Abstract
Introduction:
Gut dysbiosis is assumed to play a role in obstructive sleep apnea (OSA)-associated morbidities. Pre- and probiotics, short chain fatty acids (SCFA) and fecal matter transfer (FMT) may offer potential as novel therapeutic strategies that target this gut dysbiosis. As more mechanisms of OSA-induced dysbiosis are being elucidated, these novel approaches are being tested in preclinical and clinical development.
Areas covered:
We examine the evidence linking OSA to gut dysbiosis and discuss the effects of pre- and probiotics on associated cardiometabolic, neurobehavioral and gastrointestinal disorders. The therapeutic potential of short chain fatty acids (SCFA) and fecal matter transplantation (FMT) are also discussed. We reviewed the National Center for Biotechnology Information database, including PubMed and PubMed Central between 2000 -2020.
Expert opinion:
To date, there are no clinical trials and only limited evidence from animal studies describing the beneficial effects of pre- and probiotic supplementation on OSA-mediated dysbiosis. Thus, more work is necessary to assess whether prebiotics, probiotics and SCFA are promising future novel strategies for targeting OSA-mediated dysbiosis.
Keywords: obstructive sleep apnea, intermittent hypoxia, microbiome, gut dysbiosis, probiotics, prebiotics, short chain fatty acids, fecal matter transfer
1. Introduction
Obstructive sleep apnea (OSA) is a sleep disorder characterized by narrowing of the pharyngeal airways leading to repetitive episodes of airway collapse or increased upper airway resistance. These recurring events are usually associated with intermittent hypoxia (IH) and micro-arousals with concomitant sleep fragmentation. Although the total nocturnal sleep quantity might not be necessarily affected, sleep quality is usually poor in patients with OSA, which explains many of the symptoms seen in patients with OSA such as excessive daytime sleepiness and fatigue, poor concentration, mood alterations, and memory impairments. These symptoms can affect work productivity, increase the risk for motor vehicle and on the job accidents, as well as adversely impact social interactions, academic performance, and many other neuro-cognitive consequences [1]. IH, such as occurs in OSA, has significant impact on a variety of organ systems. Indeed, there is a strong body of literature linking IH to different diseases such as systemic hypertension, coronary artery disease, obesity, diabetes mellitus, pulmonary hypertension, cerebrovascular diseases, Alzheimer disease, mood disorders, erectile dysfunction and even cancer [2–5].
IH can directly affect cells of multiple types, or it can induce alterations in susceptible cells through the effects induced by the IH-mediated release of different mediators. It is assumed nowadays that IH plays a major role in the activation and propagation of oxidative stress and systemic inflammation by acting as a potent stimulant for the release of inflammatory mediators which contribute to many of the co-morbid disorders associated with OSA [6–9]. In this manuscript, we will briefly review the evidence linking IH, gut dysbiosis and systemic inflammation, and will focus on therapeutic interventions targeting gut dysbiosis (namely, pre- and probiotics, short chain fatty acids (SCFA) and fecal matter transplantation (FMT)), and their potential uses as novel adjuvant therapies for patients with OSA. We reviewed the National Center for Biotechnology Information database, including PubMed and PubMed Central between 2000 -2020 using the keywords: “obstructive sleep apnea, gut dysbiosis, probiotics, prebiotics, short-chain fatty acids, fecal matter transfer, cardiovascular, metabolic, cognitive, anxiety, depression, gastrointestinal” in different combinations and permutations.
2. The gut microbiome and gut dysbiosis
Microbiota is a term used to include all microbes that live in and colonize different organs (such as gastrointestinal tract, lung, upper airways, and skin). These microbes include bacteria, viruses, fungi, archaea, and eukarya [10]. The gut harbors most of the microbiota with more than 100 trillion bacteria from more than 1,000 species [11]. The mutual interactions between the host and the microbiota maintains a neutral, yet actively regulated and responsive ecosystem through which the host benefits from the microbiota and vice versa [12]. Any perturbations in the balance of the system can lead to a state of dysbiosis. The healthy gut microbiota provides the gut with many benefits, such as maintaining the integrity of the gastrointestinal epithelial cells, regulating the gut and the overall host immune system, protecting against a variety of opportunistic pathogens, and providing energy to the colonocytes, a function which plays a critical role in maintaining the gut integrity and permeability [13–16]. Gut dysbiosis invokes the presence of harmful species of bacteria that can potentially generate excessive amounts of specific compounds that are detrimental to the gut or directly produce toxic substances that can induce leakiness of the gut or traverse the leaky epithelial tight junctions into the systemic circulation. Some of these toxins (such as lipopolysaccharides produced by some gram-negative bacteria) can induce a state of local and systemic low-grade inflammation that has been shown in some studies [17] (Figure. 1). Indeed, systemic inflammation is one of the main underlying pathological mechanisms linking OSA to CVD [18].
Figure 1: Mechanisms and treatments of OSA-induce dysbiosis.
BA: bile acid, IH: intermittent hypoxia, LPS: lipopolysaccharides, OSA: obstructive sleep apnea, ROS: reactive oxygen species, TMA: trimethylamine, TMAO: trimethylamine oxide, WBCs: white blood cells
The five most common bacterial phyla that reside in the colon are Bacteroidetes, Firmicutes, Actinobacteria, Proteobacteria, and Cerrucomicrobia [19]. Some of the health promoting bacterial species include Lactobacillaceae, Bifidobacteriaceae, Erysipelotrichaceae, Ruminococcaceae, and Clostridiales which belong to the phylum Bacteroidetes. Bacteroidetes (Gram-negative) inhabit the gastrointestine (mainly the colon) and play a critical role in carbohydrate and fiber fermentation. This process produces short chain fatty acids (SCFA) such as butyrate, acetate, and propionate. SCFAs provide a major source of nutrients and energy to colonocytes [3, 20, 21]. Induction of gut dysbiosis with unhealthy diets or following IH will consume many of these SCFAs and lead to colonocyte dysfunction, manifesting as weakened intercellular tight junctions that connect the epithelial cells and preserve the gut epithelial barrier. Consequently, the gut becomes leakier. At the same time, the process of hypoxia/re-oxygenation per-se is directly toxic to tight junctions and increases the risk of leaky gut [21].
On the other hand, Prevotella, Paraprevotella, Desulfovibrio, and Lachnospiraceae species belong to the phylum Firmicutes. More than 90% of the colon bacteria consist of Bacteroidetes and Firmicutes [22]. Increased presence of Desulfovibrio induces mucin degradation and subsequently increases gut permeability [23, 24]. There is some evidence that augmentation of Lachnospiraceae in the gut can increase the risk of obesity [25]. Similarly, Prevotella is strongly linked to systemic inflammation through the production of lipopolysaccharides [26].
3. Intermittent hypoxia and gut dysbiosis (animal and human models)
The episodic nature IH associated with apnea and hypopnea events leads to fluctuation in the oxygen partial pressure (PaO2) in the gut lumen across a centrifugal gradient. These changes in PaO2 shift the gut microbiome toward a dominance of anaerobic and facultative anaerobic bacteria at the expense of obligate aerobic bacteria [27]. Of note, these findings are derived from studies conducted in this field using animal models (mice and rats) with very few human trials. Most of the animal studies used environmental hypoxic chambers, where mice are exposed to cyclical changes in the fraction of oxygen inspired (FIO2) eliciting corresponding targeted changes in blood PaO2 for a specific number of hours during the sleep period of the animals and followed by normal air at 21% FIO2 for the remained of the circadian period mimicking periods of wakefulness during which normal PaO2 is present in OSA patients. The duration of exposures varies between 1-20 weeks depending on the study design. In addition, a wide variety of IH profiles have been used, which differ in both the number of cycles of hypoxia-reoxygenation occurring every hour (simulating a specific apnea-hypopnea index) and the severity of hypoxemia targeted (usual SpO2 achieved between 60-90%) [28]. Another model used inflatable devices implanted in the mice trachea or externally applied which through periodic inflation and deflation mimic obstructive respiratory events [29–31]. The variability in study design and results using these two models has not been evaluated.
It is worth mentioning that IH is not the only sleep disorder that affects the gut microbiota. Sleep deprivation for a short period of time (48 hours) was associated with subtle changes in the gut microbiota [32]. Another study showed that chronic sleep deprivation (mean total sleep time 5.58 hours/night) who were given the chance to extend their sleep for two weeks (mean total sleep time 6.6 hours/night) did not results in changes in the gut microbiota [33]. Another example is circadian rhythm disorders. Circadian misalignment has been shown to have a bidirectional effect on the gut microbiome, where both central and peripheral clocks affect and are affected by changes in the gut microbiome [34, 35]. A common example in this regard in shift workers who exhibit alterations in the gut microbiome that can be partially contributing to the associated higher risk of cardiovascular and metabolic diseases in this group [36].
The fecal sample analysis to characterize the microbiota starts with DNA and RNA isolation followed by 16S r RNA sequencing. Nowadays, in the era of advanced technology in biomedical engineering and bioinformatic analysis, more detailed and accurate methods have emerged in the field of metagenomics. The mainstay amplicon sequencing is 16S rRNA [37, 38]. However, more specialized and unbiased databases (e.g. gunshot approaches used in whole metagenome sequencing) are being utilized nowadays [39, 40]. Another critically important tool that is used to further identify a group of bacteria is metabolomics, which analyzes different molecules and metabolites produced by different bacterial species to identify these species [41]. Ultimately, the combination of metagenomics and metabolomics provides a more comprehensive and accurate analysis of the gut microbiome (Figure 2).
Figure 2:
Pipeline for processing fecal samples for microbiome characterization.
Most of the stool samples obtained from IH-exposed animal models showed an increase in the Firmicutes/Bacteroidetes ratio, which is the hallmark of gut dysbiosis [24]. Specific bacterial species under the phylum Firmicutes include Prevotella and Desulfovibrio and both are increased with IH exposures [17, 24]. In rats, IH combined with high salt diet increased the severity of systemic hypertension likely via increasing levels of blood trimethylamine N-oxide (TMAO), enhanced release of Th1-related interferon-γ (IFN-γ) and reduced levels of the anti-inflammatory cytokine (TGF-β1). These changes coincided with changes in the gut microbiome of the exposed rats, particularly reflected by major decreases in Lactobacillus abundance [42]. Similar studies have confirmed these findings across multiple rodent species [43–48]. Xue et al [49]. suggested that it is not only IH, but rather IH and hypercapnia that plays a role in inflammation and atheroma formation in the pulmonary artery and aortic arch in mice. Although the impact of hypercapnia on the gut microbiome is still controversial, the hypothesis of hypercapnia as a common denominator between OSA and gut dysbiosis deserves expanded exploration through future experimental studies.
One of the pathological conditions that is linked to IH is inflammation. Gut dysbiosis is associated with increased abundance of Prevotella and Klebsiella which are gram negative bacteria that produce toxins such as lipopolysaccharide (LPS). When these toxins leak from the gut into the systemic circulation, they stimulate the release of other inflammatory mediators, such as interleukin-6 (IL-6) and tumor necrosis factor- α (TNF-α), that lead to systemic inflammation [17]. Inflammation plays a critical role in the pathophysiology of many co-morbid diseases (such as hypertension, coronary artery disease, obesity, and diabetes mellitus), and has been implicated in the vast majority of the morbidities associated with OSA as well [50–54].
In humans, few studies examined the association between IH and dysbiosis. The first study on these issues identified increased LPS-binding protein levels in children with OSA, and particularly among obese children with OSA, suggesting increased translocation of LPS to the circulation from the gut [55]. In a trial included pediatric age group (2 years) with history of snoring, there was evidence of increase Firmicutes/Bacteroidetes (F/B) ratio and decrease Actinobacteria/Proteobacteria ratio suggestive of inflammation [56]. In adults, one study showed an increase in the abundance of Proteobacteria, Bacteroidetes, Firmicutes, Fusobacteria, and Acidobacteria in the lungs suggestive of lung dysbiosis [57]. Another study examined the nasal microbiota in patients with OSA and found an increase in some inflammatory mediators (IL-8 and IL-6) [58]. In a small cohort of patients with OSA, three enterotypes, namely predominance of Bacteroides, Ruminococcus, and Prevotella, respectively were identified. Interestingly, arousal-related parameters and distribution of sleep stages were significantly altered among OSA patients with an apnea-hypopnea index (AHI) ≥15/hour total sleep duration among those patients in whom Prevotella predominated [59]. In another preliminary study [60], functional analysis in the fecal microbiomes of 93 OSA patients and their corresponding controls revealed significant differences in the gut microbiota profile between the two study groups. Additionally, decreased in short-chain fatty acid (SCFA)-producing bacteria and increased pathogens, accompanied by elevated levels of IL-6, while the abundance of Lactobacillus was significantly associated with homocysteine levels.
4. Pre- and probiotics as therapeutic interventions in gut dysbiosis
Probiotics refer to species of live bacteria that confer beneficial health effects on the host when ingested in adequate amounts [61]. Health benefits have been demonstrated for specific probiotic strains mainly of the following genera: Lactobacillus (e.g. prevent and decrease the severity of some allergic diseases such as atopic dermatitis), Bifidobacterium (e.g. decrease the severity of diarrhea, reduce symptoms of necrotizing enterocolitis) , Saccharomyces (e.g. reduce duration of traveler’s diarrhea and irritable bowel syndrome), Enterococcus (e.g. treatment of antibiotic-associated diarrhea), Streptococcus (e.g. reduce irritable bowel syndrome symptoms), Pediococcus, Leuconostoc, Bacillus and Escherichia coli (e.g. decrease the severity of inflammatory bowel diseases and decrease the severity of constipation) [62]. Probiotics can exert a wide range of effects such as improving metabolism and immunity through interactions with the host microbiota and also directly with the intestinal epithelium [63, 64]. They can improve mucosal defenses of the GI tract by: i) Blocking the colonization of pathogenic bacteria through decreasing luminal pH, inhibiting adhesion and invasion to epithelial cells and producing antimicrobial compounds. ii) Reinforcement of the mucosal barrier against pathogenic bacteria through enhancement of mucous production and tight junction protein phosphorylation. iii) Influencing both innate and acquired immune system locally in the gut wall [65].
