PSRwe treatment improved HFD-induced obesity in mice
Obesity mice model was successfully constructed by HFD-fed for eight weeks. PSRwe and orlistat (a well-known irreversible inhibitor of pancreatic and gastric lipases used for orally for long-term research of obesity [14]) were intragastrically administrated to the obesity mice according to their body weight (BW) once a day for 8 weeks. All the mice were sacrificed to collect samples for multi-omics examinination after the terminal of the experiment (Figure S1A). Both PSRwe and orlistat could significantly reduce the BW of HFD-induced obese mice after two weeks of therapy, and the efficacy of PSRwe was more stable and lasting than orlistat (Figure S1B). No apparent damage for liver, lung, spleen, kidney, stomach and heart of mice during the gavage of PSRwe and orlistat were observed by H&E staining (Figure S1C). Therefore, we have explored the underlying mechanism of PSRwe in treating HFD-induced obesity based on multiomics.
PSRwe treatment altered the gut microbial composition and function in obesity
PCoA analysis based on bray-curtis distance has showed that the composition of the gut microbiota was significantly different between obesity and PSRwe group at genus level (PcoA 1 = 55.6%, Fig. 1A). Shannon and Simpson indexes were used to evaluate the alpha diversity and the results revealed that PSRwe treatment could obviously increase the species diversity and richness of the gut microbiota compared with obesity mice (Fig. 1B). Differential analysis between PSRwe and obesity have identified 10 significant different genera (Fig. 1C), in which the relative abundance (RA) of Akkermansia, Parasutterella, Parabacteroides, Klebsiella, Erysipelatoclostridium, Bacteroides, Alistipes, Ileibacterium were significantly enriched while that of Firmicutes unclassified and Mucispirillum were obviously decreased in PSRwe treatment group compared with obesity mice. 21 significantly changed species were observed between PSRwe and obesity group (Fig. 1D). Among which, Akkermansia muciniphila, Alistipes indistinctus, Bacteroides uniformis, Bacteroides stercorirosoris, Bacteroides intestinalis, Clostridium cocleatum, Erysipelatoclostridium ramosum, Ileibacterium valens, Klebsiella pneumoniae, Klebsiella variicola, Klebsiella quasipneumoniae, Lachnospiraceae bacterium 10 − 1, Lactobacillus reuteri, Lachnospiraceae bacterium COE1, Lactobacillus johnsonii, Parasutterella excrementihominis, Parabacteroides gordonii, Streptococcus acidominimus were significantly enriched while that of Mucispirillum schaedleri and Firmicutes bacterium ASF500 were obviously decreased in the gut of PSRwe group.
Humann 3.0 was used to efficiently and accurately profile the abundance of microbial metabolic pathways. Significant changes of the functional modules were observed between groups (Figure S2A). Pathways including superpathway of glycolysis, pyruvate dehydrogenase, TCA, and glyoxylate bypass, superpathway of glyoxylate bypass and TCA, saturated fatty acid elongation, fatty acid beta oxidation I, TCA cycle IV, gluconeogenesis III, fucose degradation, glycolysis III (from glucose), pentose phosphate pathway, oleate biosynthesis IV (anaerobic), stearate biosynthesis II (bacteria and plants), homolactic fermentation, superpathway of fucose and rhamnose degradation, NAD de novo biosynthesis I (from aspartate) were all significantly enriched in the PSRwe group, which suggested that PSRwe treatment could significantly increase the energy metabolism in obesity mice. We then focused on fatty acid metabolism and two pathways including fatty acid elongation-saturated and superpathway of fatty acid biosynthesis initiation (E. coli) were significantly enriched in PSRwe group (Figure S2B). Two species including Bacteroides stercorirosoris and Klebsiella pneumoniae were found to contribute to fatty acid elongation-saturated pathway. Five species including Bacteroides caecimuris, Bacteroides massiliensis, Clostridium cocleatum, Erysipelatoclostridium ramosum, Parabacteroides goldsteinii were observed to participate in the superpathway of fatty acid biosynthesis initiation (E. coli). It is worth noting that A. muciniphila, a promising probiotic [15], were significantly enriched in the gut of the PSRwe group (Figure S2C). We have found that A. muciniphila is involved in a variety of biological processes such as nucleotides biosynthesis, pyruvate fermentation, folate transformations, amino acids biosynthesis, rhamnose biosynthesis, peptidoglycan biosynthesis, and superpathway of branched chain amino acid biosynthesis.
