Next Article in Journal
Standardization of the [68Ga]Ga-PSMA-11 Radiolabeling Protocol in an Automatic Synthesis Module: Assessments for PET Imaging of Prostate Cancer
Next Article in Special Issue
Magnesium in Ketamine Administration in Treatment-Resistant Depression
Previous Article in Journal
Sleep-Based Interventions in Alzheimer’s Disease: Promising Approaches from Prevention to Treatment along the Disease Trajectory
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Probiotics as a Treatment for “Metabolic Depression”? A Rationale for Future Studies

by
Oliwia Gawlik-Kotelnicka
* and
Dominik Strzelecki
Department of Affective and Psychotic Disorders, Medical University of Lodz, 90-419 Lodz, Poland
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2021, 14(4), 384; https://doi.org/10.3390/ph14040384
Submission received: 31 March 2021 / Revised: 16 April 2021 / Accepted: 18 April 2021 / Published: 20 April 2021
(This article belongs to the Special Issue Psychopharmacology of Affective Disorders)

Abstract

:
Depression and metabolic diseases often coexist, having several features in common, e.g., chronic low-grade inflammation and intestinal dysbiosis. Different microbiota interventions have been proposed to be used as a treatment for these disorders. In the paper, we review the efficacy of probiotics in depressive disorders, obesity, metabolic syndrome and its liver equivalent based on the published experimental studies, clinical trials and meta-analyses. Probiotics seem to be effective in reducing depressive symptoms when administered in addition to antidepressants. Additionally, probiotics intake may ameliorate some of the clinical components of metabolic diseases. However, standardized methodology regarding probiotics use in clinical trials has not been established yet. In this narrative review, we discuss current knowledge on the recently used methodology with its strengths and limitations and propose criteria that may be implemented to create a new study of the effectiveness of probiotics in depressive disorders comorbid with metabolic abnormalities. We put across our choice on type of study population, probiotics genus, strains, dosages and formulations, intervention period, as well as primary and secondary outcome measures.

1. Introduction

Depressive disorders and metabolic syndrome (MetS) are two of the most common and disabling civilization diseases [1]. Depression is a major risk factor for everyday disablement or suicide, and MetS may lead to cardiovascular diseases (CVD) and type 2 diabetes mellitus (2DM). Additionally, depressive disorders are often comorbid with MetS increasing mortality risks [2,3]. A former meta-analysis [4] showed that individuals with depression suffered many metabolic abnormalities and had a 1.5 times higher odds of having MetS; prevalence of MetS in depressive subjects accounted for 30%. A very recent meta-analysis of 49 studies confirmed a significant relationship between depression and MetS (the pooled Odds Ratio of MetS in patients with depression was 1.48 % in 31 cross-sectional studies and the pooled Risk Ratio 1.38 in cohort studies) [5]. Moreover, data from an 808-person sample with a current diagnosis of depression did demonstrate that persons with atypical depression had significantly higher levels of inflammatory markers, body mass index (BMI), waist circumference (WC) and triglycerides (TG), and lower high-density lipoprotein cholesterol (HDL-C) than subjects with melancholic depression [6]. Additionally, in a large, nationally representative sample, it was found that both obesity and MetS were associated with significant depressive symptoms independent of each other, and that participants with both conditions had the highest rate of depression compared to the other groups [7]. Another state, strongly associated with both depressive and MetS issues, is non-alcoholic fatty liver disease (NAFLD). NAFLD is a multisystem disease that is considered the hepatic manifestation of MetS and is characterized by excessive hepatic fat accumulation [8]. To underline its nature, recently it has been proposed to rename the syndrome as metabolic-associated fatty liver disease (MAFLD) [9]. Moreover, similarly to MetS, most deaths among NAFLD patients are attributable to CVD [10]. Altogether, the term "metabolic syndrome type II" was proposed in 2007 as a neuropsychiatric syndrome in which alterations in metabolic networks are a defining pathophysiological component [11]. In this narrative review, we use the term “metabolic depression” to describe depressive disorders comorbid with obesity, MetS or NAFLD.
Although the exact mechanisms underlying this association between depression, MetS and its liver equivalent are poorly known, several hypotheses have been proposed. First of all, antipsychotics augmentation in depression is associated with significantly higher metabolic abnormalities prevalence and increased mortality risk [12]. Secondly, lifestyle factors, especially diet and physical activity, explained more than one fifth of the association between depressive and metabolic disorders [13]. Remaining factors participating in the coexistence of depressive and metabolic diseases are poorly known; however, a possible pathophysiological overlap has been proposed [14].
The hypothalamic–pituitary–adrenal (HPA) axis dysregulation with final hypothalamic inflammation is one of the factors depression and MetS have in common [15]. In typical depression, the axis is upregulated with excess release of cortisol [16]. However, atypical depression is associated with a hypofunction of the HPA axis. In metabolic disorders, rather, neuroendocrine dysregulatory mechanisms are involved [17]. Furthermore, studies have uncovered that both depression and metabolic diseases are associated with chronic, low-grade inflammation [7,18,19]. There is also a number of studies that oxidative stress (OxS) may be involved in the pathophysiology of metabolic [20,21] as well as depressive disorders [22,23,24].
In recent years, there has been much interest in the role of microbiota changes (dysbiosis) in the development of chronic inflammation and civilization diseases [25]. Intestinal microbiota play an important role in regulating the brain functions of the host through the gut–brain axis (GBA) [26]. The gut dysbiosis has been found to play a significant role in the occurrence of mood and anxiety disorders [27,28,29,30]. Additionally, there is scientific data on the participation of the gut microbiota dysfunction in the onset of obesity-related disorders [31,32], as well as the pathogenesis of NAFLD [33,34]. Additionally, there is more and more evidence that an aberrant gut microbiota may lead to chronic inflammation [35,36] and OxS exacerbation in tissues [37] so that may serve as a link between MDs, depression and dysbiosis.
Elucidating these mechanisms linking metabolic diseases, depression, HPA dysregulation, CLGI, OxS and dysbiosis could generate potential new therapeutic means or patient-specific strategies to combat both metabolic and depressive disorders, e.g., microbiota interventions.

2. Microbiota Interventions

Several possible interventions on our microbiota have been described in the literature [38,39]. Of them, proper diet and other lifestyle factors are established ways of alleviating symptoms of metabolic diseases, as well as an adjunctive method in the treatment of depression [40,41,42]. Results of research on prebiotics have suggested favourable outcomes in metabolic disorders: decreased fasting glucose (fGlc), improved insulin sensitivity and lipid profile, reduced inflammation including neuroinflammation; however, when investigated as isolated therapies, prebiotics did not impact outcomes’ measures of depression or anxiety [43,44,45,46,47]. Interestingly, there appears to be some evidence for the treatment of psychiatric and metabolic disorders through fecal microbiota transplantation (FMT); additionally, FMT did have the potential to reduce small intestinal permeability in patients with NAFLD [48,49,50]. Another clinical intervention involving microbiota is the use of postbiotics (metabiotics). The most common types of postbiotics are microbial metabolites: short-chain fatty acids (SCFAs), peptides, enzymes, teichoic acids, and vitamins [51]. Among others, it was shown that lipoteichoic acid from B. animalis strain was responsible for its fat-reducing properties [52] and SCFAs administration alleviated symptoms of depression in mice [53]. SCFAs may also be used as a candidate agent in the prevention and treatment of obesity by inducing thermogenesis in brown adipose tissue and browning in white adipose tissue [54].
The intervention on our microbiota being the core topic of this review is supplementation with probiotics. The most accepted definition of probiotics is “live microorganisms, that when consumed in adequate amounts, confer a health effect on the host” [55].

2.1. Probiotics in Experimental Studies

Experimental studies have uncovered psychoactive properties of several probiotics formulations in different models of GBA dysfunction as well as in animal models of mental health problems, including anxiety and depression. Lactobacillus (L.) rhamnosus JB-1 was shown to prevent some of the antibiotic-induced alterations in rodents, e.g., impaired anxiety and social behaviours, as well as increased levels of aggression [56]. Moreover, Bifidobacterium (B.) longum subsp. infantis E41 and B. breve M2CF22M7 were shown to have an antidepressant effect in mice [57]. Importantly, probiotic pretreatment (L. helveticus R0052 + B. longum R0175) significantly alleviated hippocampal apoptosis induced by lipopolysaccharide in rats, suggesting that this probiotic could play a role in some neurodegenerative conditions [58,59]. Furthermore, not only stress-induced anxiety or depressive-like behaviours, but also cognitive deficits along with the reduced level of brain-derived neurotropic factor (BDNF) were alleviated by L. plantarum WLPL04 [60]. Additionally, overall, probiotics reduced anxiety-like behaviour in animals, but only among diseased ones [61]. Furthermore, the comparison of efficacy of twelve candidate probiotic strains of Bifidobacterium and Lactobacillus in a dose of 1 × 109 CFU/day on chronically stressed mice showed that L. paracasei Lpc-37, L. plantarum LP12407, L. plantarum LP12418 and L. plantarum LP12151 prevented stress-associated anxiety and depression-related behaviours [62] adding data on probiotics strain-dependence. On the contrary, L. helveticus R0052 and B. longum R0175 promoted an anti-inflammatory profile but not reductions in behavioural responses to social stress in hamsters [63].
As regards to metabolic abnormalities, L. fermentum alleviated inflammation and intestinal barrier integrity dysfunction, as well as improved insulin sensitivity in diet- and streptozotocin-induced diabetes in rats [64]. In other studies, L. fermentum CECT5716 exerted anti-obesity effects, associated with its anti-inflammatory properties and ameliorated endothelial dysfunction and gut dysbiosis in a model of high-fat diet (HFD)-induced obesity in mice [65]; and L. fermentum CRL1446 administration improved adiposity index, inflammatory, oxidative, glucose and lipid profiles and favourably modulated intestinal microbiota in mice with MetS [66]. Similarly, the administration of B. animalis subsp. lactis BB-12 and L. plantarum 299v to diet-induced obesity and MetS in rabbits demonstrated favourable effects on several metabolic abnormalities [67]. In concordance with that, the administration of L. acidophilus probiotic in rats with MetS caused by fructose reduced insulin resistance (IR) [68]. As regards MetS-associated OxS and liver function biomarkers, L. pentosus GSSK2 and L. plantarum GS26A were equally effective in HFD-induced MetS in rats [69]. Additionally, in preclinical studies, a number of probiotic genus, such as Bifidobacterium, Lactobacillus, or Bacillus, have shown beneficial effects in rodent models of NAFLD [70,71,72,73] and probiotics are now considered as a potential therapy for human NAFLD.
Interestingly, intervention with GABA-producing L. brevis DPC6108 and L. brevis DSM32386 improved both metabolic abnormalities and depressive-like behaviour associated with MetS in mice [74].
Altogether, the results have provided an experimental basis for the prophylactic and adjunct therapeutic application of probiotics on depression comorbid with lifestyle-related disorders such as obesity, MetS and its aftereffects. Furthermore, the experimental studies results may serve as an indicator for selecting specific strains and dosages of probiotics. However, animals’ results cannot always be transferred to human studies (Table 1).

