN, de Jesus VN, CozettoDATeam AQA Unesp at iGEM 2017. Engineering Microbial Living Therapeutics: The Synthetic Biology Toolbox. Trends Biotechnol 2019; 37(01):100–115 Synthetic biology has enormous potential to contribute to treating patients with neurological diseases.9393 Riglar DT, Silver PA. Engineering bacteria for diagnostic and therapeutic applications. Nat Rev Microbiol 2018;16(04):214–225

Fecal microbiota transplantation (FMT) is a new promising therapeutic option for gut dysbiosis. Numerous studies have shown that transplanting feces from a healthy control into a patient diagnosed with a neurological disorder can alleviate the pathological conditions, signs, and symptoms.9494 Zhu F, Tu H, Chen T. The Microbiota-Gut-Brain Axis in Depression: The Potential Pathophysiological Mechanisms and Microbiota Combined Antidepression Effect. Nutrients 2022;14(10):14 9595 Xiao L, Yan J, Yang T, et al. Fecal Microbiome Transplantation from Children with Autism Spectrum Disorder Modulates Tryptophan and Serotonergic Synapse Metabolism and Induces Altered Behaviors in Germ-Free Mice. mSystems 2021;6 (02):6 In contrast, experimental studies showed that when researchers transferred fecal bacteria from a donor with a neurological disorder to a healthy recipient, the method induced the appearance of similar symptoms in the healthy host.5151 Suganya K, Koo BS. Gut-Brain Axis: Role of Gut Microbiota on Neurological Disorders and How Probiotics/Prebiotics Beneficially Modulate Microbial and Immune Pathways to Improve Brain Functions. Int J Mol Sci 2020;21(20):21 9494 Zhu F, Tu H, Chen T. The Microbiota-Gut-Brain Axis in Depression: The Potential Pathophysiological Mechanisms and Microbiota Combined Antidepression Effect. Nutrients 2022;14(10):14 Nevertheless, further studies are still necessary to validate this novel therapeutic method in humans with neurological disorders, as FMT can revolutionize the treatment of conditions other than GI diseases.

In conclusion, the increasing evidence about the existence of a close and bi-directional correlation between the gut microbiota and the brain indicates that dysbiosis plays an essential role in the development, function, and disorders of the CNS. Although more studies are still necessary, our review demonstrates that avoiding dysbiosis, especially during pregnancy and early childhood, is essential to prevent neurological disorders. Moreover, using probiotics by patients with psychiatric or brain diseases is safe in most cases. It can treat gut dysbiosis, decrease depression and anxiety scores, and alleviate the symptoms and severity of the studied neurological disorders. These observations point out the potential and safety of probiotics and prebiotics as adjuvants in preventing and treating neurological diseases.

  • Support
    The present work was supported by the Hospital São Paulo and the Department of Neurology and Neurosurgery of Universidade Federal de São Paulo, Brazil.