These results suggested that PSRwe has significantly improved HFD-induced obesity in mice, as well as the gut microbial composition and function.
Colonic DEGs were correlated to the gut microbiota and their metabolites
RNA-seq was performed to depict the differentially expressed genes (DEGs) in the colon of the mice between the PSRwe and obesity group. 184 genes including Odad4, Kat2b, Abl1, Mir1932, Cop1 etc. were up-regulated and 229 genes including Mir702, Igkj1, Tspan11, Dnpep, Vsig10l etc. were down-regulated in the colon of the PSRwe mice compared with the obesity group (Fig. 2A). GO annotation (Fig. 2B) has showed that the up-regulated DEGs were dynamically localized to cellular components (CC) including photoreceptor connecting cilium, condensed chromosome outer kinetochore, cytosolic small ribosomal subunit, kinetochore, complex Sin3 complex, chromosomal region, condensed chromosome kinetochore, ciliary transition zone, polysome, polysomal ribosome, and contributed to molecular function (MF) including pre-mRNA intronic binding, pre-mRNA/mRNA binding, bHLH transcription factor binding, GTPase activator activity, D1 dopamine receptor binding, GTPase regulator activity, mRNA 3-UTR binding, acetylcholine receptor activity, histone acetyltransferase activity. The up-regulated DEGs involved in biological process (BP) mainly include cellular response to cadmium ion, positive regulation of GTPase activity, cellular response to zinc ion, response to cadmium ion, cellular response to copper ion, spinal cord motor neuron differentiation, detoxification, regulation of endothelial cell proliferation. KEGG pathway analysis revealed that serotonergic/cholinergic/glutamatergic synapse, JAK-STAT signaling pathway, PI3K-Akt signaling pathway, FoxO signaling pathway, inflammatory mediator regulation of TRP channels, and antifolate resistance were the main pathways participated by the up-regulated DEGs. For the down-regulated DEGs (Fig. 2C), they were primarily localized to inclusion body, aggresome, mitochondrial inner membrane, cullin-RING ubiquitin ligase complex, organelle inner membrane, SCF ubiquitin ligase complex, ubiquitin ligase complex, mitochondrial respirasome, lytic vacuole, lysosome. The MF involved for these DEGs were mainly centered on retinol dehydrogenase activity, oxidoreductase activity/acting on the CH-OH group of donors/NAD/NADP as acceptor, aldehyde-lyase activity, androsterone dehydrogenase activity, steroid dehydrogenase activity, oxidoreductase activity/acting, protein heterodimerization activity, carbon-carbon lyase activity, GTPase activity, transferase activity/transferring pentosyl groups. The BP involved of these DEGs were principally retinoid/ diterpenoid/ terpenoid/ pyruvate/ isoprenoid metabolic process, glycolytic process, ATP generation from ADP, response to interferon-gamma, carbohydrate catabolic process, ADP metabolic process. KEGG pathway analysis has displayed that ether lipid metabolism, glycolysis/ gluconeogenesis, pentose phosphate pathway, fructose and mannose metabolism, fat digestion and absorption, PPAR signaling pathway, alpha-Linolenic acid metabolism, and apoptosis were the main pathways participated by the down-regulated DEGs. These results revealed significant differences of the colonic gene expression as well as their distinct function between PSRwe treated and untreated obesity mice.