2.2. Probiotics in Human Studies

2.2.1. Systematic Reviews and Meta-Analyses of Randomized Clinical Trials (RCTs) of Probiotics Interventions

Systematic reviews and meta-analyses of human trials using probiotics demonstrated their usefulness in depressive or, less consistently, in anxiety outcome measures [46,47,61,75,76,77,78,79,80,81,82,83,84,85,86,87]. It has been suggested that the microorganisms can form a new group of drugs named “psychobiotics” [88].
The first meta-analysis in the field showed that probiotics significantly decreased the depression scale score. However, among five included studies only one had investigated patients with major depressive disorder (MDD) and four the healthy population. Regarding patients’ age, probiotics had an effect on the population aged under 60, but not on people aged over 65 [81]. In another study, there was no significant difference in mood between the probiotic and placebo group post-intervention; nonetheless, a subgroup analysis of studies conducted in healthy versus depressed subjects showed significant improvements in the mild-to-moderate depression group [82]. Similarly, a beneficial effect of probiotics on depressive symptoms when administered to clinical/medical samples has been shown in several meta-analyses [46,75,79,80,85,87], with a larger effect observed for psychiatric, especially MDD, samples, for longer intervention periods (more than a month) and with multiple strains formulations [46,85,87]. Moreover, a significant reduction in Hamilton Depression Rating Scale (HDRS), C-reactive protein (CRP), interleukin (IL)-10 and malondialdehyde (MDA) levels was found in patients with psychiatric disorders after probiotics supplementation [75]. According to the newest meta-analysis of RCTs in clinical depression samples, probiotics are effective in reducing depressive symptoms when administered in addition to antidepressants; however, they are not significantly advantageous over placebo when used as stand-alone treatment [78].
Additionally, with regard to anxiety, the meta-analyses revealed no significant difference between probiotics and placebo in alleviating anxiety symptoms, and did not differentially affect clinical and healthy human samples [61,76,79].
The recent systematic review and meta-analysis has confirmed the effectiveness of probiotics on the amelioration of anthropometric measures (body mass index (BMI), waist circumference (WC) and hip circumference (HC)) of overweight and obese patients with related metabolic diseases [89]. This is partly in agreement with several previous meta-analyses in obese subjects [90,91,92,93,94,95,96,97]; however, the studies lack consistency [98,99,100]. The meta-analysis assessing the effect of probiotics on risk factors of cardiometabolic diseases in healthy people revealed reduced BMI and WC and, additionally, total cholesterol (TC) in overweight subjects. Most of the genera were Lactobacillus and Bifidobacterium, the mean time of probiotic administration was 67 days and the daily probiotic dose varied between 106 and 1010 colony-forming units (CFU)/gram [101]. Another meta-analysis confirmed that the improvements in metabolic variables were mostly observed with Bifidobacterium and Lactobacillus genera, adding data on favourable effects of Streptococcus salivarius subsp. thermophilus and mixtures of probiotic strains [93]. Contrary to the previously cited results, probiotic supplements did not have favourable effects in overweight or obese children and adolescents [102], nor were they superior to placebo in overweight or obese pregnant women for the prevention of gestational diabetes mellitus [103]. Additionally, probiotics may have no-to-minor effect regarding weight loss post bariatric surgery [104].
Furthermore, it has been summarized that probiotic intake may ameliorate some of the clinical components of MetS but the results are inconclusive [93,96,97,98,105,106,107]. Analysis of patients with CVD risk factors revealed favourable effects of probiotics with longer duration of treatment (>1.5 months), higher dosage of probiotics (>1.0 × 109 CFU), diabetic patients and female populations [97]. Another systematic review of 6 RCTs of probiotic strains for modulating obesity-related microbiota dysbiosis showed that Lactobacillus genus was administered twice as often than Bifidobacterium. Additionally, the daily dose varied from 1×108 to 1.35 × 1015 CFU/day, and the time of administration varied from 4 to 24 weeks [108].
Importantly, levels of some inflammatory biomarkers associated with MetS were found to be decreased after probiotics treatment, e.g., serum CRP, the soluble vascular cell adhesion molecule 1 (sVCAM-1), IL-6, tumour necrosis factor α (TNF-α), vascular endothelial growth factor (VEGF), and thrombomodulin [100,107,109]. Similarly, an improvement in clinical features as well as in OxS biomarkers was found in type 2 diabetes mellitus (2DM) patients [110] or polycystic ovary syndrome (PCOS) patients [111] after probiotic supplementation.
Meanwhile, clinical research on probiotics conducted so far has found positive results in NAFLD subjects. Probiotics in recent systematic reviews and meta-analyses were shown to be superior to placebo regarding several anthropometric and laboratory metabolic parameters, including serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT) in both adult and paediatric patients [93,95,112], as well as inflammation markers [112,113] in NAFLD patients.
To summarize, most systematic reviews and meta-analyses have demonstrated some benefit with respect to body weight loss and BMI among adult participants; however, these changes were rather discreet and of little significance for overall health status. The results concerning other criteria of MetS are inconclusive. However, there seems to be an agreement in term of probiotics efficacy toward NAFLD severity.
Table 2 shows a brief summary of meta-analyses of RCTs with probiotics in the field of depressive and anxiety symptoms, metabolic parameters, including obesity, MetS, and its liver equivalent published in recent years.
To conclude, it seems reasonable that current probiotics formulations should only be used as a complementary treatment for both depressive and metabolic disorders. Importantly, probiotics use is associated with minor or no adverse events, thus their supplementation might be worth considering. The question arises as to whether comorbidity of depressive disorders with obesity/MetS/NAFLD may serve as a specific indication for probiotics therapy.
Additionally, one must bear in mind that, as opposed to the meta-analyses conducted in pharmacologic agents, meta-analyses conducted in nutritional interventions are not always the best method for extracting relevant information, due to the heterogeneity of formulations and protocols.
To summarize, the majority of preclinical studies and meta-analyses of clinical observations support further studies of probiotics in the treatment of depressive disorders, obesity and metabolic disorders. Considering good safety and tolerability profile, it seems worthwhile to investigate the most efficacious probiotics regimens, including interventional timing, treatment duration, strain-dependency, dosage, etc.

2.2.2. Key Features of RCTs with Probiotics

In past years, dozens of RCTs trials have been carried out to compare probiotics, including Bifidobacterium and Lactobacillus, with placebo in metabolic disorders, and, not as many, in the depressive disorders population. Table 3 presents all RCTs with probiotics that have been published so far in the depressed population and selected RCTs results in obesity, MetS and the NAFLD population.
In depressive and anxiety clinical populations research, L. acidophilus has been the most often studied probiotics species so far [114,119,127,129,133,137]. L. casei and B. bifidum are runners-up. However, it is worth noting that there have been only 10 such RCTs results published and most of them used a combination of different bacterial genera and species. Additionally, the dosages of probiotics varied from 2 × 109 to 2 × 1010 CFU per day, and the form of administration was various (cheese, yogurt, milk or probiotic capsule). Moreover, the time of intervention varied from 4 weeks to 90 days. Regarding the subjects included, one study only recruited pregnant women [137], the other depression/anxiety in irritable bowel syndrome (IBS) [135], two treatment-resistant depression [133] and the rest of the trials included patients diagnosed as being generally depressed or having MDD. The sample size varied from 12 to 82 subjects. Furthermore, probiotics supplementation has been used definitely more often as an add-on then stand-alone treatment for depression/anxiety. Moreover, the effects of probiotics have yet to be tested in a clinical sample of treatment-naïve depressed patients. Additionally, some of the studies used a prebiotic concurrently with probiotics (symbiotic formulation); recently, it has been shown that adding inulin (a prebiotic) to probiotics improved psychological and inflammatory outcomes more effectively than two supplements separately [139]. Finally, the findings have indicated that probiotic supplementation is safe and well-tolerated.
It is worth mentioning that several strains of probiotics may be especially promising for the complementary treatment of metabolic disorders. L. gasseri, originated from human breast milk, has been proven to reduce several anthropometric parameters in abdominal obesity subjects in doses as low as 2 × 108/day [117]. Interestingly, L. gasseri BNR17 received approval as a treatment for body fat reduction in South Korea [140]. Akkermansia muciniphila has been shown to be negatively correlated with obesity and its supplementation ameliorated some metabolic parameters [118]. In several studies, L. acidophilus and B. lactis given together have been shown to reduce not only obesity and MetS parameters, but also some inflammation and OxS markers connected with the syndrome [106,115,120,141].
These studies have provided necessary pilot data regarding the efficacy and safety profiles of probiotics in clinical practice and paved the way for more elaborate probiotic pharmacotherapies in the future.
Bifidobacterium and Lactobacillus strains are still the most widely used probiotics in civilization diseases complementary treatment research. Next-generation probiotics, such as Faecalibacterium prausnitzii, Akkermansia muciniphila, or Clostridia strains, supplements await further development.

2.3. Conclusions

To sum up, due to the small number and scale of studies and heterogeneity of population, probiotic strains and genus, administered doses, the period of the interventions, and of endpoints it is hard to come to firm conclusions.
Firstly, probiotics are considered evidence-based treatment for antibiotic- or Clostridium difficile-associated diarrhoea and respiratory tract infections, but not for depression nor metabolic diseases [142]. Examples of psychobiotic strains that were found to be somehow effective to counteract affective and anxiety symptoms include: L. fermentum NS8 and NS9, L. casei Shirota, L. gasseri OLL2809, L. rhamnosus JB-1, L. helveticus Rosell -52, L. acidophilus W37, L. brevis W63, L. lactis W19 and W58, B. longum Rosell-175, B. longum NCC3001, B. longum 1714, B. bifidum W23, B. lactis W52, L. plantarum 299v [143]. However, there is an urgent need for a standardised methodology in this area to determine the exact type of probiotic administered to the “metabolic depression” subpopulation.
Furthermore, it is necessary to consider possible effects of the co-administration of different types of probiotics or a probiotic and prebiotic administered as it might influence the effectiveness of the first or the latter.
Additionally, the exact dose of probiotics should be determined, as higher doses may cause adverse effects and lower ones may be ineffective. Moreover, the formulations used should be thoroughly described as different formulations may account for different absorbance rates and thus different results.
Also, the time of intervention influence outcomes of the treatment. It seems that it is reasonable to supplement the probiotic formulation for at least 6–8 weeks to reduce the depression scale score; however, it may not be enough to affect some metabolic parameters. Durability of beneficial effects is also controversial as discontinuation of the treatment may result in loss of its effectiveness after some time. Follow-up visits may explain this question.
Secondly, as regard populations studied, there is a shortage of studies in humans, and preclinical studies may not reflect human physiology. The included populations are heterogenous and this may affect the results of a trial. Moreover, sample sizes of the trials are small, causing reduced statistical power. The major drawback of current methodology in probiotics trials is a lack of personalization of treatment; each person has a unique microbiota and may need an individualized approach. Additionally, it should be underlined that different individuals can have taxonomically varied but functionally similar microbiota, which makes a functional rather than taxonomic approach to creating probiotics formulations more important [144]. One of the potential microbial function biomarkers are short-chain fatty acids (SCFAs) that are the most representative metabolites of fiber anaerobic fermentation [145]. Interestingly, a depletion of SCFAs was reported in MDD patients [146], and SCFAs can play an important role in regulating metabolic and cardiovascular health [33,147]. Therefore, SCFAs-producing bacteria may become an interesting target as a potential treatment of metabolic depression.
Finally, regarding outcome measures, the methodology needs standardization. Clinical diagnosis and professional-assessed psychometric scales are a good option in psychiatric population. When it comes to metabolic parameters, it would be advisable to use WC rather than BMI and incorporate MetS criteria into trial secondary outcome measures.
Current treatments for both depression and metabolic diseases remain suboptimal for many patients, making improvements and advances in the intervention options in great demand. Whilst microbiota interventions may have benefits for some individuals, possibly those with comorbid obesity/MetS, evidence-based probiotic treatment awaits development.
We suggest that the mechanisms of action of probiotics in relation to "metabolic depression" are: primarily decreasing chronic inflammation as well as pro-oxidative states, and additionally balancing HPA dysfunction.

3. Practical Applications

To evaluate psychobiotic potential of a microorganism or microbial formulations according to a general methodology proposed by del Toro-Barbosa [148], there are 4 steps in the procedure: 1. Formulation, 2. In vitro tests in bacteria or mammalian cells, 3. Pre-clinical tests in murine models and 4. Clinical studies. As the abovementioned formulations have been extensively studied up to the 3rd step and there is not enough evidence for their efficacy from clinical studies, it is crucial to plan appropriate clinical study protocols.
Newly constructed study protocol could include adult patients diagnosed with depressive disorders with/without the MetS and/or NAFLD. The protocol might include psychometric and anthropometric parameters, as well as laboratory tests (MetS criteria, indicators of liver fibrosis, fecal microbiota analysis and possibly biological markers such as cortisol, inflammation and OxS parameters and brain imaging studies). The study could enable to establish a subpopulation of patients sensitive to microbiota interventions, especially probiotics, as an add-on therapy, as well as to determine potential biomarkers of therapeutic efficacy of probiotics.
As for psychobiotics choice, it is well-known that the probiotic effects are strongly strain-dependent and there is a consensus that a mixture of collaborating microbes would be more beneficial than a single strain. Generally, strains from genera Lactobacillus and Bifidobacterium in combination are worth studying, e.g., well-studied probiotic mixture of bacterial strains consists of L. helveticus R0052 and B. longum R0175 [59,116,149,150,151,152,153,154,155,156]. Recently, an open-label pilot study has been published that adds evidence to the antidepressant potential of this probiotic formulation [157]. Additionally, a RCT protocol of a study assessing antidepressant properties of the formulation in the context of MetS comorbidity has been published [158].
As there are several different definitions of depression applied in clinical and research practice [159] and, according to upcoming ICD-11, depressive disorders include not only MDD, but also dysthymic and mixed depressive and anxiety disorder (MDAD), underlying their impact on patients’ everyday functioning and quality of life, as well as importance in primary care settings [160,161], it would be valuable to incorporate the whole category into probiotics trials.
Additionally, it is worth remembering that there are many factors impacting microbiota function and composition, e.g., the diet, [39,162,163,164,165,166,167,168,169,170,171,172,173,174] and they should be assessed along the study process.
Proposed features of a new randomized clinical trial protocol of probiotics efficacy in depressive patients with metabolic abnormalities are summarized in Table 4.
Based on the above, we have just registered a randomized clinical trial on the influence of probiotic supplementation on depressive symptoms, inflammation, oxidative stress and fecal microbiota in depressed patients depending on MetS comorbidity (ClinicalTrials.gov identifier: NCT04756544) [158].
Overall, the effectiveness of probiotics in the prevention and treatment of depression, obesity and metabolic diseases remains to be elucidated in future large-scale studies in clinical populations. Additionally, the ideal mixture of probiotic strains, dose, the duration of supplementation, and the durability of beneficial effects are to be established.

