The present invention relates to a composition for improving gut microbiota; control of gut enterobacteria in children with autism; reconstitution of gut microbiota and control of enterobacteria in autism and neurodegenerative diseases; a composition for improving behavioural pattern and alpha-synuclein levels, especially in autism; a composition for improving sleep pattern and serum melatonin, especially in children with autism.
Gut dysbiosis is one of the major pathologies in children with autism spectrum disorder (ASD). Evaluation of gut microbiota of the subjects in the present randomized pilot clinical study was undertaken and compared with an aim of gaining a mechanistic insight.
Autism spectrum disorders (ASDs) are a group of developmental disabilities that can cause significant impairment in social, emotional, and communication skills (cdc.gov). Several causes and underlying mechanisms have been postulated for the pathogenesis of ASD, including genetic, environmental, immune dysregulation, neuroinflammation, and oxidative stress. Neuronal synaptic imbalance and mutation in synaptic proteins and receptors have also been reported to be associated with ASD (Al-Mazeedi et al., 2020). Synucleins are small soluble proteins that are present in presynaptic terminals, and they regulate synaptic plasticity and neurotransmitter release. Synucleins are important in the context of brains and neurons (Al-Mazeedi et al., 2020; Vargas et al., 2017).
Alpha-synuclein has already been reported to be associated with several neurodegenerative disorders, collectively called synucleinopathies such as Alzheimer's disease (AD), Parkinson's disease (PD), dementia with Lewy bodies (DLBs), and multiple system atrophy (Al-Mazeedi et al., 2020). Several studies have recently reported a strong association between lower levels of α-synuclein and ASD. At present, there is no definitive cure for ASD (Al-Mazeedi et al., 2020; Kadak et al., 2015; Sriwimol et al., 2018). Interventions involve speech and behavioural therapies to improve the symptoms. According to the research, the microbiota-gut-brain axis is significant because dysbiosis has been observed in gut-related diseases and other generalized disorders, especially of the nervous system, such as AD, multiple sclerosis, PD, and ASD (Srikantha et al., 2019). Therefore, nutritional supplements are considered potential interventions in alleviating gastrointestinal and behavioural symptoms in ASD (Karhu et al., 2020).
Beta-glucans, especially yeast-derived ones, have shown a considerable positive outcome as food supplements in modulating gut microbiota (Karhu et al., 2020; Xu et al., 2020). Nichi Glucan is a black yeast-derived AFO-202 (also referred to as FO-68 [(accession number) FERM BP-19327]) beta-glucan that has been in consumption for the past two decades (Ikewaki et al., 2007) and has been shown to have potential as a nutritional supplement to balance metabolic levels of glucose, lipids, and immunomodulators (Dedeepiya et al., 2012; Ganesh et al., 2014; Ikewaki et al., 2021). The present study was undertaken to investigate the effects of Nichi Glucan as a food supplement in children with ASD, especially with relevance to the childhood autism rating scale (CARS) score and alpha-synuclein levels.
Inability to have a good quality of sleep is one of the major problems faced by people with autism spectrum disorders (ASD). This is attributed to lower melatonin levels and therefore melatonin supplementation is one of the treatments opted, reported with varying outcome. Beta Glucans having been earlier reported in animal studies to improve melatonin levels, we herein report the outcome of our pilot clinical study in which a Black yeast Aureobasidium pullulans derived Beta 1,3-1,6 Glucan food supplement was administered in children with ASD.
The present invention relates to the following:
The present invention also relates to the following:
Gut microbiota is improved and/or well-balanced by the present invention. Such a control in gut microbiota leads to an improvement of behavioural pattern, alpha-synuclein levels, sleep pattern and/or serum melatonin levels.
The present invention is effective in balancing the gut microbiota, especially, by a decrease in Bacteroides, Curli producing Enterobacteria Escherichia coli, Akkermansia muciniphila CAG:154, Blautia spp., Coprobacillus sp., and Clostridium bolteae with increase in Roseburia, Faecalibacterium prausnitzii and Prevotella copri. The other beta glucans don't produce such an effective and balanced modulation of gut bacteria with effects on diverse species (Shen R L, et al. 2012, Turunen K, et al. 2011, Zhen W, et al. 2021). While the enterobacteria may be decreased, a decrease in Akkermansia muciniphila has not been concomitantly reported. Further the concomitant increase in beneficial bacteria such as Roseburia, Faecalibacterium prausnitzii and Prevotella copri is an added advantage.
The present invention is effective for prophylactic, ameliorating and/or curative treatment of neurological disorders such as autism spectrum disorders (ASD), multiple sclerosis (MS), Alzheimer's disease (AD), Parkinson's disease (PD) and/or epilepsy. The present invention is effective for improving sleep pattern and serum melatonin, especially in children with autism spectrum disorder.
The present invention relates to a composition for improving gut microbiota. The present invention also relates to an effective control of gut enterobacteria producing Alpha synuclein and Curli amyloids after consumption of Aureobasidium pullulans derived beta 1,3-1,6 glucans in children with autism spectrum disorder in a clinical pilot study. The present invention also relates to a beneficial reconstitution of gut microbiota and control of alpha-synuclein and curli-amyloids-producing enterobacteria, by beta 1,3-1,6 glucans in a clinical pilot study of autism and potentials in neurodegenerative diseases. The present invention also relates to a gut microbiota reconstitution by Beta Glucans in autism.
Further, the present invention relates to a composition for improving behavioural pattern and alpha-synuclein levels, especially in autism spectrum disorder. The present invention also relates to an improvement of behavioural pattern and alpha-synuclein levels in autism spectrum disorder after consumption of a beta-glucan food supplement in a randomized, parallel-group pilot clinical study.
Moreover, the present invention relates to a composition for improving sleep pattern and serum melatonin, especially in children with autism spectrum disorder. The present invention also relates to an improvement of sleep pattern and serum melatonin in children with autism spectrum disorder after consumption of Beta 1,3-1,6 Glucan in a pilot clinical study.
The glucan contained in the composition of the present invention can be a glucan derived from Aureobasidium pullulans strain FO-68 (Also referred to herein as “strain AFO 202”), and preferably β-1,3-1,6 glucan derived from FO-68 (Also referred to herein simply as “glucan”, “AFO 202 glucan” or “AF 202 beta glucan”). “Aureobasidium pullulans strain FO-68” has been deposited at the Patent Biological Depository Center, National Institute of Advanced Industrial Science and Technology, under the deposit number FERMP-19327.
While the domestic deposition was made on Apr. 23, 2003, Aureobasidium pullulans strain FO-68 has then been transferred to international deposition at the International Patent Organism Depositary, National Institute of Technology and Evaluation (Room. 120, 2-5-8, Kazusa Kamatari, Kisarazu-shi, Chiba, 292-0818 Japan) on Apr. 21, 2021 with the accession number: FERM BP-19327.
Aureobasidium pullulans strain FO-68 is also called as Aureobasidium strain FERM P-18099.
This fungus produces high-molecular polysaccharide with high viscosity. This substance agglutinates easily with ethanol, making it possible to collect simply. This polysaccharide is of [beta] type, and is acidic polysaccharide having a main chain of 1,3 bond and branches from 3- and 6-positions. It contains carboxylic acids such as malic acid as organic acids and phosphoric acid. Moreover, it agglutinates easily with aluminum ions etc. This substance is also effective for the promotion of growth as a feed and the effluent treatment. It is effective as a food additive and functional food.
FO-68 forms blackish brown colonies on potato-dextrose-agar slant culturing for 7 days at 25 C. The fringe of colonies shows filamentous growth and becomes gradually light blackish brown. The cells are filamentous, and sometimes arthrospores, yeast-like budding conidiospores, oval yeast-like single cells, and, in some time, thick-walled spore cells are formed. The growth temperature is 25 C., and it decomposes hexoses such as glucose, fructose and galactose, sucrose, and starch. The medium becomes conspicuously viscous. Based on FO-68's mycological properties, it is a kind of Aureobasidium pullulans in the black fungus family of deuteromycetes.
A colony of FO-68 has a smooth surface at first and grows into a grayish white, mucous and glossy oil drop-like (fat-like), yeast-like material. The filamentous fungus body grows radially from the fringe thereof, leading to crinkled, filamentous and just dendritic growth. This filamentous fungus body grows well not only on the surface of medium, but also in the medium. In a short time, light dark brown specks appear here and there on the surface of colony, which become black specks gradually, and overall surface becomes dark black eventually. On this filamentous fungus body, a lot of light brown, elliptic or oval conidiospores are produced laterally. This conidiospore falls easily in pieces. While the surface of oil drop-like colony puts on the conidiospores here and there.
As a method for culturing FO-68 and a method for producing β-1,3-1,6 glucan using FO-68, known methods can be used, for example, see JP 2004-329077A.
In some embodiment, the present invention relates to a composition comprising a beta-glucan produced by Aureobasidium pullulans AFO-202 (FERM BP-19327) for improving gut microbiota, behavioural pattern, alpha-synuclein levels, sleep pattern and/or serum melatonin. In another aspect, the present invention also relates to use of Aureobasidium pullulans AFO-202 (FERM BP-19327) for improving gut microbiota, behavioural pattern, alpha-synuclein levels, sleep pattern and/or serum melatonin, and particularly relates to a method of improving gut microbiota, behavioural pattern, alpha-synuclein levels, sleep pattern and/or serum melatonin by administering Aureobasidium pullulans AFO-202 (FERM BP-19327) to a subject.
In the composition used in the present invention, a culture of FO-68 may be used as it is without purification, or glucan isolated from the culture or further purified as necessary may be used. In addition, for example, the culture product of the present invention was crushed into a concentrate, a paste, a spray-dried product, a freeze-dried product, a vacuum-dried product, a drum-dried product, a liquid product dispersed in a medium, a diluted product, and a dried product.
The composition of the present invention exerts its function when ingested by mammals including humans. The term “ingestion” as used herein is not limited to any administration route as long as it can enter the human body, and is realized by all known administration methods such as oral administration, tube administration, and enteral administration. Typically, oral ingestion and enteral ingestion via the digestive tract are preferable.
The dose of the present invention can be appropriately set in consideration of various factors such as administration route, age, body weight, and symptoms. The dose of the composition of the present invention is not particularly limited, but the amount of glucan is preferably 0.05 mg/kg/day or more, more preferably 0.5 mg/kg/day or more, particularly preferably 1.0 mg/kg/day. However, when ingested over a long period of time, the amount may be smaller than the preferable amount described above. In addition, the glucan used in the present invention has a sufficient dietary experience, and there is no problem in terms of safety. Therefore, an amount far exceeding the above amount (for example, 10 mg/kg/day or more) is possible.
The composition of the present invention can be used as a food or drink. The composition of the present invention, as a special-purpose food such as a food for specified health use and a nutritionally functional food, by administering to animals such as humans, can improve gut microbiota, behavioural pattern, alpha-synuclein levels, sleep pattern and/or serum melatonin.
When the composition of the present invention is used as food or drink, the type of food or drink is not particularly limited. Further, the shape of the food or drink is not particularly limited, and may be any shape of food or drink that is usually used. For example, it may be in any form such as solid form (including powder and granule form), paste form, liquid form and suspension form, and is not limited to these forms.
When used as a pharmaceutical, a dosage form that can be orally administered is preferable because the composition of the present invention reaches the intestine. Examples of preferable dosage forms of the drug according to the present invention include tablets, coated tablets, capsules, granules, powders, solutions, syrups, troches and the like. These various preparations are prepared according to a conventional method by using glucan, which is the active ingredient, an excipient, a binder, a disintegrating agent, a lubricant, a coloring agent, a flavoring agent, a solubilizing agent, a suspending agent, a coating agent, etc. It can be formulated by admixing the auxiliaries usually used in the technical field of pharmaceutical formulation.
In some embodiment, the present invention can be used in combination with other food, drink, drugs and any other substances in order to enhance the efficacy of the present invention.
Beneficial Regulation of Gut Microbiota Yielding an Advantageous Fecal Metabolome Profile by Administration of AFO-202 Biological Response Modifier Glucan in an Stam Animal Model of Non-Alcoholic Steatohepatitis, Paving Way for Effective Utilization in Human Health and Disease.
With approximately 100 trillion micro-organisms existing in the human gastrointestinal tract, the microbiome is now considered as a virtual organ of the body. The microbiome encodes over three million genes producing thousands of metabolites compared to 23,000 genes of the human genome and hence replaces many of the functions of the host influencing the host's fitness, phenotype, and health. Gut microbiota influences several aspects of human health including immune, metabolic and neurobehavioural traits [D1]. In regard to functions, the gut microbiota ferments non-digestible substrates like dietary fibres and endogenous intestinal mucus. The fermentation supports the growth of specialist microbes that produce short chain fatty acids (SCFAs) and gases. Major SCFAs produced are acetate, propionate, and butyrate.
Butryate is needed to maintain colon's cells, it helps in apoptosis of colon cancer cells, activation of intestinal gluconeogenesis, having beneficial effects on glucose and energy homeostasis and maintenance of oxygen balance in the gut, preventing gut microbiota dysbiosis. Propionate is transported to the liver, where it regulates gluconeogenesis and acetate is an essential metabolite for the growth of other bacteria, as well as playing a role in central appetite regulation [D1].
Gut dysbiosis or the altered state of the microbiota community has been associated with several diseases including but not limited to diabetes, metabolic disorders, obesity, cancers, rheumatoid arthritis, neurological disorders such as Parkinson's disease, Alzheimer's′, multiple sclerosis and autism spectrum disorders (ASD) [D2,3]. The fecal metabolome represents the functional readout of the gut microbial activity and can be considered to be an intermediate phenotype mediating host-microbiome interaction. An average 67.7% (±18.8%) of the fecal metabolome's variance represents the gut microbial composition.
Fecal metabolic profiling thus is a novel tool to explore links among microbiome composition, host phenotypes and disease states [D4]. Probiotics and pre-biotic nutritional supplements represent the major strategy other than fecal microbiota transplantation to restore the dsybiotic gut to a healthy state. Beta glucans are one of the most promising nutritional supplements with established efficacy in metabolic diseases, diabetes, cancer, cardiovascular diseases and neurological diseases. Beta glucans produced from two strains AFO-202 and N-163 of a black yeast Aureobasidium pullulans derived beta glucan has been reported with beneficial effects in diabetes [D5], dyslipidemia [D6], ASD [D7,8], Duchenne muscular dystrophy (DMD) [D9], Non-alcoholic steatohepatitis (NASH) [D10] and infectious diseases including COVID-19 [D11,12].
In a previous study, the AFO-202 beta 1,3-1,6 glucan was able to balance the gut microbiome in children with ASD [D13]. In the study on STAM™ murine model of NASH, the AFO-202 beta glucan significantly decreased inflammation-associated hepatic cell ballooning and steatosis while the N-163 beta glucan decreased fibrosis and inflammation. The combination of AFO-202 with N-163 significantly decreased the Non-alcoholic fatty liver disease (NAFLD) Activity Score (NAS) [D10]. The present study was undertaken as an extension of this NASH study to study the fecal microbiome and metabolome profile before and after administration of AFO-202 beta glucan.
The study is reported in accordance with Animal Research: Reporting of In Vivo Experiments (ARRIVE) guidelines. C57BL/6J mice were obtained from Japan SLC, Inc. (Japan). All animals used in this study were cared for under the following guidelines: Act on Welfare and Management of Animals (Ministry of the Environment, Japan, Act No. 105 of Oct. 1, 1973), standards relating to the care and management of laboratory animals and relief of pain (Notice No. 88 of the Ministry of the Environment, Japan, Apr. 28, 2006) and the guidelines for proper conduct of animal experiments (Science Council of Japan, Jun. 1, 2006). Protocol approvals were obtained from SMC Laboratories, Japan's IACUC (Study reference no: SP_SLMN128-2107-6_1). Mice were maintained in a specific pathogen-free (SPF) facility under controlled conditions of temperature (23±3° C.), humidity (50±20%), lighting (12-hour artificial light and dark cycles; light from 8:00 to 20:00) and air exchange.
The STAM model of NASH was induced as previously described [D10]. Mice were given a single subcutaneous injection of 200 μg streptozotocin (STZ, Sigma-Aldrich, USA) solution 2 days after birth and fed with a high-fat diet (HFD, 57 kcal % fat, Cat #HFD32, CLEA Japan, Inc., Japan) from 4-9 weeks of age. All mice develop liver steatosis and diabetes and at 3 weeks mice had established steatohepatitis, histologically.
Studygroups: There were five study groups, described below. Eight mice were included in each study group.
Eight NASH mice were orally administered vehicle [RO water] in a volume of 5 mL/kg once daily from 6 to 9 weeks of age.
Eight NASH mice were orally administered vehicle supplemented with AFO-202 Beta Glucan at a dose of 1 mg/kg in a volume of 5 mL/kg once daily from 6 to 9 weeks of age.
Eight NASH mice were orally administered vehicle supplemented with Telmisartan at a dose of 10 mg/kg once daily from 6 to 9 weeks of age.
AFO-202 Beta Glucan was provided by GN Corporation Co Ltd., Japan. Telmisartan (Micardis(R)) was purchased from Boehringer Ingelheim GmbH (Germany).
NASH model mice were randomized into 3 groups of 8 mice at 6 weeks of age based on their body weight the day before the start of treatment. The randomization was performed by body weight-stratified random sampling using Excel software. NASH model mice were stratified by their body weight to get SD and the difference in the mean weights among groups as small as possible.