We have also explored the gut microbial metabolites in colon and explored the association between DEGs and different metabolites. Interestingly, 165 significant different metabolites were observed between PSRwe and obesity group. Thereinto, 109 metabolites had significant high levels while 56 metabolites were obviously reduced in the gut of PSRwe treated mice compared with obesity group (Fig. 3A). The increased metabolites in the gut of PSRwe mice were mainly involved in pathways including purine/pyrimidine metabolism, cAMP signaling pathway, biosynthesis of amino acids, arginine and proline/ tyrosine/ phenylalanine/ beta-alanine metabolism, biosynthesis of cofactors, vitamin digestion and absorption, sphingolipid signaling pathway, sphingolipid metabolism, ABC transporters, neuroactive ligand-receptor interaction, morphine addiction and caffeine metabolism, all these pathways were significantly abundant in the PSRwe group (Fugure 3B, 3C). However, the decreased metabolites in PSRwe group such as LPE, LPC, PC, Carnitine C22:2 etc., participated KEGG pathways including amino and nucleotide sugar metabolism, glucagon signaling pathway, cysteine and methionine metabolism, porphyrin and chlorophyll metabolism, all of which were obviously abundant in the obesity mice (Fig. 3B, 3D).
Complex interactions have been known to exist between the colonic gene expression and gut microbiota as well as their metabolites. To unveil that, Spear-man's rank correlation was performed for the significantly different species and DEGs, as well as the evidently different gut microbial metabolites and DEGs between the PSRwe and obesity group (Fig. 4). The down-regulated DEGs involved for the top KEGG pathways were include Gpi1, Lpcat4, Agpat2, Gdpd3, Atf4, Tuba1b, Pla2g7, Aldob, Pla2g3, Acaa1b, Mpi, Mcl1, Fabp2, which were positively associated with Lachnospiraceae bacterium 3 − 2, Firmicutes bacterium ASF500, and Mucispirillum schaedleri, which were obviously decreased in the gut of PSRwe treated mice. The up-regulated DEGs contributed to the predominant KEGG pathways contained Chrm1, Homer2, Cyp2d10, Alox12, Il10, Htr2b, Chrnb2, Ep300, Gngt2, Gnas, Cacna1a, Pik3cd, were positively associated with Akkermansia muciniphila, Bacteroides stercorirosoris, Bacteroides uniformis, Alistipes indistinctus, Parasutterella excrementihominis, Parabacteroides goldsteinii etc., which were all enriched in the gut of the PSRwe treated mice. These results indicated that the gut microbiota could regulate the gene expression in the colon wall.
To further investigate the relationship between the colonic DEGs and the gut microbial metabolites, Spearman’s rank correlation between DEGs and significantly different metabolites were performed (Figure S3). the up-regulated DEGs in PSRwe group including Cacna1a, Htr2b, Ep300, Gngt2, Homer2, Chrm1, Alox12, Il10, Chrnb2, Gnas, Pik3cd were significantly positively correlated with significantly different gut microbial metabolites participated in pathways including organic acid and its derivatives, nucleotide and its metabolomics, heterocyclic compounds, fatty acyl, coEnzyme and vitamins, benzene and substituted derivatives, amino acid and its metabolomics, alcohol and amines. The down-regulated DEGs including Fabp2, Pla2g3, Acaa1b, Mpi, Aldoc, Atf4, Tuba1b, Pla2g7, Aldob, Mcl1, Gpi1, Olr1, Gdpd3, Ldha, Plin2, Lpcat4, Agpat2 were positively correlated with the gut microbial metabolites contributes to pathways including carbohydrates and its metabolites, and glycerophospholipids. The above results revealed that the colonic gene expression could affect the gut microbial metabolites.