References

  • 1
    Eckburg PB, Bik EM, Bernstein CN, et al. Diversity of the human intestinal microbial flora. Science 2005;308(5728):1635–1638
  • 2
    Integrative HMPRNCIntegrative HMP (iHMP) Research Network Consortium. The Integrative Human Microbiome Project. Nature 2019;569(7758):641–648
  • 3
    Toor D,Wsson MK, Kumar P, et al. Dysbiosis Disrupts Gut Immune Homeostasis and Promotes Gastric Diseases. Int J Mol Sci 2019;20 (10):20
  • 4
    Bravo JA, Forsythe P, Chew MV, et al. Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc Natl Acad Sci U S A 2011;108(38):16050–16055
  • 5
    Allen AP, Hutch W, Borre YE, et al. Bifidobacterium longum 1714 as a translational psychobiotic:modulation of stress, electrophysiology and neurocognition in healthy volunteers. Transl Psychiatry 2016;6(11):e939
  • 6
    Houser MC, Tansey MG. The gut-brain axis: is intestinal inflammation a silent driver of Parkinson’s disease pathogenesis? NPJ Parkinsons Dis 2017;3:3
  • 7
    Frost G, Sleeth ML, Sahuri-Arisoylu M, et al. The short-chain fatty acid acetate reduces appetite via a central homeostatic mechanism. Nat Commun 2014;5:3611
  • 8
    Tang Q, Jin G, Wang G, et al. Current Sampling Methods for Gut Microbiota: A Call for More Precise Devices. Front Cell Infect Microbiol 2020;10:151
  • 9
    Wei S, BahlMI, Baunwall SMD, Hvas CL, Licht TR. Determining Gut Microbial Dysbiosis: a Review of Applied Indexes for Assessment of Intestinal Microbiota Imbalances. Appl Environ Microbiol 2021;87(11):87
  • 10
    Brüssow H. Problems with the concept of gut microbiota dysbiosis. Microb Biotechnol 2020;13(02):423–434
  • 11
    Horiuchi Y, Kimura R, Kato N, et al. Evolutional study on acetylcholine expression. Life Sci 2003;72(15):1745–1756
  • 12
    Cattaneo A, Cattane N, Galluzzi S, et al; INDIA-FBP Group. Association of brain amyloidosis with pro-inflammatory gut bacterial taxa and peripheral inflammation markers in cognitively impaired elderly. Neurobiol Aging 2017;49:60–68
  • 13
    Chen Y, Xu J, Chen Y. Regulation of Neurotransmitters by the Gut Microbiota and Effects on Cognition in Neurological Disorders. Nutrients 2021;13(06):13
  • 14
    Cummings J, Aisen P, Apostolova LG, Atri A, Salloway S,Weiner M. Aducanumab: Appropriate Use Recommendations. J Prev Alzheimers Dis 2021;8(04):398–410
  • 15
    de Rijke TJ, Doting MHE, van Hemert S, et al. A Systematic Review on the Effects of Different Types of Probiotics in Animal Alzheimer’s Disease Studies. Front Psychiatry 2022;13:879491
  • 16
    Cirstea MS, Sundvick K, Golz E, et al. The Gut Mycobiome in Parkinson’s Disease. J Parkinsons Dis 2021;11(01):153–158
  • 17
    Sampson TR, Debelius JW, Thron T, et al. Gut Microbiota Regulate Motor Deficits and Neuroinflammation in a Model of Parkinson’s Disease. Cell 2016;167(06):1469–1480.e12
  • 18
    Lorente-Picón M, Laguna A. New Avenues for Parkinson’s Disease Therapeutics: Disease-Modifying Strategies Based on the Gut Microbiota. Biomolecules 2021;11(03):11
  • 19
    PalaciosN, Hannoun A, Flahive J, et al. Effect of Levodopa Initiation on the Gut Microbiota in Parkinson’s Disease. Front Neurol 2021; 12:574529
  • 20
    Xue LJ, Yang XZ, Tong Q, et al. Fecal microbiota transplantation therapy for Parkinson’s disease: A preliminary study. Medicine (Baltimore) 2020;99(35):e22035
  • 21
    Jangi S, Gandhi R, Cox LM, et al. Alterations of the human gut microbiome in multiple sclerosis. Nat Commun 2016; 7:12015
  • 22