The viability, clinical signs (lethargy, twitching, labored breathing) and behavior were monitored daily. Body weight was recorded daily before the treatment. Mice were observed for significant clinical signs of toxicity, moribundity and mortality before administration and after administration. The animals were sacrificed at 9 weeks of age by exsanguination through direct cardiac puncture under isoflurane anesthesia (Pfizer Inc.).
Frequency: Fecal samples were collected at the 6 weeks of age (before administration) and 9 weeks of age (before sacrifice).
Procedure: At 6 weeks of age, fecal samples were collected from each mouse by clean catch method. Handle animals with clean gloves sterilized with 70% ethanol. A sterile petri dish was placed on the bench. Gently massage the abdomen, position the bottom of mouse over a fresh petri dish, and collect 1-2 fecal pellets. At sacrifice, fecal samples were aseptically collected from cecum. The tubes with feces were placed on ice immediately. These tubes were snap frozen in liquid nitrogen and stored at −80° C. for shipping.
In this analysis, the 16S rRNA sequence data acquired by the next-generation sequencer from the fecal RNA was used to perform community analysis using the QIIME2 program for microbial community analysis. The raw read data in FASTQ format output from the next-generation sequencer was trimmed to remove adapter sequences and low QV regions that may be included in the data. Cutadapt was used to remove adapter sequences from DNA sequencing reads. Trimmomatic was used as read trimming tool for Illumina NGS data. The adapter sequence was trimmed using the adapter trimming program “cutadapt” if the Trimming of the region at the end of the read sequence overlapped the corresponding sequence by at least one base (mismatch tolerance: 20%). When reads containing N were present in at least one of Read1 and Read2, both Read1 and Read2 were removed.
Trimming of low QV regions was performed on the read data after processing using the QV trimming program “Trimmomatic” under the following conditions.
The window of 20 bases is slid from the 5′ side, and the area where the average QV is less than 20 is trimmed.
After trimming, only the reads with more than 50 bases remaining in both Read1 and Read2 were taken as output.
The microbial community analysis based on 16S rRNA sequence was performed on the sequence data trimmed in the previous section using the microbial community analysis program “QIIME2”. The annotation program “sklearn” included in QIIME2 is used to annotate the ASV (OTU) sequences.
Using “sklearn”, an annotation program included in QIIME2, the ASV (OTU) sequences obtained were annotated with taxonomy information [Kingdom (kingdom)/Phylum (phylum)/Class (class)/Order (order)/Family (family)/Genus/Species] based on the 16S rDNA database.
The data set of 16S rDNA database “greengenes” provided on the QIIME2 Resources site was used for the analysis. The ASVs (OTUs) obtained above were aggregated and graphed based on the taxonomy information a and the read counts of each specimen. Based on the composition of bacterial flora for each specimen compiled above, various index values for α-diversity were calculated.
After lyophilization of the fecal sample, about 10 mg of the sample was separated, extracted by the Bligh-Dyer method, and the resulting aqueous layer1 was collected and lyophilized. The residue was derivatized using 2-methoxyamine hydrochloride and MSTFA, and submitted to gas chromatography-mass spectrometry (GC-MS) as an analytical sample. 2-Isopropylmalic acid was used as an internal standard. In addition, an operational blank test was also conducted.
Analytical equipment used was GCMS-TQ8030 (Shimadzu Corporation); Column BPX-5 (film thickness 0.25 μm, length 30 m, inner diameter 0.25 mm, Shimadzu GC).
MS-DIAL ver.4.7 (http://prime.psc.riken.jp/compms/index.html) was used to analyze and prepare the peak list (peak height) under the conditions given in Table 2. In doing so, peaks that were detected in the QC samples and whose C.V. was less than 20% and whose intensity was more than twice that of the operating blank were treated as detected peaks.
Erysipelotrichaceae
Allobaculum
—
Turicibacteraceae
Turicibacter
—
Desulfovibrionaceae
—
—
Clostridiaceae
Other
Other
Lactobacillaceae
Lactobacillus
—
Streptococcaceae
Lactococeus
—
Bacteroidaceae
Bacteroides
—
[Paraprevotellaceae]
[Prevotella]
—
—
—
—
Deferribacteraceae
Mucispirillum
schaedleri
Bacteroidaceae
Bacteroides
acidifaciens
—
—
—
Verrucomicrobiaceae
Akkermansia
muciniphila
Rikenellaceae
—
—
Enterobacteriaceae
Other
Other
Lactobacillaceae
Lactobacillus
—
S24-7
—
—
Lachnospiraceae
—
—
Peptostreptocpccaceae
—
—
Lachnospiraceae
[Ruminococcus]
gnavus
Lachnospiraceae
Coprococeus
—
Bifidobacteriaceae
Bifidobacterium
pseudolongum
Enterobacteriaceae
Proteus
—
Lactobacillaceae
Lactobacillus
Other
S24-7
—
—
Porphyromonadaceae
Parabacteroides
distasonis
Clostridiaceae
—
—
—
—
—
Desulfovibrionaceae
Desulfovibrio
—
S24-7
—
—
Coriobacteriaceae
Adlercreutzia
—
S24-7
—
—
Erysipelotrichaceae
Clostridium
cocleatum
Ruminococcaceae
Ruminococcus
—
—
—
—
Lachnospiraceae
Other
Other
Lachnospiraceae
[Ruminococcus]
gnavus
Turicibacteraceae
Turicibacter
—
Lachnospiraceae
—
—
Bacteroidaceae
Bacteroides
—
Ruminococcaceae
Oscillospira
—
Clostridiaceae
Other
Other
17
Lachnospiraceae
Roseburia
—
Lachnospiraceae
—
—
S24-7
—
—
Lachnospiraceae
[Ruminococcus]
gnavus
—
—
—
—
—
—
Lachnospiraceae
Coprococcus
—
S24-7
—
—
S24-7
—
—
—
—
—
Lachnospiraceae
Other
Other
Lachnospiraceae
[Ruminococcus]
gnavus
Erysipelotrichaceae
Allobaculum
—
Desulfovibrionaceae
Desulfovibrio
C21_c20
S24-7
—
—
—
—
—
Streptococcaceae
Lactococcus
garvieae
—
—
—
Enterobacteriaceae
Proteus
—
Lachnospiraceae
—
—
S24-7
—
—
Lachnospiraceae
[Ruminococcus]
gnavus
—
—
—
Clostridiaceae
Clostridium
Other
Lachnospiraceae
—
—
Clostridiaceae
Other
Other
—
—
—
Clostridiaceae
Other
Other
S24-7
—
—
Lachnospiraceae
[Ruminococcus]
gnavus
Bacteroidaceae
Bacteroides
—
S24-7
—
—
—
—
—
Lachnospiraceae
[Ruminococcus]
gnavus
Ruminococcaceae
Oscillospira
—
Ruminococcaceae
Anaerotruncus
—
Alcaligenaceae
Sutterella
—
S24-7
—
—
F16
—
—
10
—
—
—
Other
Other
Other
Lachnospiraceae
[Ruminococcus]
gnavus
S24-7
—
—
Rikenellaceae
—
—
Lachnospiraceae
[Ruminococcus]
gnavus
Lachnospiraceae
—
—
Clostridiaceae
Other
Other
Lachnospiraceae
—
—
Ruminococcaceae
Oscillospira
—
Enterococcaceae
Enterococcus
—
Rikenellaceae
—
—
S24-7
—
—
Lactobacillaceae
Lactobacillus
reuteri
Bacteroidaceae
Bacteroides
acidifaciens
Ruminococcaceae
Oscillospira
—
Enterobacteriaceae
Escherichia
coli
Ruminococcaceae
Oscillospira
—
—
—
—
S24-7
—
—
—
—
—
Other
Other
Other
Lachnospiraceae
—
—
Lachnospiraceae
[Ruminococcus]
gnavus
Lachnospiraceae
—
—
Enterobacteriaceae
Klebsiella
Other
Lachnospiraceae
—
—
Enterobacteriaceae
Klebsiella
—
7
—
—
—
Lactobacillaceae
Lactobacillus
—
Other
Other
Other
—
—
—
Erysipclotrichaceae
Allobaculum
—
—
—
—
Lachnospiraceae
—
—
—
—
—
4
Other
Other
Other
Lactobacillaceae
Lactobacillus
reuteri
Ruminococcaceae
Oscillospira
—
Lachnospiraceae
—
—
S24-7
—
—
S24-7
—
—
Lachnospiraceae
—
—
S24-7
—
—
8
S24-7
—
—
Lachnospiraceae
—
—
—
—
—
Coriobacteriaceae
Adlercreutzia
—
—
—
—
Clostidiaceae
Other
Other
Turicibacteraceae
Turicibacter
—
Ruminococcaceae
Ruminococcus
—
Ruminococcaceae
Oscillospira
—
Dehalobacteriaceae
Dehalobacterium
—
Staphylococcaceae
Staphylococcus
Other
Rikenellaceae
—
—
Lachnospiraceae
—
—
—
—
—
Staphylococcaceae
Staphylococcus
sciuri
—
—
—
Ruminococcaceae
Ruminococcus
—
Coriobacteriaceae
Adlercreutzia
—
Ruminococcaceae
Oscillospira
—
Lachnospiraceae
Clostridium
Other
Lachnospiraceae
Coprococcus
—
Desulfovibrionaceae
—
—
—
—
—
Lachnospiraceae
—
—
Dehalobacteriaceae
Dehalobacterium
—
Coriobacteriaceae
Adlercreutzia
—
Lachnospiraceae
—
—
3
Lachnospiraceae
—
—
Ruminococcaceae
Oscillospira
—
S24-7
—
—
5
Streptococcaceae
Streptococcus
—
Lachnospiraceae
[Ruminococcus]
gnavus
Lachnospiraceae
—
—
Lachnospiraceae
—
—
Coriobacteriaceae
Adlercreutzia
—
9
Lachnospiraceae
—
—
Erysipelotrichaceae
Clostridium
cocleatum
S24-7
—
—
0
Lachnospiraceae
—
—
Bacteroidaceae
Bacteroides
—
Ruminococcaceae
Oscillospira
—
Ruminococcaceae
—
—
Lachnospiraceae
Other
Other
Lachnospiraceae
Dorea
—
Lachnospiraceae
—
—
S24-7
—
—
Lachnospiraceae
—
—
Lactobacillaceae
Lactobaccilus
Other
—
—
—
Ruminococcaceae
Oscillospira
—
S24-7
—
—
9
S24-7
—
—
[Mogibacteriaceae]
—
—
Peptococcaceae
—
—
—
—
—
—
—
—
S24-7
—
—
Ruminococcaceae
Oscillospira
—
5
Ruminococcaceae
Oscillospira
—
Lachnospiraceae
[Ruminococcus]
gnavus
Ruminococcaceae
Oscillospira
—
Lactobacillaceae
Lactobaccilus
reuteri
Coriobacteriaceae
Adlercreutzia
—
Lachnospiraceae
—
—
S24-7
—
—
Turicibacteraceae
Turicibacter
—
S24-7
—
—
Rikenellaceae
—
—
Enterobacteriaceae
Other
Other
Rikenellaceae
Other
Other
Porphy ceae
Parabacteroides
—
Lactobacillaceae
Lactobaccilus
Other
—
—
—
S24-7
—
—
S24-7
—
—
—
—
—
Enterobacteriaceae
Klebsiella
Other
—
—
—
Lachnospiraceae
Other
Other
Ruminococcaceae
Oscillospira
—
Ruminococcaceae
Oscillospira
—
Bacteroidaceae
Bacteroides
acidifaciens
Erysipelotrichaceae
—
—
Lachnospiraceae
Dorea
—
Enterobacteriaceae
Enterobacter
Other
Ruminococcaceae
Oscillospira
—
Ruminococcaceae
Other
Other
3
0
Ruminococcaceae
Oscillospira
—
Rikenellaceae
—
—
—
—
—
9
Lachnospiraceae
Other
Other
Other
Other
Other
Ruminococcaceae
Oscillospira
—
S24-7
—
—
Lachnospiraceae
—
—
Streptococcaceae
Staphylococcus
—
—
—
—
—
—
—
—
—
—
Other
Other
Other
0
Aerococcaceae
Aerococcus
—
Ruminococcaceae
—
—
—
—
—
Streptococcaceae
Staphylococcus
—
Ruminococcaceae
Oscillospira
—
—
—
—
Ruminococcaceae
Ruminococcus
—
—
—
—
Other
Other
Other
Ruminococcaceae
Oscillospira
—
Lachnospiraceae
[Ruminococcus]
gnavus
Lachnospiraceae
—
—
Other
Other
Other
Ruminococcaceae
Oscillospira
—
—
—
—
Other
Other
Other
Other
Other
Other
Other
Other
Other
Lachnospiraceae
Clostridium
Other
Lachnospiraceae
—
—
Other
Other
Other
Other
Other
Other
Ruminococcaceae
Oscillospira
—
Desulfovibrionaceae
—
—
Dehalobacteriaceae
Dehalobacterium
—
Porphy
Parabacteroides
—
Ruminococcaceae
Ruminococcus
—
Lachnospiraceae
Coprococcus
—
Ruminococcaceae
Oscillospira
—
Other
Other
Other
Lachnospiraceae
Coprococcus
—
Ruminococcaceae
Oscillospira
—
Christensenellaceae
—
—
S24-7
—
—
Ruminococcaceae
Oscillospira
—
Lachnospiraceae
Coprococcus
—
Lachnospiraceae
Other
Other
Enterococcaceae
Va
coccus
—
Erysipelotrichaceae
Clostridium
cocleatum
—
—
—
Desulfovibrionaceae
Bilophila
—
—
—
—
Corynebacteriaceae
Corynebacterium
stationis
Lachnospiraceae
Other
Other
Lachnospiraceae
Other
Other
Lachnospiraceae
—
—
Lachnospiraceae
—
—
—
—
—
—
—
—
—
—
—
Ruminococcaceae
—
—
—
—
—
Lachnospiraceae
—
—
Dehalobacteriaceae
Dehalobacterium
—
Other
Other
Other
Streptococcaceae
Streptococcus
luteciae
—
—
—
S24-7
—
—
Ruminococcaceae
A
otruncus
—
Ruminococcaceae
Ruminococcus
—
Lachnospiraceae
Clostridium
Other
—
—
—
Ruminococcaceae
Oscillospira
—
Lachnospiraceae
Coprococcus
—
—
—
—
S24-7
—
—
A ceae
—
Lachnospiraceae
—
—
Ruminococcaceae
Oscillospira
—
S24-7
—
—
—
—
—
Ruminococcaceae
Oscillospira
—
Bacteroidaceae
Bacteroides
acidifaciens
Bacteroidaceae
Bacteroides
acidifaciens
Lachnospiraceae
Dorea
—
Ruminococcaceae
Other
Other
Lachnospiraceae
—
—
Lachnospiraceae
—
—
S24-7
—
—
Lachnospiraceae
Other
Other
—
—
—
Ruminococcaceae
Oscillospira
—
Ruminococcaceae
Oscillospira
—
Lachnospiraceae
—
—
Erysipelotrichaceae
—
—
S24-7
—
—
Lachnospiraceae
—
—
Ruminococcaceae
Oscillospira
—
Lachnospiraceae
—
—
Other
Other
Other
F16
—
—
Christensenellaceae
—
—
Lachnospiraceae
Other
Other
Ruminococcaceae
Oscillospira
—
—
—
—
Ruminococcaceae
Other
Other
—
—
—
Lachnospiraceae
Other
Other
Other
Other
Other
Enterococcaceae
Vagococcus
—
Ruminococcaceae
Ruminococcus
—
Erysipelotrichaceae
Coprabacillus
—
Other
Other
Other
Ruminococcaceae
Oscillospira
—
Ruminococcaceae
Oscillospira
—
—
—
—
Lachnospiraceae
—
—
Ruminococcaceae
Oscillospira
—
Lachnospiraceae
Coprococcus
—
Ruminococcaceae
Other
Other
—
—
—
Erysipelotrichaceae
—
—
Rikenellaceae
—
—
—
—
—
—
—
—
—
—
—
Lachnospiraceae
—
—
—
—
—
Lachnospiraceae
Clostridium
Other
Erysipelotrichaceae
—
—
Streptococcaceae
Streptococcus
minor
Ruminococcaceae
Oscillospira
—
—
—
—
Lachnospiraceae
Other
Other
Other
Other
Other
—
—
—
—
—
—
—
—
—
—
—
—
Lachnospiraceae
—
—
Streptococcaceae
Streptococcus
—
—
—
—
S24-7
—
—
—
—
—
Other
Other
Other
[Mogibacteriaceae]
—
—
Enterobacteriaceae
Enterobacter
Other
Eubacteriaceae
A
tis
—
Erysipelotrichaceae
—
—
Aerococcaceae
Aerococcus
—
Ruminococcaceae
Oscillospira
—
Ruminococcaceae
Oscillospira
—
Other
Other
Other
—
—
—
Lachnospiraceae
Coprococcus
—
Other
Other
Other
—
—
—
Lachnospiraceae
Other
Other
Ruminococcaceae
Oscillospira
—
S24-7
—
—
Ruminococcaceae
Ruminococcus
—
Ruminococcaceae
Oscillospira
—
—
—
—
—
—
—
—
—
—
—
—
—
Ruminococcaceae
Ruminococcus
—
Lachnospiraceae
—
—
Ruminococcaceae
Oscillospira
—
—
—
—
—
—
—
Corynebacteriaceae
Corynebacterium
stationis
Ruminococcaceae
Other
Other
S24-7
—
—
Ruminococcaceae
Oscillospira
—
Ruminococcaceae
Oscillospira
—
Ruminococcaceae
Ruminococcus
—
[Mogibacteriaceae]
—
—
Erysipelotrichaceae
Coprobacillus
—
Erysipelotrichaceae
Clostridium
cocleatum
S24-7
—
—
Coriobacteriaceae
—
—
Coriobacteriaceae
Adlercreutzia
—
Leucon ceae
Weissella
parame
enteroides
Enterococcaceae
Vagococcus
—
Ruminococcaceae
Buty
coccus
pullic
um
[Mogibacteriaceae]
—
—
—
—
—
—
—
—
—
—
—
S24-7
—
—
Ruminococcaceae
Oscillospira
—
Planococcaceae
Sporosarcina
—
Lachnospiraceae
Bl
producta
Erysipelotrichaceae
Other
Other
—
—
—
Ruminococcaceae
Ruminococcus
—
—
—
—
—
—
—
—
—
—
Coriobacteriaceae
Adlercreutzia
—
Other
Other
Other
Ruminococcaceae
Other
Other
Erysipelotrichaceae
—
—
Deferribacteraceae
Mucispirillum
schaedleri
Methylobacteriaceae
Methylobacterium
Other
Methanobacteriaceae
Methanobrevibacter
—
—
—
—
Lachnospiraceae
Other
Other
Lachnospiraceae
Dorea
—
Lachnospiraceae
Dorea
—
Lachnospiraceae
—
—
Erysipelotrichaceae
—
—
—
—
—
Other
Other
Other
Ruminococcaceae
Ruminococcus
—
Lachnospiraceae
Other
Other
Erysipelotrichaceae
Clostridum
cocleatum
Clostridiaceae
—
—
S24-7
—
—
Ruminococcaceae
Oscillospira
—
—
—
—
—
—
—
Erysipelotrichaceae
—
—
Streptococcaceae
—
—
Erysipelotrichaceae
—
—
Turicibacteraceae
Turicibacter
—
S24-7
—
—
Ruminococcaceae
Oscillospira
—
Lachnospiraceae
—
—
—
—
—
Erysipelotrichaceae
—
—
Ruminococcaceae
Ruminococcus
—
Moraxellaceae
Psychrobacter
Other
Paenibacillaceae
Other
Other
Lachnospiraceae
Coprococcus
—
Bacillaceae
Bacillus
—
Enterococcaceae
Vagococcus
—
—
—
—
Lachnospiraceae
—
—
—
—
—
—
—
—
—
—
—
Ruminococcaceae
Oscillospira
—
—
—
—
Erysipelotrichaceae
—
—
Other
Other
Other
Clostridiaceae
Clostridium
Other
—
—
—
Ruminococcaceae
Buty
coccus
pulli
um
Ruminococcaceae
—
—
Deferribacteraceae
Mucispirillum
schaedleri
Turicibacteraceae
Turicibacter
—
—
—
—
Verrucomicrobiaceae
Akkermansia
phil
[Mogibacteriaceae]
—
—
Alcaligenaceae
S
ella
—
Lachnospiraceae
Dorea
longicateria
Ruminococcaceae
—
—
Lachnospiraceae
—
—
—
—
—
indicates data missing or illegible when filed
Principal component analysis (PCA) and orthogonal partial least squares-discriminant analysis (OPLS-DA) were performed to visualize the metabolic differences among the experimental groups. For principal component analysis, SIMCA-P+ ver.17 (Umetrics) was used. The normalized peak heights of the sample-derived peaks were used to perform principal component analysis using all samples and five points (F18S-12, F18S-14, F18S-16, F18S-18, and F18S-20). Transform was set to none, and Scaling was set to Pareto scaling. Differential metabolites were selected according to the statistically significant variable importance in the projection (VIP) values obtained from the OPLS-DA model. Hierarchical Cluster Analysis (HCA) and heat maps were performed using R (https://www.r-project.org/).