Author Contributions

Conceptualization, O.G.-K.; methodology, O.G.-K.; investigation, O.G.-K.; writing—original draft preparation, O.G.-K.; writing—review and editing, O.G.-K.; visualization, O.G.-K.; supervision, D.S.; funding acquisition, D.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Marazziti, D.; Rutigliano, G.; Baroni, S.; Landi, P.; Dell’Osso, L. Metabolic syndrome and major depression. CNS Spectr. 2014, 19, 293–304. [Google Scholar] [CrossRef] [PubMed]
  2. Shinkov, A.; Borissova, A.M.; Kovatcheva, R.; Vlahov, J.; Dakovska, L.; Atanassova, I.; Petkova, P. Increased prevalence of depression and anxiety among subjects with metabolic syndrome and known type 2 diabetes mellitus–a population-based study. Postgrad. Med. 2018, 130, 251–257. [Google Scholar] [CrossRef]
  3. Penninx, B.W.J.H.; Lange, S.M.M. Metabolic syndrome in psychiatric patients: Overview, mechanisms, and implications. Dialogues Clin. Neurosci. 2018, 20, 63–73. [Google Scholar]
  4. Vancampfort, D.; Correll, C.U.; Wampers, M.; Sienaert, P.; Mitchell, A.J.; De Herdt, A.; Probst, M.; Scheewe, T.W.; De Hert, M. Metabolic syndrome and metabolic abnormalities in patients with major depressive disorder: A meta-analysis of prevalences and moderating variables. Psychol. Med. 2014, 44, 2017–2028. [Google Scholar] [CrossRef] [PubMed]
  5. Moradi, Y.; Albatineh, A.N.; Mahmoodi, H.; Gheshlagh, R.G. The relationship between depression and risk of metabolic syndrome: A meta-analysis of observational studies. Clin. Diabetes Endocrinol. 2021, 7, 4. [Google Scholar] [CrossRef]
  6. Lamers, F.; Milaneschi, Y.; De Jonge, P.; Giltay, E.J.; Penninx, B.W.J.H. Metabolic and inflammatory markers: Associations with individual depressive symptoms. Psychol. Med. 2018, 48, 1102–1110. [Google Scholar] [CrossRef]
  7. Moazzami, K.; Lima, B.B.; Sullivan, S.; Shah, A.; Bremner, J.D.; Vaccarino, V. Independent and Joint Association of Obesity and Metabolic Syndrome with Depression and Inflammation. Health Psychol. 2019, 38, 586–595. [Google Scholar] [CrossRef]
  8. Soto-Angona, Ó.; Anmella, G.; Valdés-Florido, M.J.; De Uribe-Viloria, N.; Carvalho, A.F.; Penninx, B.W.J.H.; Berk, M. Non-alcoholic fatty liver disease (NAFLD) as a neglected metabolic companion of psychiatric disorders: Common pathways and future approaches. BMC Med. 2020, 18, 1–14. [Google Scholar] [CrossRef]
  9. Eslam, M.; Sanyal, A.J.; George, J.; Sanyal, A.; Neuschwander-Tetri, B.; Tiribelli, C.; Kleiner, D.E.; Brunt, E.; Bugianesi, E.; Yki-Järvinen, H.; et al. MAFLD: A Consensus-Driven Proposed Nomenclature for Metabolic Associated Fatty Liver Disease. Gastroenterology 2020, 158, 1999–2014.e1. [Google Scholar] [CrossRef] [PubMed]
  10. Targher, G.; Corey, K.E.; Byrne, C.D. NAFLD, and cardiovascular and cardiac diseases: Factors influencing risk, prediction and treatment. Diabetes Metab. 2020, 101215. [Google Scholar] [CrossRef]
  11. McIntyre, R.S.; Soczynska, J.K.; Konarski, J.Z.; Woldeyohannes, H.O.; Law, C.W.Y.; Miranda, A.; Fulgosi, D.; Kennedy, S.H. Should depressive syndromes be reclassified as ‘metabolic syndrome type II’? Ann. Clin. Psychiatry 2007, 19, 257–264. [Google Scholar] [CrossRef]
  12. Gerhard, T.; Stroup, T.S.; Correll, C.U.; Setoguchi, S.; Strom, B.L.; Huang, C.; Tan, Z.; Crystal, S.; Olfson, M. Mortality risk of antipsychotic augmentation for adult depression. PLoS ONE 2020, 15, e0239206. [Google Scholar] [CrossRef]
  13. Matta, J.; Hoertel, N.; Kesse-Guyot, E.; Plesz, M.; Wiernik, E.; Carette, C.; Czernichow, S.; Limosin, F.; Goldberg, M.; Zins, M.; et al. Diet and physical activity in the association between depression and metabolic syndrome: Constances study. J. Affect. Disord. 2019, 244, 25–32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Gawlik-Kotelnicka, O.; Strzelecki, D. Adiposity in depression or depression in adiposity? The role of immune-inflammatory-microbial overlap. Life 2021, 11, 117. [Google Scholar] [CrossRef] [PubMed]
  15. Cernackova, A.; Durackova, Z.; Trebaticka, J.; Mravec, B. Neuroinflammation and depressive disorder: The role of the hypothalamus. J. Clin. Neurosci. 2020, 75, 5–10. [Google Scholar] [CrossRef] [PubMed]
  16. Van Den Eede, F.; Claes, S.J. Mechanisms of depression: Role of the HPA axis. Drug Discov. Today Dis. Mech. 2004, 1, 413–418. [Google Scholar] [CrossRef]
  17. Pasquali, R.; Vicennati, V.; Cacciari, M.; Pagotto, U. The hypothalamic-pituitary-adrenal axis activity in obesity and the metabolic syndrome. Ann. N. Y. Acad. Sci. 2006, 1083, 111–128. [Google Scholar]
  18. Chan, K.L.; Cathomas, F.; Russo, S.J. Central and peripheral inflammation link metabolic syndrome and major depressive disorder. Physiology 2019, 34, 123–133. [Google Scholar] [CrossRef]
  19. Leonard, B.E. Inflammation and depression: A causal or coincidental link to the pathophysiology? Acta Neuropsychiatr. 2018, 30, 1–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Vona, R.; Gambardella, L.; Cittadini, C.; Straface, E.; Pietraforte, D. Biomarkers of Oxidative Stress in Metabolic Syndrome and Associated Diseases. Oxid. Med. Cell. Longev. 2019, 2019, 8267234. [Google Scholar] [CrossRef] [Green Version]
  21. Carrier, A. Metabolic Syndrome and Oxidative Stress: A Complex Relationship. Antioxid. Redox Signal. 2017, 26, 429–431. [Google Scholar] [CrossRef]
  22. Black, C.N.; Bot, M.; Scheffer, P.G.; Cuijpers, P.; Penninx, B.W.J.H. Is depression associated with increased oxidative stress? A systematic review and meta-analysis. Psychoneuroendocrinology 2015, 51, 164–175. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Rani, V.; Deep, G.; Singh, R.K.; Palle, K.; Yadav, U.C.S. Oxidative stress and metabolic disorders: Pathogenesis and therapeutic strategies. Life Sci. 2016, 148, 183–193. [Google Scholar] [CrossRef]
  24. Liu, T.; Zhong, S.; Liao, X.; Chen, J.; He, T.; Lai, S.; Jia, Y. A meta-analysis of oxidative stress markers in depression. PLoS ONE 2015, 10. [Google Scholar] [CrossRef] [PubMed]
  25. Lozupone, C.A.; Stombaugh, J.I.; Gordon, J.I.; Jansson, J.K.; Knight, R. Diversity, stability and resilience of the human gut microbiota. Nature 2012, 489, 220–230. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Codagnone, M.G.; Spichak, S.; O’Mahony, S.M.; O’Leary, O.F.; Clarke, G.; Stanton, C.; Dinan, T.G.; Cryan, J.F. Programming Bugs: Microbiota and the Developmental Origins of Brain Health and Disease. Biol. Psychiatry 2019, 85, 150–163. [Google Scholar] [CrossRef]
  27. Lach, G.; Schellekens, H.; Dinan, T.G.; Cryan, J.F. Anxiety, Depression, and the Microbiome: A Role for Gut Peptides. Neurotherapeutics 2018, 15, 36–59. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Aizawa, E.; Tsuji, H.; Asahara, T.; Takahashi, T.; Teraishi, T.; Yoshida, S.; Ota, M.; Koga, N.; Hattori, K.; Kunugi, H. Possible association of Bifidobacterium and Lactobacillus in the gut microbiota of patients with major depressive disorder. J. Affect. Disord. 2016, 202, 254–257. [Google Scholar] [CrossRef]
  29. Zheng, P.; Zeng, B.; Zhou, C.; Liu, M.; Fang, Z.; Xu, X.; Zeng, L.; Chen, J.; Fan, S.; Du, X.; et al. Gut microbiome remodeling induces depressive-like behaviors through a pathway mediated by the host’s metabolism. Mol. Psychiatry 2016, 21, 786–796. [Google Scholar] [CrossRef]
  30. Naseribafrouei, A.; Hestad, K.; Avershina, E.; Sekelja, M.; Linløkken, A.; Wilson, R.; Rudi, K. Correlation between the human fecal microbiota and depression. Neurogastroenterol. Motil. 2014, 26, 1155–1162. [Google Scholar] [CrossRef] [PubMed]
  31. Dabke, K.; Hendrick, G.; Devkota, S. The gut microbiome and metabolic syndrome. J. Clin. Investig. 2019, 129, 4050–4057. [Google Scholar] [CrossRef] [PubMed]
  32. Turnbaugh, P.J.; Hamady, M.; Yatsunenko, T.; Cantarel, B.L.; Duncan, A.; Ley, R.E.; Sogin, M.L.; Jones, W.J.; Roe, B.A.; Affourtit, J.P.; et al. A core gut microbiome in obese and lean twins. Nature 2009, 457, 480–484. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Dai, X.; Hou, H.; Zhang, W.; Liu, T.; Li, Y.; Wang, S.; Wang, B.; Cao, H. Microbial Metabolites: Critical Regulators in NAFLD. Front. Microbiol. 2020, 11, 567654. [Google Scholar] [CrossRef]
  34. Han, R.; Ma, J.; Li, H. Mechanistic and therapeutic advances in non-alcoholic fatty liver disease by targeting the gut microbiota. Front. Med. 2018, 12, 645–657. [Google Scholar] [CrossRef] [PubMed]
  35. Belkaid, Y.; Hand, T.W. Role of the microbiota in immunity and inflammation. Cell 2014, 157, 121–141. [Google Scholar] [CrossRef] [Green Version]
  36. Carlessi, A.S.; Borba, L.A.; Zugno, A.I.; Quevedo, J.; Réus, G.Z. Gut microbiota–brain axis in depression: The role of neuroinflammation. Eur. J. Neurosci. 2019, 53, 222–235. [Google Scholar] [CrossRef] [PubMed]
  37. Dumitrescu, L.; Popescu-Olaru, I.; Cozma, L.; Tulbă, D.; Hinescu, M.E.; Ceafalan, L.C.; Gherghiceanu, M.; Popescu, B.O. Oxidative Stress and the Microbiota-Gut-Brain Axis. Oxid. Med. Cell. Longev. 2018, 2018. [Google Scholar] [CrossRef] [Green Version]
  38. Quigley, E.M.M.; Gajula, P. Recent advances in modulating the microbiome. F1000Research 2020, 9, 46. [Google Scholar] [CrossRef] [Green Version]
  39. Zmora, N.; Suez, J.; Elinav, E. You are what you eat: Diet, health and the gut microbiota. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 35–56. [Google Scholar] [CrossRef]
  40. Marx, W.; Lane, M.; Hockey, M.; Aslam, H.; Berk, M.; Walder, K.; Borsini, A.; Firth, J.; Pariante, C.M.; Berding, K.; et al. Diet and depression: Exploring the biological mechanisms of action. Mol. Psychiatry 2020, 26, 134–150. [Google Scholar] [CrossRef]
  41. Velasquez, M.T. Altered gut microbiota: A link between diet and the metabolic syndrome. Metab. Syndr. Relat. Disord. 2018, 16, 321–328. [Google Scholar] [CrossRef]
  42. Firth, J.; Solmi, M.; Wootton, R.E.; Vancampfort, D.; Schuch, F.B.; Hoare, E.; Gilbody, S.; Torous, J.; Teasdale, S.B.; Jackson, S.E.; et al. A meta-review of ‘lifestyle psychiatry’: The role of exercise, smoking, diet and sleep in the prevention and treatment of mental disorders. World Psychiatry 2020, 19. [Google Scholar] [CrossRef]