    Adamczyk-Sowa M, Medrek A, Madej P, Michlicka W, Dobrakowski P. Does the Gut Microbiota Influence Immunity and Inflammation in Multiple Sclerosis Pathophysiology? J Immunol Res 2017;2017:7904821
  • 23
    Schepici G, Silvestro S, Bramanti P, Mazzon E. The Gut Microbiota in Multiple Sclerosis: An Overview of Clinical Trials. Cell Transplant 2019;28(12):1507–1527
  • 24
    Dahlin M, Prast-Nielsen S. The gut microbiome and epilepsy. EBioMedicine 2019;44:741–746
  • 25
    Ceccarani C, Viganò I, Ottaviano E, et al. Is Gut Microbiota a Key Player in Epilepsy Onset? A Longitudinal Study in Drug-Naive Children. Front Cell Infect Microbiol 2021;11:749509
  • 26
    International Classification of Sleep Disorders – Third edition (ICSD-3). 3rd ed. American Academy of Sleep Medicine; 2014
  • 27
    Veasey SC, Rosen IM. Obstructive Sleep Apnea in Adults. N Engl J Med 2019;380(15):1442–1449
  • 28
    Badran M, Mashaqi S, Gozal D. The gut microbiome as a target for adjuvant therapy in obstructive sleep apnea. Expert Opin Ther Targets 2020;24(12):1263–1282
  • 29
    Valentini F, Evangelisti M, Arpinelli M, et al. Gut microbiota composition in childrenwith obstructive sleep apnoea syndrome: a pilot study. Sleep Med 2020;76:140–147
  • 30
    Durgan DJ. Obstructive Sleep Apnea-Induced Hypertension: Role of the Gut Microbiota. Curr Hypertens Rep 2017;19(04):35
  • 31
    Liew SC, Aung T. Sleep deprivation and its association with diseases- a review. Sleep Med 2021;77:192–204
  • 32
    Reynolds AC, Paterson JL, Ferguson SA, Stanley D, Wright KP Jr, Dawson D. The shift work and health research agenda: Considering changes in gut microbiota as a pathway linking shift work, sleep loss and circadian misalignment, and metabolic disease. Sleep Med Rev 2017;34:3–9
  • 33
    Wang Z, Chen WH, Li SX, et al. Gut microbiota modulates the inflammatory response and cognitive impairment induced by sleep deprivation. Mol Psychiatry 2021;26(11):6277–6292
  • 34
    Karl JP, Hatch AM, Arcidiacono SM, et al. Effects of Psychological, Environmental and Physical Stressors on the Gut Microbiota. Front Microbiol 2018;9:2013
  • 35
    Smith RP, Easson C, Lyle SM, et al. Gut microbiome diversity is associated with sleep physiology in humans. PLoS One 2019;14 (10):e0222394
  • 36
    Li Y, Zhang B, Zhou Y, et al. Gut Microbiota Changes and Their Relationship with Inflammation in Patients with Acute and Chronic Insomnia. Nat Sci Sleep 2020;12:895–905
  • 37
    Park YS, Kim SH, Park JW, et al. Melatonin in the colon modulates intestinal microbiota in response to stress and sleep deprivation. Intest Res 2020;18(03):325–336
  • 38
    Hernández-García J, Navas-Carrillo D, Orenes-Piñero E. Alterations of circadian rhythms and their impact on obesity, metabolic syndrome and cardiovascular diseases. Crit Rev Food Sci Nutr 2020;60(06):1038–1047
  • 39
    Voigt RM, Forsyth CB, Green SJ, Engen PA, Keshavarzian A. Circadian Rhythm and the Gut Microbiome. Int Rev Neurobiol 2016;131:193–205
  • 40
    Parekh PJ, Oldfield EC IV, Johnson DA. The Effects of Sleep on the Commensal Microbiota: Eyes Wide Open? J Clin Gastroenterol 2018;52(03):204–209
  • 41
    Anderson JR, Carroll I, Azcarate-Peril MA, et al. A preliminary examination of gut microbiota, sleep, and cognitive flexibility in healthy older adults. Sleep Med 2017;38:104–107
  • 42
    Hua X, Zhu J, Yang T, et al. The Gut Microbiota and Associated Metabolites Are Altered in Sleep Disorder of Children With Autism Spectrum Disorders. Front Psychiatry 2020;11:855