Prebiotics can be defined as “a substrate utilized selectively by host microorganisms conferring a health benefit” [66]. Dietary fiber is one of the main classes of prebiotics that includes inulin, fructooligosaccharides (FOS), galactooligosaccharides (GOS) and resistant starch [67]. Prebiotics can prevent intestinal invasion of pathogens due to their anti-adhesive properties [68] and their ability to stimulate the growth of favorable probiotic bacteria such as Lactobacillus and Bifidobacterium [69]. Byproducts produced through prebiotic digestion and fermentation by gut microbiota (such as SCFAs) can positively modulate local and systemic effects of the immune system [70]. In addition to the role of dietary fibers as a source of SCFA and probiotics, they were found to be negatively correlated with the severity of OSA. Stelmach-Mardas et al [71]. concluded that alcohol consumption is positively correlated with the severity of OSA and high fiber diet is negatively correlated with OSA severity.
The past few decades have seen a surge in experimental and clinical studies on gut dysbiosis and its role in a wide spectrum of diseases. This surge was accompanied by multiple interventions that includes a large variety of pre- and probiotics. Although, many of the beneficial effects of pre- and probiotics can be seen in general population, in the next sections, we will discuss the beneficial effects of pre- and probiotics in cardiometabolic, neurobehavioral and gastrointestinal diseases in the context of sleep related breathing disorders.
4.1. Cardiometabolic Disease
Cardiovascular disease (CVD) continues to be the leading cause of death worldwide with increasing prevalence of common metabolic risk factors such as obesity, type 2 diabetes and the metabolic syndrome [72]. Inadequate effectiveness of pharmaceutical therapies for the management of atherosclerosis, a hallmark of CVD, combined with the recent developments in understanding of the gut microbiome - host interactions have fueled the interest in pre- and probiotics as a potential add-ons therapy for CVD [73]. In gut dysbiosis, compromised intestinal barrier can lead to LPS and other bacterial components leakage triggering inflammatory responses through monocyte recruitment and activation of Toll-like receptors [74]. Immune cell recruitment, production of pro-inflammatory factors and oxidative stress are the main underlying mechanisms promoting the initiation and propagation of endothelial dysfunction and atherosclerosis [75]. Furthermore, increased TMAO, an oxidized form of the bacterial metabolite trimethylamine from dietary choline and L-carnitine, can also promote inflammation, thrombosis, macrophage cell formation and disrupt lipid homeostasis [76]. Primary bile acids synthesized from cholesterol can be deconjugated by certain microbiota that have bile salt hydroxylase (BSH) activity to secondary bile acid salts, which are less soluble and less efficiently reabsorbed from the intestine, resulting in more cholesterol demand to synthesize more bile acids. This cholesterol lowering-effect can be greatly affected by dysbiosis [77]. It is also well documented that gut dysbiosis can reduce SCFA-producing bacteria [78]. Taken together, dysbiosis-mediated atherosclerosis is a promising target for treatment of CVD. Indeed, OSA induces systemic inflammation, oxidative stress and increased sympathetic activity leading to endothelial dysfunction and atherosclerosis [18, 79], and is considered as an independent risk factor for CVD [80]. It is becoming an irrefutable fact that OSA-mediated gut dysbiosis contributes to CVD.
A multitude of recent randomized control trials (RCTs) have been conducted to examine the effects of pre-, pro-, and symbiotic supplements in patients with cardiovascular and metabolic diseases (Tables 1 and 2). Meta-analyses of such interventional studies revealed beneficial effects on cardiometabolic parameters. Zheng et al. identified 13 trials that included 671 patients for analysis and showed that pre-, pro- and synbiotic therapies significantly reduced C- reactive protein (CRP), malondialdehyde (MDA), total cholesterol (TC) and low-density lipoprotein (LDL) levels; they also increased glutathione (GSH), total antioxidant capacity (TAC) and high-density lipoprotein (HDL) in patients with chronic kidney disease (CKD) compared to placebo-treated groups. Subgroup analyses showed that duration of intervention, body mass index (BMI) and age had an effect of therapy on outcomes [81]. Another meta-analysis conducted by the same group on 16 RCTs with 1,060 patients reported that microbial treatment improved biomarkers of inflammation and oxidative stress in diabetic patients [82]. A total of 19 RCTs that included 967 participants with hypercholesterolemia were analyzed by Mo and colleagues and examined the effects of probiotics on serum lipid concentrations. The study found that probiotic interventions reduced TC and LDL levels compared to control with no significant effects on either TG or HDL concentrations. The lipid lowering effects were greater for longer intervention times, specific probiotic strains and were also more beneficial among younger participants [83]. Zhang et al. included 10 RCTs with myocardial infarction (n=1,392), coronary artery disease (n=4,490), stroke (n=7,078), and uncategorized CVD (n=51,707) that examined the effects of fermented dairy food intake on CVD risk in a recent meta-analysis. There was a significant decrease in CVD risk associated with fermented dairy food intake. In a subgroup analysis, cheese and yogurt were associated with decreased CVD risk [84]. Although clinical and observational studies suggest that natural pre- and probiotics have potential benefits on cardiometabolic disease, there is still not enough evidence to routinely and conclusively recommend them at this time, due to the heterogeneity of the approaches used and the lack of specific well circumscribed therapeutic targets and risk groups [85].
Table 2: Pre- and probiotic treatment effects on metabolic parameters.
FBG: fasting blood glucose, HbA1C: hemoglobin A1C, HDL: high-density lipoprotein, HOMA-IR: Homeostatic model of assessment of insulin resistance, hs-CRP: high sensitivity C-reactive protein, INF-λ: interferon-λ, LBP: lipopolysaccharide binding protein, LDL: low-density lipoprotein, NAFLD: non-alcoholic fatty liver disease, TC: total cholesterol, TG: triglycerides, TNF-α: tumor necrosis factor-α.
Pre-/Probiotic | Condition | Dose | Effects | Ref |
---|---|---|---|---|
Ecologic® Barrier (Bifidobacterium and Lactobacillus strains) | obese postmenopausal women | 2 g sachets, 2 sachets/ day (1010/day) for 12 weeks | ↓ Homocysteine, TNF-α, TC, LDL, and TG | Majewska et al.[182] 2020 |
UB316 (contains Bifidobacterium and Lactobacillus strains) | type 2 diabetics | 5 x 109 CFU twice a day for 12 weeks | ↓ HbA1c ↓ Weight ↔ FBG, HOMA-IR, insulin, TC, TG, HDL, and LDL levels. ↑ Quality of life |
Madempudi et al.[183] 2019 |
whole-grain pasta containing barley β-glucans and Bacillus coagulans BC30, 6086 | healthy sedentary overweight and obese subjects | 1 serving/day of innovative pasta for 12 weeks | ↓ hs-CRP and plasma LDL/HDL cholesterol ratio ↓ Plasma resistin |
Angelino et al.[184] 2019 |
Symbiter Omega (multistrain probiotic with flax and wheat germ oil) | type-2 diabetics with NAFLD | 1 sachet (10g) of (250 mg omega-3 fatty acids 1-5%) with probiotics for 8 weeks | ↓ Fatty liver index ↔ Liver stiffness ↓ Serum gamma-glutamyl transpeptidase, TG, and TC, IL-1β, TNF-α, IL-8, IL-6, and INF-γ. |
Kobyliak et al.[185] 2018 |
Bifidobacterium animalis subsp. lactis CECT 8145 (Ba8145) and heat-killed form (h-k Ba8145) | Abdominally obese subjects | 1 capsule/day (1010 CFU of Ba8145, 1010 CFU of h-k Ba8145 for 3 months. | Ba8145 and h-k Ba8145: ↓ Waist circumference, waist circumference/height ratio, Conicity index and BMI h-k Ba8145: ↓ Visceral fat area, diastolic blood pressure and HOMA index. |
Pedret et al.[186] 2019 |
Symbiotic supplement (contains Bifidobacterium, Lactobacillus strains) + oat β-glucan | Specific-pathogen free mice fed HFD | Oral gavage (108 CFU/day) + 1g/kg body weight/day of oat β-glucan for 12 weeks | ↓ Body weight gain ↑ HOMA-IR score ↓ Fasting insulin, TC and LBP ↓ Hepatic steatosis and adipocyte size |
Ke et al.[187] 2019 |
Lactobacillus plantarum TAR4, Lactobacillus acidophilus ATCC 4356 | Sprague-Dawley rats fed high-cholesterol diet | Oral gavage (2 ml (109 CFU/ml) for 4 weeks | ↓ TC, TG, liver TG | Lim et al.[188] 2020 |
Microencapsulated Lactobacillus plantarum LIP-1 | Wistar rats fed HFD | Intragastric administration (20 mL supplement/kg BW (2 × 109 CFU/ml) for 4 weeks | ↓ TC ↑ cholesterol excretion |
Yao et al.[189] 2020 |
Bifidobacterium adolescentis, B. bifidum or Lactobacillus rhamnosus | C57BL/6J mice fed HFD and sucrose | intragastric administration (5 × 109 CFU/mL) for 12 weeks | ↓Fasting and postprandial glucose levels ↑ Glucose tolerance ↓ Pancreatic damage L. rhamnosus >> Bifidobacterium strains in the regulation of blood lipid levels |
Wang et al.[190] 2020 |
In animal models (Tables 1 and 2), probiotic supplementation (VSL#3) reduced high fat diet (HFD)-induced lesion development in apolipoprotein-E knockout (ApoE−/−) mice in addition to reduced vascular inflammation, adhesion molecules, oxidized LDL, TNF-α, plasma TMAO, and TMAO-induced atherosclerosis [86]. A similar study in ApoE −/− mice examined the effects of two Lactobacillus strains (Lactobacillus acidophilus 4356 and 4962) on atherosclerosis. L.4356 significantly reduced atherosclerotic lesion, plasma TC and LDL via inhibition of intestinal cholesterol absorption, but no effects were observed in the L.4962 group.[87] In another study, plasma TMAO levels and TMAO-induced atherosclerosis in ApoE −/− mice were attenuated after Lactobacillus plantarum ZDY04 supplementation [88]. Probiotics have been shown to attenuate endothelial dysfunction by increasing the bioavailability of nitric oxide (NO), reducing oxidative stress and inflammation, and recruitment of progenitor cells [89]. VSL#3 supplementation attenuated oxidative stress-mediated endothelial dysfunction in rat mesentery arteries following bile duct ligation [90]. Furthermore, kefir yogurt (which has a high concentration of Lactobacillus and Bifidobacterium) consumption improved endothelial function in spontaneous hypertensive rats by improving NO bioavailability and reducing oxidative stress [91]. Another study revealed that rats fed with GOS and HFD had significant decreases in serum TG, TC, LDL and VLDL levels along with increased levels of HDL [92]. Although a large body of evidence supports a beneficial role of pre- and probiotics use in cardiovascular and metabolic disease, there are some studies that show no effect or even the opposite, i.e., pro-atherogenic effects [74, 93]. More mechanism-based studies of pre- and probiotics with multiple concentrations and strains are needed to address these discrepancies in the literature.
4.2. Neurobehavioral disorders
In the past decades, the gut microbiota emerged as a key regulator of the gut-brain axis. Evidence from germ free mice showed that myelination, dendritic growth, neurogenesis, blood brain barrier (BBB), microglia, neurotransmitters, and synaptic plasticity are all affected in the absence of microbiota [94]. Alterations in behavior in animals and humans given specific strains of bacteria, and the long-lasting effects of antibiotics on the brain in early life indicate that the microbiota can exert a considerable influence over host cognitive, mood and behavioral processes [94]. Pathways of communication between the gut microbiota and the brain are bidirectional and include: i) the vagus nerve [95] - Vagotomy can decrease the benefits of L. rhamnosus [96] while enteroendocrine cells can release glutamate that activates the vagal pathways [97]; ii) Intestinal microbiota can generate bioactive molecules including SCFAs that regulate microglial inflammatory responses[98], and modulate neurotransmitter release and activity like dopamine, λ-aminobutyric acid (GABA) and norepinephrine, as well as serotonin (through tryptophan metabolism) [94]; iii) the immune system is considered the most important communication pathway since the intestines contains a condensed concentration of immune cells that produces a variety of inflammatory cytokines, which can be transmitted to the central nervous system (CNS) via the circulatory and nervous systems [99]. Pro-inflammatory cytokines can reach the CNS through the humoral route (via saturable transporters at the BBB), neural route (via afferent neurons), and cellular route (immune cells migrating to the brain vasculature and parenchyma).[100] CNS inflammation is initiated when the microglia become activated and release pro-inflammatory cytokines including IL-1β, which eventually induces the production of the stress hormone cortisol [101, 102]. It is well-documented that inflammation is the main underlying mechanism in many neurodegenerative and neuropsychiatric disorders such as Parkinson’s disease (PD), Alzheimer’s disease (AD), and major depressive disorder (MDD).[103] Thus, gut dysbiosis can be linked to neurobehavioral disorders [104]. Pre- and probiotics that influence the gut-brain -axis are called “psychobiotics” and a significant number of studies have recently shown anxiolytic, anti-depressant, emotional, and systemic effects, and also beneficial cognitive effects [105].
Some of the most recent RCTs evaluating the effects of pre- and probiotics in neurobehavioral disorders are discussed in Table 3. A systematic review of 24 RCTs examining the effects of pre- and probiotics on depression and anxiety showed that probiotics, but not supplementation with prebiotics exerted significant beneficial effects on depression and anxiety with a larger effect observed for clinical referral samples than community-based cohorts [106]. Another meta-analysis that evaluated the effects of probiotics on depressive symptoms in 19 RCTs with a total of 1,901 participants found that probiotics treatment led to significant improvements in depressive symptoms compared to placebo. The effects however were in participants with major depressive disorders (MDD), but not in those with other clinical conditions or in the general population.[107] On the other hand, other systematic reviews found no beneficial effects of probiotics in patients with dementia [108] or schizophrenia [109].