  43. Barengolts, E. Gut microbiota, prebiotics, probiotics, and synbiotics in management of obesity and prediabetes: Review of randomized controlled trials. Endocr. Pract. 2016, 22, 1224–1234. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Paiva, I.H.R.; Duarte-Silva, E.; Peixoto, C.A. The role of prebiotics in cognition, anxiety, and depression. Eur. Neuropsychopharmacol. 2020, 34, 1–18. [Google Scholar] [CrossRef] [PubMed]
  45. Noonan, S.; Zaveri, M.; Macaninch, E.; Martyn, K. Food & mood: A review of supplementary prebiotic and probiotic interventions in the treatment of anxiety and depression in adults. BMJ Nutr. Prev. Health 2020, 3, 351–362. [Google Scholar] [CrossRef]
  46. Liu, R.T.; Walsh, R.F.L.; Sheehan, A.E. Prebiotics and probiotics for depression and anxiety: A systematic review and meta-analysis of controlled clinical trials. Neurosci. Biobehav. Rev. 2019, 102, 13–23. [Google Scholar] [CrossRef] [PubMed]
  47. Vaghef-Mehrabany, E.; Maleki, V.; Behrooz, M.; Ranjbar, F.; Ebrahimi-Mameghani, M. Can psychobiotics “mood” ify gut? An update systematic review of randomized controlled trials in healthy and clinical subjects, on anti-depressant effects of probiotics, prebiotics, and synbiotics. Clin. Nutr. 2019. [Google Scholar] [CrossRef] [PubMed]
  48. Craven, L.; Rahman, A.; Nair Parvathy, S.; Beaton, M.; Silverman, J.; Qumosani, K.; Hramiak, I.; Hegele, R.; Joy, T.; Meddings, J.; et al. Allogenic Fecal Microbiota Transplantation in Patients with Nonalcoholic Fatty Liver Disease Improves Abnormal Small Intestinal Permeability: A Randomized Control Trial. Am. J. Gastroenterol. 2020, 115, 1055–1065. [Google Scholar] [CrossRef] [PubMed]
  49. Chinna Meyyappan, A.; Forth, E.; Wallace, C.J.K.; Milev, R. Effect of fecal microbiota transplant on symptoms of psychiatric disorders: A systematic review. BMC Psychiatry 2020, 20. [Google Scholar] [CrossRef]
  50. Proença, I.M.; Allegretti, J.R.; Bernardo, W.M.; de Moura, D.T.H.; Ponte Neto, A.M.; Matsubayashi, C.O.; Flor, M.M.; Kotinda, A.P.S.T.; de Moura, E.G.H. Fecal microbiota transplantation improves metabolic syndrome parameters: Systematic review with meta-analysis based on randomized clinical trials. Nutr. Res. 2020, 83, 1–14. [Google Scholar] [CrossRef]
  51. Vallianou, N.; Stratigou, T.; Christodoulatos, G.S.; Tsigalou, C.; Dalamaga, M. Probiotics, Prebiotics, Synbiotics, Postbiotics, and Obesity: Current Evidence, Controversies, and Perspectives. Curr. Obes. Rep. 2020, 9, 179–192. [Google Scholar] [CrossRef]
  52. Balaguer, F.; Enrique, M.; Llopis, S.; Barrena, M.; Navarro, V.; Álvarez, B.; Chenoll, E.; Ramón, D.; Tortajada, M.; Martorell, P. Lipoteichoic acid from Bifidobacterium animalis subsp. lactis BPL1: A novel postbiotic that reduces fat deposition via IGF-1 pathway. Microb. Biotechnol. 2021. [Google Scholar] [CrossRef]
  53. Yamawaki, Y.; Yoshioka, N.; Nozaki, K.; Ito, H.; Oda, K.; Harada, K.; Shirawachi, S.; Asano, S.; Aizawa, H.; Yamawaki, S.; et al. Sodium butyrate abolishes lipopolysaccharide-induced depression-like behaviors and hippocampal microglial activation in mice. Brain Res. 2018, 1680, 13–38. [Google Scholar] [CrossRef]
  54. Canfora, E.E.; Meex, R.C.R.; Venema, K.; Blaak, E.E. Gut microbial metabolites in obesity, NAFLD and T2DM. Nat. Rev. Endocrinol. 2019, 15, 261–273. [Google Scholar] [CrossRef] [PubMed]
  55. 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] [CrossRef] [PubMed] [Green Version]
  56. Leclercq, S.; Mian, F.M.; Stanisz, A.M.; Bindels, L.B.; Cambier, E.; Ben-Amram, H.; Koren, O.; Forsythe, P.; Bienenstock, J. Low-dose penicillin in early life induces long-term changes in murine gut microbiota, brain cytokines and behavior. Nat. Commun. 2017, 8. [Google Scholar] [CrossRef] [PubMed]
  57. Tian, P.; Wang, G.; Zhao, J.; Zhang, H.; Chen, W. Bifidobacterium with the role of 5-hydroxytryptophan synthesis regulation alleviates the symptom of depression and related microbiota dysbiosis. J. Nutr. Biochem. 2019, 66, 43–51. [Google Scholar] [CrossRef]
  58. Mohammadi, G.; Dargahi, L.; Naserpour, T.; Mirzanejad, Y.; Alizadeh, S.A.; Peymani, A.; Nassiri-Asl, M. Probiotic mixture of Lactobacillus helveticus R0052 and Bifidobacterium longum R0175 attenuates hippocampal apoptosis induced by lipopolysaccharide in rats. Int. Microbiol. 2019, 22, 317–323. [Google Scholar] [CrossRef] [PubMed]
  59. Mohammadi, G.; Dargahi, L.; Peymani, A.; Mirzanejad, Y.; Alizadeh, S.A.; Naserpour, T.; Nassiri-Asl, M. The Effects of Probiotic Formulation Pretreatment (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) on a Lipopolysaccharide Rat Model. J. Am. Coll. Nutr. 2019, 38, 209–217. [Google Scholar] [CrossRef]
  60. Sun, X.; Zhang, H.-F.; Ma, C.-L.; Wei, H.; Li, B.-M.; Luo, J. Alleviation of Anxiety/Depressive-Like Behaviors and Improvement of Cognitive Functions by Lactobacillus plantarum WLPL04 in Chronically Stressed Mice. Can. J. Infect. Dis. Med. Microbiol. 2021, 2021, 1–11. [Google Scholar] [CrossRef]
  61. Reis, D.J.; Ilardi, S.S.; Punt, S.E.W. The anxiolytic effect of probiotics: A systematic review and meta-analysis of the clinical and preclinical literature. PLoS ONE 2018, 13, e0199041. [Google Scholar] [CrossRef] [Green Version]
  62. Stenman, L.K.; Patterson, E.; Meunier, J.; Roman, F.J.; Lehtinen, M.J. Strain specific stress-modulating effects of candidate probiotics: A systematic screening in a mouse model of chronic restraint stress. Behav. Brain Res. 2020, 379. [Google Scholar] [CrossRef] [PubMed]
  63. Partrick, K.A.; Rosenhauer, A.M.; Auger, J.; Arnold, A.R.; Ronczkowski, N.M.; Jackson, L.M.; Lord, M.N.; Abdulla, S.M.; Chassaing, B.; Huhman, K.L. Ingestion of probiotic (Lactobacillus helveticus and Bifidobacterium longum) alters intestinal microbial structure and behavioral expression following social defeat stress. Sci. Rep. 2021, 11. [Google Scholar] [CrossRef]
  64. Archer, A.C.; Muthukumar, S.P.; Halami, P.M. Lactobacillus fermentum MCC2759 and MCC2760 Alleviate Inflammation and Intestinal Function in High-Fat Diet-Fed and Streptozotocin-Induced Diabetic Rats. Probiotics Antimicrob. Proteins 2021. [Google Scholar] [CrossRef] [PubMed]
  65. Molina-Tijeras, J.A.; Diez-Echave, P.; Vezza, T.; Hidalgo-García, L.; Ruiz-Malagón, A.J.; Rodríguez-Sojo, M.J.; Romero, M.; Robles-Vera, I.; García, F.; Plaza-Diaz, J.; et al. Lactobacillus fermentum CECT5716 ameliorates high fat diet-induced obesity in mice through modulation of gut microbiota dysbiosis. Pharmacol. Res. 2021, 105471. [Google Scholar] [CrossRef] [PubMed]
  66. Russo, M.; Marquez, A.; Herrera, H.; Abeijon-Mukdsi, C.; Saavedra, L.; Hebert, E.; Gauffin-Cano, P.; Medina, R. Oral administration of: Lactobacillus fermentum CRL1446 improves biomarkers of metabolic syndrome in mice fed a high-fat diet supplemented with wheat bran. Food Funct. 2020, 11, 3879–3894. [Google Scholar] [CrossRef] [PubMed]
  67. Bouaziz, A.; Dib, A.L.; Lakhdara, N.; Kadja, L.; Espigares, E.; Moreno, E.; Bouaziz, O.; Gagaoua, M. Study of Probiotic Effects of Bifidobacterium animalis subsp. lactis BB-12 and Lactobacillus plantarum 299v Strains on Biochemical and Morphometric Parameters of Rabbits after Obesity Induction. Biology 2021, 10, 131. [Google Scholar] [CrossRef] [PubMed]
  68. Kapar, F.S.; Ciftci, G. The effects of curcumin and Lactobacillus acidophilus on certain hormones and insulin resistance in rats with metabolic syndrome. J. Diabetes Metab. Disord. 2020, 19, 907–914. [Google Scholar] [CrossRef]
  69. Khanna, S.; Walia, S.; Kondepudi, K.K.; Shukla, G. Administration of indigenous probiotics modulate high-fat diet-induced metabolic syndrome in Sprague Dawley rats. Antonie Leeuwenhoek Int. J. Gen. Mol. Microbiol. 2020, 113, 1345–1359. [Google Scholar] [CrossRef]
  70. Jena, P.K.; Sheng, L.; Nagar, N.; Wu, C.; Barile, D.; Mills, D.A.; Wan, Y.J.Y. Synbiotics Bifidobacterium infantis and milk oligosaccharides are effective in reversing cancer-prone nonalcoholic steatohepatitis using western diet-fed FXR knockout mouse models. J. Nutr. Biochem. 2018, 57, 246–254. [Google Scholar] [CrossRef]
  71. Park, S.S.; Lee, Y.J.; Song, S.; Kim, B.; Kang, H.; Oh, S.; Kim, E. Lactobacillus acidophilus NS1 attenuates diet-induced obesity and fatty liver. J. Endocrinol. 2018, 237, 87–100. [Google Scholar] [CrossRef]
  72. Kim, B.; Kwon, J.; Kim, M.S.; Park, H.; Ji, Y.; Holzapfel, W.; Hyun, C.K. Protective effects of Bacillus probiotics against high-fat diet-induced metabolic disorders in mice. PLoS ONE 2018, 13. [Google Scholar] [CrossRef] [Green Version]
  73. Oh, J.-H.; Schueler, K.L.; Stapleton, D.S.; Alexander, L.M.; Yen, C.-L.E.; Keller, M.P.; Attie, A.D.; van Pijkeren, J.-P. Secretion of Recombinant Interleukin-22 by Engineered Lactobacillus reuteri Reduces Fatty Liver Disease in a Mouse Model of Diet-Induced Obesity. mSphere 2020, 5. [Google Scholar] [CrossRef]
  74. Patterson, E.; Ryan, P.M.; Wiley, N.; Carafa, I.; Sherwin, E.; Moloney, G.; Franciosi, E.; Mandal, R.; Wishart, D.S.; Tuohy, K.; et al. Gamma-aminobutyric acid-producing lactobacilli positively affect metabolism and depressive-like behaviour in a mouse model of metabolic syndrome. Sci. Rep. 2019, 9. [Google Scholar] [CrossRef] [PubMed]
  75. Amirani, E.; Milajerdi, A.; Mirzaei, H.; Jamilian, H.; Mansournia, M.A.; Hallajzadeh, J.; Ghaderi, A. The effects of probiotic supplementation on mental health, biomarkers of inflammation and oxidative stress in patients with psychiatric disorders: A systematic review and meta-analysis of randomized controlled trials. Complement. Ther. Med. 2020, 49, 102361. [Google Scholar] [CrossRef]
  76. Liu, B.; He, Y.; Wang, M.; Liu, J.; Ju, Y.; Zhang, Y.; Liu, T.; Li, L.; Li, Q. Efficacy of probiotics on anxiety-A meta-analysis of randomized controlled trials. Depress. Anxiety 2018, 35, 935–945. [Google Scholar] [CrossRef] [PubMed]
  77. Yang, B.; Wei, J.; Ju, P.; Chen, J. Effects of regulating intestinal microbiota on anxiety symptoms: A systematic review. Gen. Psychiatry 2019, 32, 100056. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Nikolova, V.L.; Cleare, A.J.; Young, A.H.; Stone, J.M. Updated Review and Meta-Analysis of Probiotics for the Treatment of Clinical Depression: Adjunctive vs. Stand-Alone Treatment. J. Clin. Med. 2021, 10, 647. [Google Scholar] [CrossRef]