  • 43
    Lecomte A, Barateau L, Pereira P, et al. Gutmicrobiota composition is associated with narcolepsy type 1. Neurol Neuroimmunol Neuroinflamm 2020;7(06):7
  • 44
    Wagner-Skacel J, Dalkner N, Moerkl S, et al. Sleep and Microbiome in Psychiatric Diseases. Nutrients 2020;12(08):12
  • 45
    Organization WH. Depression and Other Common Mental Disorders Global Health Estimates.World Health Organization; 2017
  • 46
    Canet-Juric L, Andrés ML, Del Valle M, et al. A Longitudinal Study on the Emotional Impact Cause by the COVID-19 Pandemic Quarantine on General Population. Front Psychol 2020; 11:565688
  • 47
    Bet PM, Hugtenburg JG, Penninx BW, Hoogendijk WJ. Side effects of antidepressants during long-term use in a naturalistic setting. Eur Neuropsychopharmacol 2013;23(11):1443–1451
  • 48
    Bowers HM, Williams SJ, Geraghty AWA, et al. Helping people discontinue long-term antidepressants: views of health professionals in UK primary care. BMJ Open 2019;9(07):e027837
  • 49
    Dinan TG, Cryan JF. Brain-Gut-Microbiota Axis andMental Health. Psychosom Med 2017;79(08):920–926
  • 50
    Foster JA, Baker GB, Dursun SM. The Relationship Between the Gut Microbiome-Immune System-Brain Axis and Major Depressive Disorder. Front Neurol 2021;12:721126
  • 51
    Suganya K, Koo BS. Gut-Brain Axis: Role of Gut Microbiota on Neurological Disorders and How Probiotics/Prebiotics Beneficially Modulate Microbial and Immune Pathways to Improve Brain Functions. Int J Mol Sci 2020;21(20):21
  • 52
    Misiak B, Łoniewski I, MarliczW, et al. The HPA axis dysregulation in severe mental illness: Can we shift the blame to gut microbiota? Prog Neuropsychopharmacol Biol Psychiatry 2020; 102:109951
  • 53
    Waclawiková B, El Aidy S. Role of Microbiota and Tryptophan Metabolites in the Remote Effect of Intestinal Inflammation on Brain and Depression. Pharmaceuticals (Basel) 2018;11(03):11
  • 54
    Kim DY, Camilleri M. Serotonin: a mediator of the brain-gut connection. Am J Gastroenterol 2000;95(10):2698–2709
  • 55
    Strandwitz P. Neurotransmitter modulation by the gut microbiota. Brain Res 2018;1693(Pt B):128–133
  • 56
    Whitehead WE, Palsson O, Jones KR. Systematic review of the comorbidity of irritable bowel syndrome with other disorders: what are the causes and implications? Gastroenterology 2002; 122(04):1140–1156
  • 57
    Faravelli C, Lo Sauro C, Godini L, et al. Childhood stressful events, HPA axis and anxiety disorders. World J Psychiatry 2012;2(01): 13–25
  • 58
    Huo R, Zeng B, Zeng L, et al. Microbiota Modulate Anxiety-Like Behavior and Endocrine Abnormalities in Hypothalamic-Pituitary-Adrenal Axis. Front Cell Infect Microbiol 2017;7:489
  • 59
    O’Mahony SM, Clarke G, Borre YE, Dinan TG, Cryan JF. Serotonin, tryptophan metabolism and the brain-gut-microbiome axis. Behav Brain Res 2015;277:32–48
  • 60
    Huang F, Wu X. Brain Neurotransmitter Modulation by Gut Microbiota in Anxiety and Depression. Front Cell Dev Biol 2021; 9:649103
  • 61
    Song Y, Liu C, Finegold SM. Real-time PCR quantitation of clostridia in feces of autistic children. Appl Environ Microbiol 2004;70 (11):6459–6465
  • 62
    Iglesias-Vázquez L, Van Ginkel Riba G, Arija V, Canals J. Composition of Gut Microbiota in Children with Autism Spectrum Disorder: A Systematic Review and Meta-Analysis. Nutrients 2020;12 (03):12
  • 63