In summary, the gut microbiota and their metabolites could regulate the colonic gene expression related to lipid metabolism, fat digestion and absorption, glycolysis, Pentose phosphate pathway, JAK-STAT signaling pathway, FoxO signaling pathway, PPAR signaling pathway to affect energy absorption and metabolism in obesity, which in return, the gene expression might affect the composition and function of the gut microbiota.
Differential analysis of the liver DEGs and metabolites
Liver is the main organ for fatty acids biosynthesis and metabolism; thus, we investigated the differences for liver gene expression and metabolites between PSRwe and obesity group.
RNA-seq was used to study the liver gene expression between PSRwe and obesity group. PcoA analysis has revealed that the liver gene expression was significantly different (PcoA 1, P = 0.00067, Fig. 5A). Differential analysis has unveiled 2791 significant DEGs in the liver between the PSRwe and obesity group. Among them, 2278 DEGs were up-regulated while 513 DEGs were down-regulated in the PSRwe group (Fig. 5B). GO annotation has revealed that the up-regulated DEGs were mainly localized to peroxisome, microbody, peroxisomal membrane, microbody membrane, lytic vacuole, lysosome, cytosolic ribosome, vacuolar membrane, autophagosome membrane, and ribosomal subunit. MF of these DEGs were primarily ATPase activity, oxidoreductase activity/acting on the CH-CH group of donors/NAD or NADP as acceptor, manganese ion binding, nucleoside-diphosphatase activity, structural constituent of ribosome, transferase activity/transferring acyl groups, carboxylic ester hydrolase activity, fatty acid binding, and monocarboxylic acid binding. The BP involved for these DEGs were principally fatty acid catabolic process, fatty acid beta − oxidation, fatty acid metabolic process, monocarboxylic acid catabolic process, lipid and cellular lipid catabolic process, lipid and fatty acid oxidation, small molecule catabolic process, sulfur compound metabolic process. KEGG pathway analysis showed that fatty acid degradation, peroxisome, fatty acid metabolism, PPAR signaling pathway, biosynthesis of unsaturated fatty acids, fatty acid elongation, lysosome, ribosome, other glycan degradation, and valine/leucine and isoleucine degradation were the main pathways contributed by the up-regulated DEGs (Fig. 5C). However, for the down-regulated DEGs, they mainly function at host cellular component, peroxisome, microbody, integral component of organelle membrane, intrinsic and integral component of endoplasmic reticulum membrane. The MF of these DEGs were mainly focused on oxidoreductase activity/acting on paired donors/ with incorporation or reduction of molecular oxygen, iron ion binding, monooxygenase activity, steroid hydroxylase activity, tetrapyrrole and heme binding, estrogen 16-alpha-hydroxylase activity, retinoic acid 4-hydroxylase activity, sulfotransferase activity, aromatase activity. The BP for down-regulated DEGs were majorly sterol/steroid metabolic process, sterol/steroid biosynthesis process, cholesterol biosynthetic and metabolic process, alcohol and secondary alcohol biosynthetic and metabolic process, and organic hydroxy compound biosynthetic process. The KEGG pathways involved for the DEGs were concentrated on chemical carcinogenesis-DNA adducts, steroid hormone and steroid biosynthesis, linoleic acid metabolism, PPAR signaling pathway, terpenoid backbone biosynthesis, retinol metabolism, antigen processing and presentation, cholesterol metabolism, metabolism of xenobiotics by cytochrome P450 (Fig. 5D). Taken together, the up-regulated DEGs were mainly focused on fatty acids biosynthesis and metabolism, and lipid metabolism while the down-regulated DEGs were centered on cholesterol, steroid, and sterol metabolism.