Statistical data were analysed using Microsoft Excel statistics package analysis software. Graphs were prepared using Origin Lab's Originb 2021 software. For normally distributed variables, t-test or ANOVA with Tukey HSD was used and P values<0.05 were considered significant. For OPLS-DA, values from two-tailed Student's t-tests were applied on the normalized peak areas; metabolites with VIP values>1 and P values<0.05 were included. The Euclidean distance and Ward's method were used to analyze the heat map. The mean and variance were normalized so that the mean is 0 and the variance is 1.
There were no significant differences in mean body weights at any day during the treatment period between the control group and the other treatment groups. There were no significant differences in mean body weights on the day of sacrifice between the treatment groups.
With regard to the taxonomic profiling, firmicutes represented the most abundant phyla followed by Bacteroidetes (
When individual taxa were analysed in each of the beta glucan groups comparing it to Telmisartan (standard), decrease in enterobacteria was highest in AFO-202 group (
The resulting score plot of the principal component analysis using the normalized peak heights of the 10 samples (Pre- and post intervention of five groups), is shown in
Peak height of all the detected metabolite compounds after normalization is available in Table 3. The values which showed decrease post-intervention are highlighted in bold in the different groups. The number of peaks detected in the QC samples was 108, of which 53 peaks were qualitatively determined and 55 peaks were unknown.
Differential abundance analysis, log 2 fold change results are shown in
2.36
−46030.14
−33556.73
−4608.17
4
−182525.46
.94
.59
.14
.14
.2
−2877.12
−12323.49
.79
−726073.07
−23560.
3
.59
−65
28.4
822.23
.00
−170827.12
55701.34
61041.65
−28538.33
−48551.21
−35605.88
−14542.52
−16354.66
−14587.00
−116837.33
4923.32
−9328.70
thine-
467.68
6.02
−86133.86
−25379.22
−15626.91
−795.07
−1490.65
−125466.79
13904.61
−1866.22
−5760.44
−149.10
51487.07
−3276.22
−35249.
8
indicates data missing or illegible when filed
Score plots of PCA and compounds with a VIP value of 1 or higher in the OPLS-DA are shown in
In all the groups except telmisartan, phosphoric acid shoed the highest log 2 fold change in terms of increase. Putrescine showed the highest decrease. In regard to specific compounds, increase in succinic acid was highest in AFO-202 (P-value=0.06) with statistical significance (
This is the first study to investigate profiles of fecal gut microbiome and metabolome in a NASH model of mice with relevance to beta glucans specially so for two different beta glucans produced by different strains of same species of black yeast A. pullulans. Beta glucans are obtained from different sources and the functionality depends on the source and extraction/purification processes [D14]. The beta glucans described in the study from AFO-202 strain of the A. pullulans black yeast are unique as they are produced as an exopolysaccharide without the need for extraction/purification and hence the biological actions are superior [D15].
The AFO-202 beta glucan has been reported to have superior metabolic benefits by regularization of blood glucose levels [D5] apart from immune enhancement in immune-infectious illnesses such as COVID-19 [D11,12] and has been reported to produce positive effects on melatonin and alpha-synuclein neurotransmitters apart from improving sleep and behaviour in neurodevelopmental disorders such as ASD [D7,8]. In the NASH animal study, AFO-202 beta glucan has been able to significantly decrease inflammation-associated hepatic cell ballooning and steatosis [D10].
The N-163 beta glucan has been able to produce immune-modulatory benefits in terms of regulating dyslipidaemia evident from balance of the levels of non-esterified fatty acids [D16] and decrease in fibrosis and inflammation in NASH [D10]. The combination of AFO-202 and N-163 beta glucans has been able to decrease in pro-inflammatory markers and increase in anti-inflammatory markers in an advantageous manner in healthy human volunteers [D17], decrease the NAFLD Activity Score (NAS) in the NASH model [D10] and significantly control of immune-mediated dysregulated levels of IL-6, CRP and Ferritin in Covid-19 patients [D11,12]. In the study done on gut microbiome analysis in ASD subjects, there was efficient control of enterobacteria apart from beneficial reconstitution of the gut microbiome favourable for producing benefits in ASD by AFO-202 beta glucan [D13]. In the current study, we sought to evaluate the benefits of AFO-202 and N-163 individually and in combination in the NASH animal model.
The STAM model of NASH used in the study is a disease model, the Stelic Animal model [D10,18,19], in which mice are allowed to develop liver steatosis by injection streptozotocin solution 2 days after birth and fed with a high-fat diet. This model recapitulates most of the features of metabolic syndrome which occurs in humans wherein obesity and a high fat diet gives rise to diabetes, dyslipidaemia and liver steatosis.
Therefore, the gut microbiome profiles and fecal metabolite profiles that are present at baseline can be considered to recapitulate that which is present in metabolic syndrome [D20,21] which over time will produce pathophysiological problems in different organ systems of the body including the heart, liver, kidney apart from immune-metabolic interactions leading to a declined immune system with aging and its associated complications.
Therefore, the present study will serve as a forerunner to study the effects of the beta glucans on different aspects of metabolic syndrome associated pathologies as well as conditions associated with immune-metabolic interactions including neurological disorders in which such immune-metabolic interactions have profound implications [D20].
An abundance of bacterial species, such as Proteobacteria, Enterobacteria, and Escherichia coli has been reported in humans with NAFLD[D21, 22]. In the current study, a decrease in Enterobacteria with AFO-202 was reported.
We have earlier reported the decrease in Enterobacteria, Escherichia coli, Akkermansia muciniphila CAG:154, Blautia spp., Coprobacillus sp., and Clostridium bolteae CAG:59, with an increase of Faecalibacterium prausnitzii and Prevotella copri after AFO-202 consumption in children with ASD [D13]. In the present study as well, the decrease in enterobacteria was highest in AFO-202 group (
Use of Steroids has been reported to cause increase in E-Coli, enterococcus while decrease in Bacteroides [D25]. In the current study, the control of enterobacteria with increase in Bacteroides by AFO-202 make it worthy adjuncts for medications such as steroids as well.
In summary, black yeast A. pullulans' strain AFO-202 produced beta glucans increase the gut microbial diversity, control the harmful bacteria, promote healthy ones apart from producing beneficial differences in fecal metabolites, all indicative of a healthy profile both individually and in combination in this NASH animal model. AFO-202 will serve as a worthy treatment adjunct for neurodevelopmental conditions such as ASD and neurodegenerative conditions such as PD. With further validation on the correlation between gut microbiome and fecal metabolites in specific conditions, these safety proven prebiotics have potential applications in promoting a healthy life.
There is increasing evidence that gut dysbiosis plays a critical role in the development and progression of several neurodevelopmental conditions such as autism spectrum disorders (ASD), neuroinflammatory conditions such as multiple sclerosis (MS) and neurodegenerative diseases such as Alzheimers (AD) and Parkinson's disease (PD). The biofilms and byproducts of the bacteria especially the Gram-negative enteric bacteria mediate the effects of the altered gut microbiome in these disease conditions. Amyloid proteins constitute the major part of the biofilms especially the Curli amyloid whose characteristics have been reported to be similar to pathological and immunomodulatory human amyloids such as Alzheimer's disease associated amyloid-β, ASD and PD associated α-synuclein [A1].
Though these reports discuss on the correlation of the levels of these amyloids, enteric bacteria and the neural diseases, there has been no simple and safe intervention with subjective and objective correlation to a clinical benefit associated by altering the gut microbiota to a beneficial advantage. We herein report the outcome of beneficial reconstitution of gut microbiota especially those of the bacteria which are associated with Alpha synuclein and Curli amyloids after consumption of Beta 1,3-1,6 Glucans in children with autism spectrum disorder in a clinical pilot study.
The emerging evidence of constant communication and interaction between the gut and the brain through the gut-brain axis has begun to unravel its significance associated with the health of the central nervous system. Any dysbiosis of the gut microbiota has been shown to influence the development and progression of neurological pathologies of developmental (autism spectrum disorder [ASD]), inflammatory (multiple sclerosis [MS]), and degenerative (Alzheimer's disease [AD] and Parkinson's disease [PD]) disorders (Kang et al., 2019). The mechanisms involve activation of the immune system; production of inflammatory cytokines and chemokines (e.g., IL-6 and TNF-α); and alteration of the gut barrier permeability, which in turn is due to the increased levels of circulating lipopolysaccharide in these neurological disorders. These mechanisms modulate the neurotrophic factors, activity of the central and peripheral nervous system, and the endocrine pathways, all of which contribute to the onset or the phenotypic expression of neuropsychiatric and neurodevelopmental disorders (Santocchi et al., 2020).
Another important player is the amyloid protein, which has self-aggregation properties. Even non-identical amyloid proteins can accelerate reciprocal amyloid aggregation in a prion-like fashion. Nearly 30 amyloidogenic proteins are encoded by humans, whereas some functional amyloids are produced by the gut microbiome (Werner et al., 2020). Of importance are cell-surface amyloid proteins called curli, which are produced by certain enterobacteria that in turn accelerate formation of α-synuclein (αSyn), which is a presynaptic neurotransmitter that is crucial in the initiation and pathogenesis of neurological disorders such as ASD, PD, AD, and MS (Sampson et al., 2020). Though these reports (Sampson et al., 2020; Werner et al., 2020; Santocchi et al., 2020) discuss the correlation of the levels of these amyloids, enteric bacteria, and neural diseases, there has been no simple and safe intervention with subjective and objective correlation to a clinical benefit that can be derived based on an associated balancing of the gut microbiota.
We herein report the outcome of beneficial reconstitution of gut microbiota, especially those of the bacteria associated with αSyn and curli amyloids after consumption of beta 1,3-1,6 glucans in children with ASD in a clinical pilot study. The beta-glucan studied (Nichi Glucan) was obtained from the AFO-202 strain of a black yeast called Aureobasidium pullulans that has beneficial advantages in metabolic disorders by alleviating glucotoxicity (Dedeepiya et al., 2012), lipotoxicity (Ganesh et al., 2014; Ikewaki et al., 2021a), lipidemia-induced hepatic fibrosis (Ikewaki et al., 2021b), inflammation (Ikewaki et al., 2021b) apart from immune-enhancement (Ikewaki et al., 2021c), and modulation in COVID-19 (Raghavan et al., 2022), which have been reported in translational and clinical studies.
The effects of this AFO-202 beta glucan in terms of behavioural (Raghavan et al., 2021a) and sleep pattern improvement (Raghavan et al., 2021b), increase in levels of plasma αSyn (Raghavan et al., 2021a), and serum melatonin (Raghavan et al., 2021b) levels in children with ASD has been reported. We report the effects of AFO-202 beta glucan on the gut microbiome in children with ASD who participated in our pilot study.
The beta-glucan studied (Nichi Glucan) was obtained from the AFO-202 strain of a black yeast called Aureobasidium pullulans that has beneficial advantages in metabolic disorders by alleviating glucotoxicity [B5], lipotoxicity [B6, B7], lipidemia-induced hepatic fibrosis [B8], inflammation [B8] apart from immune-enhancement [B9], and modulation in COVID-19 [B10], which have been reported in translational and clinical studies. The effects of this AFO-202 beta glucan in terms of behavioural [E11] and sleep pattern improvement [B12], increase in levels of plasma αSyn [E11], and serum melatonin [B12] levels in children with ASD has been reported. We report the effects of AFO-202 beta glucan on the gut microbiome in children with ASD who participated in our pilot study.
In this randomized pilot clinical study, we have evaluated the gut microbiota of subjects with autism spectrum disorder (ASD) after consumption of Aureobasidium pullulans (black yeast) AFO-202 strain-produced beta glucan, Nichi Glucan.
The study was conducted in 18 subjects who were randomly allocated; six subjects (n=6) to the control group (Gr.1) who underwent conventional treatment comprising remedial behavioural therapies and L-carnosine 500 mg per day, and twelve subjects (n=12) Gr. 2 underwent supplementation with an Aureobasidium black yeast AFO-202 (Aureobasidium pullulans strain AFO-202 (also referred to as FO-68 [accession number: FERM BP-19327])) derived beta glucan, Nichi Glucan 0.5 g twice daily along with the conventional treatment. Stool samples from the subjects was collected at baseline and after the intervention. The samples were sequenced using Novaseq 6000 with a read length of 151 bp and taken for whole genome metagenome analysis.
Thirteen subjects (four in control (Gr.1) and nine in Nichi Glucan (Gr.2) completed the study. The sample reads were filtered for human DNA contamination. The alignment to the human genome was around 18.98%. The filtered reads were then aligned to bacterial, fungal, viral and archea genomes. The overall alignment to the bacterial genome was around 40%. However, the alignment to the viral, fungi and archea genomes was around 0.05-0.2%. De novo assembly was carried out using the pre-processed reads to obtain the scaffolds. These scaffolds were then used for gene prediction. Bacterial abundance was analysed.
The abundance of Enterobacter was decreased almost to nil in the Nichi Glucan (Gr.2) group after intervention while it increased from 0.36% to 0.85% in the control group (
Earlier reports have indicated a lower abundance of Prevotella, higher abundance of Lactobacillus and Bacteroides in children with autism spectrum disorders [A2]. MS patients also have been reported to have a lower abundance of Prevotella [A4]. Enterobacter produce functional amyloid proteins termed curli which promotes human amyloid α-synuclein (αSyn) pathology and the aggregation of curli and αSyn stimulates pathological and immunological processes that lead to the neurodevelopmental and neurodegenerative diseases such as ASD, MS, PD and AD [A1]. The results of this study which have produced favorable results after Nichi Glucan consumption in the gut microbiota by alleviate the pathological processes behind such neurodevelopmental and neurodegenerative diseases involving amyloid accumulations warrant larger clinical studies to recommend this as a routine food supplement in patients with neurodegenerative and neuroinflammatory conditions apart from research to explore their mechanisms and further potentials.