Table 3: Pre- and probiotic treatment effects on neurobehavioral parameters.
BDNF: brain-derived neurotrophic factor, CEBPD: CCAAT/enhancing-binding protein delta, IL: interleukin, IkBα: nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor-α, SCFA: short-chain fatty acid.
Pre-/Probiotic | Condition | Dose | Effects | Ref |
---|---|---|---|---|
Multi-strain probiotic (Bifidobacterium longum, Lactobacillus acidophilus, and Enterococcus faecalis) | Elderly patients scheduled for elective orthopedic or colorectal surgery | probiotic capsules (0.84 g) twice daily from admission until discharge (≥107 CFU/capsule each) | ↓ Incidence of postoperative cognitive impairment ↓ IL-6 and cortisol levels 5-7 days after surgery. ↔ Postoperative intensity, sleep quality and GI function recovery |
Wang et al.[191] 2020 |
Bifidobacterium bifidum BGN4 and Bifidobacterium longum BORI | Healthy elders | 4 capsules/day (109 CFU/d) for 12 weeks | ↑ Mental flexibility test and stress score ↑ Serum BDNF levels |
Kim et al.[192] 2020 |
Multi-strain probiotic (contains Bifidobacterium and Lactobacillus strains) + glutamine | Students | 2 capsules/day (109 CFU/d) for 28 days | ↓ Perceived stress scale, depression anxiety stress scale, and state-trait anxiety inventory scores ↓ Serum cortisol levels |
Venkataraman et al.[193] 2020 |
Bifidobacterium breve A1 (MCC1274) | healthy older adults with mild cognitive impairment | 2 capsules/day (1010 CFU/d) for 16 weeks | ↑ Repeatable Battery for the Assessment of Neuropsychological Status total score ↑ Domain scores of immediate memories, visuospatial/constructional, and delayed memory in both intention-to-treat analysis and per-protocol analysis. |
Xiao et al.[194] 2020 |
Lactobacillus Plantarum 299v (LP299v) | patients with major depressive disorder | 2 capsules/day (109 CFU/d) for 28 days for 8 weeks | ↑ Attention and Perceptivity test and the California Verbal Learning Test total recall of trials 1-5 ↓ Kynurenine levels |
Rudzki et al.[195] 2019 |
VSL#3 (multi-strain probiotic) | Ferrets | Syringe-fed 10 ml Kitty Milk Replacement with (4.5 × 109 CFU of freeze-dried VSL#3) for 6 days | ↔ Microbiome composition ↑ Preference for novelty (object and conspecific) |
Dugyala et al.[196] 2020 |
Inulin | Mouse model of Alzheimer’s disease) | Dietary (8% inulin) for 16 weeks | ↑ Beneficial microbiota and decreased harmful microbiota ↑ metabolism in the cecum, periphery, and brain (increased the levels of SCFAs, tryptophan-derived metabolites, bile acids and glycolytic metabolites) ↓ Inflammatory gene expression in the hippocampus. |
Hoffman et al.[197] 2019 |
Lactobacillus plantarum (Lp 286) and (Lp 81) | Swiss mice | oral gavage solution (109/0.1 ml CFU each) for 30 days | ↔ Locomotor activity or learning and memory Lp 286: ↑ Forced swim test and plus-maze discriminative avoidance task |
Barros-Santos et al.[198] 2020 |
multi-strain probiotic (contains Bifidobacterium and Lactobacillus strains) | Mouse model of Parkinson disease | Dietary (1010 CFU/mouse/day) for 16 weeks | ↓ Motor impairments in gait pattern, balance function, and motor coordination. ↑ Tyrosine hydroxylase-positive cell in the substantia nigra |
Hsieh et al.[199] 2020 |
Lactobacillus acidophilus and Bifidobacterium infantis | Specific pathogen free (SPF) pregnant C57/BL6J mice. Pups challenged with IL-6 injection (postnatal systematic inflammation model) | fed daily from E16 to weaning with either a combination of 109 L. acidophilus and 109 B. infantis | ↓ Postnatal systemic inflammation associated with reduced astrocyte/microglia activation and downregulation of the transcriptional regulators CEBPD and IκBα) ↓ Blood-brain barrier permeability and tight junction protein expression in the offspring at pre-weaned age. ↓ Markers associated with leukocyte transendothelial migration, extracellular matrix injury and neuroinflammation. ↑ Neuronal and oligodendrocyte progenitor cell development in the offspring. |
Lu et al.[200] 2020 |
A growing body of evidence from experimental studies has shown that psychobiotics can reduce anxiety and depression (Table 3). In a mouse model of early life-stress, administration of Lactobacillus plantarum PS128 decreased time spent immobile in the Forced Swim Test (FST) (a classic test of depression in rodents) and increased the time spent in the center of the arena in the Open Field Test (OFT), and more entries in the open arms of the Elevated Plus Maze (EPM) (both of these test evaluate anxiety levels). PS128 also increased levels of dopamine, serotonin in the prefrontal cortex and striatum, as well as decreased inflammation and corticosterone levels [110]. In a rat model of AD, multi-strain probiotic administration (Lactobacillus and Bifidobacterium) significantly improved spatial memory acquisition and retention measured by Morris Water Maze (MWM). Treatment also reduced Aβ plaques, MDA, IL-1β, TNF-α and other inflammatory markers in the hippocampus [111]. Prebiotics such as FOS have been shown to increase acetylcholine, serotonin, and adrenaline, as well as decreased damage to the CA1 region of the hippocampus in a rat model of AD [112]. FOS also improved cognition in a mouse model of AD by improving performance in cognitive tests and synaptic plasticity in the hippocampus possibly by regulating levels of glucagon-like peptide-1 (GLP-1) [113]. Increased expression of brain-derived neurotrophic factor (BDNF) and N-Methyl-d-aspartate receptor (NMDAR) subunits in the hippocampus and frontal cortex was observed in mice fed FOS and GOS. In healthy animals, dietary FOS and GOS improved parameters in OFT, FST and EPM [114]. Although our understanding of the gut-brain-axis is still in its early stages, much work is still required to prove that changes in microbiota are central to the pathophysiology of neural system-related disorders. Also, large-scale studies testing optimal doses, strains and timing of application with causative and mechanistic analysis are essential to the field of pre- and probiotics as therapeutic interventions.
From another perspective, many studies have pointed to a higher prevalence of OSA in many neurodegenerative disorders. In Alzheimer dementia, IH induces neuro-inflammation, releases of reactive oxygen species, and subsequently cellular apoptosis [115]. The inflammatory process leads to the accumulation of β amyloid protein in the central nervous system and hyperphosphorylation of tau protein [116, 117]. In Parkinson disease (PD), the exact correlation between the disease and OSA remains unclear. However, dysfunction in the upper airway dilator muscles has been shown in patients with PD which is probably part of the tremor, bradykinesia, and rigidity that is noticed in the disease in general [118]. This is a very similar picture to what is seen in patients with multiple systemic atrophy [119]. Interestingly, many studies confirmed the cognitive improvements in patients with neurogenerative diseases and OSA after using PAP therapy [120–124]. Moreover, a close association was noted between many mental diseases and OSA. A systematic review by Gupta et al [125]. concluded that OSA prevalence maybe increased in patients with MDD and post-traumatic stress disorders (PTSD) and treatment with PAP therapy improved many of psychiatric symptoms in these patients.
3. Gastrointestinal disorders
Inflammatory Bowel Disease (IBD) is characterized by chronic immune-mediated intestinal inflammatory condition driven by environmental, genetic and microbial factors.[126] IBD includes Ulcerative Colitis (UC) and Crohn’s Disease (CD) and is considered a major risk factor for malignancy development, such as colorectal cancer [127]. Recent metagenomic and metabolomic studies revealed changes in microbial composition and metabolites in patients with IBD [128]. Indeed, the depleted metabolites and related species were of anti-inflammatory nature while the enriched metabolites were pro-inflammatory [129]. For instance, SCFA-producing bacteria such as Faecalibacterium prausnitzii[128] and Roseburia intestinalis and hominis[130] were decreased in stool samples from patients with UC and CD. In addition to decreased SCFA levels, tryptophan metabolism (which promotes mucus production and down-regulates inflammation) is also reduced in IBD [131]. It is well documented that secondary bile acids play an important role in modulating host T regulatory function [132]. In patients with IBD, fecal primary bile acids and its conjugates (glycine and taurine) were increased, while secondary bile acids (lithocholate and deoxycholate) were decreased [126]. Bacteria associated with bile acid metabolism, such as Roseburia, Subdoligranulum, and Clostridium leptum were all reduced in IBD [126]. On the other hand, invasive virulent bacteria such as adherent invasive Escherichia coli (AIEC), Enterococcus faecium and Campylobacter concisus, and pro-inflammatory metabolites such as nictotinuric acid, taurine and acylcarnitines were all increased in IBD [129].
Many recent RCTs described the benefits of pre- and probiotics in the management of gastrointestinal disorders (Table 4). A recent meta-analysis included 5 RCTs examining the efficacy of prebiotics and probiotics in functional dyspepsia and found that treatment improved symptoms scores of functional dyspepsia [133]. Another meta-analysis identified 11 RCTs with 729 participants and assessed the effects of prebiotics in patients with IBS but found no differences for severity of abdominal pain, bloating and flatulence and quality of life score. Only flatulence improved by prebiotic doses <6 grams/day and by non-inulin type fructan prebiotics. However, there was a significant increase in Bifdobacteria [134]. One study included 18 RCTs with 1,491 patients with UC and reported that remission efficacy was improved only when probiotics containing Bifidobacterium were used [135]. Another meta-analysis included 10 studies with 1,049 patients evaluated the effects of probiotics on IBD. Results showed no differences in remission, relapse, and complication rates between Escherichia coli Nissle and mesalazine groups. Subgroup analysis suggested that VSL#3 presented a higher remission rate and lower relapse rate [136].
Table 4: Recent interventional studies using pre- and probiotics in gastrointestinal disorders.
COX-2: cyclooxygenase-2, CRP: C-reactive protein, hs-CRP: high-sensitivity C-reactive protein, IBD: inflammatory bowel disease, IBS: irritable bowel syndrome, IL: interleukin, PCNA: proliferating cell nuclear antigen, SCFA: short-chain fatty acid.
Pre-/Probiotic | Condition | Dose | Effects | Ref |
---|---|---|---|---|
Bifico® (Bacillus Coagulans GBI30 6086) | patients with IBD | 2 tablets (2 x 1010 CFU/day) with 2 tablets of mesalazine | ↓ Levels of fecal lactoferrin, 1-antitrypsin, β2-microglobulin, hs-CRP, and IL-6 ↓ Activity scores and recurrence rate |
Fan et al.[201] 2019 |
Symprove™ (Lactobacillus and Enterococcus strains) | patients with ulcerative colitis and Crohn’s disease | Oral suspension 50ml/dose (1010 CFU/day) for 4 weeks | ↔ IBD-quality of life scores ↓ Fecal calprotectin in patients with ulcerative colitis but not Crohn’s disease. |
Bjarnason et al.[202] 2019 |
Multi-strain probiotic (contains Bifidobacterium and Lactobacillus strains) | patients with colorectal adenocarcinoma | 2 capsules/day (109 CFU/d) for one month then 1 capsule/day for up to one year | ↓ Reduction in postoperative complications in the localization of tumors on the rectum and the ascending colon | Bajramagic et al.[203] 2019 |
Xyloglucan (XG), pea protein and tannins (PPT) from grape seed extract, and xylo-oligosaccharides (XOS) | 60 patients with diarrhea-predominant IBS | one capsule twice daily | ↑ Stool consistency ↓ Abdominal pain and bloating ↑ Quality of life |
Trifan et al.[204] 2019 |
Multi-strain probiotic (contains Bifidobacterium and Lactobacillus strains) | colorectal cancer patients at 4 weeks after surgery | orally twice daily (30 × 1010) for 6 months | ↓ Plasma pro-inflammatory cytokine (TNF-α, IL-6, IL-10, IL-12, IL-17A, IL-17C and IL-22) | Zaharuddin et al.[205] 2019 |
Vivomix™ plus Bacillus subtilis | Mouse model of colitis | Oral gavage (50 × 109 probiotic CFU/kg) for 7 days | ↓ Development of signs and symptoms of colitis ↓ Expression of pro-inflammatory cytokines (IL-6 and TNF-α) in colon ↑ Expression of IL-10 mRNA and the number of T regulatory cells in colon |
Biagioli et al.[206] 2020 |
Lactobacillus rhamnosus 1.0320 + inulin | Mouse model of colitis | Intragastric (2 x 108 cfu/0.2ml) + 20mg inulin for 28 days | ↓ Signs of colitis ↓ Disease Activity Index of colon tissue ↓ Myeloperoxidase activity ↑ Hemoglobin content ↓ Expression levels of inflammatory cytokines IL-1β, IL-6, TNF-α ↑ Expression of IL-10. |
Liu et al.[207] 2020 |
Bacillus coagulans MTCC5856 plus green banana resistant starch (GBRS) | Mouse model of colitis | Dietary (2 × 109 CFU/day/mouse) and GBRS (400 mg/day/mouse) for 7 days followed by 7 days of colitis induction | ↓ Disease Activity Index score ↑ Expressions of tight junction proteins ↓ Expression of IL-1β and CRP ↔ SCFA levels across the whole length of the colon. |
Shinde et al.[208] 2020 |
Multi-strain probiotic (contains Bifidobacterium and Lactobacillus strains) | Mouse model of colorectal cancer | Oral gavage (5 x 1011 cfu/0.2ml) for 3 weeks | ↓ Disease activity ↓ Incidence and progress of tumors to higher stages and grades |
Parisa et al.[209] 2020 |
Lactobacillus acidophilus + Djulis | Rat model of colorectal cancer | Dietary (10% djulis + 5 × 106 CFU/g) for 10 weeks | ↓ Numbers of total aberrant crypt foci, sialomucin-producing aberrant crypt foci, and mucin-depleted foci in the distal colon ↓ Expressions of PCNA and COX-2 ↓ Apoptosis-related proteins |
Lee et al.[210] 2019 |
In murine models of colitis, several probiotics such as Bifidobacterium (infantis, bifidum PRL2010 and bifidum 231), Lactobacillus (plantarum 299, animalis BB12 and reuteri 5454), Faecalibacterium prausnitzii and VSL#3 reduced symptoms of disease, enhanced mucosal barrier and exhibited anti-inflammatory effects [137]. Furthermore, Lactobacillus fermentum V3 and plantarum YYC-3 ameliorated colitis-associated tumorigenesis in mice mainly through downregulation of the expression of IL-6, IL-17 f and IL-22, and suppression of the activation of nuclear factor kappa B (Nf-Kb) and Wnt transcriptional pathways [138, 139]. Moreover, in a mouse model of intestinal adenoma, administration of the SCFA-producing Clostridium butyricum exhibited decreased proliferation and increased apoptosis of intestinal tumor cells mainly through suppression of Wnt signaling. Additionally, C. butyricum modulated gut microbiota composition and increased cecal SCFA levels [140]. Symbiotic treatment of rats with colitis (Lactobacillus casei 01 and chitosan-Ca-alignate microparticles) reduced colonic damage and increased lactobacilli counts in feces. Treatment also reduced inflammation and lesions associated with a significant reduction in myeloperoxidase (MPO) activity [141]. Considering the pivotal role of the gut microbiota in IBD and the negative effects of conventional therapy, the supplementation of pre- and probiotics open a potential avenue in managing IBD. Also, standardizing therapeutic protocols could improve future studies and minimize potential bias.