PcoA analysis has revealed that the liver metabolites were significantly different between the PSRwe and obesity group (Fig. 6A). Differential analysis has revealed 131 metabolites significantly enriched in the liver of PSRwe mice and 88 metabolites including carnitine C10:1, carnitine C16:2, etc., which were obviously higher in the liver of obesity mice (Fig. 6B). We have then analyzed the top abundant pathways involved by these different metabolites in PSRwe and obesity group and revealed significant differences between them. Specifically, the top abundant enriched pathways in obesity mice include ABC transporters, purine metabolism, biosynthesis of cofactors, vitamin digestion and absorption, mineral absorption, amino and nucleotide sugar metabolism, D-glutamine and D-glutamate metabolism, inositol phosphate metabolism, starch and sucrose metabolism, fructose and mannose/galactose metabolism, carbohydrate digestion and absorption, drug metabolism-other enzymes, bile secretion, taurine and hypotaurine metabolism, insulin secretion, AMPK signaling pathway, cAMP signaling pathway, cGMP-PKG signaling pathway, FoxO signaling pathway, prolactin signaling pathway, parathyroid hormone synthesis/secretion and action, C-type lectin receptor signaling pathway, sulfur relay system, butirosin and neomycin biosynthesis, lysosome, neuroactive ligand-receptor interaction, taste transduction, olfactory transduction (Fig. 6C). Meanwhile, the top abundant pathways in PSRwe group contained pyrimidine metabolism, protein digestion and absorption, biosynthesis of amino acids (arginine, lysine ), glycine-serine-threonine/ phenylalanine/ tryptophan metabolism, valine, leucine and isoleucine biosynthesis and degradation, glycerophospholipid metabolism, sphingolipid metabolism, glycosylphosphatidylinositol(GPI)-anchor biosynthesis, sphingolipid signaling pathway, ether lipid metabolism, fatty acid biosynthesis and metabolism, steroid hormone biosynthesis, carbon metabolism, 2-oxocarboxylic acid metabolism, pantothenate and CoA biosynthesis, axon regeneration, axon regeneration, and monobactam biosynthesis (Fig. 6D).
The metabolites contribute to the above KEGG pathways were showed in Figure S4 and it is easy to see that most amino acids, sugars, most of LPEs and carnitine isomers, choline were significantly higher in PSRwe group, however, cytosine, adenosine, cAMP, organic acids including ippuric acid, uric acid, maleic acid, epinephrine, sphingosine, FFA, most of LPCs etc. had higher levels in the liver of obesity mice, which were consistent with the metabolic pathways. In summary, the metabolic pathways of the obese mice were mainly focused on active transport and absorption of minerals, sugars, lipids, sterols, peptides, and proteins, hormone secretion such as insulin and parathyroid hormone, bile and taurine and hypotaurine secretion and metabolism, while that of the PSRwe treated mice were mainly focused on amino acids and lipid metabolism.
Gut microbiota affect liver gene expression and its metabolites
To further characterize the differences in lipid and fatty acids metabolism between the PSRwe and obesity groups, 12 pathways including fatty acid biosynthesis/ degradation/ elongation, glycerophospholipid metabolism, glycine, serine and threonine metabolism, peroxisome, arachidonic acid metabolism, PI3K-AKT signaling pathway, FoxO signaling pathway, Jak-STAT signaling pathway, AMPK signaling pathway, and PPAR signaling pathway were precisely analyzed and the genes involved in these pathways were analyzed in detail (Fig. 7). Interestingly, most of the genes participated in these pathways (marked as red in Fig. 7) were significantly highly expressed in liver of the PSRwe treated mice, suggesting that PSRwe treatment could evidently increase the lipid and fatty acids metabolism to alleviate the symptoms of HFD-obesity.