Eighteen subjects with ASD were randomly allocated: six subjects in the control group (Group 1): conventional treatment comprising remedial behavioural therapies and L-carnosine 500 mg per day, and 12 subjects (Group 2) underwent supplementation with Nichi Glucan 0.5 g twice daily along with the conventional treatment for 90 days.
Whole genome metagenome (WGM) sequencing of the stool samples at baseline and after intervention, showed that among genera of relevance, the abundance of Enterobacteria was decreased almost to zero in Group 2 after intervention, whereas it increased from 0.36% to 0.85% in Group 1. The abundance of Bacteroides increased in Group 1, whereas it decreased % in Group 2. The abundance of Prevotella increased in both Group 1 and Group 2. The decrease in abundance of Lactobacillus was significant in Group 2 compared to Group 1. Among species, a decrease was seen in Escherichia coli, Akkermansia muciniphila CAG:154, Blautia spp., Coprobacillus sp., and Clostridium bolteae CAG:59, with an increase of Faecalibacterium prausnitzii and Prevotella copri, which are both beneficial.
AFO-202 beta 1,3-1,6 glucan apart from balancing the gut microbiome in children with ASD, its role in effective control of curli-producing enterobacteria that leads to α-synuclein (αSyn) misfolding and accumulation, may have a prophylactic role in Parkinson's and Alzheimer's diseases as well.
The study was registered in India's clinical trial registry CTRI, Ref no:
This study was approved by the institutional ethics committee of the hospital in which the study took place and was registered as a clinical trial in the national clinical trial registry. The caregivers of each subject gave their informed consent for inclusion before participation in the study. The study was conducted in accordance with the Declaration of Helsinki.
Patients were involved in the design and conduct of this research. During the feasibility stage, priority of the research question, choice of outcome measures, and methods of recruitment were informed by discussions with patients through a focus group session and structured interviews. Once the trial has been published, participants will be informed of the results through a study newsletter suitable for a non-specialist audience.
The subjects enrolled in the study had received a diagnosis of ASD from a developmental pediatrician, which was verified by a psychologist using a clinical interview for a behavioural pattern that incorporated the Childhood Autism Rating Scale score.
Eighteen subjects with ASD were enrolled in this prospective, open-label, pilot clinical trial comprised of two arms. The CONSORT flow diagram is presented in
Arm 1 or Group 1 (control group): Six subjects with ASD underwent conventional treatment comprising remedial behavioural therapies and L-carnosine 500 mg per day.
Arm 2 or Group 2 (Nichi Glucan group): 12 subjects underwent supplementation with Nichi Glucan food supplement along with conventional treatment (remedial behavioural therapies and L-carnosine 500 mg per day). Each subject consumed two sachets (0.5 g each) of Nichi Glucan daily—one sachet with a meal twice daily—for 90 days.
The inclusion and exclusion criteria along with the assessment of behavioural and sleep pattern apart from evaluation of levels of αSyn and melatonin are available in the results of the clinical trial reported earlier (Raghavan et al., 2021a,b)
Faecal samples were collected at baseline and 90 days after the intervention using a sterile faecal collection kit and the samples were kept at −20° C. until they were transferred to the laboratory and processed. Samples for DNA extraction were stored at −80° C. until needed for analysis.
Total microbial DNA was extracted from faeces of each specimen using the QIAAmp DNA Mini Kit (Qiagen) according to manufacturer's instructions. Each batch of specimens were extracted with negative buffer control (extraction control).
Whole-genome metagenome sequencing libraries were prepared. In brief, the DNA was sheared using a Covaris ultrasonicator. Sheared DNA was subjected to a sequence of enzymatic steps for repairing the ends and tailing with dA by ligation of indexed adapter sequences. These adapter-ligated fragments were then cleaned up using SPRI beads. Next, the clean fragments are indexed using limited cycle PCR to enrich the adapter-ligated molecules. Finally, the amplified products were purified and checked before sequencing.
Prepared libraries were sequenced using Novaseq 6000 with a read length of 151 bp. The samples were taken for whole genome metagenome analysis. Initially, the reads were filtered for human DNA contamination (
The following bioinformatics pipeline was used to perform whole-genome-sequencing metagenomic analysis. The quality of the raw data was analysed and the adapters were trimmed. The low-processed reads were first aligned to human genome to remove unaligned reads that were then assembled using METASPADES de novo assembler for metagenomics. After assembly, the gene prediction was performed using PRODIGAL. The predicted genes were then searched against existing genes in the NCBI database using the DIAMOND MEGAN5 program. The occurrence of dominant microbial population was studied at various levels (phylum, class, order, family, and genus) based on the taxonomic abundance in the given samples. The dominance was calculated based on the amount of sequence obtained from samples, community composition, and the contig size distribution. Chimeric sequences were identified and filtered from the analysis.
Statistical data were analysed using Microsoft Excel statistics package analysis software. Paired t tests were also calculated using this package, and P values<0.05 were considered significant.
Eighteen patients who fulfilled all the selection criteria and none of the exclusion criteria were selected to begin the study. During enrolment, one participant in the treatment group (Group 2) dropped out before the study began. During the study, four subjects were lost to follow-up: two in Group 1 (one dropped out due to social problems in the family, and the other relocated to another city) and two in Group 2 (one dropped out due to social problems in the family, and the other relocated to another city). After excluding these four subjects, 13 subjects were included in the analysis.
The pre-processed reads were first aligned with the human genome (hg19) using BWA-MEM aligner to remove human genome contamination from the samples. The uncontaminated sequences were then taken for further alignment with known bacteria, fungi, virus, and archaea bacteria genomes using BWA MEM aligner. Around 7-12% of the reads mapped to the human genome, and the bacterial genome with 30-60% mapped reads.
Regarding the SEED average, there was several fold decrease in all the gene annotations (metabolites and metabolic functions) in the AFO-202 Nichi Glucan treatment group including Carbohydrates, Fatty acids, lipids, virulence, metabolite damage and nitrogen metabolism (
Bacterial kingdom was the most abundant organism type one. In both Group 1 (control) and Group 2 (Nichi Glucan), both before and after intervention, phylum Firmicutes was the most abundant followed by Bacteroidetes (
Among the genera of relevance, the abundance of Enterobacter was decreased to almost zero in Group 2 after intervention, while it increased from 0.36% to 0.85% in Group 1 (
The data of the genus and species level abundance in Gr. 1 and Gr. 2 are presented in Tables 5-9.
Bacteroides
Prevotella
Clostridium
Faecalibacterium
Bifidobacterium
Blautia
Roseburia
Eubacterium
Ruminococcus
Lachnoclostridium
Hungatella
Dialister
Klebsiella
Alistipes
Enterococcus
Veillonella
Akkermansia
Streptococcus
Megasphaera
Anaerostipes
Parabacteroides
Lactobacillus
Coprococcus
Dorea
Butyricicoccus
Flavonifractor
Escherichia
Odoribacter
Enterobacter
355
757
0.36
0.85
Subdoligranulum
Oscillibacter
Bilophila
Eggerthella
Collinsella
Turicibacter
Mitsuokella
Catenibacterium
Fusicatenibacter
Romboutsia
Haemophilus
Gemmiger
Sutterella
Lachnospira
Mycoplasma
Holdemanella
Parasutterella
Weissella
Ruminiclostridium
Chlamydia
Dakarella
Lactococcus
Clostridioides
Terrisporobacter
Coprobacillus
Adlercreutzia
Butyrivibrio
Anaerotruncus
Bacillus
Agathobaculum
Shigella
Tyzzerella
Oribacterium
Intestinimonas
Citrobacter
Fusobacterium
Acholeplasma
Pseudoflavonifractor
Raoultella
Muribaculum
Acidiphilium
Paenibacillus
Acidaminococcus
Salmonella
Anaeromassilibacillus
Pantoea
Intestinibacter
Eisenbergiella
Prevotellamassilia
Anaerotignum
Selenomonas
Actinomyces
Paraprevotella
Porphyromonas
Barnesiella
Anaerococcus
Stenotrophomonas
Finegoldia
Pediococcus
Solobacterium
Peptoniphilus
Urinacoccus
Enterorhabdus
Slackia
Achromobacter
Lysinibacillus
Olsenella
Comamonas
Ruthenibacterium
Viridibacillus
Rummeliibacillus
Gordonibacter
Drancourtella
Monoglobus
Lagierella
Peptococcus
Senegalimassilia
Libanicoccus
Urmitella
Paraclostridium
Acinetobacter
Anaerocolumna
Marvinbryantia
Robinsoniella
Neglecta
Butyricimonas
Bacteroides
Clostridium
Bifidobacterium
Faecalibacterium
Enterococcus
Eubacterium
Ruminococcus
Blautia
Alistipes
Collinsella
Parabacteroides
Oscillibacter
Prevotella
Lactobacillus
Roseburia
Trichosporon
Dorea
Lachnoclostridium
Olsenella
Escherichia
Klebsiella
Gemmiger
Streptococcus
Coprococcus
Subdoligranulum
Eggerthella
Dialister
Hungatella
Lysinibacillus
Acinetobacter
Pediococcus
Holdemanella
Butyricicoccus
Catenibacterium
Cloacibacillus
Pichia
Odoribacter
Rummeliibacillus
Weissella
Flavonifractor
Senegalimassilia
Megasphaera
Romboutsia
Desulfovibrio
Fusicatenibacter
Sutterella
Mycoplasma
Peptoniphilus
Ruminiclostridium
Anaerotruncus
Butyrivibrio
Methanobrevibacter
Megamonas
Bacillus
Butyricimonas
Anaerostipes
Phascolarctobacterium
Akkermansia
Gordonibacter
Coprobacillus
Coraliomargarita
Methanosphaera
Enterobacter
257
17
0.22
0.01
Pyramidobacter
Acidaminococcus
Paenibacillus
Acidiphilium
Adlercreutzia
Actinomyces
Succinatimonas
Libanicoccus
Angelakisella
Pseudoflavonifractor
Chlamydia
Anaeromassilibacillus
Intestinimonas
Duodenibacillus
Oribacterium
Clostridioides
Barnesiella
Slackia
Eisenbergiella
Acidovorax
Lactococcus
Paraprevotella
Shigella
Oxalobacter
Selenomonas
Turicibacter
Haemophilus
Pygmaiobacter
Urinacoccus
Bilophila
Allisonella
Ruthenibacterium
Raoultibacter
Comamonas
Neglecta
Atopabium
Christensenella
Massilimaliae
Finegoldia
Treponema
Robinsoniella
Enterorhabdus
Viridibacillus
Peptococcus
Fusobacterium
Terrisporobacter
Tyzzerella
Erysipelatoclostridium
Sporobacter
Mitsuokella
Cutaneotrichosporon
Marvinbryantia
Corallococcus
Anaerotignum
Prevotellamassilia
Anaerofilum
Synergistes
Alloprevotella
Isoptericola
Anaerococcus
Propionibacterium
Leuconostoc
Brachyspira
Lachnospira
Lagierella
Mogibacterium
Agathobaculum
Pseudobutyrivibrio
Hydrogenoanaerobacterium
Holdemania
Desulfotomaculum
Veillonella
Succinivibrio
Anaerocolumna
Mobilibacterium
Arabia
Enorma
Candida <Debaryomycetaceae>
Intestinibacter
Kurthia
Paeniclostridium
Paraclostridium
Listeria
Kwoniella
Cryptococcus
Fournierella
Sellimonas
Blastocystis
Acholeplasma
Azospirillum
Porphyromonas
Emergencia
Acetobacter
Dakarella
Parasutterella
Elusimicrobium
Drancourtella
Anaerovorax
Caldicoprobacter
Prevotella copri
Bifidobacterium longum
Hungatella hathewayi
Bacteroides fragilis
Bacteroides thetaiotaomicron
Blautia producta
Bifidobacterium bifidum
Roseboria intestinalis
Bacteroides vulgatus
Bacteroides ovatus
Clostridium neonatale
Escherichia coli
Akkermansia muciniphila
Prevotella copri CAG: 164
Flavonifractor plautii
Anaerostipes sp. BG01
Dialister sp. CAG: 357
Lactobacillus ruminis
Bacteroides uniformis
Roseburia faecis
Prevotella sp. CAG: 386
Eubacterium sp. CAG: 252
Bifidobacterium adolescentis
Klebsiella pneumoniae
Prevotella sp. CAG: 1092
Dialister succinatiphilus
Clostridium sp. AT4
Prevotella sp. CAG: 604
Bacteroides xylanisolvens
Bilophila wadsworthia
Blautia obeum
Prevotella sp. 885
Fusicatenibacter saccharivorans
Megasphaera elsdenii
Bacteroides nordii
Faecalibacterium sp. CAG: 82
Ruminococcus sp. CAG: 177
Dorea longicatena
Mitsuokella multacida
Blautia wexlerae
Eubacterium sp. CAG: 251
Prevotella multisaccharivorax
Odoribacter splanchnicus
Dialister sp. CAG: 486
Gemmiger formicilis
Streptococcus thermophilus
Romboutsia timonensis
Subdoligranulum sp. 60_17
Bacteroides oleiciplenus
Subdoligranulum variabile
Bacteroides plebeius
Roseburia hominis
Anaerostipes hadrus
Alistipes putredinis
Eggerthella lenta
Catenibacterium sp. CAG: 290
Mycoplasma sp. CAG: 956
Veillonella dispar
Oscillibacter sp. ER4
Clostridium sp. CAG: 81
Collinsella aerofaciens
Bacteroides timonenasis
Turicibacter sanguinis
Bifidobacterium sp. N5G01
Bacteroides caccae
Bacteroides cellulosilyticus
Clostridium sp. CAG: 7
Parabacteroides merdae
Clostridium sp. CAG: 389
Blautia sp. CAG: 37
Coprococcus eutactus
Veillonella atypica
Ruminococcus callidus
Holdemanella biformis
Bacteroides stercoris
Bifidobacterium catenulatum
Prevotella sp. CAG: 520
Roseburia sp. CAG: 18
Alistipes sp. HGB5
Haemophilus parainfluenzae
Streptococcus salvarius
Coprococcus catus
Bacteroides plebeius CAG: 211
Ruminococcus bromii
Megasphaera massiliensis
Enterobacter hormaechei
Roseburia intestinalis CAG: 13
Catenibacterium mitsuokai
Dakarella massiliensis
Chlamydia trachomatis
Enterobacter cloacae
Bifidobacterium pseudocatenulatum
Veillenella parvula
Roseburia sp. CAG: 18_43_25
Bacteroides sp. HPS0048
Weissella confusa
Clostridium sp. CAG: 122
Prevotella stercorea
Ruminococcus sp. CAG: 254
Bacteroides sp. 2_2_4
Enterococcus asini
Blautia sp. KLE 1732
Clostridioides difficile
Bifidobacteriam kashiwanohense
Roseburia inulinivorans
Prevotella sp. CAG: 732
Clostridium sp. CAG: 12237_41
Clostridium bolteae CAG: 59
Clostridium sp. ATCC BAA-442
Laetobacillus ruminis CAG: 367
Alistipes senegalensis
Alistipes finegoldii
Adlercreutzia equolifaciens
Sutterella sp. CAG: 351