  79. Chao, L.; Liu, C.; Sutthawongwadee, S.; Li, Y.; Lv, W.; Chen, W.; Yu, L.; Zhou, J.; Guo, A.; Li, Z.; et al. Effects of probiotics on depressive or anxiety variables in healthy participants under stress conditions or with a depressive or anxiety diagnosis: A meta-analysis of randomized controlled trials. Front. Neurol. 2020, 11, 421. [Google Scholar] [CrossRef] [PubMed]
  80. Nikolova, V.; Zaidi, S.Y.; Young, A.H.; Cleare, A.J.; Stone, J.M. Gut feeling: Randomized controlled trials of probiotics for the treatment of clinical depression: Systematic review and meta-analysis. Ther. Adv. Psychopharmacol. 2019, 9, 204512531985996. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Huang, R.; Wang, K.; Hu, J. Effect of probiotics on depression: A systematic review and meta-analysis of randomized controlled trials. Nutrients 2016, 8, 483. [Google Scholar] [CrossRef] [PubMed]
  82. Ng, Q.X.; Peters, C.; Ho, C.Y.X.; Lim, D.Y.; Yeo, W.S. A meta-analysis of the use of probiotics to alleviate depressive symptoms. J. Affect. Disord. 2018, 228, 13–19. [Google Scholar] [CrossRef] [PubMed]
  83. Herman, A. Probiotics Supplementation in Prophylaxis and Treatment of Depressive and Anxiety Disorders—A Review of Current Research. Psychiatr. Pol. 2019, 53, 459–473. [Google Scholar] [CrossRef]
  84. Slykerman, R.F.; Hood, F.; Wickens, K.; Thompson, J.M.D.; Barthow, C.; Murphy, R.; Kang, J.; Rowden, J.; Stone, P.; Crane, J.; et al. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Goh, K.K.; Liu, Y.W.; Kuo, P.H.; Chung, Y.C.E.; Lu, M.L.; Chen, C.H. Effect of probiotics on depressive symptoms: A meta-analysis of human studies. Psychiatry Res. 2019, 282. [Google Scholar] [CrossRef]
  86. Chen, C.; Shan, W. Pharmacological and non-pharmacological treatments for major depressive disorder in adults: A systematic review and network meta-analysis. Psychiatry Res. 2019, 281, 112595. [Google Scholar] [CrossRef] [PubMed]
  87. Sanada, K.; Nakajima, S.; Kurokawa, S.; Barceló-Soler, A.; Ikuse, D.; Hirata, A.; Yoshizawa, A.; Tomizawa, Y.; Salas-Valero, M.; Noda, Y.; et al. Gut microbiota and majore depressive disorder: A systematic review and meta-analysis. J. Affect. Disord. 2020, 266, 1–13. [Google Scholar] [CrossRef]
  88. Dinan, T.G.; Stanton, C.; Cryan, J.F. Psychobiotics: A novel class of psychotropic. Biol. Psychiatry 2013, 74, 720–726. [Google Scholar] [CrossRef]
  89. Perna, S.; Ilyas, Z.; Giacosa, A.; Gasparri, C.; Peroni, G.; Faliva, M.A.; Rigon, C.; Naso, M.; Riva, A.; Petrangolini, G.; et al. Is probiotic supplementation useful for the management of body weight and other anthropometric measures in adults affected by overweight and obesity with metabolic related diseases? A systematic review and meta-analysis. Nutrients 2021, 13, 666. [Google Scholar] [CrossRef] [PubMed]
  90. Zhang, Q.; Wu, Y.; Fei, X. Effect of probiotics on body weight and body-mass index: A systematic review and meta-analysis of randomized, controlled trials. Int. J. Food Sci. Nutr. 2016, 67, 571–580. [Google Scholar] [CrossRef]
  91. John, G.K.; Wang, L.; Nanavati, J.; Twose, C.; Singh, R.; Mullin, G. Dietary alteration of the gut microbiome and its impact on weight and fat mass: A systematic review and meta-analysis. Genes 2018, 9, 167. [Google Scholar] [CrossRef] [Green Version]
  92. Borgeraas, H.; Johnson, L.K.; Skattebu, J.; Hertel, J.K.; Hjelmesæth, J. Effects of probiotics on body weight, body mass index, fat mass and fat percentage in subjects with overweight or obesity: A systematic review and meta-analysis of randomized controlled trials. Obes. Rev. 2018, 19, 219–232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Koutnikova, H.; Genser, B.; Monteiro-Sepulveda, M.; Faurie, J.M.; Rizkalla, S.; Schrezenmeir, J.; Clement, K. Impact of bacterial probiotics on obesity, diabetes and non-alcoholic fatty liver disease related variables: A systematic review and meta-analysis of randomised controlled trials. BMJ Open 2019, 9, e017995. [Google Scholar] [CrossRef]
  94. Wang, Z.B.; Xin, S.S.; Ding, L.N.; Ding, W.Y.; Hou, Y.L.; Liu, C.Q.; Zhang, X.D. The Potential Role of Probiotics in Controlling Overweight/Obesity and Associated Metabolic Parameters in Adults: A Systematic Review and Meta-Analysis. Evidence-Based Complement. Altern. Med. 2019, 2019. [Google Scholar] [CrossRef]
  95. .Xiao, M.W.; Lin, S.X.; Shen, Z.H.; Luo, W.W.; Wang, X.Y. Systematic review with meta-analysis: The effects of probiotics in nonalcoholic fatty liver disease. Gastroenterol. Res. Pract. 2019, 2019. [Google Scholar] [CrossRef] [PubMed]
  96. Companys, J.; Pla-Pagà, L.; Calderón-Pérez, L.; Llauradó, E.; Solà, R.; Pedret, A.; Valls, R.M. Fermented Dairy Products, Probiotic Supplementation, and Cardiometabolic Diseases: A Systematic Review and Meta-analysis. Adv. Nutr. 2020, 11, 834–863. [Google Scholar] [CrossRef] [PubMed]
  97. Dixon, A.; Robertson, K.; Yung, A.; Que, M.; Randall, H.; Wellalagodage, D.; Cox, T.; Robertson, D.; Chi, C.; Sun, J. Efficacy of Probiotics in Patients of Cardiovascular Disease Risk: A Systematic Review and Meta-analysis. Curr. Hypertens. Rep. 2020, 22, 74. [Google Scholar] [CrossRef]
  98. Dong, Y.; Xu, M.; Chen, L.; Bhochhibhoya, A. Probiotic Foods and Supplements Interventions for Metabolic Syndromes: A Systematic Review and Meta-Analysis of Recent Clinical Trials. Ann. Nutr. Metab. 2019, 74, 224–241. [Google Scholar] [CrossRef]
  99. Park, S.; Bae, J.H. Probiotics for weight loss: A systematic review and meta-analysis. Nutr. Res. 2015, 35, 566–575. [Google Scholar] [CrossRef]
  100. Kazemi, A.; Soltani, S.; Ghorabi, S.; Keshtkar, A.; Daneshzad, E.; Nasri, F.; Mazloomi, S.M. Effect of probiotic and synbiotic supplementation on inflammatory markers in health and disease status: A systematic review and meta-analysis of clinical trials. Clin. Nutr. 2019. [Google Scholar] [CrossRef]
  101. Skonieczna-Żydecka, K.; Kaźmierczak-Siedlecka, K.; Kaczmarczyk, M.; Śliwa-Dominiak, J.; Maciejewska, D.; Janda, K.; Stachowska, E.; Łoniewska, B.; Malinowski, D.; Borecki, K.; et al. The Effect of Probiotics and Synbiotics on Risk Factors Associated with Cardiometabolic Diseases in Healthy People—A Systematic Review and Meta-Analysis with Meta-Regression of Randomized Controlled Trials. J. Clin. Med. 2020, 9, 1788. [Google Scholar] [CrossRef]
  102. Mohammadi, H.; Ghavami, A.; Hadi, A.; Askari, G.; Symonds, M.; Miraghajani, M. Effects of pro-/synbiotic supplementation on anthropometric and metabolic indices in overweight or obese children and adolescents: A systematic review and meta-analysis. Complement. Ther. Med. 2019, 44, 269–276. [Google Scholar] [CrossRef] [PubMed]
  103. Chatzakis, C.; Goulis, D.G.; Mareti, E.; Eleftheriades, M.; Zavlanos, A.; Dinas, K.; Sotiriadis, A. Prevention of gestational diabetes mellitus in overweight or obese pregnant women: A network meta-analysis. Diabetes Res. Clin. Pract. 2019, 158, 107924. [Google Scholar] [CrossRef]
  104. Swierz, M.J.; Storman, D.; Staskiewicz, W.; Gorecka, M.; Jasinska, K.W.; Swierz, A.M.; Tobola, P.; Skuza, A.; Bala, M.M. Efficacy of probiotics in patients with morbid obesity undergoing bariatric surgery: A systematic review and meta-analysis. Surg. Obes. Relat. Dis. 2020, 16, 2105–2116. [Google Scholar] [CrossRef]
  105. Sáez-Lara, M.J.; Robles-Sanchez, C.; Ruiz-Ojeda, F.J.; Plaza-Diaz, J.; Gil, A. Effects of probiotics and synbiotics on obesity, insulin resistance syndrome, type 2 diabetes and non-alcoholic fatty liver disease: A review of human clinical trials. Int. J. Mol. Sci. 2016, 17, 928. [Google Scholar] [CrossRef] [Green Version]
  106. Szulińska, M.; Łoniewski, I.; van Hemert, S.; Sobieska, M.; Bogdański, P. Dose-dependent effects of multispecies probiotic supplementation on the lipopolysaccharide (LPS) level and cardiometabolic profile in obese postmenopausal women: A 12-week randomized clinical trial. Nutrients 2018, 10, 773. [Google Scholar] [CrossRef] [Green Version]
  107. Tenorio-Jiménez, C.; Martínez-Ramírez, M.J.; Gil, Á.; Gómez-Llorente, C. Effects of probiotics on metabolic syndrome: A systematic review of randomized clinical trials. Nutrients 2020, 12, 124. [Google Scholar] [CrossRef] [Green Version]
  108. López-Moreno, A.; Suárez, A.; Avanzi, C.; Monteoliva-Sánchez, M.; Aguilera, M. Probiotic strains and intervention total doses for modulating obesity-related microbiota dysbiosis: A systematic review and meta-analysis. Nutrients 2020, 12, 1921. [Google Scholar] [CrossRef]
  109. Kazemi, A.; Noorbala, A.A.; Azam, K.; Eskandari, M.H.; 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. [Google Scholar] [CrossRef] [PubMed]
  110. Ardeshirlarijani, E.; Tabatabaei-Malazy, O.; Mohseni, S.; Qorbani, M.; Larijani, B.; Baradar Jalili, R. Effect of probiotics supplementation on glucose and oxidative stress in type 2 diabetes mellitus: A meta-analysis of randomized trials. DARU J. Pharm. Sci. 2019, 27, 827–837. [Google Scholar] [CrossRef]
  111. Tabrizi, R.; Ostadmohammadi, V.; Akbari, M.; Lankarani, K.B.; Vakili, S.; Peymani, P.; Karamali, M.; Kolahdooz, F.; Asemi, Z. The Effects of Probiotic Supplementation on Clinical Symptom, Weight Loss, Glycemic Control, Lipid and Hormonal Profiles, Biomarkers of Inflammation, and Oxidative Stress in Women with Polycystic Ovary Syndrome: A Systematic Review and Meta-analysis of Randomized Controlled Trials. Probiotics Antimicrob. Proteins 2019. [Google Scholar] [CrossRef]
  112. Tang, Y.; Huang, J.; Zhang, W.y.; Qin, S.; Yang, Y.X.; Ren, H.; Yang, Q.B.; Hu, H. Effects of probiotics on nonalcoholic fatty liver disease: A systematic review and meta-analysis. Therap. Adv. Gastroenterol. 2019, 12. [Google Scholar] [CrossRef]
  113. Pan, X.; Wen, S.W.; Kaminga, A.C.; Liu, A. Gut metabolites and inflammation factors in non-alcoholic fatty liver disease: A systematic review and meta-analysis. Sci. Rep. 2020, 10. [Google Scholar] [CrossRef]
  114. Akkasheh, G.; Kashani-Poor, Z.; Tajabadi-Ebrahimi, M.; Jafari, P.; Akbari, H.; Taghizadeh, M.; Memarzadeh, M.R.; Asemi, Z.; Esmaillzadeh, A. Clinical and metabolic response to probiotic administration in patients with major depressive disorder: A randomized, double-blind, placebo-controlled trial. Nutrition 2016, 32, 315–320. [Google Scholar] [CrossRef] [PubMed]