5. Potential novel therapeutic interventions targeting the gut microbiome for the treatment of OSA
Continuous positive airway pressure (CPAP) is the mainstay treatment for OSA. However, the main challenge facing CPAP therapy is adherence. CPAP adherence is overall low and estimated to be around 35% over the last twenty years, with no significant improvements despite improved machines and patient-machine interfaces [142]. Mandibular advancement devices (MAD) are another option of treating OSA, however, they are not optimal for every OSA patient (for example severe OSA patients are unlikely to benefit) and can cause temporomandibular joint discomfort long-term [143]. The remaining options (including surgery and hypoglossal nerve stimulator) are invasive and may carry a higher risk for OSA patients (especially the elderly). We should not forget the role of conservative management (e.g. weight loss and exercise) in the armamentarium aimed at minimizing OSA severity and consequences. Peppard et al [144]. concluded that 10% weight loss reduced the AHI by 26% and 10% weight gain increases 6-fold the risk of developing OSA. Moreover, weight loss (dietary or through bariatric surgery) has been shown to impact the gut microbiome [145].
Therapies that target gut dysbiosis are potential novel therapies that can be adjuvant to OSA standard treatment and can potentially translate into benefits regarding the morbidities associated with OSA.
5.1. Pre- and probiotics
As stated above, experimental and clinical studies using pre- and probiotics treatments point to favorable outcomes in dysbiosis-related conditions. However, very few studies have explored the effects of pre- and probiotics in OSA-mediated dysbiosis. Ganesh et al. treated an rats with OSA (endotracheal obstruction device implantation) and fed HFD with either one billion CFU of Clostridium butyricum or 20% high amylose corn starch Hylon VII and reported that both treatments prevented OSA-induced hypertension, altered the gut microbiota composition, reduced OSA-induced damage to the gut wall, normalized cecal acetate levels and prevented neuroinflammation associated with OSA. They also reported that continuous infusion of exogenous acetate for 2 weeks prevented OSA-induced hypertension [146].
In another study, Liu et al. investigated the effect of Lactobacillus rhamnosus GG strain (LGG) on the development of OSA-induced hypertension in rats exposed to IH. As mentioned above, they found that IH combined with high salt content in the diet exacerbated the severity of hypertension by increasing levels of plasma TMAO, IFN-λ release from inflammatory cells, and reductions in TGF-β1, while also displaying depleted Lactobacillus content in the gut. They also reported increased expression of phosphorylated extracellular signal-regulated kinase1/2 (ERK1/2), phosphorylated protein kinase B (Akt) and phosphorylated mammalian target of rapamycin (mTOR) in the aorta. Treatment with LGG prevented increase in hypertension, modulated Th1/Th2 cytokine imbalance and blunted the phosphorylation of ERK1/2, Akt and mTOR.[42] Another study found that LGG treatment in obese mice exposed to IH for 15 weeks prevented cardiac remodeling and inflammation. The cardioprotective effect was associated with up-regulation of nuclear factor erythroid2-related factor (Nrf2) – mediated antioxidant pathways [147].
A very recent study by Liu et al. reported glucose intolerance, insulin resistance, increased adipose tissue inflammation and adipocyte size and reduced circulation levels of adiponectin in obese mice exposed to IH for 15 weeks, all parameters were restored by LGG treatment and was associated with increased hepatic fibroblast growth factor 21 (FGF21) mRNA expression and circulating FGF21 protein levels. The increased FGF21 was associated with upregulated hepatic peroxisome proliferator-activated receptor-α (PPAR-α) expression and fecal butyrate concentration. Furthermore, LGG treatment significantly reduced IH-induced hepatic fat accumulation and apoptosis [148]. Collectively, the data from these experimental studies show metabolic, neuronal and cardiovascular benefits which indicate to a potential adjuvant therapy in patients with OSA. Nevertheless, large RCTs need to be performed using multiple species of probiotics and types of prebiotics to determine their health benefits in OSA-induced dysbiosis.
5.2. Fecal matter transplantation or fecal microbiota transfer (FMT)
FMT simply implies the transfer of stool from healthy donor’s colon to a recipient’s colon [149]. This procedure is utilized in patients with difficult to treat or resistant Clostridium difficile with or without inflammatory bowel disease [150]. This is the only approved clinical indication for the use of FMT as per the European consensus conference on fecal microbiota transplantation in clinical practice [151]. However, FMT has also shown very promising results in the treatment of obesity and diabetes mellitus. In obesity, FMT trials in animal and human models showed an increase in Bacteroidetes and decrease in Firmicutes [152], increase in butyrate-producing bacteria (e.g., Roseburia intestinalis or Eubacterium hallii) [153], and increase in Verrucomicrobia, Akkermansia, and Alistipes [154]. In type II diabetes mellitus, FMT has been shown to correct insulin resistance and promote the repair and recovery of islet cells [155]. FMT plays an important therapeutic intervention in systemic hypertension in both animal and human models. Adnan et al. reported that FMT of cecal microbiota from spontaneous hypertensive and stroke prone rats to normotensive WKY rats resulted in an elevation in blood pressure in the latter [156]. In humans, Li et al. [157] examined the fecal microbiota in three groups in humans (Normal, pre-hypertensive and hypertensive) and noticed significant increases in the relative abundance of Prevotella and Klebsiella in both pre-hypertensive and hypertensive groups compared to the normotensive group. They also showed that hypertension is transferrable through FMT. Fecal microbiota from three groups were transferred to germ-free normotensive mice which led to increases in blood pressure in the mice who received fecal microbiota from the hypertensive and pre-hypertensive groups.
FMT has been utilized to test the transferability of OSA-induced hypertension in animal models. Durgan et al. [47] investigated the effect of high fat diet (HFD) alone, OSA alone, and both HFD and OSA on systemic hypertension in a rat model implanted with an endotracheal device that can be inflated/deflated mimicking an apnea/hypopnea at a rate of 60 times per hour for 8 hours every day. They concluded that HFD and OSA caused significant elevation in systolic blood pressure compared to HFD alone or OSA alone suggesting a synergistic effect of both on blood pressure. To further prove this hypothesis, they transferred the gut microbiota from donor rats fed (HFD) with no OSA and rats fed HFD with OSA into recipient rats fed normal chow diet with OSA and found that recipients who received gut microbiota from OSA-HFD donors developed significant elevation in systolic blood pressure compared to HFD alone donors. They also noticed that treating rats with oral antibiotics to deplete the gut microbiota prevented the elevation in systolic blood pressure. Such findings support the role of OSA- induced gut dysbiosis in inducing or exacerbating systemic hypertension. Different parameters of gut dysbiosis were tested in this trial. First, the F/B ratio which is the signature of gut dysbiosis was the highest in HFD rats alone. Combined HFD and OSA showed lower increases in F/B ratio but still higher than sham rats fed normal chow diet. Second, the Chao index (which reflects the total number of distinct genera) was decreased in rats fed HFD. Third, Shannon index (which reflects the relative abundance and richness of genera) was not statistically significant in rate fed HFD. It was noticed also that rats that are fed HFD and have OSA have significant reduction in the relative abundance of SCFA-producing bacteria, especially butyrate. Ruminococcaceae are prominent producers of butyrate and their abundance was reduced from 20% to 10% in this study. At the same time, there was an increase in the relative abundance of lactate-producing bacteria, namely (Lactococcus and Coprobacillus). Of note, lactate in gut is known to increase blood pressure [158].
Currently, the only clinical indication for FMT in humans is severe and resistant Clostridium difficile to antibiotic therapy. The procedure per se is effective and relatively safe. Cammarota et al.[159] conducted a systematic review which included one randomized controlled trial and showed resolution of diarrhea in 87% of total participants with infusion of the fecal suspension matter in the colon using colonoscopy being the most effective site of infusion. Up to date, FMT is not indicated for any other GI or non-GI diseases in humans.
5.3. Short chain fatty acids (SCFAs)
SCFAs include butyrate, propionate, and acetate. They are produced by the gut microbiota and have many beneficial effects locally (in the colon) and extracolonic. SCFAs, especially butyrate, are the major source of energy and nutrients for colonocytes [160]. Butyrate plays a role in the oxidative phosphorylation process that takes place in the mitochondria of the colonic epithelial cells [161]. Butyrate has anti-inflammatory features via the inhibition of IL-12, upregulation of IL-10 in monocytes [162], inhibition of many proinflammatory mediators (such as IL-1β, TNF-α), and suppression of NF-κB pathway which plays an important role in inflammation.[163] Similarly, acetate has anti-inflammatory features in vitro by enhancing the release of reactive oxygen species that have a bactericidal activity against pathogens [164]. Butyrate showed an anti-tumorigenic activity on variable human cancer cell lines (such has human hepatoma cells and human colon cancer cells).[165, 166] Locally, SCFAs play a vital role in maintaining the gut integrity. The mucus layer that separates the colonocytes from the lumen consists of secretory and epithelial membrane-bound mucin glycoproteins [167]. Butyrate and propionate can induce the secretion of some of these mucin glycoprotein to protect the mucus layer in some diseases (e.g., mucositis) [168]. In addition to the mucus layer, tight junction proteins (e.g. ZO-1 and occludin) are located between the epithelial cells laterally [169]. In vitro, butyrate was shown to enhance the assembly of ZO-1 and occludin proteins which increase trans-epithelial resistance (TER) and consequently maintain the gut integrity and prevent gut permeability [170]. Another SCFA, acetate, was shown in vitro to prevent the translocation of shiga toxin from the lumen into the blood stream [171] (Table 5).
Table 5. The use of SCFAs in some diseases (Human and animal models).
SCFA= short chain fatty acids; DBP= diastolic blood pressure; SBP= systolic blood pressure; DOCA= deoxycorticosterone acetate; HDAC= histone deacetylase; HFD= high fat diet; GLP-1= Glucagon-like peptide-1; GPR= G-protein-coupled Receptor.
SCFA | Condition | Effects | Reference |
---|---|---|---|
Cardiovascular disorders | |||
Propionate | Hypertensive mice | ↓ Blood pressure ↓ Vascular inflammation and Atherosclerosis ↓ Cardiac immune cell infiltration and remodeling ↓ Susceptibility to Ventricular Arrythmias |
Bartolomaeus et al.[211] 2019 |
Formate | Patients with hypertension | ↓ Systolic and diastolic blood pressure | Holmes et al.[212] 2008 |
Acetate | Hypertensive mice (DOCA) | ↓ Blood pressure ↓ Cardiac hypertrophy ↓ Cardiac and renal fibrosis |
Marques et al.[213] 2017 |
Metabolic disorders | |||
Butyrate and propionate | Diabetic mice | Both: ↑ Pancreatic β-cells by incretins ↑ plasma insulin ↑ Oral glucose tolerance test Propionate: ↓ Fasting blood glucose |
Lin et al.[214] 2012 |
Butyrate | Diabetic mice | ↓ Weight gain ↓ Insulin resistance ↑ energy expenditure ↑ Mitochondrial function |
Gao et al.[215] 2009 |
Butyrate | Diabetic rats | ↑ β-cell proliferation ↓ β-cell apoptosis ↓ HbA1c and plasma glucose ↑ Plasma insulin |
Khan et al.[216] 2014 |
Butyrate and propionate | Obese mice | Butyrate: ↓ Food intake Propionate: ↔ Food intake |
Lin et al.[214] 2012 |
Butyrate | Obese mice | ↓ Total body, liver and epididymal fat pad weights ↑ Adiponectin pathway ↑ Mitochondrial function ↑ Fatty acid β-oxidation ↓ Lipid deposition in the muscle |
Hong et al.[217] 2016 |
Butyrate | Obese mice | ↓ Weight gain ↓ Food intake ↑ release of GLP-1 ↑ glucose tolerance |
Yadav et al.[218] 2013 |
Neoplastic disorders | |||
Butyrate | Human liver cancer cell lines | ↓ telomerase activity ↔ levels of reverse transcriptase component (as HDAC inhibitor) |
Nakamura et al.[165] 2001 |
Butyrate | Human colon cancer cell line | ↑ cancer cell apoptosis as a GPR109A ligand | Thangaraju et al.[219] 2009 |
Since OSA is associated with gut dysbiosis, theoretically, supplementation with SCFAs could protect against developing OSA-induced complications. To test this theory, Ganesh et al. [146] conducted an animal trial in which rats were randomized into three groups (HFD fed rats, HFD + Hylon “prebiotic”, and HFD + C. butyricum “probiotic”). Since its known that OSA alter the gut microbiota profile, the next step was to test if the metabolomic measures (i.e. the metabolites produced by altered gut microbiota) and they found that OSA decreased acetate concentration in the rat’s colon in HFD group. The addition of Hylon and C. butyricum increased acetate concentration independent of OSA and OSA did not have any impact on butyrate or propionate levels. After concluding that OSA reduced acetate concentration in the cecum, the next step was to examine the effect of supplementing acetate. They continuously replaced acetate in the colon using an indwelling catheter to see if that can prevent the development of HTN in OSA group. Cecal infusion of 20 μmol/(kg/min) sodium acetate or phosphate buffered saline (PBS) started and continued for two weeks in sham and OSA rats. After two weeks, OSA rats treated with PBS showed reduction in acetate in the cecum while OSA rats treated with acetate showed 2-fold increase in acetate concentration compared to sham. Acetate also prevented any increase in IL-1α and IL-6 in OSA group.