Parabacteroides distasonis
Enterococcus avium
Bacteroides dorei
Bacteroides stercorirosoris
Collinsella sp. CAG: 66
Lactococcus garvieae
Prevotella sp. CAG: 474
Terrisporobacter glycolicus
Osdillibacter sp. 57_20
Veillonella sp. DORA_A_3_16_22
Ruminococcus sp. CAG: 90
Prevotella sp. CAG: 592
Lachnospira pactinoschiza
Bacteroides intestinalis
Bacteroides caccae CAG: 21
Bacteroides sp. 3_1_23
Faecalibacterium sp. CAG: 82-related_59_9
Klebsiella michiganensis
Bifidobacterium sp. N4G05
Eubacterium sp. CAG: 248
Dorea formicigenerans
Megasphaera sp. D1SK 18
Coprobacillus sp. CAG: 235
Prevotella stercorea CAG: 629
Blautia sp. CAG: 37
Alistipes finegoldii CAG: 68
Dorea sp. CAG: 105
Ruminococcus obeum CAG: 39
Bacteroides cellulosilyticus CAG: 158
Ruminococcus sp. CAG: 17
Eubacterium sp. CAG: 38
Clostridium sp. CAG: 91
Klebsiella oxytoca
Bacteroides sp. 43_46
Prevotella sp. P3-122
Roseburia sp. CAG: 471
Agathobaculum desmolans
Sutterella parvirubra
Eubacterium rectale CAG: 36
Collinsella sp. 4_8_47FAA
Blantia sp. Marseille-P3201T
Bacteroide sp. 4_1_36
Parasutterella excrementihominis CAG: 233
Bilophila sp. 4_1_30
Eubacterium elugens CAG: 72
Parabacteroides merdae CAG: 48
Blautia sp. CAG: 37_48_57
Bifidobacterium breve
Ruminococcus sp. CAG: 108
Bacteroides sp. 41_26
Anaerostipes caccae
Enterococcus sp. HMSC05C03
Butyricicoccus sp. BB10
Alistipes putredinis CAG: 67
Bacteroides sp. D20
Lachnoclostridium sp. An196
Dorea longicatena CAG: 42
Klebsiella variicola
Akkermansia muciniphila CAG: 154
Streptococcus pneumoniae
Eubacterium sp. 45_250
Bacteroides sp. D22
Eubacterium sp. CAG: 76
Oscillibacter sp. CAG: 241
Blautia massiliensis
Blautia sp. SF-50
Parabacteroides sp. D13
Enterococcus faecium
Mitsuokella jalaludinii
Sutterella wadsworthensis
Coprococcus sp. CAG: 131
Lachnoclostridium sp. An14
Blautia sp. Marseille-P2398
Eubacterium ballii CAG: 12
Bacteroides sp. 1_1_30
Acholeplasma sp. CAG: 878
Blautia sp. CAG: 52
Veillonella sp. HPA0037
Alistipes indistinctus
Ruminococcus sp. SR1/5
Roseburia sp. CAG: 50
Erwinia phage vB_EamM_V3
Bacteroides sp. D2
Eubacterium ramulus
Veillonella sp. oral taxon 158
Turicibacter sp. H121
Fusobacterium sp. CAG: 815
Alistipes sp. 58_9_plus
Prevotella sp. CAG: 873
Clostridium sp. CAG: 448
Clostridium sp. CAG: 492
Veillonella sp. ACP1
Bacteroides sp. 14(A)
Ruminococcus sp. CAG: 108_related_41_35
Bacteroides intestinalis CAG: 315
Lactobacillus rogosae
Alistipes timonensis
Anaerotruncus sp. CAG: 390
Prevotella sp. P4-65
Blautia sp. An81
Bacteroides fragilis CAG: 558
Tyzzerella nexilis
Romboutsia ilealis
Prevotella sp. P5-108
Prevotella sp. P4-76
Acidiphilium sp. CAG: 727
Muribaculum intestinale
Eubacterium sp. 41_20
Shigella sonnei
Terrisporobacter othiniensis
Bifidobacterium adolescentis CAG: 119
Veillonella sp. ICM51a
Prevotella sp. AGR2160
Bacteroides sp. 3_1_13
Prevotella sp. CAG: 279
Intestinimonas butyriciproducens
Prevotella bryantii
Streptococcus parasanguinis
Clostridium sp. CAG: 43
Klebsiella aerogenes
Ruminococcus sp. CAG: 330
Clostridium sp. SS2/1
Prevotella sp. P5-64
Oscillibacter sp. CAG: 241_62_21
Megashaera sp. MJR8396C
Coprococcus sp. CAG: 131-related_45_246
Ruminococcus sp. CAG: 9
Prevotella sp. P5-60
Bacteroides sp. CAG: 927
Megashaera sp. BL7
Odoribacter splanchnicus CAG: 14
Bacteroides sp. 43_108
Citrobacter koseri
Clostridium sp. CAG: 221
Veillonella tobetsuensis
Collinsella sp. TF06-26
Prevotella sp. P2-180
Bacteroides intestinalis CAG: 564
Lachnoclostridium edouardi
Eggerthella sp. 1_3_56FAA
Klebsiella sp. MS 92-3
Ruminococcus faecis
Bacteroides sp. 3_1_19
Bacteroides stercoris CAG: 120
Bacteroides sp. 1_1_14
Parabacteroides johnsonii
Weissella cibaria
Prevotella sp. P4-67
Intestinibacter bartlettii
Eubacterium siraeum CAG: 80
Salmonella enterica
Prevotella sp. P4-51
Clostridium botulinum
Clostridium nexile CAG: 348
Prevotellamassilia timonensis
Prevotella sp. P5-125
Shigella flexneri
Eubacterium sp. CAG76_36_125
Prevotella sp. P5-119
Prevotella ruminicola
Prevotella lascolanii
Prevotella buccae
Clostridium butyricum
Blautia sp. An46
Eggerthella sp. HGA1
Coprococcus comes
Prevotella sp. CAG: 1185
Bifidobacterium ruminantium
Bacteroides finegoldii
Bacteroides sp. 4_3_47FAA
Bacteroides sp. 3_1_40A
Bacteroides sp. CAG: 530
Prevotella sp. CAG: 1124
Bacteroides vulgatus CAG: 6
Prevotella baroniae
Anaerotignum lactatifermentans
Blautia hansenii
Prevotella sp. CAG: 487
Prevotella timonensis
Ruminococcus gnavus CAG: 126
Megasphaera sp. NM10
Prevotella buccalis
Prevotella sp. P5-92
Streptococcus infantarius
Bacteroides sp. AR20
Enterobacter sp. BIDMC 29
Bacteroides uniformis CAG: 3
Prevotella sp. CAG: 5226
Bacteroides sartorii
Blautia schinkii
Prevotella intermedia
Bacteroides mediterraneensis
Veillonella sp. 6_1_27
Lactoccoccus lactis
Bacteroides faecis
Prevotella histicola
Sutterella sp. KLE1602
Prevotella oralis
Bifidobacterium bifidum CAG: 234
Prevotella sp. P4-119
Prevotella paludivivens
Prevotella sp. tc2-28
Prevotella sp. P5-126
Prevotella sp. 109
Prevotella brevis
Prevotella oris
Prevotella sp. DNF00663
Prevotella oryzae
Prevotella sp. CAG: 255
Prevotella sp. S7-1-8
Prevotella dentalis
Prevotella sp. KH2C16
Prevotella sp. CAG: 1058
Prevotella maculosa
Prevotella bergensic
Dorea formicigenerans CAG:28
Rummeliibacillus stabekisii
Bifidobacterium pseudolongum
Clostridium sp. CAG: 138
Blautia sp. CAG: 257
Enterococcus sp. HMSC072H05
Bacteroides dorei CAG: 222
Bacteroides sp. CAG: 189
Ruminococcus sp. CAG: 382
Peptococcus niger
Oribacterium sp. C9
Olsenella provencensis
Collinsella sp. CAG: 289
Ruthenibacterium lactatiformans
Acinetobacter sp. NIPH 899
Oribacterium sp. WCC10
Clostridium sp. CAG: 609
Clostridium sp. CAG: 571
Butyricimonas virosa
Slackia piriformis
Achromobacter xylosoxidans
Lactobacillus mucosae
Clostridium sp. CAG: 433
Clostridium sp. CAG: 226
Solobacterium moorei
Anaerococcus prevotii
Olsenella sp. An188
Clostridium minihomine
Acinetobacter sp. NIPH 2171
Anaeromassilibacillus senegalensis
Dialister invisus
Gordonibacter pamelaeae
Parabacteroides sp. HGS0025
Anaeromassilibacillus sp. Marseille-P3371
Peptoniphilus senegalensis
Clostridium sp. 7_2_43FAA
Peptoniphilus duerdenii
Comamonas testosteroni
Anaeromassilibacillus sp. An200
Lysinibacillus sp. ZYM-1
Mycoplasma sp. CAG: 472
Clostridium sp. L2-50
Collinsella sp. MS5
Bacillus kochii
Faecalibacterium sp. CAG: 74_58_120
Clostridium sp. CAG: 793
Neglecta timonensis
Clostridium sp. ASB-410
Enterococcus cassaliflavus
Eggerthella timonensis
Faecalibacterium sp. CAG: 74
Anaerocococcus vaginalis
Anaeromassilibacillus sp. An250
Acinetobacter sp. CIP 101934
Barnesiella intestinihominis
Enterococcus gallinarum
Lysinibacillus sp. FJAT-14222
Alistipes shahii
Peptoniphilus timonensis
Akkermansia sp. CAG: 344
Lagierella massiliensis
Acinetobacter sp. LCT-H3
Drancourtella massiliensis
Subdoligranulum sp. 4_3_54A2FAA
Peptoniphilus harei
Ruminococcus sp. CAG:9-related_41_34
Ruminococcus lactaris
Enterorhabdus caecimuris
Alistipes sp. Marseille-P2431
Bacteroides thetaiotaomicron CAG: 40
Ruminococcus flavefaciens
Blantis sp. Marseille-P3087
Oribacterium sp. P6A1
Clostridium sp. C105KSO15
Achromobacter sp. Root170
Clostridium sp. CAG: 264
Lysinibacillus sphaerieus
Clostridium sp. CAG: 1024
Urinacoccus sp. Marseille-P3926
Enterococcus sp. FDAARGOS_375
Lysinibacillus boronitolerans
Collinsella bouchesdurhonensis
Rummeliibacillus pyenus
Collinsella sp. 60_9
Alistipes obesi
Dialister invisus CAG: 218
Eubacterium sp. CAG: 161
Bacteroides sp. 3_1_33FAA
Paraclostridium bifermentans
Monoglobus pectinilyticus
Oribacterium sp. NK2B42
Lactobacillus brevis
Senegalimassilia anaerobia
Acinetobacter baumannii
Clostidium sp. CAG: 567
Coprococcus sp. ART55/1
Peptoniphilus sp. HMSC075B08
Enterococcus pallens
Finegoldia magna
Lysinibacillus sp. FJAT-14745
Coprobacillus sp. 8_1_38FAA
Peptoniphilus sp. oral taxon 375
Gordonibacter massiliensis
Olsenella sp. An290
Peptoniphilus grossensis
Bacteroides sp. 9_1_42FAA
Enterococcus faecalis
Lactobacillus plantarum
Lysinibacillus fusiformis
Eubacterium sp. 38_16
Peptoniphilus sp. HMSC062D09
Alistipes sp. AG:53
Peptoniphilus phoceensis
Peptoniphilus sp. BV3AC2
Clostridium sp. CAG: 302
Gordonibacter urolithinfaciens
Acinetobacter sp. YZS-X1-1
Lysinibacillus xylanilyticus
Acinetobacter schindleri
Bacteroides sp. CAG: 20
Clostridium sp. CAG: 269
Alistipes sp. cv1
Roseburia inulinivorans CAG: 15
Urmitella timonensis
Olsenella sp. An293
Eisenbergiella tayi
Enterococcus saccharolyticus
Parabacteroides goldsteinii
Marvinbryantia formatexigens
Lysinibacillus macroides
Bacteroides salyersiae
Eubacterium sp. CAG: 146
Peptoniphilus coxii
Libanicoccus massiliensis
Roseburia sp. CAG: 182
Clostridium sp. CAG: 413
Dorea sp. AGR2135
Acinetobacter lwoffii
Faecalibacterium prausnitzii
Bifidobacterium longum
Trichosporon asahii
Bifidobacterium adolescentis
Escherichia coli
Collinsella aerofaciens
Ruminococcus sp. CAG: 177
Bacteroides fragilis
Blautia obeum
Gemmiger formicilis
Dorea longicatena
Clostridium sp. CAG: 226
Clostridium sp. CAG: 138
Bacteroides uniformis
Alistipes sp. CAG: 435
Hungatella hathewayi
Holdemanella biformis
Oscillibacter sp. CAG: 241
Subdoligranulum sp. 60_17
Pichia kudriavzevii
Bacteroides thetaiotaomicron
Prevotella copri
Eubacterium sp. CAG: 202
Enterococcus faecium
Klebsiella pneumoniae
Rummeliibacillus pycnus
Senegalimassilia anaerobia
Bifidobacterium bifidum
Bacteroides ovatus
Bacteroides vulgatus
Blautia wexlerae
Lactobacillus ruminis
Fusicatenibacter saccharivorans
Olsenella umbonata
Desulfovibrio piger
Dialister sp. CAG: 486
Enterococcus avium
Faecalibacterium sp. CAG: 74
Bacteroides caccae
Oscillibacter sp. CAG: 241_62_21
Romboutsia timonensis
Eubacterium sp. CAG: 180
Parabacteroides merdae
Pediococcus pentosaceus
Enterococcus faecalis
Clostridium sp. CAG: 221
Butyricicoccus sp. BB10
Prevotella sp. CAG: 279
Bifidobacterium sp. N5G01
Cloacibacillus porcorum
Clostridium sp. CAG: 1024
Prevotella copri CAG: 164
Catenibacterium mitsuokai
Eggerthella lenta
Mycoplasma sp. CAG: 956
Oscillibacter sp. ER4
Roseburia faecis
Coraliomargarita sp. CAG: 312
Catenibacterium sp. CAG: 290
Pediococcus acidilactici
Bacteroides dorei
Alistipes putredinis
Odoribacter splanchnicus
Parabacteroides sp. SN4
Alistipes indistinctus
Alistipes obesi
Clostridium sp. CAG: 510
Dialister sp. CAG: 357
Faecalibacterium sp. CAG: 74_58_120
Lactobacillus brevis
Bifidobacterium ruminantium
Anaerostipes hadrus
Enterococcus casseliflavus
Cloacibacillus sp. An23
Bacteroides plebeius
Weissella confusa
Lactobacillus plantarum
Bacteroides sp. CAG: 545
Clostridium sp. CAG: 452
Collinsella sp. 4_8_47FAA
Ruminococcus sp. CAG: 724
Roseburia inulinivorans
Bacteroides xylanisolvens
Butyricimonas virosa
Bacteroides intestinalis
Collinsella sp. CAG: 166
Ruminococcus sp. CAG: 488
Klebsiella aerogenes
Phascolarctobacterium sp. CAG: 207
Acidiphilium sp. CAG: 727
Parabacteroides gordonii
Adlercreutzia equolifaciens
Butyrivibrio sp. CAG: 318
Blautia sp. CAG: 37
Clostridium sp. CAG: 448
Bacteroides salyersiae
Coprobacillus sp. CAG: 235
Bifidobacterium pseudocatenulatum
Collinsella sp. TF06-26
Alistipes sp. CAG: 53
Clostridium sp. CAG: 571
Sutterella sp. CAG: 397
Libanicoccus massiliensis
Succinatimonas sp. CAG: 777
Subdoligranulum variabile
Anaerotruncus sp. CAG: 390
Bifidobacterium angulatum
Angelakisella massiliensis
Alistipes senegalensis
Megamonas funiformis
Ruminococcus obeum CAG: 39
Acidaminococcus fermentans
Lactobacillus mucosae
Chlamydia trachomatis
Ruminococcus bromii
Eubacterium sp. CAG: 581
Clostridium sp. CAG: 433
Weissella cibaria
Methanobrevibacter smithii
Enterococcus gallinarum
Clostridium sp. CAG: 302
Bacteroides plebeius CAG: 211
Eubacterium limosum
Clostridium sp. CAG: 349
Clostridium sp. CAG: 43
Parabacteroides sp. HGS0025
Akkermansia muciniphila
Clostridium sp. CAG: 245
Enterococcus hirae
Duodenibacillus massiliensis
Coprococcus eutactus
Faecalibacterium sp. CAG: 82
Lysinibacillus xylanilyticus
Peptoniphilus coxii
Lysinibacillus sp. FJAT-14222
Bifidobacterium breve
Clostridium sp. CAG: 245_30_32
Clostridium sp. CAG: 451
Sutterella wadsworthensis
Parabacteroides goldsteinii
Streptococcus mutans
Roseburia hominis
Streptococcus salivarius
Alistipes sp. CAG: 514
Roseburia sp. CAG: 18
Ruminococcus sp. 5_1_39BFAA
Clostridioides difficile
Flavonifractor plautii
Gordonibacter urolithinfaciens
Pyramidobacter sp. C12-8
Ruminococcus sp. CAG: 17
Bifidobacterium sp. N4G05
Streptococcus thermophilus
Clostridium sp. CAG: 568
Blautia sp. KLE 1732
Parabacteroides merdae CAG: 48
Dorea formicigenerans
Barnesiella intestinihominis
Intestinimonas butyriciproducens
Olsenella scatoligenes
Clostridium sp. CAG: 413
Clostridium sp. CAG: 343
Alistipes shahii
Pyramidobacter piscolens
Collinsella sp. CAG: 289
Bacteroides caccae CAG: 21
Acinetobacter baumannii
Bacteroides sp. 2_2 4
Eubacterium sp. CAG: 841
Ruminococcus sp. CAG: 382
Olsenella sp. kh2p3
Phascolarctobacterium succinatutens
Lactobacillus fermentum
Mycoplasma sp. CAG: 877
Bacteroides intestinalis CAG: 564
Oxalobacter formigenes
Eisenbergiella tayi
Parabacteroides distasonis
Dorea longicatena CAG: 42
Ruminococcus sp. CAG: 563
Olsenella sp. KH3B4
Lactobacillus ruminis CAG: 367
Blautia sp. CAG: 37_48_57
Alistipes sp. 56_sp_Nov_56_25
Enterococcus thailandicus
Alistipes sp. HGB5
Blautia sp. CAG: 237
Bacteroides stercoris
Clostridium sp. CAG: 1193
Roseburia sp. CAG: 18_43_25
Oscillibacter sp. 57_20
Olsenella provencensis
Olsenella sp. An188
Anaerotruncus colihominis
Clostridium sp. CAG: 524
Bacteroides coprocola CAG: 162
Coprococcus comes
Ruminococcus sp. CAG: 90
Clostridium sp. CAG: 492
Pygmaiobacter massiliensis
Lysinibacillus sphaericus
Roseburia intestinalis
Ruminococcus flavefaciens
Lactococcus lactis
Gordonibacter pamelaeae
Bacteroides eggerthii
Bifidobacterium kashiwanohense
Collinsella sp. 60_9
Allisonella histaminiformans
Megasphaera elsdenii
Lysinibacillus sp. FJAT-14745
Ruminococcus sp. CAG: 9
Urinacoccus sp. Marseille-P3926
Bacteroides coprocola
Olsenella sp. An285