  115. Szulińska, M.; Łoniewski, I.; Skrypnik, K.; Sobieska, M.; Korybalska, K.; Suliburska, J.; Bogdański, P. Multispecies Probiotic Supplementation Favorably Affects Vascular Function and Reduces Arterial Stiffness in Obese Postmenopausal Women—A 12-Week Placebo-Controlled and Randomized Clinical Study. Nutrients 2018, 10, 1672. [Google Scholar] [CrossRef] [Green Version]
  116. Romijn, A.R.; Rucklidge, J.J.; Kuijer, R.G.; Frampton, C. A double-blind, randomized, placebo-controlled trial of Lactobacillus helveticus and Bifidobacterium longum for the symptoms of depression. Aust. N. Z. J. Psychiatry 2017, 51, 810–821. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Kadooka, Y.; Sato, M.; Ogawa, A.; Miyoshi, M.; Uenishi, H.; Ogawa, H.; Ikuyama, K.; Kagoshima, M.; Tsuchida, T. Effect of Lactobacillus gasseri SBT2055 in fermented milk on abdominal adiposity in adults in a randomised controlled trial. Br. J. Nutr. 2013, 110, 1696–1703. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Depommier, C.; Everard, A.; Druart, C.; Plovier, H.; Van Hul, M.; Vieira-Silva, S.; Falony, G.; Raes, J.; Maiter, D.; Delzenne, N.M.; et al. Supplementation with Akkermansia muciniphila in overweight and obese human volunteers: A proof-of-concept exploratory study. Nat. Med. 2019, 25, 1096–1103. [Google Scholar] [CrossRef]
  119. Ghorbani, Z.; Nazari, S.; Etesam, F.; Nourimajd, S.; Ahmadpanah, M.; Razeghi Jahromi, S. The Effect of Synbiotic as an Adjuvant Therapy to Fluoxetine in Moderate Depression: A Randomized Multicenter Trial. Arch. Neurosci. 2018, 5. [Google Scholar] [CrossRef] [Green Version]
  120. Rezazadeh, L.; Gargari, B.P.; Jafarabadi, M.A.; Alipour, B. Effects of probiotic yogurt on glycemic indexes and endothelial dysfunction markers in patients with metabolic syndrome. Nutrition 2019, 62, 162–168. [Google Scholar] [CrossRef] [PubMed]
  121. Miyaoka, T.; Kanayama, M.; Wake, R.; Hashioka, S.; Hayashida, M.; Nagahama, M.; Okazaki, S.; Yamashita, S.; Miura, S.; Miki, H.; et al. Clostridium butyricum MIYAIRI 588 as Adjunctive Therapy for Treatment-Resistant Major Depressive Disorder: A Prospective Open-Label Trial. Clin. Neuropharmacol. 2018, 41, 151–155. [Google Scholar] [CrossRef]
  122. Leber, B.; Tripolt, N.J.; Blattl, D.; Eder, M.; Wascher, T.C.; Pieber, T.R.; Stauber, R.; Sourij, H.; Oettl, K.; Stadlbauer, V. The influence of probiotic supplementation on gut permeability in patients with metabolic syndrome: An open label, randomized pilot study. Eur. J. Clin. Nutr. 2012, 66, 1110–1115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Tripolt, N.J.; Leber, B.; Blattl, D.; Eder, M.; Wonisch, W.; Scharnagl, H.; Stojakovic, T.; Obermayer-Pietsch, B.; Wascher, T.C.; Pieber, T.R.; et al. Short communication: Effect of supplementation with Lactobacillus casei Shirota on insulin sensitivity, β-cell function, and markers of endothelial function and inflammation in subjects with metabolic syndrome-A pilot study. J. Dairy Sci. 2013, 96, 89–95. [Google Scholar] [CrossRef]
  124. Stadlbauer, V.; Leber, B.; Lemesch, S.; Trajanoski, S.; Bashir, M.; Horvath, A.; Tawdrous, M.; Stojakovic, T.; Fauler, G.; Fickert, P.; et al. Lactobacillus casei Shirota Supplementation Does Not Restore Gut Microbiota Composition and Gut Barrier in Metabolic Syndrome: A Randomized Pilot Study. PLoS ONE 2015, 10, e0141399. [Google Scholar] [CrossRef] [Green Version]
  125. Rudzki, L.; Ostrowska, L.; Pawlak, D.; Małus, A.; Pawlak, K.; Waszkiewicz, N.; Szulc, A. Probiotic Lactobacillus Plantarum 299v decreases kynurenine concentration and improves cognitive functions in patients with major depression: A double-blind, randomized, placebo controlled study. Psychoneuroendocrinology 2019, 100, 213–222. [Google Scholar] [CrossRef]
  126. Sharafedtinov, K.K.; Plotnikova, O.A.; Alexeeva, R.I.; Sentsova, T.B.; Songisepp, E.; Stsepetova, J.; Smidt, I.; Mikelsaar, M. Hypocaloric diet supplemented with probiotic cheese improves body mass index and blood pressure indices of obese hypertensive patients—A randomized double-blind placebo-controlled pilot study. Nutr. J. 2013, 12. [Google Scholar] [CrossRef] [Green Version]
  127. Chahwan, B.; Kwan, S.; Isik, A.; van Hemert, S.; Burke, C.; Roberts, L. Gut feelings: A randomised, triple-blind, placebo-controlled trial of probiotics for depressive symptoms. J. Affect. Disord. 2019, 253, 317–326. [Google Scholar] [CrossRef]
  128. Barreto, F.M.; Colado Simão, A.N.; Morimoto, H.K.; Batisti Lozovoy, M.A.; Dichi, I.; Helena da Silva Miglioranza, L. Beneficial effects of Lactobacillus plantarum on glycemia and homocysteine levels in postmenopausal women with metabolic syndrome. Nutrition 2014, 30, 939–942. [Google Scholar] [CrossRef]
  129. Reininghaus, E.Z.; Platzer, M.; Kohlhammer-Dohr, A.; Hamm, C.; Mörkl, S.; Bengesser, S.A.; Fellendorf, F.T.; Lahousen-Luxenberger, T.; Leitner-Afschar, B.; Schöggl, H.; et al. Provit: Supplementary probiotic treatment and vitamin b7 in depression—a randomized controlled trial. Nutrients 2020, 12, 3422. [Google Scholar] [CrossRef]
  130. Bernini, L.J.; Simão, A.N.C.; Alfieri, D.F.; Lozovoy, M.A.B.; Mari, N.L.; de Souza, C.H.B.; Dichi, I.; Costa, G.N. Beneficial effects of Bifidobacterium lactis on lipid profile and cytokines in patients with metabolic syndrome: A randomized trial. Effects of probiotics on metabolic syndrome. Nutrition 2016, 32, 716–719. [Google Scholar] [CrossRef] [PubMed]
  131. Majeed, M.; Nagabhushanam, K.; Arumugam, S.; Majeed, S.; Ali, F. Bacillus coagulans MTCC 5856 for the management of major depression with irritable bowel syndrome: A randomised, double-blind, placebo controlled, multi-centre, pilot clinical study. Food Nutr. Res. 2018, 62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Cicero, A.F.G.; Fogacci, F.; Bove, M.; Giovannini, M.; Borghi, C. Impact of a short-term synbiotic supplementation on metabolic syndrome and systemic inflammation in elderly patients: A randomized placebo-controlled clinical trial. Eur. J. Nutr. 2020, 60. [Google Scholar] [CrossRef]
  133. Bambling, M.; Edwards, S.C.; Hall, S.; Vitetta, L. A combination of probiotics and magnesium orotate attenuate depression in a small SSRI resistant cohort: An intestinal anti-inflammatory response is suggested. Inflammopharmacology 2017, 25, 271–274. [Google Scholar] [CrossRef]
  134. Behrouz, V.; Aryaeian, N.; Zahedi, M.J.; Jazayeri, S. Effects of probiotic and prebiotic supplementation on metabolic parameters, liver aminotransferases, and systemic inflammation in nonalcoholic fatty liver disease: A randomized clinical trial. J. Food Sci. 2020, 85, 3611–3617. [Google Scholar] [CrossRef] [PubMed]
  135. Pinto-Sanchez, M.I.; Hall, G.B.; Ghajar, K.; Nardelli, A.; Bolino, C.; Lau, J.T.; Martin, F.P.; Cominetti, O.; Welsh, C.; Rieder, A.; et al. Probiotic Bifidobacterium longum NCC3001 Reduces Depression Scores and Alters Brain Activity: A Pilot Study in Patients With Irritable Bowel Syndrome. Gastroenterology 2017, 153, 448–459.e8. [Google Scholar] [CrossRef] [PubMed]
  136. Abhari, K.; Saadati, S.; Yari, Z.; Hosseini, H.; Hedayati, M.; Abhari, S.; Alavian, S.M.; Hekmatdoost, A. The effects of Bacillus coagulans supplementation in patients with non-alcoholic fatty liver disease: A randomized, placebo-controlled, clinical trial. Clin. Nutr. ESPEN 2020, 39, 53–60. [Google Scholar] [CrossRef] [PubMed]
  137. Browne, P.D.; Bolte, A.C.; Besseling-van der Vaart, I.; Claassen, E.; de Weerth, C. Probiotics as a treatment for prenatal maternal anxiety and depression: A double-blind randomized pilot trial. Sci. Rep. 2021, 11. [Google Scholar] [CrossRef] [PubMed]
  138. Scorletti, E.; Afolabi, P.R.; Miles, E.A.; Smith, D.E.; Almehmadi, A.; Alshathry, A.; Childs, C.E.; Del Fabbro, S.; Bilson, J.; Moyses, H.E.; et al. Synbiotics Alter Fecal Microbiomes, But Not Liver Fat or Fibrosis, in a Randomized Trial of Patients With Nonalcoholic Fatty Liver Disease. Gastroenterology 2020, 158, 1597–1610.e7. [Google Scholar] [CrossRef]
  139. Moludi, J.; Khedmatgozar, H.; Nachvak, S.M.; Abdollahzad, H.; Moradinazar, M.; Sadeghpour Tabaei, A. The effects of co-administration of probiotics and prebiotics on chronic inflammation, and depression symptoms in patients with coronary artery diseases: A randomized clinical trial. Nutr. Neurosci. 2021. [Google Scholar] [CrossRef]
  140. Jung, S.P.; Lee, K.M.; Kang, J.H.; Yun, S.I.; Park, H.O.; Moon, Y.; Kim, J.Y. Effect of Lactobacillus gasseri BNR17 on overweight and obese adults: A randomized, double-blind clinical trial. Korean J. Fam. Med. 2013, 34, 80–89. [Google Scholar] [CrossRef] [Green Version]
  141. Rezazadeh, L.; Alipour, B.; Jafarabadi, M.A.; Behrooz, M.; Gargari, B.P. Daily consumption effects of probiotic yogurt containing Lactobacillus acidophilus La5 and Bifidobacterium lactis Bb12 on oxidative stress in metabolic syndrome patients. Clin. Nutr. ESPEN 2021, 41, 136–142. [Google Scholar] [CrossRef]
  142. Rondanelli, M.; Faliva, M.A.; Perna, S.; Giacosa, A.; Peroni, G.; Castellazzi, A.M. Using probiotics in clinical practice: Where are we now? A review of existing meta-analyses. Gut Microbes 2017, 8, 521–543. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Skonieczna-Żydecka, K.; Marlicz, W.; Misera, A.; Koulaouzidis, A.; Łoniewski, I. Microbiome—The Missing Link in the Gut-Brain Axis: Focus on Its Role in Gastrointestinal and Mental Health. J. Clin. Med. 2018, 7, 521. [Google Scholar] [CrossRef] [Green Version]
  144. Cheung, S.G.; Goldenthal, A.R.; Uhlemann, A.C.; Mann, J.J.; Miller, J.M.; Sublette, M.E. Systematic review of gut microbiota and major depression. Front. Psychiatry 2019, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Tan, J.; McKenzie, C.; Potamitis, M.; Thorburn, A.N.; Mackay, C.R.; Macia, L. The Role of Short-Chain Fatty Acids in Health and Disease. Adv. Immunol. 2014, 121, 91–119. [Google Scholar] [PubMed]