6. Conclusions
OSA is a chronic disease associated with gut dysbiosis. Recent findings reveal complex interactions between the microbiota and other biological systems, whereby perturbations in the microbiota structure and metabolites are linked to cardiometabolic, neurobehavioral and gastrointestinal diseases. Gut dysbiosis is characterized by compromising the intestinal barrier and induction of low systemic inflammation. Dysbiosis treatment with pre,pro-biotics, SCFAs and FMT have shown to mitigate dysbiosis-induced pathophysiology in many disorders. Current pre-clinical studies show beneficial effects of pre- and probiotics in animal models of OSA. However, randomized control trials in OSA patients and more mechanistic studies are needed to justify recommending those interventions as adjuvant therapies in OSA with the aim of preventing, abrogating or at least attenuating OSA-induced morbidities.
7. Expert opinion
The field of the gut microbiome and gut dysbiosis is rapidly evolving. Since the launch of the Human Microbiome Project by the National Institutes of Health in 2007 and with the current advances in metagenomics, metatranscriptomics, and metabolomics, our understanding of the human microbiome and its complicated interactions with the host has enormously progressed [172]. Recent studies indicate that perturbations in the gut microbiome can lead to a plethora of disorders across biological systems [173]. However, more mechanistic pre-clinical studies and large cohort studies are required to further elucidate these interactions. Questions have also been raised regarding the reproducibility of investigations on the gut microbiome research in experimental animals [174]. Environmental factors, rodent husbandry, and treatment protocols should be standardized and show the desired reproducibility. Although many of the microbes have been identified, many others (including viruses and fungi) are still undetermined [175]. Defining the host microbiome is not a straightforward process. For instance, High F:B ratio is usually associated with pathological states [176] but this is not always the case [177, 178]. In this context, the “pathobiome” is still not well defined.
OSA is associated with alterations in the gut microbiome profile and metabolites [179]. Linking OSA to gut dysbiosis might open new avenues for targeting chronic illnesses associated with OSA, however, the literature so far does not prove causal relationship. For example, Moreno et al. examined the impact of reversing IH in mice for six weeks and surprisingly this did not completely reverse gut dysbiosis [45]. Accordingly, we need large cohort studies to help establish a correlation between OSA and gut dysbiosis, and whether treating OSA can restore the gut microbiota, notwithstanding the potential confounders. In this regard, we need to tackle numerous issues such as sample collection, storage, processing, and potential contaminations [74]. Although the exact mechanisms behind OSA-induced dysbiosis are still not fully identified, it is probable that OSA-induced IH and sleep fragmentation can foster inflammation and oxidative stress, the latter playing a role in initiating gut dysbiosis, all the while dysbiosis can exacerbate inflammation and oxidative stress in a vicious cycle. Furthermore, gut dysbiosis and OSA share similar bidirectional relationships with chronic conditions such as obesity, type 2 diabetes mellitus, and systemic hypertension, which further complicates the OSA-dysbiosis relationships, and makes therapeutic interventions more difficult to delineate. Large interventional studies in OSA patients using CPAP and its effect on the microbiome with consistent follow-up, may provide a better insight on the efficacy of CPAP in targeting dysbiosis and may also suggest whether adjuvant interventions are worthwhile pursuing.
Current evidence from clinical and experimental studies show beneficial effects of pre- and probiotics treating gut dysbiosis in many conditions [180]. While no studies have been conducted in OSA patients, animal models of OSA treated with pre- and probiotics could modulate metabolism, blood pressure and immunity [146]. Given the useful effects of pre-and probiotics in mitigating dysbiosis-related inflammation in cardiometabolic, gastrointestinal and neurobehavioral conditions, it is plausible that pre- and probiotics treatment can target OSA-induced dysbiosis. Additionally, the anti-inflammatory effects of pre- and probiotics may be able to modify risk factors of OSA (such as obesity and hypertension) and in turn reduce the incidence or severity of OSA (Figure 3). However, there is minimal guidance from regulatory entities with conflicting information on probiotics in the literature sources used for clinical guidelines [181]. Furthermore, more studies in OSA patients and animal OSA models with multiple species and doses of pre- and probiotics are needed to better tailor treatments for OSA patients. SCFAs (especially butyrate and acetate) are very promising interventions targeting OSA-induced dysbiosis and OSA-induced co-morbidities especially that SCFAs have been shown to ameliorate OSA-induced inflammation and OSA-induced hypertension in animal models. Finally, FMT is another promising approach to ameliorate OSA-induced morbid conditions, yet it is too early to consider this option due to the lack of sufficient evidence. FMT has proven that some pathological features and diseases (e.g. hypertension and obesity) are transferrable by the gut microbiome in animal models. More animal studies are required to test the transferability of OSA symptoms (e.g. excessive daytime sleepiness) and OSA-induced co-morbidities by the gut microbiome.
Figure 3: Treatment of gut dysbiosis may ameliorate chronic diseases and reduce the development or progression of OSA.
IH: intermittent hypoxia, FMT: fecal matter transplantation, OSA: obstructive sleep apnea, SCFA: short chain fatty acid, SF: sleep fragmentation
Additional mechanistic and large cohort studies are needed to establish cause and effect relationships. Pre-and probiotics, beneficial bacterial metabolites and fecal matter transplantation from healthy subjects are novel therapeutic options that may benefit patients with OSA and OSA-induced co-morbidities. However, we are still in the early stages of establishing the recommendations of such use. Yet, we could envision a scenario soon where a well-defined microbiota profile associated with the production of toxic molecules would be appropriately modulated to lessen the disease risk of OSA, and that such approaches may open the way for precision-based interventions, one patient at a time.
Article Highlights.
Obstructive sleep apnea (OSA) is a chronic condition associated with multiple chronic disorders affecting multiple systems
The gut microbiome is integral to human health and imbalance in gut commensal bacteria is linked to many diseases
OSA can alter the gut microbiome leading to dysbiosis
Pre- and probiotics and short chain fatty acids (SCFA) can protect against dysbiosis by strengthening the gut barrier integrity and production of immunomodulatory compounds
Given the useful effects of pre-and probiotics in mitigating dysbiosis-related inflammation in cardiometabolic, gastrointestinal and neurobehavioral conditions, it is plausible that pre- and probiotic treatment can target OSA-induced dysbiosis
Acknowledgments
Funding
This paper was not funded
List of abbreviations:
- AD
Alzheimer’s disease
- AHI
apnea-hypopnea index
- Akt
protein kinase B
- ApoE−/−
apolipoprotein-E knockout
- BA
bile acid
- BBB
blood brain barrier
- BDNF
brain-derived neurotrophic factor
- CD
Crohn’s disease
- CKD
chronic kidney disease
- CNS
central nervous system
- CPAP
continuous positive airway pressure
- CRP
c- reactive protein
- EPM
elevated plus maze
- ERK1/2
extracellular signal-regulated kinase1/2
- F/B ratio
Firmicutes/Bacteroidetes ratio
- FGF21
fibroblast growth factor 21
- FIO2
fraction of oxygen inspired
- FMT
fecal matter transfer/fecal microbiota transplantation
- FOS
fructooligosaccharides
- FST
forced swim test
- GABA
λ-aminobutyric acid
- GLP-1
glucagon-like peptide-1
- GOS
galactooligosaccharides
- GSH
glutathione
- HDL
high-density lipoprotein
- HFD
High fat diet
- HTN
hypertension
- IBD
inflammatory bowel disease
- IH
intermittent hypoxia
- LDL
low-density lipoprotein
- LPS
lipopolysaccharides
- MAD
Mandibular advancement devices
- MDA
malondialdehyde
- MDD
major depressive disorder
- MPO
myeloperoxidase
- mTOR
mammalian target of rapamycin
- Nf-ƘB
nuclear factor kappa B
- NO
nitric oxide
- NMDAR
N-Methyl-d-aspartate receptor
- Nrf2
nuclear factor erythroid2-related factor
- OSA
obstructive sleep apnea
- OFT
open field test
- PaO2
oxygen partial pressure
- PD
Parkinson’s disease
- PPAR-α
proliferator-activated receptor-α
- RCT
randomized control trail
- ROS
reactive oxygen species
- SCFA
short-chain fatty acids
- TAC
total antioxidant capacity
- TC
total cholesterol
- TER
trans-epithelial resistance
- TMA
trimethylamine
- TMAO
trimethylamine N-oxide
- WBCs
white blood cells
- UC
ulcerative colitis
Footnotes
Declaration of Interest
The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants, or patents received or pending, or royalties.
Reviewer disclosures
Peer reviewers on this manuscript have no relevant financial or other relationships to disclose
References
Papers of special note have been highlighted as either of interest (•) or of considerable interest (••) to readers
- 1.Remmers JE, et al. , Pathogenesis of upper airway occlusion during sleep. J Appl Physiol Respir Environ Exerc Physiol, 1978. 44(6): p. 931–8. [DOI] [PubMed] [Google Scholar]
- 2.Young T, et al. , Burden of sleep apnea: rationale, design, and major findings of the Wisconsin Sleep Cohort study. Wmj, 2009. 108(5): p. 246–9. [PMC free article] [PubMed] [Google Scholar]
- 3.Burt VL, et al. , Trends in the prevalence, awareness, treatment, and control of hypertension in the adult US population. Data from the health examination surveys, 1960 to 1991. Hypertension, 1995. 26(1): p. 60–9. [DOI] [PubMed] [Google Scholar]
- 4.Marshall NS, et al. , Is sleep apnea an independent risk factor for prevalent and incident diabetes in the Busselton Health Study? J Clin Sleep Med, 2009. 5(1): p. 15–20. [PMC free article] [PubMed] [Google Scholar]
- 5.Brunetti V, et al. , Sleep apneas trigger epilepsy. Sleep Med, 2017. 39: p. 23–24. [DOI] [PubMed] [Google Scholar]
- 6.Xu W, et al. , Increased oxidative stress is associated with chronic intermittent hypoxia-mediated brain cortical neuronal cell apoptosis in a mouse model of sleep apnea. Neuroscience, 2004. 126(2): p. 313–23. [DOI] [PubMed] [Google Scholar]
- 7.Row BW, et al. , Intermittent hypoxia is associated with oxidative stress and spatial learning deficits in the rat. Am J Respir Crit Care Med, 2003. 167(11): p. 1548–53. [DOI] [PubMed] [Google Scholar]
- 8.Kheirandish-Gozal L and Gozal D, Obstructive Sleep Apnea and Inflammation: Proof of Concept Based on Two Illustrative Cytokines. Int J Mol Sci, 2019. 20(3). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Kheirandish-Gozal L, et al. , Effects of adenotonsillectomy on plasma inflammatory biomarkers in obese children with obstructive sleep apnea: A community-based study. Int J Obes (Lond), 2015. 39(7): p. 1094–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Neish AS, Microbes in gastrointestinal health and disease. Gastroenterology, 2009. 136(1): p. 65–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Kamada N, et al. , Role of the gut microbiota in immunity and inflammatory disease. Nat Rev Immunol, 2013. 13(5): p. 321–35. [DOI] [PubMed] [Google Scholar]
- 12.Bäckhed F, et al. , Host-bacterial mutualism in the human intestine. Science, 2005. 307(5717): p. 1915–20. [DOI] [PubMed] [Google Scholar]
- 13.Natividad JM and Verdu EF, Modulation of intestinal barrier by intestinal microbiota: pathological and therapeutic implications. Pharmacol Res, 2013. 69(1): p. 42–51. [DOI] [PubMed] [Google Scholar]
- 14.den Besten G, et al. , The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J Lipid Res, 2013. 54(9): p. 2325–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Bäumler AJ and Sperandio V, Interactions between the microbiota and pathogenic bacteria in the gut. Nature, 2016. 535(7610): p. 85–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Gensollen T, et al. , How colonization by microbiota in early life shapes the immune system. Science, 2016. 352(6285): p. 539–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Poroyko VA, et al. , Chronic Sleep Disruption Alters Gut Microbiota, Induces Systemic and Adipose Tissue Inflammation and Insulin Resistance in Mice. Sci Rep, 2016. 6: p. 35405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Wu ZH, et al. , The role of nitric oxide (NO) levels in patients with obstructive sleep apnea-hypopnea syndrome: a meta-analysis. Sleep Breath, 2020. [DOI] [PubMed] [Google Scholar]
- 19.Qin J, et al. , A human gut microbial gene catalogue established by metagenomic sequencing. Nature, 2010. 464(7285): p. 59–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Canani RB, et al. , Potential beneficial effects of butyrate in intestinal and extraintestinal diseases. World J Gastroenterol, 2011. 17(12): p. 1519–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Vinolo MA, et al. , Regulation of inflammation by short chain fatty acids. Nutrients, 2011. 3(10): p. 858–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Dominguez-Bello MG, et al. , Development of the human gastrointestinal microbiota and insights from high-throughput sequencing. Gastroenterology, 2011. 140(6): p. 1713–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Payne AN, Chassard C, and Lacroix C, Gut microbial adaptation to dietary consumption of fructose, artificial sweeteners and sugar alcohols: implications for host-microbe interactions contributing to obesity. Obes Rev, 2012. 13(9): p. 799–809. [DOI] [PubMed] [Google Scholar]