Gut microbiota has been reported to play an important role in obesity, thus we have explored the relationship between the significant gut microbial species and the lipid and fatty acids related DEGs in liver (Fig. 8). Interestingly, the highly expressed DEGs in liver of the obese mice including scd1, scd2, pmvk, pltp, fabp5, cyp7a1, Foxo3, cyp27a1, fabp2, fabp4, elovl6 were positively associated with the obese mice enriched species such as Mucispirillum schaedleri. However, the highly expressed DEGs in liver of PSRwe treated obese mice including stk11, chdh, acaa1b, pias2, ptgr2, cyp4a31, acadm, pck2, pla2g12a, egfr, ppt1, g6pc etc. were positively correlated with species enriched in the gut of PSRwe treated mice including Akkermansia muciniphila, Bifidobacterium pseudolongum, Lactobacillus hominis, Lactobacillus johnsonii, Lactobacillus reuteri, Klebsiella quasipneumoniae, Klebsiella variicola, Klebsiella pneumoniae, Bacteroides uniformis, Bacteroides intestinalis, Parabacteroides gordonii et al.
We have subsequently analyzed the correlation between the significantly different gut microbial species and the different metabolites in liver (Figure S5). It’s worth noting that 5-Aminolevulina, Phosphocholine, O-Acetylcholine, which were highly in the obese mice, were negatively correlated with the obese mice enriched species such as Mucispirillum schaedleri. However, the highly expressed metabolites in the PSRwe mice including betaine, choline, Threonine, L-Serine, L-Aspartate etc. were negatively associated the PSRwe enriched species such as Akkermansia muciniphila, Lactobacillus johnsonii, Lactobacillus reuteri, Lactobacillus hominis, Parabacteroides gordonii, Lachnospiraceae bacterium COE1, Klebsiella pneumoniae, Parasutterella excrementihominis.
These above results suggesting that the gut microbiota could regulate the liver gene expression related to lipid metabolism, which finally changed the liver metabolites such as betaine and choline.
Differential metabolite analysis in plasma, liver tissue and PSR water extract
The plasma metabolites in PSRwe and obesity group were significantly different (Fig. 9A). Totally 212 different metabolites were detected and 58 metabolites were significantly high while154 metabolites were obviously reduced in PSRwe group compared with the obesity mice (Fig. 9B). These different metabolites were mainly involved in pathways highly abundant in PSRwe group such as thyroid hormone signaling pathway, bile secretion, fatty acid biosynthesis, biosynthesis of unsaturated fatty acids, alpha-Linolenic acid metabolism, as well as the significantly abundant pathways in obesity such as glycine, serine and threonine metabolism, tyrosine metabolism, biosynthesis of cofactors, taurine and hypotaurine metabolism, nicotinate and nicotinamide metabolism, pyrimidine metabolism, and neuroactive ligand-receptor interaction (Fig. 9C). Venn diagram was used to illustrate the similarities, differences, and relationship of metabolites among plasma, liver and water extracts of PSR and a total of 42 common metabolites were obtained (Fig. 9D). Interestingly, among these metabolites, seven metabolites including betaine, uridine, 5-Aminoimidazole ribonucleotide, Thymidine, Cytosine, N,N-Dimethylglycine, and 5-Methyluridine were significantly enriched in liver of PSRwe-treated mice compared with obesity mice. However, betaine was the only compound that enriched in the plasma of PSRwe treated mice compared to obesity group (Fig. 9E). Betaine, can be either endogenous or exogenous, was reported to enhances lipid metabolism and improves insulin resistance [16] through gut microbiota-drived microRNA-378a family [17] in HFD-induced obese mice. Exogenous betaine might be primarily derived from PSR water extract and was absorbed into the blood to circulate to liver to affect lipid metabolism while the endogenous betaine was mainly generated from choline under the function of chdh and aldh7a1 in liver. Both choline and betaine were detected in PSR water extract. As choline can be metabolized into betaine in liver, we then analyzed the expression of chdh and aldh7a1 in liver and found that chdh was significantly highly expressed in liver of the PSRwe mice but no significant difference for Aldh7a1(Fig. 9F), which indicated that chdh encoded the necessary enzyme or enzymes to metabolize choline into betaine in liver. Taken together, betaine was significantly higher in liver of the PSRwe mice which might due to either direct absorption from PSR water extract or choline metabolism in liver.