  146. Skonieczna-żydecka, K.; Grochans, E.; Maciejewska, D.; Szkup, M.; Schneider-Matyka, D.; Jurczak, A.; Łoniewski, I.; Kaczmarczyk, M.; Marlicz, W.; Czerwińska-Rogowska, M.; et al. Faecal short chain fatty acids profile is changed in Polish depressive women. Nutrients 2018, 10, 1939. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Chambers, E.S.; Preston, T.; Frost, G.; Morrison, D.J. Role of Gut Microbiota-Generated Short-Chain Fatty Acids in Metabolic and Cardiovascular Health. Curr. Nutr. Rep. 2018, 7, 198–206. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Del Toro-Barbosa, M.; Hurtado-Romero, A.; Garcia-Amezquita, L.E.; García-Cayuela, T. Psychobiotics: Mechanisms of action, evaluation methods and effectiveness in applications with food products. Nutrients 2020, 12, 3896. [Google Scholar] [CrossRef]
  149. Ait-Belgnaoui, A.; Payard, I.; Rolland, C.; Harkat, C.; Braniste, V.; Théodorou, V.; Tompkins, T.A. Bifidobacterium longum and Lactobacillus helveticus synergistically suppress stress-related visceral hypersensitivity through hypothalamic-pituitary-adrenal axis modulation. J. Neurogastroenterol. Motil. 2018, 24, 138–146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Arseneault-Bréard, J.; Rondeau, I.; Gilbert, K.; Girard, S.A.; Tompkins, T.A.; Godbout, R.; Rousseau, G. Combination of Lactobacillus helveticus R0052 and Bifidobacterium longum R0175 reduces post-myocardial infarction depression symptoms and restores intestinal permeability in a rat model. Br. J. Nutr. 2012, 107, 1793–1799. [Google Scholar] [CrossRef] [Green Version]
  151. Gilbert, K.; Arseneault-Bréard, J.; Monaco, F.F.; Beaudoin, A.; Bah, T.M.; Tompkins, T.A.; Godbout, R.; Rousseau, G. Attenuation of post-myocardial infarction depression in rats by n-3 fatty acids or probiotics starting after the onset of reperfusion. Br. J. Nutr. 2013, 109, 50–56. [Google Scholar] [CrossRef] [Green Version]
  152. Girard, S.A.; Bah, T.M.; Kaloustian, S.; Lada-Moldovan, L.; Rondeau, I.; Tompkins, T.A.; Godbout, R.; Rousseau, G. Lactobacillus helveticus and Bifidobacterium longum taken in combination reduce the apoptosis propensity in the limbic system after myocardial infarction in a rat model. Br. J. Nutr. 2009, 102, 1420–1425. [Google Scholar] [CrossRef] [Green Version]
  153. Zareie, M.; Johnson-Henry, K.; Jury, J.; Yang, P.C.; Ngan, B.Y.; McKay, D.M.; Soderholm, J.D.; Perdue, M.H.; Sherman, P.M. Probiotics prevent bacterial translocation and improve intestinal barrier function in rats following chronic psychological stress. Gut 2006, 55, 1553–1560. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Messaoudi, M.; Lalonde, R.; Violle, N.; Javelot, H.; Desor, D.; Nejdi, A.; Bisson, J.-F.; Rougeot, C.; Pichelin, M.; Cazaubiel, M.; et al. Assessment of psychotropic-like properties of a probiotic formulation (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) in rats and human subjects. Br. J. Nutr. 2011, 105, 755–764. [Google Scholar] [CrossRef] [Green Version]
  155. Messaoudi, M.; Violle, N.; Bisson, J.-F.; Desor, D.; Javelot, H.; Rougeot, C. Beneficial psychological effects of a probiotic formulation (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) in healthy human volunteers. Gut Microbes 2011, 2, 256–261. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Diop, L.; Guillou, S.; Durand, H. Probiotic food supplement reduces stress-induced gastrointestinal symptoms in volunteers: A double-blind, placebo-controlled, randomized trial. Nutr. Res. 2008, 28, 1–5. [Google Scholar] [CrossRef]
  157. Wallace, C.J.K.; Milev, R.V. The Efficacy, Safety, and Tolerability of Probiotics on Depression: Clinical Results From an Open-Label Pilot Study. Front. Psychiatry 2021, 12. [Google Scholar] [CrossRef]
  158. Gawlik-Kotelnicka, O.; Skowrońska, A.; Margulska, A.; Czarnecka-Chrebelska, K.H.; Łoniewski, I.; Skonieczna-Żydecka, K.; Strzelecki, D. The Influence of Probiotic Supplementation on Depressive Symptoms, Inflammation, and Oxidative Stress Parameters and Fecal Microbiota in Patients with Depression Depending on Metabolic Syndrome Comorbidity—PRO-DEMET Randomized Study Protocol. J. Clin. Med. 2021, 10, 1342. [Google Scholar] [CrossRef] [PubMed]
  159. Sjöberg, L.; Karlsson, B.; Atti, A.R.; Skoog, I.; Fratiglioni, L.; Wang, H.X. Prevalence of depression: Comparisons of different depression definitions in population-based samples of older adults. J. Affect. Disord. 2017, 221, 123–131. [Google Scholar] [CrossRef] [PubMed]
  160. Reed, G.M.; First, M.B.; Kogan, C.S.; Hyman, S.E.; Gureje, O.; Gaebel, W.; Maj, M.; Stein, D.J.; Maercker, A.; Tyrer, P.; et al. Innovations and changes in the ICD-11 classification of mental, behavioural and neurodevelopmental disorders. World Psychiatry 2019, 18, 3–19. [Google Scholar] [CrossRef] [Green Version]
  161. Ziebold, C.; Goldberg, D.P.; Reed, G.M.; Minhas, F.; Razzaque, B.; Fortes, S.; Robles, R.; Lam, T.P.; Bobes, J.; Iglesias, C.; et al. Dimensional analysis of depressive, anxious and somatic symptoms presented by primary care patients and their relationship with ICD-11 PHC proposed diagnoses. Psychol. Med. 2019, 49, 764–771. [Google Scholar] [CrossRef] [Green Version]
  162. Lee, S.H.; Yun, Y.; Kim, S.J.; Lee, E.-J.; Chang, Y.; Ryu, S.; Shin, H.; Kim, H.-L.; Kim, H.-N.; Lee, J.H. Association between Cigarette Smoking Status and Composition of Gut Microbiota: Population-Based Cross-Sectional Study. J. Clin. Med. 2018, 7, 282. [Google Scholar] [CrossRef] [Green Version]
  163. Requena, T.; Martínez-Cuesta, M.C.; Peláez, C. Diet and microbiota linked in health and disease. Food Funct. 2018, 9, 688–704. [Google Scholar] [CrossRef] [Green Version]
  164. Westfall, S.; Pasinetti, G.M. The Gut Microbiota Links Dietary Polyphenols with Management of Psychiatric Mood Disorders. Front. Neurosci. 2019, 13, 1196. [Google Scholar] [CrossRef] [Green Version]
  165. Yang, Q.; Liang, Q.; Balakrishnan, B.; Belobrajdic, D.P.; Feng, Q.J.; Zhang, W. Role of dietary nutrients in the modulation of gut microbiota: A narrative review. Nutrients 2020, 12, 381. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Paoli, A.; Mancin, L.; Bianco, A.; Thomas, E.; Mota, J.F.; Piccini, F. Ketogenic Diet and Microbiota: Friends or Enemies? Genes 2019, 10, 534. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Salvucci, E. The disappearing microbiota: Diseases of the Western civilization. In How Fermented Foods Feed a Healthy Gut Microbiota: A Nutrition Continuum; Springer International Publishing: Cham, Switzerland, 2019; pp. 325–347. ISBN 9783030287375. [Google Scholar]
  168. Vich Vila, A.; Collij, V.; Sanna, S.; Sinha, T.; Imhann, F.; Bourgonje, A.R.; Mujagic, Z.; Jonkers, D.M.A.E.; Masclee, A.A.M.; Fu, J.; et al. Impact of commonly used drugs on the composition and metabolic function of the gut microbiota. Nat. Commun. 2020, 11. [Google Scholar] [CrossRef]
  169. Bretler, T.; Weisberg, H.; Koren, O.; Neuman, H. The effects of antipsychotic medications on microbiome and weight gain in children and adolescents. BMC Med. 2019, 17, 112. [Google Scholar] [CrossRef]
  170. Cao, T.T.B.; Wu, K.-C.; Hsu, J.-L.; Chang, C.-S.; Chou, C.; Lin, C.-Y.; Liao, Y.-M.; Lin, P.-C.; Yang, L.-Y.; Lin, H.-W. Effects of Non-insulin Anti-hyperglycemic Agents on Gut Microbiota: A Systematic Review on Human and Animal Studies. Front. Endocrinol. 2020, 11. [Google Scholar] [CrossRef] [PubMed]
  171. Elbere, I.; Kalnina, I.; Silamikelis, I.; Konrade, I.; Zaharenko, L.; Sekace, K.; Radovica-Spalvina, I.; Fridmanis, D.; Gudra, D.; Pirags, V.; et al. Association of metformin administration with gut microbiome dysbiosis in healthy volunteers. PLoS ONE 2018, 13. [Google Scholar] [CrossRef]
  172. Guo, J.; Tang, J.; Kang, T.; Xiong, Y.; Xiang, Z.; Qin, C. Different immunization methods lead to altered gut flora and varied responses to Mycobacterium tuberculosis infection in mice. J. Infect. Dev. Ctries. 2020, 14, 1170–1177. [Google Scholar] [CrossRef]
  173. Mir, R.A.; Schaut, R.G.; Allen, H.K.; Looft, T.; Loving, C.L.; Kudva, I.T.; Sharma, V.K. Cattle intestinal microbiota shifts following Escherichia coli O157:H7 vaccination and colonizationtravel. PLoS ONE 2019, 14. [Google Scholar] [CrossRef] [PubMed]
  174. Sandhu, K.V.; Sherwin, E.; Schellekens, H.; Stanton, C.; Dinan, T.G.; Cryan, J.F. Feeding the microbiota-gut-brain axis: Diet, microbiome, and neuropsychiatry. Transl. Res. 2017, 179, 223–244. [Google Scholar] [CrossRef] [PubMed]
Table 1. Summary of the most important data from pre-clinical studies on probiotics in some mental health and metabolic disorders models.
Table 1. Summary of the most important data from pre-clinical studies on probiotics in some mental health and metabolic disorders models.
DataMental Health Problems ModelsMetabolic Disorders Models
Main clinical features findingsPrevention of anxiety and depression, antidepressant and antianxiety effect, alleviation of cognitive deficitsAnti-obesity effect
Main laboratory findingsPrevention of hippocampal apoptosis, reduction of brain-derived neurotropic factor (BDNF) level, promotion of an anti-inflammatory profileAlleviation of inflammation, oxidation, endothelial dysfunction and intestinal barrier integrity dysfunction, improvement in insulin sensitivity, glucose and lipid profiles, liver function biomarkers
Commonly studied probioticsLactobacillus (e.g., L. plantarum) and Bifidobacterium generaLactobacillus (e.g., L. fermentum) and Bifidobacterium genera
Table 2. Meta-analyses of RCTs with probiotics efficacy towards depressive and anxiety symptoms and recent meta-analyses of RCTs with probiotics efficacy toward metabolic health parameters.
Table 2. Meta-analyses of RCTs with probiotics efficacy towards depressive and anxiety symptoms and recent meta-analyses of RCTs with probiotics efficacy toward metabolic health parameters.
Depressive and Anxiety Symptoms
Amirani et al. 2020 [75]12 RCTs656 subjectsReduced the HDRS score by 9.60. Reduced CRP by 1.59 mg/L, TNF-α by 0.12 pg/mL, and MDA by 0.38 μmol/L.
Chao et al. 2020 [79]10 RCTs685 subjectsReduced the depression scale score by 0.47.
No significant impact on anxiety symptoms.
Goh et al. 2019 [85]19 RCTs1901 subjectsReduced the depression scale score by 0.31.