    Socała K, Doboszewska U, Szopa A, et al. The role of microbiotagut-brain axis in neuropsychiatric and neurological disorders. Pharmacol Res 2021;172:105840
  • 64
    Bhadra R, Cobb DA, Weiss LM, Khan IA. Psychiatric disorders in toxoplasma seropositive patients–the CD8 connection. Schizophr Bull 2013;39(03):485–489
  • 65
    Kim YK, Shin C. The Microbiota-Gut-Brain Axis in Neuropsychiatric Disorders: Pathophysiological Mechanisms and Novel Treatments. Curr Neuropharmacol 2018;16(05):559–573
  • 66
    Painold A, Mörkl S, Kashofer K, et al. A step ahead: Exploring the gut microbiota in inpatients with bipolar disorder during a depressive episode. Bipolar Disord 2019;21(01):40–49
  • 67
    Yolken R, Adamos M, Katsafanas E, et al. Individuals hospitalized with acute mania have increased exposure to antimicrobial medications. Bipolar Disord 2016;18(05):404–409
  • 68
    Parkin K, Christophersen CT, Verhasselt V, Cooper MN,Martino D. Risk Factors for Gut Dysbiosis in Early Life.Microorganisms 2021; 9(10):9
  • 69
    Mörkl S, Butler MI, Holl A, Cryan JF, Dinan TG. Probiotics and the Microbiota-Gut-Brain Axis: Focus on Psychiatry. Curr Nutr Rep 2020;9(03):171–182
  • 70
    Bousquet M, Calon F, Cicchetti F. Impact of ω-3 fatty acids in Parkinson’s disease. Ageing Res Rev 2011;10(04):453–463
  • 71
    Modi SR, Collins JJ, Relman DA. Antibiotics and the gutmicrobiota. J Clin Invest 2014;124(10):4212–4218
  • 72
    Cankaya S, Cankaya B, Kilic U, Kilic E, Yulug B. The therapeutic role of minocycline in Parkinson’s disease. Drugs Context 2019; 8:212553
  • 73
    Tait C, Sayuk GS. The Brain-Gut-Microbiotal Axis: A framework for understanding functional GI illness and their therapeutic interventions. Eur J Intern Med 2021;84:1–9
  • 74
    Long-Smith C, O’Riordan KJ, Clarke G, Stanton C, Dinan TG, Cryan JF. Microbiota-Gut-Brain Axis: New Therapeutic Opportunities. Annu Rev Pharmacol Toxicol 2020;60:477–502
  • 75
    TamtajiOR, Taghizadeh M, Daneshvar Kakhaki R, et al. Clinical and metabolic response to probiotic administration in people with Parkinson’s disease: A randomized, double-blind, placebo-controlled trial. Clin Nutr 2019;38(03):1031–1035
  • 76
    Agahi A, Hamidi GA, Daneshvar R, et al. Does Severity of Alzheimer’s Disease Contribute to Its Responsiveness to Modifying Gut Microbiota? A Double Blind Clinical Trial. Front Neurol 2018; 9:662
  • 77
    Barichella M, Pacchetti C, Bolliri C, et al. Probiotics and prebiotic fiber for constipation associated with Parkinson disease: An RCT. Neurology 2016;87(12):1274–1280
  • 78
    Rahimlou M, Nematollahi S, Husain D, Banaei-Jahromi N, Majdinasab N, Hosseini SA. Probiotic supplementation and systemic inflammation in relapsing-remitting multiple sclerosis: A randomized, double-blind, placebo-controlled trial. Front Neurosci 2022;16:901846
  • 79
    Kouchaki E, Tamtaji OR, Salami M, et al. Clinical and metabolic response to probiotic supplementation in patients with multiple sclerosis: A randomized, double-blind, placebo-controlled trial. Clin Nutr 2017;36(05):1245–1249
  • 80
    Lee HJ, Hong JK, Kim JK, et al. Effects of Probiotic NVP-1704 on Mental Health and Sleep in Healthy Adults: An 8-Week Randomized, Double-Blind, Placebo-Controlled Trial. Nutrients 2021;13 (08):13
  • 81
    Ho YT, Tsai YC, Kuo TBJ, Yang CCH. Effects of Lactobacillus plantarum PS128 on Depressive Symptoms and Sleep Quality in Self-Reported Insomniacs: A Randomized, Double-Blind, Placebo-Controlled Pilot Trial. Nutrients 2021;13(08):13
  • 82