Acinetobacter sp. NIPH 2171
Clostridium sp. CAG: 7
Bifidobacterium catenulatum
Olsenella sp. An290
Eubacterium hallii CAG: 12
Streptococcus pneumoniae
Collinsella sp. MSS
Eggerthella sp. CAG: 209
Bacteroides sp. CAG: 20
Clostridium sp. C105KSO15
Bacteroides sp. CAG: 189
Clostridium sp. CAG: 594
Bacteroides sp. AR29
Subdoligranulum sp. 4_3_54A2FAA
Ruminococcus bicirculans
Oscillibacter sp. 1-3
Blautia sp. Marseille-P3087
Turicibacter sanguinis
Peptoniphilus sp. oral taxon 375
Olsenella sp. An293
Bacteroides intestinalis CAG: 315
Acinetobacter sp. YZS-X1-1
Ruthenibacterium lactatiformans
Coprococcus eutactus CAG: 665
Bacteroides sp. D20
Clostridium sp. CAG: 264
Blautia producta
Bacteroides sp. 1_1_14
Peptoniphilus sp. HMSC075B08
Selenomonas bovis
Bacteroides sp. 4_1_36
Slackia piriformis
Enterobacter cloacae
Ruminococcus sp. CAG: 254
Alistipes finegoldii
Haemophilus parainfluenzae
Alistipes onderdonkii
Actinomyces sp. HPA0247
Roseburia sp. CAG: 182
Parabacteroides sp. merdae-related_45_40
Collinsella vaginalis
Faecalibacterium sp. CAG: 82-related_59_9
Acinetobacter schindleri
Bacteroides oleiciplenus
Megamonas rupellensis
Blautia sp. Marseille-P2398
Clostridium sp. CAG: 81
Comamonas kerstersii
Eggerthella sp. 1_3_56FAA
Enterobacter hormaechei
Rummeliibacillus stabekisii
Acinetobacter bereziniae
Lactobacillus pentosus
Eubacterium eligens CAG: 72
Oscillibacter valericigenes
Bacteroides nordii
Clostridium sp. CAG: 127
Eggerthella sp. HGA1
Ruminococcus sp. CAG: 108
Olsenella mediterranea
Oscillibacter sp. PC13
Subdoligranulum sp. CAG: 314
Oscillibacter ruminantium
Roseburia inulinivorans CAG: 15
Finegoldia magna
Bacteroides sp. 3_1_23
Peptococcus niger
Bacteroides sp. HPS0048
Oribacterium sp. WCC10
Paraprevotella clara CAG: 116
Prevotella sp. CAG: 604
Intestinimonas massiliensis
Clostridium sp. L2-50
Clostridium sp. CAG: 1000
Enterococcus sp. FDAARGOS_375
Prevotella sp. CAG: 891
Parabacteroides sp. D13
Enterococcus sp. HMSC05C03
Eubacterium siraeum CAG: 80
Megasphaera sp. BL7
Bacteroides sp. D22
Lysinibacillus macroides
Klebsiella sp. MS 92-3
Sporobacter termitidis
Acinetobacter sp. NIPH 899
Pseudoflavonifractor capillosus
Lysinibacillus fusiformis
Peptoniphilus grossensis
Ruminococcus albus
Cutaneotrichosporon oleaginosum
Marvinbryantia formatexigens
Collinsella bouchesdurhonensis
Acidovorax sp. 12322-1
Corallococcus sp. CAG: 1435
Bacteroides sp. 3_1_40A
Anaerotruncus rubiinfantis
Ruminococcus champanellensis
Clostridium sp. CAG: 349_48_7
Eubacterium rectale CAG: 36
Clostridium sp. CAG: 91
Butyricimonas sp. An62
Alistipes finegoldii CAG: 68
Flavonifractor sp. An10
Lactobacillus salivarius
Butyricimonas synergistica
Prevotella sp. CAG: 5226
Romboutsia ilealis
Eubacterium sp. CAG: 146
Clostridium sp. CAG: 609
Blautia schinkii
Methanosphaera stadtmanae
Alistipes timonensis
Lysinibacillus sp. ZYM-1
Bacteroides sp. CAG: 709
Terrisporobacter glycolicus
Bacteroides sp. 3_1_19
Bacteroides stercorirosoris
Peptoniphilus phoceensis
Oribacterium sp. P6A1
Prevotellamassilia timonensis
Ruminococcus faecis
Anaerofilum sp. An201
Dorea sp. 42_8
Eubacterium sp. CAG: 251
Clostridium sp. CAG: 389
Blautia sp. SF-50
Eubacterium sp. CAG: 76
Bacteroides bouchesdurhonensis
Eubacterium sp. 38_16
Clostridium sp. HGF2
Bacteroides sp. 9_1_42FAA
Mitsuokella jalaludinii
Paraprevotella clara
Blautia massiliensis
Bacteroides sp. 43_108
Olsenella sp. An270
Isoptericola variabilis
Clostridium sp. KNHs209
Streptococcus parasanguinis
Gordonibacter massiliensis
Bilophila wadsworthia
Clostridium sp. SS2/1
Parabacteroides johnsonii
Flavonifractor sp. An100
Propionibacterium acidifaciens
Clostridium sp. CAG: 914
Clostridium sp. 26_22
Bacteroides cellulosilyticus
Butyricicoccus pullicaecorum
Actinomyces sp. ICM47
Olsenella sp. Marseille-P2300
Collinsella tanakaei
Gemmiger sp. An120
Pseudoflavonifractor sp. An184
Peptoniphilus sp. HMSC062D09
Bacteroides sp. 3_1_13
Flavonifractor sp. An306
Peptoniphilus timonensis
Eubacterium ventriosum
Cloacibacillus evryensis
Akkermansia muciniphila CAG: 154
Oribacterium sp. C9
Olsenella uli
Prevotella sp. CAG: 1092
Slackia heliotrinireducens
Lagierella massiliensis
Oribacterium sp. NK2B42
Bacteroides eggerthii CAG: 109
Eggerthella timonensis
Clostridium sp. CAG: 762
Brachyspira sp. CAG: 484
Eggerthella sp. 51_9
Ruminococcus sp. CAG: 379
Bacteroides timonensis
Oscillibacter sp. CAG: 155
Peptoniphilus senegalensis
Lachnospira pectinoschiza
Megamonas sp. Calf98-2
Massilimaliae massiliensis
Anaeromassilibacillus sp. An200
Odoribacter sp. 43_10
Bacteroides mediterraneensis
Bacteroides dorei CAG: 222
Bacteroides sp. 4_3_47FAA
Clostridium disporicum
Neglecta timonensis
Bacteroides uniformis CAG: 3
Clostridium bolteae CAG: 59
Agathobaculum desmolans
Bacteroides sartorii
Bacteroides sp. D2
Lachnoclostridium sp. An196
Acidaminococcus massiliensis
Raoultibacter massiliensis
Clostridium sp. SN20
Eggerthella sp. YY7918
Bacteroides sp. 43_46
Alistipes sp. AL-1
Ruminococcus sp. SR1/5
Bacteroides fragilis CAG: 558
Hydrogenoanaerobacterium saccharovorans
Alistipes sp. 58_9_plus
Eubacterium sp. CAG: 86
Faecalibacterium sp. CAG: 1138
Actinomyces oris
Dorea sp. CAG: 105
Succinivibrio dextrinosolvens
Lactococcus garvieae
Bacteroides sp. 1_1_30
Ruminococcus sp. CAG: 9-related 41_34
Enterococcus sp. HMSC072H05
Clostridium sp. CAG: 440
Coprobacillus sp. 8_1_38FAA
Alloprevotella rava
Collinsella ihuae
Coprobacillus sp. CAG: 235_29_27
Megasphaera sp. NM10
Enterorhabdus caecimuris
Peptoniphilus harei
Prevotella sp. CAG: 755
Ruminococcus sp. CAG: 579
Paraprevotella xylaniphila
Enterococcus sp. 5B7_DIV0075
Tyzzerella nexilis
Ruminococcus sp. CAG: 57
Parabacteroides sp. Marseille-P3763
Raoultibacter timonensis
Bacteroides thetaiotaomicron CAG: 40
Eubacterium sp. 41_20
Mobilibacterium timonense
Arabia massiliensis
Bacteroides vulgatus CAG: 6
Lysinibacillus boronitolerans
Ruminococcus sp. CAG: 108-related_41_35
Acinetobacter sp. LCT-H3
Odoribacter splanchnicus CAG: 14
Synergistes jonesii
Olsenella profusa
Synergistes sp. 3_1_syn1
Bilophila sp. 4_1_30
Clostridium sp. CAG: 798
Bacteroides sp. 2_1_33B
Prevotella sp. 885
Intestinibacter bartlettii
Enterorhabdus mucosicola
Bacteroides cellulosilyticus CAG: 158
Enterococcus sp. 3H8_DIV0648
Clostridium sp. CAG: 269
Candida parapsilosis
Clostridium nexile CAG: 348
Weissella sp. DD23
Actinomyces dentalis
Tissierellia bacterium SS-A11
Clostridium sp. ASBs410
Roseburia sp. CAG: 471
Paeniclostridium sordellii
Collinsella stercoris
Bacteroides coprophilus
Bacteroides sp. 3_1_33FAA
Enterococcus gilvus
Acidaminococcus sp. CAG: 542
Alistipes sp. CAG: 29
Prevotella sp. CAG: 617
Leuconostoc lactis
Collinsella sp. An2
Actinomyces sp. oral taxon 175
Listeria monocytogenes
Clostridium sp. 7_2_43FAA
Paraclostridium bifermentans
Enterococcus pallens
Anaerococcus prevotii
Actinomyces viscosus
Bacteroides finegoldii
Clostridium celatum
Bacteroides stercoris CAG: 120
Enterococcus saccharolyticus
Enterococcus malodoratus
Clostridium sp. CAG: 470
Actinomyces odontolyticus
Peptoniphilus duerdenii
Actinomyces sp. ICM58
Ruminococcus sp. 37_24
Alistipes putredinis CAG: 67
Fusobacterium mortiferum
Collinsella phocaeensis
Dialister succinatiphilus
Enterococcus sp. kppr-6
Eubacterium callanderi
Sutterella wadsworthensis CAG: 135
Prevotella lascolaii
Collinsella intestinalis
Actinomyces sp. ICM39
Acinetobacter sp. CIP 101934
Acinetobacter lwoffii
Peptoniphilus sp. BV3AC2
Bifidobacterium bifidum CAG: 234
Bifidobacterium pseudocatenulatum CAG: 263
Lactobacillus rhamnosus
Prevotella sp. CAG: 732
Megamonas funiformis CAG: 377
Megamonas hypermegale
Eubacterium sp. CAG76_36_ 125
Bifidobacterium adolescentis CAG: 119
Roseburia sp. CAG: 197
Bacteroides sp. CAG: 98
Parasutterella excrementihominis CAG: 233
Clostridium sp. CAG: 122
Elusimicrobium sp. An273
Bifidobacterium dentium
Tyzzerella sp. Marseille-P3062
Acidaminococcus intestini
Prevotella sp. CAG: 386
Clostridium sp. M62/1
Sellimonas intestinalis
Clostridium sp. CAG: 780
Clostridium sp. AT4
Roseburia sp. CAG: 380
Ruminococcus sp. CAG: 330
Clostridium sp. CAG: 575
Clostridium sp. CAG: 62
Eubacterium sp. CAG: 603
Akkermansia sp. CAG: 344
Clostridium sp. 44_14
Clostridium sp. CAG: 628
Ruminococcus sp. CAG: 624
Roseburia sp. CAG: 303
Lactobacillus rogosae
Roseburia sp. CAG: 309
Acholeplasma sp. CAG: 878
Shigella sonnei
Megasphaera elsdenii CAG: 570
Fusobacterium sp. CAG: 439
Bifidobacterium pseudolongum
Bacteroides sp. CAG: 770
Bacteroides sp. 14(A)
Anaerovorax odorimutans
Bifidobacterium merycicum
Ruminococcus callidus
Eubacterium sp. CAG: 252
Clostridium sp. CAG: 253
Clostridium sp. ATCC BAA-442
Coprobacillus sp. 28_7
Clostridium sp. SCN 57-10
Ruminococcus sp. CAG: 403
Clostridium sp. 42_12
Sutterella parvirubra
Eubacterium sp. 45_250
Clostridium sp. 29_15
Veillonella dispar
Bacteroides sp. CAG: 530
Flavonifractor sp. An82
Eubacterium ramulus
Clostridium sp. CAG: 75
Shigella flexneri
Coprococcus comes CAG: 19
Clostridium sp. CAG: 813
Coprobacillus sp. CAG: 698
Prevotella stercorea
Gemmiger sp. An50
Clostridium sp. 26_21
Clostridium sp. CAG: 217
Butyrivibrio crossotus
Coprococcus sp. CAG: 782
Prevotella sp. P3-122
Blautia sp. CAG: 52
Roseburia sp. 499
Clostridium sp. CAG: 277
Mitsuokella multacida
Azospirillum sp. 51_20
Ruminococcus sp. DSM 100440
Clostridium sp. CAG: 62_40_43
Butyrivibrio crossotus CAG: 259
Azospirillum sp. CAG: 239
Prevotella sp. CAG: 520
Bacteroides massiliensis
Eubacterium sp. CAG: 248
Eubacterium sp. 36_13
Veillonella atypica
Sutterella sp. 54_7
Dakarella massiliensis
Clostridium ventriculi
Sutterella sp. CAG: 351
Bacteroides acidifaciens
Roseburia sp. CAG: 10041_57
Veillonella sp. DORA_A_3_16_22
Selenomonas ruminantium
Emergencia timonensis
Roseburia sp. CAG: 45
Azospirillum sp. 47_25
Fournierella massiliensis
Blastocystis sp. subtype 1
Prevotella bryantii
Roseburia sp. 831b
Bacteroides sp. CAG: 754
Roseburia sp. 40_7
Roseburia sp. CAG: 100
Bacteroides congonensis
Clostridium sp. CAG: 12237_41
Eubacterium sp. CAG: 38
Azospirillum sp. CAG: 260
Bacteroides ovatus CAG: 22
Veillonella parvula
Clostridium sp. CAG: 265
Coprococcus sp. ART55/1
Parasutterella excrementihominis
Roseburia intestinalis CAG: 13
Roseburia sp. CAG: 50
Akkermansia glycaniphila
Mycoplasma sp. CAG: 611
Eggerthella sp. CAG: 298
Clostridium sp. CAG: 632
Prevotella ruminicola
Drancourtella massiliensis
Lachnoclostridium sp. An14
Eubacterium sp. CAG: 115
Bacteroides sp. CAG: 1076
Coprococcus sp. CAG: 131
Veillonella sp. oral taxon 158
Phascolarctobacterium sp. CAG: 266
Lachnoclostridium edouardi
Prevotella stercorea CAG: 629
Alloprevotella tannerae
Olsenella sp. oral taxon 807
Metabolism
and
Metabolism
repair or mitigation
acquisition and
and
lation
sugars
surfaces
Stress Response
electron
indicates data missing or illegible when filed
In this study, following earlier reports of clinical improvement in terms of sleep (Raghavan et al., 2021a), behavioural pattern (Raghavan et al., 2021b), plasma αSyn (Raghavan et al., 2021a) and serum melatonin increase (Raghavan et al., 2021b), gut dysbiosis has been shown to have a strong correlation with the severity of symptoms in ASD (Grimaldi et al., 2018). We evaluated and compared the gut microbiota of the subjects who were supplemented with AFO-202-derived 1,3-1,6 beta glucan with those who did not take the supplement.
Several studies have reported the differences in the gut microbiota between children with ASD and neurotypical children. Reduced number of bifidobacterial and increased Clostridium spp., Desulfovibrio spp., Sutterella spp., and/or Veillonellacea was reported by Souza et al (2012). Tomova et al (2015) reported a change in Bacteroidetes/Firmicutes ratio and an increase in bifidobacterial numbers after probiotic administration.
An exclusion diet and a 6-week prebiotic intervention demonstrated lower abundance of Bifidobacterium spp. and Veillonellaceae family and higher abundance of Faecalibacterium prausnitzii and Bacteroides spp. (Grimaldi et al., 2018). Faecalibacterium, Ruminococcus, and Bifidobacterium were relatively less abundant, whereas Caloramator, Sarcina, Sutterella ceae, and Enterobacteriaceae were more abundant in children with ASD (De Angelis et al., 2013). In addition, lower abundances of the genera Prevotella, Coprococcus, and unclassified Veillonellaceae have been reported (Kang et al., 2013). Among these bacteria, increased Bacteroidaceae, Prevotellaceae, and Ruminococcaceae and decreased Prevotella copri, Faecalibacterium prausnitzii, and Haemophilus parainfluenzae have been reported (Kang et al., 2018; Oh et al., 2020).
In the present study, in line with these reports, the shift of the gut microbiome was towards a beneficial spectrum in Group 2 (Nichi Glucan) because there was a decrease in Enterobacter, Lactobacillus, Escherichia coli, Akkermansia muciniphila CAG:154, Blautia spp., Coprobacillus sp., several clostridium spp., and Clostridium bolteae CAG:59, with an increase in the abundance of Bacteroides, Prevotella, Faecalibacterium prausnitzii, and Prevotella copri. Desulfovibrio Bacteria which have been reported to be associated with PD (Murros et al., 2021) decreased in the Gr. 2.
In particular, Enterobacteria and E. coli significantly decreased in Group 2 compared to Group 1 after the intervention. Gram-negative enteric bacteria such as the Enterobacter and E. coli secrete the amyloid curli that constitutes 85% of the extracellular matrix of enteric biofilms. The curli has similarities and associations with pathological and immunomodulatory human amyloids such as amyloid-3 implicated in AD, αSyn involved in ASD and PD, and serum amyloid A associated with neuroinflammation (Miller et al., 2021). Curli causes misfolding (Al-Mazidi et al., 2021) and accumulation of the neuronal protein αSyn in the form of insoluble amyloid aggregations, leading to inflammation and neuronal dysfunction that is central to pathogenesis of Lewy-body-associated synucleinopathies, including PD and AD.