Huang et al. 2016 [81]5 RCTs365 adult subjectsReduced the depression scale score by 0.30.
Liu et al. 2018 [76]12 RCTs1551 subjectsNo significant impact on anxiety symptoms.
Liu et al. 2019 [46]29 RCTs?Reduced the depression scale score by 0.24 and the anxiety scale score by 0.10.
Ng et al. 2018 [82]10 RCTs1349 subjectsReduced the depression scale score by 0.684 in mild/moderate depression. No significant difference in mood overall (healthy and clinical population).
Nikolova et al 2021 [78]7 RCTs404 subjectsReduced the depression scale score by 0.83 as an add-on. No significant impact as a standalone treatment.
Nikolova et al. 2019 [80]3 RCTs229 subjectsReduced the depression scale score by 1.371.
Reis et al. 2018 [61]14 RCTs1527 subjectsNo significant impact on anxiety symptoms.
Sanada et al. 2020 [87]6 RCTs302 subjectsReduced the depression scale score by 1.62.
Obesity, MetS, NAFLD, and metabolic parameters in healthy subjects
Borgeraas H et al. 2018 [92]15 RCTs957 subjectsReduced BW by 0.60 kg, BMI by 0.27 kg/m2 and fat percentage by 0.60%
Chatzakis et al. 2019 [103]5 RCTs1235 overweight or obese pregnant womenNo significant impact on GDM risk, nor gestational weight gain.
Companys et al. 2020 [96]52 RCTsOverweight/obese/hypercholesterolemia/MetS subjectsReduced BW, BMI, WC, BFP. Improved lipids profile.
Dixon et al. 2020 [97]34 RCTs2177 hypertension, obesity, CVD, MetS, T2D or hypercholesterolaemia subjectsReduced SBP by 1.31 mmHg, DBP by 1.87 mmHg, TC by 6.05 mg/dL, LDL-C by 8.77 mg/dL, fGlc by 4.92 mg/dL, HbA1C by 0.18%, BMI by 0.31 kg/m2. Increased HDL-C by 1.05 mg/dL.
No significant effect on TG.
Dong et al. 2019 [98]18 RCTs1544 subjectsReduced BFP by 0.3% and LDL-c by 0.18 mg/dL;
No significant differences of BMI, BFM, WC, HC, WHR, SBP, DBP, fGlc, fasting insulin, TC, HDL-C, HbA1c, or TG.
Kazemi et al. 2019 [100]29 RCTsMetabolic disorders (e.g., NAFLD and MetS) subjectsNo significant impact on BMI. Reduced CRP by 0.32 mg/L.
Koutnikova H et al. 2019 [93]111 RCTs6826 (e.g., obese, NAFLD) subjectsReduced body weight by 0.94 kg, BMI by 0.55 kg/m2, WC by 1.31 cm, BFM by 0.96 kg, and visceral adipose tissue mass by 6.30 cm2
Kunnackal et al. 2018 [91]22 RCTs?Reduced BW by 0.65 kg, BFM by 0.94 kg and BMI by 0.33 kg/m2
Mohammadi et al. 2019 [102]9 RCTs410 overweight or obese children and adolescentsNo significant changes in BMI, WC, BW, BFM, fGlc and lipid profiles.
Pan et al. 2020 [113]11 RCTsNAFLD subjects Reduced TNF-α by 0.52 pg/mL and CRP by 0.62 mg/L.
Park S et al. 2015 [99]4 RCTs449 adult subjectsNo significant effect on body weight and BMI
Perna et al. 2021 [89]20 RCTs1411 subjectsReduced BMI by 0.73 kg/m2, WC by 0.71 cm and HC by 0.73 cm. No significant effect on body weight.
Skonieczna-Żydecka et al. 2020 [101]61 RCTs5422 healthy subjects (including overweight/obese ones)Reduced BMI by 0.45 kg/m2, WC by 1.21 cm in healthy persons. Reduced TC in overweight/obese subjects. No significant impact on carbohydrate and lipid metabolism
Swierz et al. 2020 [104]5 RCTs Morbid obesity undergoing bariatric surgery subjects No significant effect on body weight.
Tang et al. 2019 [112]18 RCTsNAFLD subjectsReduced weight by 2.31 kg, and BMI by 1.08 kg/m2. Reduced ALT by 7.22 U/L, AST by 7.22 U/L, AP by 25.87 U/L, GTP by 5.76 U/L. Reduced TC by 0.73, LDL-C by 0.54, TG by 0.36 mg/dL. Reduced fGlc by 4.45 mg/dL, insulin by 0.63 µIU/mL. Reduced TNF-α by 0.62 pg/mL, and leptin by 1.14 ng/mL.
Wang ZB et al. 2019 [94]12 RCTs821 adult subjectsReduced BW by 0.55 kg, BMI by 0.30 kg/m2, WC by 1.20 cm, BFM by 0.91 kg, and BFP by 0.92%
Xiao et al. 2019 [95]28 RCTs1555 NAFLD subjectsReduced BMI by 1.46 kg/m2, ALT by 13.40 U/L, AST by 13.54 U/L, GTP by 9.88 U/L, insulin by 1.32 𝜇IU/mL, and TC by 15.38 mg/dL;
No significant effect on fGlc, lipid profile or TNFα.
Zhang Q et al. 2016 [90]25 RCTs1931 adult subjectsReduced BW by 0.59 kg and BMI by 0.49 kg/m2
Abbreviations: ALT: alanine transaminase; AP: alkaline phosphatase; AST: aspartate transaminase; BMI: body mass index; BFM: body fat mass; BFP: body fat percentage; CRP: C-reactive protein; CVD: cardiovascular disease; DBP: diastolic blood pressure; fGlc: fasting glucose; GDM: gestational diabetes mellitus; GTP: gamma-glutamyl transferase; HbA1c: haemoglobin A1c; HC: hip circumference; HDL-C: high-density lipoprotein cholesterol; HDRS: Hamilton Depression Rating Scale; LDL-C: low-density lipoprotein cholesterol; MDA: malondialdehyde; MetS: metabolic syndrome; NAFLD: non-alcoholic fatty liver disease; SBP: systolic blood pressure; TNF-α: tumour necrosis factor α; RCT: randomized clinical trial; T2D: type 2 diabetes mellitus; TC: total cholesterol; TG: triglycerides; WC: waist circumference; WHR: waist-to-hip ratio.
Table 3. The selected recent randomized clinical trials with probiotics formulations in the field of depressive and metabolic disorders.
Table 3. The selected recent randomized clinical trials with probiotics formulations in the field of depressive and metabolic disorders.
Clinical trials: size; type; duration; probiotic formulationDepressionObesity, MetS and NAFLD
Akkasheh, 2016 [114]: MDD; 40, add-on, 8 weeks; L. acidophilus (2 × 109), L. casei (2 × 109), B. bifidum (2 × 109).Szulińska 2018 [106,115]: 81; 12 weeks; B. bifidum W23, B. lactis W51, B. lactis W52, L. acidophilus W37, L. brevis W63, L. casei W56, L. salivarius W24, L. lactis W19, L. lactis W58, lyophilisate powder, low dose (2.5 × 109 CFU/day) or high dose (1 × 1010 CFU/day)
Romijn et al. 2017 [116]: Depressive and anxiety disorders; 79; standalone; 8 weeks; L. helveticus R0052, B. longum R0175 (≥3 × 109/day)Kadooka et al. 2013 [117]: Obesity; 210; 12 weeks; L. gasseri SBT2055 2 × 108/day
Kazemi, 2019 [109]: MDD; 74; add-on; 8 weeks; L. helveticus + B. longum; ≥10 × 109/day.Depommier et al. 2019 [118]: overweight/obese insulin-resistant subjects; 40; 3 months; Akkermansia muciniphila 1010 CFU/day.
Ghorbani, 2018 [119]: MDD; 40; add-on; 6 weeks; L. casei 3 × 108, L. acidofilus 2 × 108, L. bulgaricus 2 × 109, L. rhamnosus 3 × 108, B. breve 2 × 108, B. longum 1 × 109, Streptococcus thermophilus 3 × 108 (plus prebiotic).Rezazadeh 2019 [120]: 44; 8 weeks; yogurt containing
6.45 × 106 CFU/g of L. acidophilus La5 and 4.94 × 106 of B. lactis Bb12
Miyaoka,2018 [121]: TRD; 40; add-on; 8 weeks; C. butyricum, 60 mg daily.Leber 2012 [122], Tripolt 2013 [123], Stadlbauer 2012 [124]: 28; add-on; 12 weeks; L. casei Shirota, milk (65 mL bottles × 3/day) 108 cells/mL.
Rudzki, 2019 [125]: MDD; 60; add-on; 8 weeks; L. plantarum (10 × 109).Sharafedtinov 2013 [126]: 40; 3 weeks; add-on; L. plantarum TENSIA, cheese (50 g/day) 1.5 × 1011 CFU/g.
Chahwan, 2019 [127]: depressive disorders; 71; stand-alone; 8 weeks; B. bifidum, B. lactis W51 & W52, L. acidophilus, L. brevis, L. casei, L. salivarius, and Lactococcus lactis W19 & W58; 2.5 × 109.Barreto 2014 [128]: 24; 12 weeks; L. plantarum, milk (80 mL bottles × 1/day) 107 CFU/g.
Reininghaus 2020 [129]: MDD; 82; add-on; 4 weeks; B. bifidum W23, B. lactis W51, B. lactis W52, L. acidophilus W22, L. casei W56, L. paracasei W20, L. plantarum W62, L. salivarius W24 and Lactococcus lactis W19; 7.5 × 109 CFU/day.Bernini 2016 [130]: 51; 6 weeks; B. lactis HN019, milk (80 mL bottle × 1/day) 3.4 × 108 CFU/mL.
Majeed, 2018 [131]: MDD in IBS; 40; stand-alone; 90 days; Bacillus coagulans 2 × 109Cicero et al. 2020 [132] MetS elderly patients; 60 days; L. plantarum PBS067, L. acidophilus PBS066 and L. reuteri PBS072 (plus prebiotic).
Bambling et al. 2017 [133]: TRD; 12; add-on; 8 weeks; L. acidophilus, B. bifidum, Streptoccocus thermophilus; 2 × 1010 CFU/day.Behrouz et al. 2020 [134]: NAFLD; 71; add-on; 12 weeks; L. casei, L. rhamnosus, L. acidophilus, B. longum, and B. breve; 5 × 109 CFU/day.
Pinto-Sanchez et al. 2017 [135]: depression/anxiety in IBS; 44; 6 weeks; B. longum NCC 3001; 3 × 109 CFUAbhari et al. 2020 [136]: NAFLD; 53; 12 weeks; Bacillus coagulans (GBI-30) 109 spore/day (plus inulin).
Browne et al. 2021 [137]: depression/anxiety in pregnancy; stand-alone; 40; 8 weeks; B. bifidum W23, B. lactis W51, B. lactis W52, L. acidophilus W37, L. brevis W63, L. casei W56, L. salivarius W24, Lactococcus lactis W19 and Lactococcus lactis W58; 5 × 109 CFU/dayScorletti et al. 2020 [138]: NAFLD; 104; stand-alone; 10–14 months; fructo-oligosaccharides, 4 g twice per day, plus B. animalis subsp. lactis BB-12 10 × 109 CFU/day (plus fructo-oligosaccharides).
Abbreviations: B.: Bifidobacterium; CFU: colony-forming unit; IBS: irritable bowel syndrome; L.: Lactobacillus; MDD: major depressive disorder; MetS: metabolic syndrome; NAFLD: non-alcoholic fatty liver disease; TRD: treatment-resistant depression.
Table 4. Proposed key points of a randomized clinical trial protocol of probiotics efficacy in depressive patients with metabolic abnormalities.
Table 4. Proposed key points of a randomized clinical trial protocol of probiotics efficacy in depressive patients with metabolic abnormalities.
PopulationDepressive Disorders with Comorbid Obesity/MetS/NAFLD
ProbioticsLactobacillus and Bifidobacterium strains mixture
Probiotic dose per daymin. 109 CFU/day
Formulationcapsule
Intervention period8 weeks
Primary outcomedepressive symptoms
Secondary outcomesanthropometric parameters, MetS criteria, indicators of liver fibrosis, fecal microbiota composition and function analysis
Tertiary outcomescortisol, inflammation and oxidative stress parameters
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Gawlik-Kotelnicka, O.; Strzelecki, D. Probiotics as a Treatment for “Metabolic Depression”? A Rationale for Future Studies. Pharmaceuticals 2021, 14, 384. https://doi.org/10.3390/ph14040384

AMA Style

Gawlik-Kotelnicka O, Strzelecki D. Probiotics as a Treatment for “Metabolic Depression”? A Rationale for Future Studies. Pharmaceuticals. 2021; 14(4):384. https://doi.org/10.3390/ph14040384

Chicago/Turabian Style

Gawlik-Kotelnicka, Oliwia, and Dominik Strzelecki. 2021. "Probiotics as a Treatment for “Metabolic Depression”? A Rationale for Future Studies" Pharmaceuticals 14, no. 4: 384. https://doi.org/10.3390/ph14040384

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