    Kazemi A, Noorbala AA, AzamK, EskandariMH, Djafarian K. Effect of probiotic and prebiotic vs placebo on psychological outcomes in patients with major depressive disorder: A randomized clinical trial. Clin Nutr 2019;38(02):522–528
  • 83
    Sanchez M, Darimont C, Panahi S, et al. Effects of a Diet-Based Weight-Reducing Program with Probiotic Supplementation on Satiety Efficiency, Eating Behaviour Traits, and Psychosocial Behaviours in Obese Individuals. Nutrients 2017;9(03):9
  • 84
    Qin Q, Liu H, Yang Y, et al. Probiotic Supplement Preparation Relieves Test Anxiety by Regulating Intestinal Microbiota in College Students. Dis Markers 2021;2021:5597401
  • 85
    Slykerman RF, Hood F, Wickens K, et al; Probiotic in Pregnancy Study Group. Effect of Lactobacillus rhamnosus HN001 in Pregnancy on Postpartum Symptoms of Depression and Anxiety: A Randomised Double-blind Placebo-controlled Trial. EBioMedicine 2017;24:159–165
  • 86
    Shaaban SY, El Gendy YG,Mehanna NS, et al. The role of probiotics in children with autism spectrum disorder: A prospective, openlabel study. Nutr Neurosci 2018;21(09):676–681
  • 87
    Santocchi E, Guiducci L, Prosperi M, et al. Effects of Probiotic Supplementation on Gastrointestinal, Sensory and Core Symptoms in Autism Spectrum Disorders: A Randomized Controlled Trial. Front Psychiatry 2020;11:550593. Doi: 10.3389/fpsyt.2020.550593
    » https://doi.org/10.3389/fpsyt.2020.550593
  • 88
    Ghaderi A, Banafshe HR, Mirhosseini N, et al. Clinical and metabolic response to vitamin D plus probiotic in schizophrenia patients. BMC Psychiatry 2019;19(01):77
  • 89
    Tomasik J, Yolken RH, Bahn S, Dickerson FB. Immunomodulatory Effects of Probiotic Supplementation in Schizophrenia Patients: A Randomized, Placebo-Controlled Trial. Biomark Insights 2015; 10:47–54
  • 90
    Reininghaus EZ, Wetzlmair LC, Fellendorf FT, et al. The Impact of Probiotic Supplements on Cognitive Parameters in Euthymic Individuals with Bipolar Disorder: A Pilot Study. Neuropsychobiology 2018;•••:1–8
  • 91
    Del Toro-Barbosa M, Hurtado-Romero A, Garcia-Amezquita LE, García-Cayuela T. Psychobiotics: Mechanisms of Action, Evaluation Methods and Effectiveness in Applications with Food Products. Nutrients 2020;12(12):12
  • 92
    Pedrolli DB, RibeiroNV, Squizato PN, de Jesus VN, CozettoDATeam AQA Unesp at iGEM 2017. Engineering Microbial Living Therapeutics: The Synthetic Biology Toolbox. Trends Biotechnol 2019; 37(01):100–115
  • 93
    Riglar DT, Silver PA. Engineering bacteria for diagnostic and therapeutic applications. Nat Rev Microbiol 2018;16(04):214–225
  • 94
    Zhu F, Tu H, Chen T. The Microbiota-Gut-Brain Axis in Depression: The Potential Pathophysiological Mechanisms and Microbiota Combined Antidepression Effect. Nutrients 2022;14(10):14
  • 95
    Xiao L, Yan J, Yang T, et al. Fecal Microbiome Transplantation from Children with Autism Spectrum Disorder Modulates Tryptophan and Serotonergic Synapse Metabolism and Induces Altered Behaviors in Germ-Free Mice. mSystems 2021;6 (02):6

Publication Dates

  • Publication in this collection
    28 Aug 2023
  • Date of issue
    2023

History

  • Received
    13 Oct 2022
  • Reviewed
    01 Dec 2022
  • Accepted
    04 Dec 2022
Academia Brasileira de Neurologia - ABNEURO R. Vergueiro, 1353 sl.1404 - Ed. Top Towers Offices Torre Norte, 04101-000 São Paulo SP Brazil, Tel.: +55 11 5084-9463 | +55 11 5083-3876 - São Paulo - SP - Brazil
E-mail: revista.arquivos@abneuro.org