- 24.Moreno-Indias I, et al. , Intermittent hypoxia alters gut microbiota diversity in a mouse model of sleep apnoea. Eur Respir J, 2015. 45(4): p. 1055–65. [DOI] [PubMed] [Google Scholar]
- 25.de la Cuesta-Zuluaga J, et al. , Gut microbiota is associated with obesity and cardiometabolic disease in a population in the midst of Westernization. Sci Rep, 2018. 8(1): p. 11356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Scher JU, et al. , Expansion of intestinal Prevotella copri correlates with enhanced susceptibility to arthritis. Elife, 2013. 2: p. e01202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Albenberg L, et al. , Correlation between intraluminal oxygen gradient and radial partitioning of intestinal microbiota. Gastroenterology, 2014. 147(5): p. 1055–63. e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Farre R, et al. , Intermittent Hypoxia Severity in Animal Models of Sleep Apnea. Front Physiol, 2018. 9: p. 1556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Schoorlemmer GH, et al. , A new method to produce obstructive sleep apnoea in conscious unrestrained rats. Exp Physiol, 2011. 96(10): p. 1010–8. [DOI] [PubMed] [Google Scholar]
- 30.Crossland RF, et al. , A new rodent model for obstructive sleep apnea: effects on ATP-mediated dilations in cerebral arteries. Am J Physiol Regul Integr Comp Physiol, 2013. 305(4): p. R334–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Carreras A, et al. , Non-invasive system for applying airway obstructions to model obstructive sleep apnea in mice. Respir Physiol Neurobiol, 2011. 175(1): p. 164–8. [DOI] [PubMed] [Google Scholar]
- 32.Benedict C, et al. , Gut microbiota and glucometabolic alterations in response to recurrent partial sleep deprivation in normal-weight young individuals. Mol Metab, 2016. 5(12): p. 1175–1186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Reutrakul S, et al. , No changes in gut microbiota after two-week sleep extension in chronically sleep-deprived individuals. Sleep Med, 2020. 68: p. 27–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Matenchuk BA, Mandhane PJ, and Kozyrskyj AL, Sleep, circadian rhythm, and gut microbiota. Sleep Med Rev, 2020. 53: p. 101340. [DOI] [PubMed] [Google Scholar]
- 35.Mashaqi S and Gozal D, “Circadian misalignment and the gut microbiome. A bidirectional relationship triggering inflammation and metabolic disorders”- a literature review. Sleep Med, 2020. 72: p. 93–108. [DOI] [PubMed] [Google Scholar]
- 36.Reynolds AC, et al. , The shift work and health research agenda: Considering changes in gut microbiota as a pathway linking shift work, sleep loss and circadian misalignment, and metabolic disease. Sleep Med Rev, 2017. 34: p. 3–9. [DOI] [PubMed] [Google Scholar]
- 37.Chaffron S, et al. , A global network of coexisting microbes from environmental and whole-genome sequence data. Genome Res, 2010. 20(7): p. 947–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Faust K, et al. , Microbial co-occurrence relationships in the human microbiome. PLoS Comput Biol, 2012. 8(7): p. e1002606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Sharpton TJ, An introduction to the analysis of shotgun metagenomic data. Front Plant Sci, 2014. 5: p. 209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Nelson KE, et al. , A catalog of reference genomes from the human microbiome. Science, 2010. 328(5981): p. 994–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Fiehn O, Metabolomics--the link between genotypes and phenotypes. Plant Mol Biol, 2002. 48(1–2): p. 155–71. [PubMed] [Google Scholar]
- 42.Liu J, et al. , Lactobacillus rhamnosus GG strain mitigated the development of obstructive sleep apnea-induced hypertension in a high salt diet via regulating TMAO level and CD4(+) T cell induced-type I inflammation. Biomed Pharmacother, 2019. 112: p. 108580. [DOI] [PubMed] [Google Scholar]
- 43.Tripathi A, et al. , Intermittent Hypoxia and Hypercapnia Reproducibly Change the Gut Microbiome and Metabolome across Rodent Model Systems. mSystems, 2019. 4(2). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.O’Connor KM, et al. , Bugs, breathing and blood pressure: microbiota-gut-brain axis signalling in cardiorespiratory control in health and disease. J Physiol, 2020. [DOI] [PubMed] [Google Scholar]
- 45.Moreno-Indias I, et al. , Normoxic Recovery Mimicking Treatment of Sleep Apnea Does Not Reverse Intermittent Hypoxia-Induced Bacterial Dysbiosis and Low-Grade Endotoxemia in Mice. Sleep, 2016. 39(10): p. 1891–1897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Lucking EF, et al. , Chronic intermittent hypoxia disrupts cardiorespiratory homeostasis and gut microbiota composition in adult male guinea-pigs. EBioMedicine, 2018. 38: p. 191–205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Durgan DJ, et al. , Role of the Gut Microbiome in Obstructive Sleep Apnea-Induced Hypertension. Hypertension, 2016. 67(2): p. 469–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Almendros I and Farre N, Obstructive Sleep Apnea and Atherosclerosis: Both the Gut Microbiome and Hypercapnia Matter. Am J Respir Cell Mol Biol, 2017. 57(5): p. 501–503. [DOI] [PubMed] [Google Scholar]
- 49.Xue J, et al. , Intermittent Hypoxia and Hypercapnia Accelerate Atherosclerosis, Partially via Trimethylamine-Oxide. Am J Respir Cell Mol Biol, 2017. 57(5): p. 581–588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Turnbull CD, et al. , Transcriptomics Identify a Unique Intermittent Hypoxia-mediated Profile in Obstructive Sleep Apnea. Am J Respir Crit Care Med, 2020. 201(2): p. 247–250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Sanderson JE, Fang F, and Wei Y, Obstructive sleep apnoea and inflammation in age-dependent cardiovascular disease. Eur Heart J, 2020. 41(26): p. 2503. [DOI] [PubMed] [Google Scholar]
- 52.Campos-Rodriguez F, et al. , Interleukin 6 as a marker of depression in women with sleep apnea. J Sleep Res, 2020: p. e13035. [DOI] [PubMed] [Google Scholar]
- 53.Arnaud C, et al. , Obstructive sleep apnoea and cardiovascular consequences: Pathophysiological mechanisms. Arch Cardiovasc Dis, 2020. 113(5): p. 350–358. [DOI] [PubMed] [Google Scholar]
- 54.Wali SO, et al. , The utility of proinflammatory markers in patients with obstructive sleep apnea. Sleep Breath, 2020. [DOI] [PubMed] [Google Scholar]
- 55.Kheirandish-Gozal L, et al. , Lipopolysaccharide-binding protein plasma levels in children: effects of obstructive sleep apnea and obesity. J Clin Endocrinol Metab, 2014. 99(2): p. 656–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Collado MC, et al. , Dysbiosis in Snoring Children: an Interlink to Comorbidities? J Pediatr Gastroenterol Nutr, 2018. [DOI] [PubMed] [Google Scholar]
- 57.Lu D, et al. , Profiling of lung microbiota in the patients with obstructive sleep apnea. Medicine (Baltimore), 2018. 97(26): p. e11175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Wu BG, et al. , Severe Obstructive Sleep Apnea is Associated with Alterations in the Nasal Microbiome and Increase in Inflammation. Am J Respir Crit Care Med, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Ko CY, et al. , Disruption of sleep architecture in Prevotella enterotype of patients with obstructive sleep apnea-hypopnea syndrome. Brain Behav, 2019. 9(5): p. e01287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Ko CY, et al. , Gut microbiota in obstructive sleep apnea-hypopnea syndrome: disease-related dysbiosis and metabolic comorbidities. Clin Sci (Lond), 2019. 133(7): p. 905–917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Butel MJ, Probiotics, gut microbiota and health. Med Mal Infect, 2014. 44(1): p. 1–8. [DOI] [PubMed] [Google Scholar]
- 62.Fijan S, Microorganisms with claimed probiotic properties: an overview of recent literature. Int J Environ Res Public Health, 2014. 11(5): p. 4745–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.El Aidy S, Dinan TG, and Cryan JF, Gut Microbiota: The Conductor in the Orchestra of Immune-Neuroendocrine Communication. Clin Ther, 2015. 37(5): p. 954–67. [DOI] [PubMed] [Google Scholar]
- 64.Patterson E, et al. , Gut microbiota, obesity and diabetes. Postgrad Med J, 2016. 92(1087): p. 286–300. [DOI] [PubMed] [Google Scholar]
- 65.Bermudez-Brito M, et al. , Probiotic mechanisms of action. Ann Nutr Metab, 2012. 61(2): p. 160–74. [DOI] [PubMed] [Google Scholar]
- 66.Gibson GR, et al. , Expert consensus document: The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics. Nat Rev Gastroenterol Hepatol, 2017. 14(8): p. 491–502. [DOI] [PubMed] [Google Scholar]
- 67.Bindels LB, et al. , Towards a more comprehensive concept for prebiotics. Nat Rev Gastroenterol Hepatol, 2015. 12(5): p. 303–10. [DOI] [PubMed] [Google Scholar]
- 68.Douellou T, Montel MC, and Thevenot Sergentet D, Invited review: Anti-adhesive properties of bovine oligosaccharides and bovine milk fat globule membrane-associated glycoconjugates against bacterial food enteropathogens. J Dairy Sci, 2017. 100(5): p. 3348–3359. [DOI] [PubMed] [Google Scholar]
- 69.Gibson GR, et al. , Dietary modulation of the human colonic microbiota: updating the concept of prebiotics. Nutr Res Rev, 2004. 17(2): p. 259–75. [DOI] [PubMed] [Google Scholar]
- 70.McLoughlin RF, et al. , Short-chain fatty acids, prebiotics, synbiotics, and systemic inflammation: a systematic review and meta-analysis. Am J Clin Nutr, 2017. 106(3): p. 930–945. [DOI] [PubMed] [Google Scholar]
- 71.Stelmach-Mardas M, et al. , Dietary and cardio-metabolic risk factors in patients with Obstructive Sleep Apnea: cross-sectional study. PeerJ, 2017. 5: p. e3259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Flora GD and Nayak MK, A Brief Review of Cardiovascular Diseases, Associated Risk Factors and Current Treatment Regimes. Curr Pharm Des, 2019. 25(38): p. 4063–4084. [DOI] [PubMed] [Google Scholar]
- 73.Sanchez-Rodriguez E, et al. , The Gut Microbiota and Its Implication in the Development of Atherosclerosis and Related Cardiovascular Diseases. Nutrients, 2020. 12(3). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Tang WHW, et al. , Intestinal Microbiota in Cardiovascular Health and Disease: JACC State-of-the-Art Review. J Am Coll Cardiol, 2019. 73(16): p. 2089–2105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Libby P, et al. , Atherosclerosis. Nat Rev Dis Primers, 2019. 5(1): p. 56. [DOI] [PubMed] [Google Scholar]
- 76.Ufnal M, Zadlo A, and Ostaszewski R, TMAO: A small molecule of great expectations. Nutrition, 2015. 31(11–12): p. 1317–23. [DOI] [PubMed] [Google Scholar]
- 77.O’Morain VL and Ramji DP, The Potential of Probiotics in the Prevention and Treatment of Atherosclerosis. Mol Nutr Food Res, 2020. 64(4): p. e1900797. [DOI] [PubMed] [Google Scholar]
- 78.Parada Venegas D, et al. , Short Chain Fatty Acids (SCFAs)-Mediated Gut Epithelial and Immune Regulation and Its Relevance for Inflammatory Bowel Diseases. Front Immunol, 2019. 10: p. 277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Badran M, Ayas N, and Laher I, Insights into obstructive sleep apnea research. Sleep Med, 2014. 15(5): p. 485–95. [DOI] [PubMed] [Google Scholar]
- 80.Yacoub M, et al. , Cardiovascular Disease Risk in Obstructive Sleep apnea: An Update. J Sleep Disord Ther, 2017. 7(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Zheng HJ, et al. , Probiotics, prebiotics, and synbiotics for the improvement of metabolic profiles in patients with chronic kidney disease: A systematic review and meta-analysis of randomized controlled trials. Crit Rev Food Sci Nutr, 2020: p. 1–22. [DOI] [PubMed] [Google Scholar]
- 82.Zheng HJ, et al. , The effect of probiotic and synbiotic supplementation on biomarkers of inflammation and oxidative stress in diabetic patients: A systematic review and meta-analysis of randomized controlled trials. Pharmacol Res, 2019. 142: p. 303–313. [DOI] [PubMed] [Google Scholar]
- 83.Mo R, Zhang X, and Yang Y, Effect of probiotics on lipid profiles in hypercholesterolaemic adults: A meta-analysis of randomized controlled trials. Med Clin (Barc), 2019. 152(12): p. 473–481. [DOI] [PubMed] [Google Scholar]
- 84.Zhang K, et al. , Fermented dairy foods intake and risk of cardiovascular diseases: A meta-analysis of cohort studies. Crit Rev Food Sci Nutr, 2020. 60(7): p. 1189–1194. [DOI] [PubMed] [Google Scholar]
- 85.Freeman AM, et al. , A Clinician’s Guide for Trending Cardiovascular Nutrition Controversies: Part II. J Am Coll Cardiol, 2018. 72(5): p. 553–568. [DOI] [PubMed] [Google Scholar]
- 86.Chan YK, et al. , Probiotic mixture VSL#3 reduce high fat diet induced vascular inflammation and atherosclerosis in ApoE(−/−) mice. AMB Express, 2016. 6(1): p. 61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Huang Y, et al. , Lactobacillus acidophilus ATCC 4356 prevents atherosclerosis via inhibition of intestinal cholesterol absorption in apolipoprotein E-knockout mice. Appl Environ Microbiol, 2014. 80(24): p. 7496–504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Qiu L, et al. , Lactobacillus plantarum ZDY04 exhibits a strain-specific property of lowering TMAO via the modulation of gut microbiota in mice. Food Funct, 2018. 9(8): p. 4299–4309. [DOI] [PubMed] [Google Scholar]