Curli-producing bacteria also increase the production and aggregation of the amyloid protein αSyn, which has been shown to propagate in a prion-like fashion from the gut to the brain via the vagus nerve and/or spinal cord, thus culminating in the neurological disorders such as ASD (Al-Mazidi et al., 2021). In this study, the significant decrease in Enterobacter and E-coli will thus be of benefit in these synculeopathies. Before the start of this study, the objective to study αSyn was to understand the effects of the beta glucan supplementation on Synaptic imbalance in presynaptic terminals, observed in ASD.
The study results showed that plasma levels of αSyn increased in Group 2 compared with Group 1 along with improvement in Childhood Autism Rating Scale score and sleep pattern which to our knowledge is the first of its kind intervention producing an observable change in the plasma synuclein levels (Al-Mazidi et al., 2021).
In a study by Ding et al., it was reported that 14 functional properties displayed differences between the ASD and healthy control groups. Four functions, including galactose metabolism, glycosyltransferase activity, glutathione metabolism, and antifolate resistance, were enriched in the ASD group. In another study by Lindefeldt et al, the relative abundance of 26 metabolic pathways was diminished after 3 months on a ketogenic diet in children with epilepsy and 3 became more relative abundant.
The group with most pathways changed was carbohydrate metabolism, showing reduction of fructooligosaccharides (FOS) and raffinose utilization, sucrose utilization, glycogen metabolism, lacto-N-biose I and galacto-N-biose metabolic pathway. The SEED average findings of the present study also reflect this beneficial outcome (
The study of the gut microbiome has offered further new insights wherein Enterobacter increasing curli protein and αSyn deposition in the enteric nervous system having been controlled by the beta glucan food supplement, the increase in plasma αSyn levels point out to the disintegration of the amyloid deposits leading to these αSyn entering the blood stream. Indeed, natural killer (NK) cells have been shown to act as efficient scavengers of abnormal α-Syn aggregates (Earls et al., 2020), and the AFO-202 beta glucan has a proven capability to increase and activate NK cells (Ikewaki et al., 2007), which could be another probable mechanism contributing to the increased αSyn levels in the plasma, apart from positive clinical outcomes in these children with ASD.
This result highlights NK cell's potential as a promising therapeutic strategy for prophylaxis and prevention of brain disorders, and they are likely to be used for such αSyn accumulation and propagation, after relevant research on their specific pathways and variations in their capability. The NK cells have been proven to clear the amyloid deposits peripherally though not macrophages (Raghavan et al., 2021c). In the central nervous system, such a role is played by the microglia (Bartels et al., 2020). βeta-glucans also rejuvenate microglia (Luna et al., 2015) that have been shown to scavenge amyloid deposits in the brain and CNS (Morato Torres et al., 2020), thus proving to be a wholesome therapeutic strategy for neurodevelopmental and neurodegenerative diseases.
Altered α-Syn protein misfolding spreading to anatomically connected regions in a prion-like manner and mediating neurodegenerative diseases such as PD (Terajima et al., 2020) and the increased risk of children with ASD in developing PD at a later stage (Swirski et al., 2014) suggests further research into these converging pathogenic pathways of neurodevelopmental and neurodegenerative diseases is needed as well as suggests that this safety-proven food supplement is a preventive strategy in subjects with ASD against PD.
Other than research into normal and abnormal α-syn being warranted, studying the implications of soluble and insoluble α-syn (Boziki et al., 2020) is important because the proportion of insoluble α-syn that was phosphorylated at Ser129 was reported to be significantly higher in brain tissue from PD patients. In addition, cell lines such as the SH-SYHY neuroblastoma (Boziki et al., 2020) will reveal the correlation of the various α-syn with the severity of symptoms and pathogenesis in these neurodegenerative diseases, apart from helping to develop novel disease-modifying strategies employing such simple nutritional supplementation.
The microbiota reconstituted in a beneficial manner in the present study with AFO-202 beta glucan must be further progressed into research to study the effects of other variants of A. pullulans beta glucan that have been shown to be anti-inflammatory. Such research could lead to mechanistic insight into the molecular pathways from the local immune responses in the gut leading to systemic inflammation and, eventually, to organ-specific autoimmunity of the CS in neuroinflammatory conditions such as MS.
Favourable reconstitution of the gut microbiota after consumption of AFO-202 beta glucan in children with ASD has been demonstrated in this study, apart from the clinical improvement already reported. The decrease in Enterobacteria demonstrates the potential of this beta-glucan supplementation for neurodevelopmental conditions such as ASD as well as neurodegenerative disorders such as PD and AD, with converging pathways of amyloid accumulations and propagation, warranting larger clinical studies and research to recommend this as a routine food supplement or an adjunct to existing therapies for prevention and management of both neurodevelopmental and neurodegenerative diseases.
Autism spectrum disorders (ASDs) are a wide range of disabilities in which the neurosynaptic biomarkers and mechanisms remain elusive. As there are no definite interventional modalities available to improve the behavioural pattern, remedial therapies are the only option and have varying outcomes. Based on our earlier study on the improvement of melatonin in ASD children when supplemented with a biological response modifier beta-glucan food supplement, we have evaluated the childhood autism rating scale (CARS) and alpha-synuclein levels in this randomized, parallel-group, multiple-arm clinical trial. Six subjects with ASD (n=6) Gr. 1 underwent conventional treatment comprising remedial behavioural therapies and L-Carnosine 500 mg per day, and 12 subjects (n=12) Gr. 2 underwent supplementation with the Nichi Glucan food supplement 0.5 g twice daily along with the conventional treatment.
There was a significant decrease in the CARS score in all of the children of the Nichi Glucan Gr.2 compared to the control (p-value=0.034517), by an average of 3 points in the improvement of autism's signs and symptoms, whereas the improvement was very mild or nil in Gr.1. Plasma levels of alpha-synuclein were significantly higher in Gr. 2 (Nichi Glucan) than in the control group Gr. 1 (p-value=0.091701). Improvement of the behavioural pattern CARS score and a correlating alpha-synuclein level, followed by a safe beta-glucan food supplement, warrants further research on other parameters, such as gut-microbiota evaluation, and relevant neuronal biomarkers which is likely to cast light on novel solutions.
Behavioural pattern in children with Autism Spectrum Disorder has been observed to improve following consumption of Beta 1,3-1,6 Glucan food supplement. The CARS score has also shown improved, compared to the control group in this pilot clinical study along with increase of a neuronal marker Alpha-Synuclein which is usually lower in affected children compared to normal age matched controls.
This study was approved by the institutional ethics committee of Kenmax Medical Service Private Limited, Madurai, India and was registered as a randomized, parallel-group, and multiple-arm clinical trial in the Clinical Trials Registry of India (CTRI/2020/10/028322). The caregiver of all the subjects gave their informed consent for inclusion before participation in the study. The study was conducted in accordance with the Declaration of Helsinki.
The subjects enrolled in the study had received a diagnosis of ASD by a developmental paediatrician and were verified by a psychologist using a clinical interview for a behavioural pattern that incorporated CARS.
Eighteen subjects (n=18) with ASD in total were enrolled in this prospective, open-label, pilot clinical trial comprising of two arms. The CONSORT flow diagram is presented as
Arm 1 or Gr. 1: Control: Six subjects with ASD (n=6) underwent conventional treatment comprising remedial behavioural therapies and L-Carnosine 500 mg per day.
Arm 2 or Gr. 2: Treatment arm: 12 subjects (n=12) underwent supplementation with Nichi Glucan (Aureobasidium pullulans strain AFO-202 (also referred to as FO-68 [(accession number) FERM BP-19327]) derived Beta 1,3-1,6 Glucan) food supplement along with conventional treatment. Each subject consumed two sachets (0.5 g each) of Nichi Glucan daily—one sachet with a meal twice daily—for a period of 90 days.
All data were analysed using Excel software statistics package analysis software (Microsoft Office Excel(R)); Student's paired t-tests were also calculated using this package; and p-values<0.05 were considered significant.
During enrolment, six subjects with ASD (n=6) could be enrolled in the control group (Gr. 1), whereas in the treatment group (Gr. 2), one of them dropped out before the start of the study. During the study, four subjects were lost to follow-up: two in Gr. 1 (one dropped out due to social problems in the family, and the other relocated to another city) and two in Gr. 2 (one dropped out due to social problems in the family, and the other relocated to another city). A total of 13 subjects (four in Gr. 1 and nine in Gr. 2) completed the study. One female subject was in both Gr. 1 and Gr. 2. The rest were male.
Only one child exhibited possible mild adverse effects related to increased bowel movements in Gr. 2 for one week after supplementation with Nichi Glucan, which settled on its own. No adverse effects were found in any of the other children.
Among the children in the control group (Gr.1), all four were in the category of severe autism, and their score at baseline ranged from 37 to 52 (mean=42.75±5.76). Among the nine children in Gr.2, two were in the mild-to-moderate category of autism (mean=33.5±2.5), whereas the remaining seven were in the category of severe autism (mean=43.71±4.80).
After the intervention, the mean CARS score in the four children of the control group was 42.5±5.4, while in Gr.2 (Nichi Glucan), the mean of the CARS score in the two children with mild-to-moderate autism was 32.5±0.5. In the remaining seven children, the CARS score after Nichi Glucan intervention had a mean of 40.1±5.96. Thus, there was a significant decrease in the CARS score in all of the children in the Nichi Glucan Gr.2 group compared to the control (p-value=0.034517), with an average of 3 points in the improvement of autism's signs and symptoms, whereas the improvement was very mild or nil in Gr.1 (
Among the various parameters assessed on the CARS, there was visible subjective improvement in the emotional response, including reduction in irritability and anger (88%), sleep improvement (88%), speech characteristics with improvement in finger pointing and monosyllables in 77%, and improved responses to the caregiver in 77% of the children in Nichi Glucan Gr. 2, but these improvements were very mild or nil in Gr.1.
Plasma levels of alpha-synuclein ranged between 0.12 and 20.41 ng/dl (mean=9.73 ng/dl) in the control group and between 0.45 and 41.12 ng/dl (mean=9.39 ng/dl) in the treatment group at baseline. After the intervention, plasma levels of alpha-synuclein increased, with a mean increase in levels of 26.72 ng/dl in the treatment (Nichi Glucan) Gr.2 group compared to the control group Gr. 1 (mean increase=10.56 ng/dl) (p-value=0.091701) (
In this study of 13 subjects, the behavioural pattern evaluated by the CARS score improved in all nine subjects of Gr.2 (Nichi Glucan) (
Further, alpha-synuclein has recently been considered one of the important biomarkers for the diagnosis of autism and ASD, wherein the levels are low compared to age-matched controls (Kadak et al., 2015; Sriwimol et al., 2018; Siddique et al. 2020). In regard to neurodegenerative diseases such as PD, the reports have been varied, with some reporting lower than normal levels and others higher. In a correlating hypothesis of the plasma alpha-synuclein level, between autism and neurodegenerative diseases, it has been proposed that alpha-synuclein aggregation in the neural synapse may lead to lower plasma levels (Sriwimol et al., 2018).
Whether the increase in alpha-synuclein levels in plasma in the ASD patients after Nichi Glucan supplementation is due to regulation/prevention of alpha-synuclein's aggregation in the neural synapse must be investigated because an earlier study on beta-glucan from yeast showed reduction in alpha-synuclein expression on the brain substantia nigra in Parkinson's rat model (Masruroh et al., 2017). However, no single mechanism, intervention, or therapy has proven its ability to regulate alpha-synuclein levels, especially in children with ASD. In our study, which is the first of its kind, the plasma alpha-synuclein levels showed significant increase after Nichi Glucan supplementation, and the levels were in line with those that were reported for children without ASD (Kadak et al., 2015; Sriwimol et al., 2018).
Studies on children with ASD have indicated there is an underlying neuroinflammatory process occurring in different regions of the brain involved in microglial activation, thus resulting in a loss of connections or underconnectivity of neurons and leading to behavioural manifestations (Shah et al., 2009). MCP-1, IL-6, IL-10, and TNF-α have been shown to be expressed in higher levels in children with autism (Shah et al., 2009). Beta-glucan has been proven to reduce the expression of inflammatory and proinflammatory markers, including II-6 and TNF-α (Ikewaki et al., 2007), apart from having a neuroprotective effect by attenuating inflammatory cytokine production through microglia (Alp et al., 2012). This mechanism of counteracting ASD inflammation by Nichi Glucan supplementation deserves further research.
In another study, beta-glucan reduced induced microglia activation and its phagocytosis of synaptic puncta and upregulation of proinflammatory cytokine (TNF-α, IL-1β, and IL-6) mRNA expression apart from promoting Tau signalling and improving cognition and brain function via the gut-brain axis (Shi et al., 2020). The mechanism by which the beta-glucan promoted behavioural improvement in the present study and correlated with the regulation of alpha-synuclein levels needs further in-depth research, not only for ASD but also for neurodegenerative diseases such as AD, PD, and so on, especially with regard to its effects on the gut-microbial ecosystem. The evolving data on the gut-brain axis and gut microbiota indicate there are two major approaches to balancing gut microbiota: probiotic and prebiotic.
Probiotic approaches, such as nutritional probiotics, faecal transplantation, and so on, involve direct administration of the beneficial microorganisms that have to colonise the gut (Peng et al., 2020). However, the gut environment must be conducive for such probiotic supplementation. This is where prebiotic approaches come in, such as Nichi Glucan, which help in regulating the gut-microbial ecosystem and preventing chronic inflammatory status Peng et al., 2020); this must be validated by future studies in terms of the effects of Nichi Glucan and gut microbiota in their relevance to ASD.
The limitation of the study is the limited number of participants, the unequal distribution of genders, and the number of participants between the groups. However, this is only a pilot study, and larger randomized, multi-centric clinical trials are warranted. Nevertheless, the study is significant as it has identified a simple nutritional supplemental intervention based on a naturally derived beta-glucan, the Nichi Glucan, which could stimulate endogenous alpha-synuclein secretion, promote better synaptic regulation, and improve the behaviour symptoms of children with autism. However, the results suggest that the benefits will be considerable when evaluated in terms of social and emotional well-being and alleviation of caregiver stress, which is extremely significant.
Patients with ASD showed improvement in behavioural symptoms and improved levels of plasma alpha-synuclein; thus, this pilot clinical study of nutritional supplementation with an AFO-202 strain of black yeast Aureobasidium pullulans produced the biological response modifier beta-glucan (Nichi Glucan). Evaluation as per the CARS score has also shown significant beneficial effects. Although further validations need to be performed, the study definitely confirms the potential of Nichi Glucan as a simple but effective food supplement to be considered as a routine in children with ASD. Further research on the mechanisms of its action in improving alpha-synuclein levels and balancing the immune system in the context of managing chronic inflammation and gut-microbiota regulation as a prebiotic is likely to improve understanding of other diseases caused by neuroinflammation such as PD and AD.
Comparison of Different Beta Glucans with AFO-202
Alpha-Synuclein Suppression Data from F26S Study
Neuroblastoma cell line (SHSY-5Y Tet-On: SCC291) (MERCK) was suspended in 10% FCS-DMEM/F12 medium and seeded into 96-well microplates to achieve a cell count of 5×104 cells/well. After 24 hours of incubation, the cells were washed with 10% FCS-DMEM/F12 medium and new 10% FCS-DMEM/F12 medium was added.
Then, the different beta glucans (Product 1: Micelle Glucan(R) (gel or liquid type) purchased from RL-JP Co., Japan; Product. 2: Beta-glucan NEW EX (gel or liquid type) purchased from Aureo BIS, Japan; Product 3: Yeast Glucan (capsule, inside powder?) purchased from Shell Life Japan Co., Japan) were added to the medium, and the cells were cultured for 1 day. After incubation, the wells were washed three times with PBS and fixed in 0.25% glutaraldehyde-PBS for 1 hour at room temperature. After fixation, the wells were washed three times with PBS-0.05% Tween (PBS-T). After washing the wells with PBS-T, α-synuclein polyclonal antibody (proteintech Co. 10842-1-AP; x1,000 dilution) was added to the wells and incubated at room temperature for 1 h. Then, 2% BSA-PBS-T was added and left for 1 h (to block non-specific reactions).
After 1 hour, the wells were washed with PBS-T and 50 μL of biotin-labeled anti-rabbit IgG antibody (Cosmo Bio) diluted 5,000-fold was added to the wells and the reaction was carried out for 40 minutes at room temperature. After washing the wells, 50 μL of 10,000-fold diluted peroxidase-labeled streptavidin (Cosmo Bio) was added and the reaction was carried out for 20 minutes at room temperature. After washing the wells again, 50 μL of TMB (Cosmo Bio) was added to the wells, and the reaction was stopped with 0.5 M-HCl for 10 minutes.
After that, the absorbance (OD value) was measured with a microplate reader (450 nm) (Tosoh). Data were expressed as ΩOD value (sample OD value−blank OD value). The expression rate was calculated based on the control (None).
Results are found in Table 10.
indicates data missing or illegible when filed
Decrease in α-synuclein expression was highest in AFO-202 beta glucan. Since these cell lines produced α-synuclein are considered to be abnormal/misfolded and capable of aggregation, their decrease in expression is considered to be a suppression of production of abnormal α-synuclein and hence an advantage.
Sleep problems are reported in 50 to 80% of children with Autism and Autism Spectrum disorders (ASD) [C1]. In adolescents and older children with ASD, sleep problems are higher including delayed sleep onset, shorter sleep duration and daytime sleepiness whereas in younger children bedtime resistance, sleep anxiety, parasomnias and night waking are predominant [C2]. Problems in sleep exacerbate the other features of autism such as tantrums, aggression, self-injury, inattention, hyperactivity, social interactions and repetitive behaviours, adding to the parental stress and the entire family's well-being [C1, C3].