- 89.Vasquez EC, et al. , Probiotics as Beneficial Dietary Supplements to Prevent and Treat Cardiovascular Diseases: Uncovering Their Impact on Oxidative Stress. Oxid Med Cell Longev, 2019. 2019: p. 3086270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Rashid SK, et al. , Probiotics (VSL#3) prevent endothelial dysfunction in rats with portal hypertension: role of the angiotensin system. PLoS One, 2014. 9(5): p. e97458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Friques AG, et al. , Chronic administration of the probiotic kefir improves the endothelial function in spontaneously hypertensive rats. J Transl Med, 2015. 13: p. 390. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Hashmi A, et al. , Effect of Prebiotic Galacto-Oligosaccharides on Serum Lipid Profile of Hypercholesterolemics. Probiotics Antimicrob Proteins, 2016. 8(1): p. 19–30. [DOI] [PubMed] [Google Scholar]
- 93.Fak F and Backhed F, Lactobacillus reuteri prevents diet-induced obesity, but not atherosclerosis, in a strain dependent fashion in Apoe−/− mice. PLoS One, 2012. 7(10): p. e46837. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Cryan JF, et al. , The Microbiota-Gut-Brain Axis. Physiol Rev, 2019. 99(4): p. 1877–2013. [DOI] [PubMed] [Google Scholar]
- 95.Fulling C, Dinan TG, and Cryan JF, Gut Microbe to Brain Signaling: What Happens in Vagus. Neuron, 2019. 101(6): p. 998–1002. [DOI] [PubMed] [Google Scholar]
- 96.Bravo JA, et al. , Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc Natl Acad Sci U S A, 2011. 108(38): p. 16050–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Reinshagen M, [A gut-brain neural circuit for nutrient sensory transduction]. Z Gastroenterol, 2019. 57(3): p. 335. [DOI] [PubMed] [Google Scholar]
- 98.Huuskonen J, et al. , Regulation of microglial inflammatory response by sodium butyrate and short-chain fatty acids. Br J Pharmacol, 2004. 141(5): p. 874–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Long-Smith C, et al. , Microbiota-Gut-Brain Axis: New Therapeutic Opportunities. Annu Rev Pharmacol Toxicol, 2020. 60: p. 477–502. [DOI] [PubMed] [Google Scholar]
- 100.Miller AH and Raison CL, The role of inflammation in depression: from evolutionary imperative to modern treatment target. Nat Rev Immunol, 2016. 16(1): p. 22–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Goshen I and Yirmiya R, Interleukin-1 (IL-1): a central regulator of stress responses. Front Neuroendocrinol, 2009. 30(1): p. 30–45. [DOI] [PubMed] [Google Scholar]
- 102.DiSabato DJ, et al. , Interleukin-1 receptor on hippocampal neurons drives social withdrawal and cognitive deficits after chronic social stress. Mol Psychiatry, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Bauer ME and Teixeira AL, Inflammation in psychiatric disorders: what comes first? Ann N Y Acad Sci, 2019. 1437(1): p. 57–67. [DOI] [PubMed] [Google Scholar]
- 104.Paiva IHR, Duarte-Silva E, and Peixoto CA, The role of prebiotics in cognition, anxiety, and depression. Eur Neuropsychopharmacol, 2020. 34: p. 1–18. [DOI] [PubMed] [Google Scholar]
- 105.Sarkar A, et al. , Psychobiotics and the Manipulation of Bacteria-Gut-Brain Signals. Trends Neurosci, 2016. 39(11): p. 763–781. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Liu RT, Walsh RFL, and Sheehan AE, Prebiotics and probiotics for depression and anxiety: A systematic review and meta-analysis of controlled clinical trials. Neurosci Biobehav Rev, 2019. 102: p. 13–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Goh KK, et al. , Effect of probiotics on depressive symptoms: A meta-analysis of human studies. Psychiatry Res, 2019. 282: p. 112568. [DOI] [PubMed] [Google Scholar]
- 108.Kruger JF, et al. , Probiotics for dementia: a systematic review and meta-analysis of randomized controlled trials. Nutr Rev, 2020. [DOI] [PubMed] [Google Scholar]
- 109.Ng QX, et al. , A Systematic Review of the Effect of Probiotic Supplementation on Schizophrenia Symptoms. Neuropsychobiology, 2019. 78(1): p. 1–6. [DOI] [PubMed] [Google Scholar]
- 110.Liu YW, et al. , Psychotropic effects of Lactobacillus plantarum PS128 in early life-stressed and naive adult mice. Brain Res, 2016. 1631: p. 1–12. [DOI] [PubMed] [Google Scholar]
- 111.Mehrabadi S and Sadr SS, Assessment of Probiotics Mixture on Memory Function, Inflammation Markers, and Oxidative Stress in an Alzheimer’s Disease Model of Rats. Iran Biomed J, 2020. 24(4): p. 220–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Chen D, et al. , Prebiotic Effect of Fructooligosaccharides from Morinda officinalis on Alzheimer’s Disease in Rodent Models by Targeting the Microbiota-Gut-Brain Axis. Front Aging Neurosci, 2017. 9: p. 403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Greiner TU and Backhed F, Microbial regulation of GLP-1 and L-cell biology. Mol Metab, 2016. 5(9): p. 753–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Burokas A, et al. , Targeting the Microbiota-Gut-Brain Axis: Prebiotics Have Anxiolytic and Antidepressant-like Effects and Reverse the Impact of Chronic Stress in Mice. Biol Psychiatry, 2017. 82(7): p. 472–487. [DOI] [PubMed] [Google Scholar]
- 115.Daulatzai MA, Death by a thousand cuts in Alzheimer’s disease: hypoxia--the prodrome. Neurotox Res, 2013. 24(2): p. 216–43. [DOI] [PubMed] [Google Scholar]
- 116.Zhang X and Le W, Pathological role of hypoxia in Alzheimer’s disease. Exp Neurol, 2010. 223(2): p. 299–303. [DOI] [PubMed] [Google Scholar]
- 117.Gozal D and Kheirandish L, Sleepiness and neurodegeneration in sleep-disordered breathing: convergence of signaling cascades. Am J Respir Crit Care Med, 2005. 171(12): p. 1325–7. [DOI] [PubMed] [Google Scholar]
- 118.Vincken WG, et al. , Involvement of upper-airway muscles in extrapyramidal disorders. A cause of airflow limitation. N Engl J Med, 1984. 311(7): p. 438–42. [DOI] [PubMed] [Google Scholar]
- 119.Shimohata T, et al. , Daytime hypoxemia, sleep-disordered breathing, and laryngopharyngeal findings in multiple system atrophy. Arch Neurol, 2007. 64(6): p. 856–61. [DOI] [PubMed] [Google Scholar]
- 120.Zimmerman ME, et al. , Normalization of memory performance and positive airway pressure adherence in memory-impaired patients with obstructive sleep apnea. Chest, 2006. 130(6): p. 1772–8. [DOI] [PubMed] [Google Scholar]
- 121.Kushida CA, et al. , Effects of continuous positive airway pressure on neurocognitive function in obstructive sleep apnea patients: The Apnea Positive Pressure Long-term Efficacy Study (APPLES). Sleep, 2012. 35(12): p. 1593–602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Canessa N, et al. , Obstructive sleep apnea: brain structural changes and neurocognitive function before and after treatment. Am J Respir Crit Care Med, 2011. 183(10): p. 1419–26. [DOI] [PubMed] [Google Scholar]
- 123.Ancoli-Israel S, et al. , Cognitive effects of treating obstructive sleep apnea in Alzheimer’s disease: a randomized controlled study. J Am Geriatr Soc, 2008. 56(11): p. 2076–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Neikrug AB, et al. , Continuous positive airway pressure improves sleep and daytime sleepiness in patients with Parkinson disease and sleep apnea. Sleep, 2014. 37(1): p. 177–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Gupta MA and Simpson FC, Obstructive sleep apnea and psychiatric disorders: a systematic review. J Clin Sleep Med, 2015. 11(2): p. 165–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Ni J, et al. , Gut microbiota and IBD: causation or correlation? Nat Rev Gastroenterol Hepatol, 2017. 14(10): p. 573–584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Kim ER and Chang DK, Colorectal cancer in inflammatory bowel disease: the risk, pathogenesis, prevention and diagnosis. World J Gastroenterol, 2014. 20(29): p. 9872–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Lloyd-Price J, et al. , Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature, 2019. 569(7758): p. 655–662. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Liu S, et al. , The microbiome in inflammatory bowel diseases: from pathogenesis to therapy. Protein Cell, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Vich Vila A, et al. , Gut microbiota composition and functional changes in inflammatory bowel disease and irritable bowel syndrome. Sci Transl Med, 2018. 10(472). [DOI] [PubMed] [Google Scholar]
- 131.Schirmer M, et al. , Microbial genes and pathways in inflammatory bowel disease. Nat Rev Microbiol, 2019. 17(8): p. 497–511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Song X, et al. , Microbial bile acid metabolites modulate gut RORgamma(+) regulatory T cell homeostasis. Nature, 2020. 577(7790): p. 410–415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Zhang J, et al. , Efficacy of prebiotics and probiotics for functional dyspepsia: A systematic review and meta-analysis. Medicine (Baltimore), 2020. 99(7): p. e19107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Wilson B, et al. , Prebiotics in irritable bowel syndrome and other functional bowel disorders in adults: a systematic review and meta-analysis of randomized controlled trials. Am J Clin Nutr, 2019. 109(4): p. 1098–1111. [DOI] [PubMed] [Google Scholar]
- 135.Asto E, et al. , The Efficacy of Probiotics, Prebiotic Inulin-Type Fructans, and Synbiotics in Human Ulcerative Colitis: A Systematic Review and Meta-Analysis. Nutrients, 2019. 11(2). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Jia K, et al. , The clinical effects of probiotics for inflammatory bowel disease: A meta-analysis. Medicine (Baltimore), 2018. 97(51): p. e13792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Jakubczyk D, Leszczynska K, and Gorska S, The Effectiveness of Probiotics in the Treatment of Inflammatory Bowel Disease (IBD)-A Critical Review. Nutrients, 2020. 12(7). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Chou YC, et al. , Lactobacillus fermentum V3 ameliorates colitis-associated tumorigenesis by modulating the gut microbiome. Am J Cancer Res, 2020. 10(4): p. 1170–1181. [PMC free article] [PubMed] [Google Scholar]
- 139.Yue Y, et al. , Probiotic strain Lactobacillus plantarum YYC-3 prevents colon cancer in mice by regulating the tumour microenvironment. Biomed Pharmacother, 2020. 127: p. 110159. [DOI] [PubMed] [Google Scholar]
- 140.Chen D, et al. , Clostridium butyricum, a butyrate-producing probiotic, inhibits intestinal tumor development through modulating Wnt signaling and gut microbiota. Cancer Lett, 2020. 469: p. 456–467. [DOI] [PubMed] [Google Scholar]
- 141.Ivanovska TP, et al. , Synbiotic loaded chitosan-Ca-alginate microparticles reduces inflammation in the TNBS model of rat colitis. Int J Pharm, 2017. 527(1–2): p. 126–134. [DOI] [PubMed] [Google Scholar]
- 142.Rotenberg BW, Murariu D, and Pang KP, Trends in CPAP adherence over twenty years of data collection: a flattened curve. J Otolaryngol Head Neck Surg, 2016. 45(1): p. 43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Ramar K, et al. , Clinical Practice Guideline for the Treatment of Obstructive Sleep Apnea and Snoring with Oral Appliance Therapy: An Update for 2015. J Clin Sleep Med, 2015. 11(7): p. 773–827. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Peppard PE, et al. , Longitudinal study of moderate weight change and sleep-disordered breathing. JAMA, 2000. 284(23): p. 3015–21. [DOI] [PubMed] [Google Scholar]
- 145.Seganfredo FB, et al. , Weight-loss interventions and gut microbiota changes in overweight and obese patients: a systematic review. Obes Rev, 2017. 18(8): p. 832–851. [DOI] [PubMed] [Google Scholar]
- 146.Ganesh BP, et al. , Prebiotics, Probiotics, and Acetate Supplementation Prevent Hypertension in a Model of Obstructive Sleep Apnea. Hypertension, 2018. 72(5): p. 1141–1150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Xu H, et al. , Protective Effect of Lactobacillus rhamnosus GG and its Supernatant against Myocardial Dysfunction in Obese Mice Exposed to Intermittent Hypoxia is Associated with the Activation of Nrf2 Pathway. Int J Biol Sci, 2019. 15(11): p. 2471–2483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Liu Q, et al. , Probiotic culture supernatant improves metabolic function through FGF21-adiponectin pathway in mice. J Nutr Biochem, 2020. 75: p. 108256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Gupta A and Khanna S, Fecal Microbiota Transplantation, in Jama. 2017: United States. p. 102. [DOI] [PubMed] [Google Scholar]
- 150.Kelly CR, et al. , Update on Fecal Microbiota Transplantation 2015: Indications, Methodologies, Mechanisms, and Outlook. Gastroenterology, 2015. 149(1): p. 223–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Cammarota G, et al. , European consensus conference on faecal microbiota transplantation in clinical practice. Gut, 2017. 66(4): p. 569–580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Turnbaugh PJ, et al. , An obesity-associated gut microbiome with increased capacity for energy harvest. Nature, 2006. 444(7122): p. 1027–31. [DOI] [PubMed] [Google Scholar]
- 153.Vrieze A, et al. , Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndrome. Gastroenterology, 2012. 143(4): p. 913–6. e7. [DOI] [PubMed] [Google Scholar]
- 154.Liou AP, et al. , Conserved shifts in the gut microbiota due to gastric bypass reduce host weight and adiposity. Sci Transl Med, 2013. 5(178): p. 178ra41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Wang H, et al. , Promising Treatment for Type 2 Diabetes: Fecal Microbiota Transplantation Reverses Insulin Resistance and Impaired Islets. Front Cell Infect Microbiol, 2019. 9: p. 455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Adnan S, et al. , Alterations in the gut microbiota can elicit hypertension in rats. Physiol Genomics, 2017. 49(2): p. 96–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Li J, et al. , Gut microbiota dysbiosis contributes to the development of hypertension. Microbiome, 2017. 5(1): p. 14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Juraschek SP, et al. , Plasma lactate and incident hypertension in the atherosclerosis risk in communities study. Am J Hypertens, 2015. 28(2): p. 216–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Cammarota G, Ianiro G, and Gasbarrini A, Fecal microbiota transplantation for the treatment of Clostridium difficile infection: a systematic review. J Clin Gastroenterol, 2014. 48(8): p. 693–702. [DOI] [PubMed] [Google Scholar]