Melatonin, a neurohormone secreted by the pineal gland which regulates circadian rhythms including sleep patterns has been shown to be released at lower levels in individuals with autism and has been shown to have a positive effect on sleep in autism by acting on relieving Anxiety, improving sensory processing, possess anti-nociceptive effects on pain, and also gastro-intestinal dysfunction or gut dysbiosis [C3]. A significant proportion of children with ASD have chronic gastrointestinal problems such as diarrhea and/or constipation, irritable bowel syndrome etc. These GIT symptoms have been related cortisol response to stress and gut dysbiosis induced chronic inflammation in ASD [C2] which in turn has associations with altered melatonin levels in autism [C2]. Thus, melatonin supplementation [C2,C4,C5] is one of the main pharmacological approaches under consideration for ASD.
Clinical studies of supplemental melatonin in ASD children have shown to improve sleep latency and quality [C2,C4,C5] in varying degrees [C3]. It is also to be noted that melatonin, though the side effects have been reported to be minimal, it has been found to be effective mostly in short term treatment of sleep disorders and the positive effects have waned during follow up (6-12 months) in specific clinical studies [C6]. Beta (β)-glucans which are naturally occurring compounds have been shown to have a wide range of biological response modifying beneficial effects in metabolism, anti-cancer as well as in reducing the stress and mental disorders by acting on the immune system related pathways [C7]. An animal study has earlier shown that melatonin levels were upregulated in the blood serum of rats in the presence of rice bran (RB) and Beta (β)-glucan present in a mushroom Sarcodon aspratus (S)'s extracts [C7,C8]. We and other research teams have earlier reported the beneficial effects of Nichi Glucan, a black yeast (Aureobasidium pullulans) AFO-202 derived 1,3-,16 beta glucan in metabolic disorders [C9,C10], cancer [C11,C12] in human clinical studies and as a suggested vaccine adjuvant for COVID-19 [C13]. Herein we undertook to study the effects of Nichi Glucan on sleep pattern and serum melatonin levels of ASD children in this pilot clinical study.
Thirteen children with ASD, four in the control group (Gr.1) and nine in the treatment group (Gr.2) age range 2.5 to 13 years were included in the study. The subjects of Gr.2 consumed 1 gram of Nichi glucan (Aureobasidium pullulans strain AFO-202 (also referred to as FO-68 [(accession number) FERM BP-19327]) derived Beta 1,3-1,6 Glucan) as food supplement along with conventional therapies while Gr. 1 underwent conventional therapies alone for a duration of 90 days. The serum melatonin levels were evaluated before and after the study along with assessment of the subjective parameters in sleep pattern by means of a questionnaire to the caregiver in both the groups.
In the Nichi Glucan supplementation group (Gr. 2), the serum melatonin increased on an average from 238.85 ng/dl pre-intervention to 394.72 ng/dl post-intervention which was greater than the control group (Gr.1). All the children in the Nichi Glucan group (Gr.2) showed improvement in sleep pattern and quality.
Aureobasidium pullulans derived Beta 1,3-1,6 Glucan after 90-days consumption has shown visible improvement in sleep quality, pattern and serum melatonin levels in this first of its kind report in the literature which warrants a larger multicentric study for validation and in-depth research on the mechanisms to recommend this as a routine supplementation in kids with ASD to improve their quality of sleep.
This study was approved by our Institutional ethics committee of Kenmax Medical Service Private Limited, Madurai, India and registered in the Clinical trial registry of India (CTRI/2020/10/028322).
The subjects enrolled in the study had clinical diagnosis of ASD by a developmental paediatrician using standard assessment verified using a clinical interview that incorporated CARS (Childhood Autism Rating Scale).
Eighteen subjects (n=18) with ASD in total were enrolled in this prospective open label pilot clinical trial comprising of two arms,
Arm 1 or Group (Gr.) 1: Control: Six subjects with ASD (n=6) underwent conventional treatment which comprised of remedial behavioural therapies and L-Carnosine 500 mg per day.
Arm 2 or Group (Gr.) 2: Treatment arm: Twelve subjects (n=12) underwent supplementation with Nichi Glucan (Aureobasidium pullulans strain AFO-202 (also referred to as FO-68 [(accession number) FERM BP-19327]) derived Beta 1,3-1,6 Glucan) food supplement along with conventional treatment. The subjects consumed 2 sachets (0.5 g each) of Nichi Glucan, one sachet with a meal twice daily for a period of 90 days.
All data were analysed using Excel software statistics package analysis software (Microsoft Office Excel(R)); Student's paired t-tests were also calculated using this package; P-values<0.05 were considered significant.
During enrolment, six subjects with ASD (n=6) could be enrolled in the control Gr.1 while in treatment group (Gr. 2), one of them dropped out even before start of the study. During the study, three subjects were lost to follow-up, one in Gr.1 (subject relocated to another city) and 2 in Gr. 2 (one due to social problems in the family and other relocated to another city). Totally 13 subjects (4 in Gr.1 and 9 in Gr.2) completed the study. There was one female subject in both Gr.1 and Gr. 2. The rest were male.
On the Children's Sleep Habits related Questionnaire (CSHQ), there was significant reduction in the total score especially in terms of decrease in bedtime resistance and time of onset of sleep in the Gr.2 compared to Gr.1 (Table 11). The total sleep score ranged from 66 to 67 in Gr.1 (Mean=66.25±0.5) in the Gr.1 Control group while it ranged from 62 to 75 in Gr.2 at baseline (Mean=72±5.02) in Gr.2 (Nichi Glucan) at baseline. At the end of the study the total sleep score ranged from 58 to 66 in Gr.1 (Mean=64±4) in the Gr.1 Control group while it ranged from 51 to 70 in Gr.2 (Mean=64.22±7.47) in Gr.2 (Nichi Glucan). The reduction in sleep score after intervention indicating improvement in sleep behaviour was statistically significant in Gr.2 (p value=0.009879) indicating a significant improvement in the sleep patterns of the subject in the Nichi Glucan arm while the difference in sleep score did not show any statistically significant improvement in the control arm (p value=0.153494). The total sleep score also decreased well in the Nichi Glucan group compared to the control (
In the control group (Gr.1), the serum Melatonin increased on an average from 110.585 to only 114.11 post-intervention (
Only one child exhibited possible mild adverse effects related to increased bowel movements in Gr. 2 for one week after supplementation with Nichi Glucan which settled on its own. There were no adverse effects in any of the other children.
In this open-label clinical trial of supplementation with Nichi Glucan, we found that majority of the children in the Nichi Glucan group (Gr.2), 8 out of 9 subjects (88%) had an improvement in sleep pattern and quality of sleep observed by decrease in sleep score after Nichi Glucan supplementation. The serum melatonin increased to a greater extent in Gr. 2 compared to Gr.1. The sleep score significantly decreased in Gr. 2 compared to gr.1 (
There were only minimal adverse effects. This is the first of its kind study, in which a nutritional supplement that is not a pharmacological drug has been able to improve sleep pattern with evidence in laboratory evaluation of corresponding serum melatonin and in children with ASD.
Sleep difficulties are a major problem in children with ASD with 53% having been reported to have difficulty in sleep onset (53%), 40% restless sleep, 34% night-time awakening and 32% difficulty in arousal from sleep [C14]. Lack of good sleep also affects emotional and functioning ability in turn leading to impairment in academic and social functioning and maintaining relationships in these children. Therefore, ensuring good quality sleep becomes an essential part of therapy for ASD. Among pharmacological interventions, melatonin [C2,C4,C5], trazodone, benzodiazepines, and SSRI antidepressants represent the most commonly used medications in the paediatric population [C15]. Melatonin supplementation remains the treatment of choice, given the side effects of other interventions and clinical trials having showed positive outcome of its supplementation [C15]. Nevertheless, there are reports that melatonin is more effective as short term rather than long term though those studies have mostly been in individuals without ASD [C6].
A nutritional supplement which can be simple, easy to administer and has minimal or no adverse effects will be an ideal alternative to melatonin. In the current study, Nichi Glucan which has been in consumption as a food supplement for several decades [C16] with proven benefits in metabolic disorders, cancer etc. [C9-12] has been shown to be a promising strategy based on the current study's results in terms of improvement in quality of sleep and increase in daytime serum melatonin levels.
It has been postulated that low melatonin levels in ASD children could have its etiologic origin in melatonin deficiency in mothers of these children exerting its effects during neurodevelopment in embryo [C17]. Another study has reported the clear correlation between gut microbiome profiles of children with ASD and their mothers suggesting the importance for assessing the microbiome during the early stage in mothers during pregnancy and planning of personalized treatment and prevention of ASD via microbiota modulation [C17]. Beta glucan has also been shown to reduce the underlying chronic inflammation due to gut dysbiosis and helping to modulate towards a healthy microbiome, which will be further advantageous in ASD as chronic inflammation has been shown to be associated with severity of ASD symptoms [C18], Thus, with the current study showing that beta glucans can enhance melatonin and sleep quality in children with ASD, the ability of Beta glucans to modulate gut microbiota and reverse gut dysbiosis as the possible mechanism behind the increase in levels of melatonin [C6,C19,C20], thereby improving sleep, needs further research.
This is only a pilot study and the limitation with the very less ample number is planned to be overcome by additional large-scale studies apart from studying the possible beneficial effects of Nichi Glucan on the behavioural aspects and other symptoms in patients with ASD.
Patients with ASD have shown improvement in quality of sleep and improved levels of serum melatonin, in this open label pilot clinical study of nutritional supplementation with an AFO-202 strain of black yeast Aureobasidium pullulans produced 1,3-1,6 beta Glucan (Nichi-Glucan). The efficacy of Nichi Glucan in terms of behavioural improvement and other parameters observed in this pilot study in children with ASD, when confirmed in a larger study with long-term follow-up, it is worth recommending it as a supplementary food in such children. Further in-depth evaluation of the mechanisms and their correlation with other neurological parameters is recommended, which may throw light on novel solutions and drug candidates from such findings.
The study involved 18 subjects with ASD who were randomly allocated: six subjects in the control group (Group 1) underwent conventional treatment comprising remedial behavioural therapies and L-carnosine 500 mg per day, and 12 subjects (Group 2) underwent supplementation with Nichi Glucan 0.5 g twice daily along with the conventional treatment for 90 days. The subjects' stool samples were collected at baseline and after the intervention.
Whole genome metagenome (WGM) sequencing was performed.
The results are shown in
All the species, R. Hominis, R. intestinalis, R. inulinivorans and R. faecis increased greatly post-intervention in AFO-202 treatment group. R. inulinivorans and R. faecis decreased in the control group. The increase in melatonin and improved sleep reported in the study (doi: 10.21203/rs.3.rs-701988/v1) can be attributed to the increase in abundance of Roseburia.
In 2004, the SEED (http://pubseed.theseed.org/) was created to provide consistent and accurate genome annotations across thousands of genomes and as a platform for discovering and developing de novo annotations. The SEED is a constantly updated integration of genomic data with a genome database, web front end, API and server scripts. It is used by many scientists for predicting gene functions and discovering new pathways
Eighteen subjects with ASD were enrolled in this prospective, open-label, pilot clinical trial comprised of two arms. Arm 1 or Group 1 (control group): Six subjects with ASD underwent conventional treatment comprising remedial behavioural therapies and L-carnosine 500 mg per day. Arm 2 or Group 2 (Nichi Glucan group): 12 subjects underwent supplementation with Nichi Glucan food supplement along with conventional treatment (remedial behavioural therapies and L-carnosine 500 mg per day). Each subject consumed two sachets (0.5 g each) of Nichi Glucan daily—one sachet with a meal twice daily—for 90 days.
Faecal samples were collected at baseline and 90 days after the intervention using a sterile faecal collection kit and the samples were kept at −20° C. until they were transferred to the laboratory and processed. Samples for DNA extraction were stored at −80° C. until needed for analysis.
The samples were then taken for whole genome metagenome analysis. Initially, the reads were filtered for human DNA contamination. The filtered reads were then aligned to bacterial, fungal, viral and archea genomes. De novo assembly was carried out using the pre-processed reads to obtain the scaffolds. These scaffolds were then used for gene prediction. The abundances in terms of SEED annotations were analysed.
The results are shown in
There is several fold decrease in all the gene annotations (metabolites and metabolic functions) in the AFO-202 Nichi Glucan treatment group including Carbohydrates, Fatty acids, lipids, virulence, metabolite damage, nitrogen metabolism, mitochondrial electron transport system etc.
Neuroblastoma cell line (SHSY-5Y Tet-On: SCC291) (MERCK) was suspended in 10% FCS-DMEM/F12 medium and seeded into 96-well microplates to achieve a cell count of 5×104 cells/well. After 24 hours of incubation, the cells were washed with 10% FCS-DMEM/F12 medium and new 10% FCS-DMEM/F12 medium was added.
Then, β-glucan AFO-202 (50 μg/mL) and PMA (500 ng/mL) (Sigma) were added to the medium, and the cells were cultured for 1 to 3 days. After incubation, the wells were washed three times with PBS and fixed in 0.25% glutaraldehyde-PBS for 1 hour at room temperature. After fixation, the wells were washed three times with PBS-0.05% Tween (PBS-T). After washing the wells with PBS-T, 50 μL of 500-fold diluted α-synuclein polyclonal rabbit antibody (proteintech Co. 10842-1-AP) was added to the wells and incubated at room temperature for 1 h. Then, 2% BSA-PBS-T was added and left for 1 h (to block non-specific reactions).
After 1 hour, the wells were washed with PBS-T and 50 μL of biotin-labeled anti-rabbit IgG antibody (Cosmo Bio) diluted 5,000-fold was added to the wells and the reaction was carried out for 40 minutes at room temperature. After washing the wells, 50 μL of 10,000-fold diluted peroxidase-labeled streptavidin (Cosmo Bio) was added and the reaction was carried out for 20 minutes at room temperature. After washing the wells again, 50 μL of TMB (Cosmo Bio) was added to the wells, and the reaction was stopped with 0.5 M-HCl for 10 minutes.
After that, the absorbance (OD value) was measured with a microplate reader (450 nm) (Tosoh). Data were expressed as ΩOD value (sample OD value−blank OD value). The expression rate was calculated based on the control (None).
The results are shown in
The neuroblastoma cell line (SHSY-5Y Tet-On: SCC291) was stimulated with BGs or PMA for 3 days. An α-synuclein polyclonal antibody (proteintech Co. 10842-1-AP) was used in this experiment (glutaraldehyde-fixed cellular ELISA).
Alpha synuclein expression is decreased by AFO-202 in cell lines. Since these cell lines produced ASN are abnormal/misfolded and capable of aggregation, their decrease in expression is considered to be a suppression of production of abnormal ASN and hence an advantage.
Beta Glucans having been able to control ROS and mitochondrial Stress and hence the above suppression of abnormal αSyn could be attributed due to that mechanism.
It is the abnormal ASN causing aggregates which can show transmission from cell to cell and prone to propagation like prions through gut brain axis and therefore AFO-202 making their production lesser at cellular level is helping to address the issue at the root cause itself.
Since AFO-202 can regulate dyslipidemia and because alpha-synuclein binding with oxidized lipid metabolites can lead to mitochondrial dysfunction, leading to neuronal disorders, AFO-202 (i) decreasing misfolded alpha-synuclein production and (ii) regulating lipids has an advantage.
The increase in plasma levels of Alpha-Synuclein in the autism children (http://dx.doi.org/10.1136/bmjno-2021-000203) can be attributed to the clearing of the deposits by NK cells activated by AFO-202 beta glucan.
Capacity of beta glucans to activate microglia will be of advantage in clearing the aggregates in the CNS therefore attributing to the behaviour and sleep pattern improvement of AFO-202 in the autism study (http://dx.doi.org/10.1136/bmjno-2021-000203)
Various modifications and variations of the described glucan products, compositions and methods as well as the concept of the invention will be apparent to those skilled in the art without departing from the scope and spirit of the invention. Although the invention has been described in connection with specific preferred embodiments, it should be understood that the invention as claimed is not intended to be limited to such specific embodiments. Various modifications of the described modes for carrying out the invention which are obvious to those skilled in the chemical, biological, medical, environmental, cosmetic or food arts or related fields are intended to be within the scope of the following claims.
Number | Date | Country | Kind |
---|---|---|---|
2021-065938 | Apr 2021 | JP | national |
2021-076579 | Apr 2021 | JP | national |
2021-174365 | Oct 2021 | JP | national |
This application is a national phase application filed under 35 USC 371 of International Application No. PCT/JP2022/017237, filed Apr. 7, 2022, and claims the benefit of the filing dates of Japanese Application No. 2021-65938, entitled “A COMPOSITION FOR IMPROVING SLEEP PATTERN AND SERUM MELATONIN”, filed Apr. 8, 2021; Japanese Application No. 2021-76579, entitled “A COMPOSITION FOR IMPROVING BEHAVIOURAL PATTERN AND ALPHA-SYNUCLEIN LEVELS”, filed Apr. 28, 2021; and Japanese Application No. 2021-174365, entitled “A COMPOSITION FOR IMPROVING GUT MICROBIOTA”, filed Oct. 26, 2021; the contents of each of which are incorporated herein by reference in their entirety.
Filing Document | Filing Date | Country | Kind |
---|---|---|---|
PCT/JP2022/017237 | 4/7/2022 | WO |