- 160.Suzuki T, Yoshida S, and Hara H, Physiological concentrations of short-chain fatty acids immediately suppress colonic epithelial permeability. Br J Nutr, 2008. 100(2): p. 297–305. [DOI] [PubMed] [Google Scholar]
- 161.Donohoe DR, et al. , The microbiome and butyrate regulate energy metabolism and autophagy in the mammalian colon. Cell Metab, 2011. 13(5): p. 517–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Säemann MD, et al. , Anti-inflammatory effects of sodium butyrate on human monocytes: potent inhibition of IL-12 and up-regulation of IL-10 production. Faseb j, 2000. 14(15): p. 2380–2. [DOI] [PubMed] [Google Scholar]
- 163.Ni YF, et al. , Histone deacetylase inhibitor, butyrate, attenuates lipopolysaccharide-induced acute lung injury in mice. Respir Res, 2010. 11(1): p. 33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Maslowski KM and Mackay CR, Diet, gut microbiota and immune responses. Nat Immunol, 2011. 12(1): p. 5–9. [DOI] [PubMed] [Google Scholar]
- 165.Nakamura M, et al. , Reduction of telomerase activity in human liver cancer cells by a histone deacetylase inhibitor. J Cell Physiol, 2001. 187(3): p. 392–401. [DOI] [PubMed] [Google Scholar]
- 166.Borthakur A, et al. , A novel nutrient sensing mechanism underlies substrate-induced regulation of monocarboxylate transporter-1. Am J Physiol Gastrointest Liver Physiol, 2012. 303(10): p. G1126–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Cherbut C, et al. , Short-chain fatty acids modify colonic motility through nerves and polypeptide YY release in the rat. Am J Physiol, 1998. 275(6): p. G1415–22. [DOI] [PubMed] [Google Scholar]
- 168.Burger-van Paassen N, et al. , The regulation of intestinal mucin MUC2 expression by short-chain fatty acids: implications for epithelial protection. Biochem J, 2009. 420(2): p. 211–9. [DOI] [PubMed] [Google Scholar]
- 169.Balda MS and Matter K, Tight junctions at a glance. J Cell Sci, 2008. 121(Pt 22): p. 3677–82. [DOI] [PubMed] [Google Scholar]
- 170.Tolhurst G, et al. , Short-chain fatty acids stimulate glucagon-like peptide-1 secretion via the G-protein-coupled receptor FFAR2. Diabetes, 2012. 61(2): p. 364–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Xiong Y, et al. , Short-chain fatty acids stimulate leptin production in adipocytes through the G protein-coupled receptor GPR41. Proc Natl Acad Sci U S A, 2004. 101(4): p. 1045–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Ji B and Nielsen J, From next-generation sequencing to systematic modeling of the gut microbiome. Front Genet, 2015. 6: p. 219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Round JL and Mazmanian SK, The gut microbiota shapes intestinal immune responses during health and disease. Nat Rev Immunol, 2009. 9(5): p. 313–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Omary MB, et al. , Not All Mice Are the Same: Standardization of Animal Research Data Presentation. Cell Mol Gastroenterol Hepatol, 2016. 2(4): p. 391–393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Eisenstein M, The hunt for a healthy microbiome. Nature, 2020. 577(7792): p. S6–S8. [DOI] [PubMed] [Google Scholar]
- 176.Ley RE, et al. , Microbial ecology: human gut microbes associated with obesity. Nature, 2006. 444(7122): p. 1022–3. [DOI] [PubMed] [Google Scholar]
- 177.De Filippo C, et al. , Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc Natl Acad Sci U S A, 2010. 107(33): p. 14691–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Chen Y, et al. , Characterization of fecal microbial communities in patients with liver cirrhosis. Hepatology, 2011. 54(2): p. 562–72. [DOI] [PubMed] [Google Scholar]
- 179.Mashaqi S and Gozal D, Obstructive Sleep Apnea and Systemic Hypertension: Gut Dysbiosis as the Mediator? J Clin Sleep Med, 2019. 15(10): p. 1517–1527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Sanders ME, et al. , Probiotics and prebiotics in intestinal health and disease: from biology to the clinic. Nat Rev Gastroenterol Hepatol, 2019. 16(10): p. 605–616. [DOI] [PubMed] [Google Scholar]
- 181.Dronkers TMG, et al. , The ascent of the blessed: regulatory issues on health effects and health claims for probiotics in Europe and the rest of the world. Benef Microbes, 2018. 9(5): p. 717–723. [DOI] [PubMed] [Google Scholar]
- 182.Majewska K, et al. , The Multispecies Probiotic Effectively Reduces Homocysteine Concentration in Obese Women: A Randomized Double-Blind Placebo-Controlled Study. J Clin Med, 2020. 9(4). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Madempudi RS, et al. , Efficacy of UB0316, a multi-strain probiotic formulation in patients with type 2 diabetes mellitus: A double blind, randomized, placebo controlled study. PLoS One, 2019. 14(11): p. e0225168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Angelino D, et al. , Glucose- and Lipid-Related Biomarkers Are Affected in Healthy Obese or Hyperglycemic Adults Consuming a Whole-Grain Pasta Enriched in Prebiotics and Probiotics: A 12-Week Randomized Controlled Trial. J Nutr, 2019. 149(10): p. 1714–1723. [DOI] [PubMed] [Google Scholar]
- 185.Kobyliak N, et al. , Beneficial effects of probiotic combination with omega-3 fatty acids in NAFLD: a randomized clinical study. Minerva Med, 2018. 109(6): p. 418–428. [DOI] [PubMed] [Google Scholar]
- 186.Pedret A, et al. , Effects of daily consumption of the probiotic Bifidobacterium animalis subsp. lactis CECT 8145 on anthropometric adiposity biomarkers in abdominally obese subjects: a randomized controlled trial. Int J Obes (Lond), 2019. 43(9): p. 1863–1868. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Ke X, et al. , Synbiotic-driven improvement of metabolic disturbances is associated with changes in the gut microbiome in diet-induced obese mice. Mol Metab, 2019. 22: p. 96–109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Lim PS, et al. , Cholesterol homeostasis associated with probiotic supplementation in vivo. J Appl Microbiol, 2020. [DOI] [PubMed] [Google Scholar]
- 189.Yao C, et al. , Antihyperlipidaemic effect of microencapsulated Lactobacillus plantarum LIP-1 on hyperlipidaemic rats. J Sci Food Agric, 2020. 100(5): p. 2007–2017. [DOI] [PubMed] [Google Scholar]
- 190.Wang G, et al. , Lactic acid bacteria reduce diabetes symptoms in mice by alleviating gut microbiota dysbiosis and inflammation in different manners. Food Funct, 2020. [DOI] [PubMed] [Google Scholar]
- 191.Wang P, et al. , Perioperative probiotic treatment decreased the incidence of postoperative cognitive impairment in elderly patients following non-cardiac surgery: A randomised double-blind and placebo-controlled trial. Clin Nutr, 2020. [DOI] [PubMed] [Google Scholar]
- 192.Kim CS, et al. , Probiotic supplementation improves cognitive function and mood with changes in gut microbiota in community-dwelling elderly: A randomized, double-blind, placebo-controlled, multicenter trial. J Gerontol A Biol Sci Med Sci, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Venkataraman R, et al. , Effect of Multi-strain Probiotic Formulation on Students Facing Examination Stress: a Double-Blind, Placebo-Controlled Study. Probiotics Antimicrob Proteins, 2020. [DOI] [PubMed] [Google Scholar]
- 194.Xiao J, et al. , Probiotic Bifidobacterium breve in Improving Cognitive Functions of Older Adults with Suspected Mild Cognitive Impairment: A Randomized, Double-Blind, Placebo-Controlled Trial. J Alzheimers Dis, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Rudzki L, et al. , 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: p. 213–222. [DOI] [PubMed] [Google Scholar]
- 196.Dugyala S, et al. , Putative modulation of the gut microbiome by probiotics enhances preference for novelty in a preliminary double-blind placebo-controlled study in ferrets. Anim Microbiome, 2020. 2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Hoffman JD, et al. , Dietary inulin alters the gut microbiome, enhances systemic metabolism and reduces neuroinflammation in an APOE4 mouse model. PLoS One, 2019. 14(8): p. e0221828. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Barros-Santos T, et al. , Effects of chronic treatment with new strains of Lactobacillus plantarum on cognitive, anxiety- and depressive-like behaviors in male mice. PLoS One, 2020. 15(6): p. e0234037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Hsieh TH, et al. , Probiotics Alleviate the Progressive Deterioration of Motor Functions in a Mouse Model of Parkinson’s Disease. Brain Sci, 2020. 10(4). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Lu J, et al. , Maternal administration of probiotics promotes brain development and protects offspring’s brain from postnatal inflammatory insults in C57/BL6J mice. Sci Rep, 2020. 10(1): p. 8178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Fan H, et al. , Effects of pentasa-combined probiotics on the microflora structure and prognosis of patients with inflammatory bowel disease. Turk J Gastroenterol, 2019. 30(8): p. 680–685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Bjarnason I, Sission G, and Hayee B, A randomised, double-blind, placebo-controlled trial of a multi-strain probiotic in patients with asymptomatic ulcerative colitis and Crohn’s disease. Inflammopharmacology, 2019. 27(3): p. 465–473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Bajramagic S, et al. , Usage of Probiotics and its Clinical Significance at Surgically Treated Patients Sufferig from Colorectal Carcinoma. Med Arch, 2019. 73(5): p. 316–320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Trifan A, et al. , Efficacy and safety of Gelsectan for diarrhoea-predominant irritable bowel syndrome: A randomised, crossover clinical trial. United European Gastroenterol J, 2019. 7(8): p. 1093–1101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Zaharuddin L, et al. , A randomized double-blind placebo-controlled trial of probiotics in post-surgical colorectal cancer. BMC Gastroenterol, 2019. 19(1): p. 131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Biagioli M, et al. , Discovery of a Novel Multi-Strains Probiotic Formulation with Improved Efficacy toward Intestinal Inflammation. Nutrients, 2020. 12(7). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Liu Z, et al. , Study of the alleviation effects of a combination of Lactobacillus rhamnosus and inulin on mice with colitis. Food Funct, 2020. 11(5): p. 3823–3837. [DOI] [PubMed] [Google Scholar]
- 208.Shinde T, et al. , Synbiotic supplementation with prebiotic green banana resistant starch and probiotic Bacillus coagulans spores ameliorates gut inflammation in mouse model of inflammatory bowel diseases. Eur J Nutr, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Parisa A, et al. , Anti-cancer effects of Bifidobacterium species in colon cancer cells and a mouse model of carcinogenesis. PLoS One, 2020. 15(5): p. e0232930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Lee CW, et al. , Synbiotic Combination of Djulis (Chenopodium formosanum) and Lactobacillus acidophilus Inhibits Colon Carcinogenesis in Rats. Nutrients, 2019. 12(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Bartolomaeus H, et al. , Short-Chain Fatty Acid Propionate Protects From Hypertensive Cardiovascular Damage. Circulation, 2019. 139(11): p. 1407–1421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Holmes E, et al. , Human metabolic phenotype diversity and its association with diet and blood pressure. Nature, 2008. 453(7193): p. 396–400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Marques FZ, et al. , High-Fiber Diet and Acetate Supplementation Change the Gut Microbiota and Prevent the Development of Hypertension and Heart Failure in Hypertensive Mice. Circulation, 2017. 135(10): p. 964–977. [DOI] [PubMed] [Google Scholar]
- 214.Lin HV, et al. , Butyrate and propionate protect against diet-induced obesity and regulate gut hormones via free fatty acid receptor 3-independent mechanisms. PLoS One, 2012. 7(4): p. e35240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Gao Z, et al. , Butyrate improves insulin sensitivity and increases energy expenditure in mice. Diabetes, 2009. 58(7): p. 1509–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Khan S and Jena GB, Protective role of sodium butyrate, a HDAC inhibitor on beta-cell proliferation, function and glucose homeostasis through modulation of p38/ERK MAPK and apoptotic pathways: study in juvenile diabetic rat. Chem Biol Interact, 2014. 213: p. 1–12. [DOI] [PubMed] [Google Scholar]
- 217.Hong J, et al. , Butyrate alleviates high fat diet-induced obesity through activation of adiponectin-mediated pathway and stimulation of mitochondrial function in the skeletal muscle of mice. Oncotarget, 2016. 7(35): p. 56071–56082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.Yadav L, et al. , Matrix metalloproteinases and cancer - roles in threat and therapy. Asian Pac J Cancer Prev, 2014. 15(3): p. 1085–91. [DOI] [PubMed] [Google Scholar]
- 219.Thangaraju M, et al. , GPR109A is a G-protein-coupled receptor for the bacterial fermentation product butyrate and functions as a tumor suppressor in colon. Cancer Res, 2009. 69(7): p. 2826–32. [DOI] [PMC free article] [PubMed] [Google Scholar]