Eligibility criteria for selecting studies and pretreatment of fecal samples
Patients with T2DM, defined according to the 1999 WHO criteria based on oral glucose tolerance test, were recruited from five hospitals in Henan province (Fig. 1 and Supplementary Data 1). All patients were treated with acarbose and have voluntarily signed informed consent before enrollment. Those with one or more of the following characteristics were excluded: severe gastrointestinal disease; history of intestinal surgery or abdominal surgery; participants who had been treated within the last three months with immunosuppressive agents, steroids, antidiarrhea agents, antibiotics, and/or other gastrointestinal motility agents; severe liver dysfunction (serum alanine aminotransferase concentration more than 2.5 times above upper limit of normal range); abnormal renal function (eGFR < 60 ml/min/1.73m2); history or presence of cancer; other severe conditions which would put the patients in high risk during the study. A total of 499 patients were screened for eligibility, in which 393 patients (57.5% male, mean age 57.1) were included in the primary analysis based on exclusion criteria. A flowchart for the inclusion of the participants is shown in Fig. 1. The samples were obtained with informed consent from all participants or their legal guardians. The study was approved by the Ethics Committee of the First Affiliated Hospital of Zhengzhou University (2021-KY-1180-002).
General characteristics of the patients with T2DM who received acarbose treatment are listed in Supplementary Data 1. Groups are mainly based on the levels of blood glucose and HbA1c after acarbose treatment. Patients with acarbose treatment-sensitive (high-response, N = 93) and non-sensitive (low-response, N = 217) were subjected to the analysis of their stool samples. All participants with diabetes have signed an informed consent form. Freshly collected human fecal material from the volunteers (< 10 min from collection, transported on ice) was immediately brought into an anaerobic chamber (75% N2, 10% CO2, 15%H2) (Thermo Forma, Inc., Waltham, MA, USA). Three grams of the sample were suspended in 25 mL of sterile phosphate buffer (8 g/L of NaCl, 0.2 g/L of KCl, 1.44 g/L of Na2HPO4, and 0.24 g/L of KH2HPO4) supplemented with 0.1% l-cysteine (PBSc) in a 50 mL sterile tube. The mixture was left standing still for 5 min to let insoluble particles settle. The supernatant was further mixed with an equal volume of 40% glycerol in PBSc. Aliquots (1 mL) of this suspension were placed in sterile cryogenic vials and frozen at − 80 ℃ for the following experiments15.
Evaluation of acarbose degradation by the fecal microbiome from diabetics
The ex vivo culturing of the gut microbiome from patient samples was carried out as described previously15. All the manipulations were carried out in an anaerobic chamber (Thermo Forma, Inc., Waltham, MA, USA). Briefly, 200 µL of each glycerol stock from the abovementioned fecal samples was inoculated into 5 mL of modified GAM Broth Medium (mGAM) for inoculum preparation. After 12 h of cultivation at 37 ℃, 200 µL of inoculum was transferred into 5 mL of mGAM medium supplemented with acarbose (1.8 mg/mL). The culture was incubated at 37 ℃ for 24 h and then centrifuged at 14,000 g for 10 min at 4 ℃. The obtained supernatant was used for HPLC analysis to determine the residual acarbose amount.
HPLC analysis of acarbose and other sugars
The concentration of acarbose was determined using high-performance liquid chromatography (HPLC) (Agilent 1260 infinity HPLC system) with a ZORBAX SB-C18 column (particle size, 5 µm; 250 mm × 4.6 mm) (Agilent Technologies, Santa Clara, USA) and a UV diode array detector (Agilent Technologies, Santa Clara, USA). The analysis was carried out under the following conditions: the column temperature was maintained at 50 ℃. Solution A (H2O) and solution B (methanol) served as the mobile phases to form a gradient as follows: 3–50% solution B for 0–25 min, 50–3% solution B for 25–25.1 min, and 3% solution B for 25.1–30 min. The flow rate was 1 mL/min. The injection volume was 10 µl. The UV detection wave length was 210 nm. The concentrations of samples were calculated according to peak area-based calibration curves of various concentrations of acarbose standards.
The concentrations of glucose, xylose, and fructose were determined using a high-performance liquid chromatography (HPLC) (Agilent 1100 series) equipped with a Bio-Rad Aminex HPx-87H column (1, 300 mm × 7.8 mm) and a refractive index detector. The analysis was carried out with 5 mM H2SO4 as the mobile phase at a rate of 0.6 mL/min. The column temperature was set to 35 ℃.
Enrichment cultivation and isolation of acarbose-degrading strains
A fecal sample from an acarbose-resistant patient was pretreated as described above. All the manipulations were carried out in an anaerobic chamber (Thermo Forma, Inc., Waltham, MA, USA). Briefly, 200 µL of glycerol stock from the abovementioned fecal samples was transferred into mGAM medium containing 0.01 mg/mL acarbose. The culture was incubated at 37 ℃ for 12 h and then passaged twice by a 1: 20 dilution into fresh mGAM medium containing 0.01 mg/mL acarbose. Each successive passage was incubated for 24 hours. Next, the culture was transferred into mGAM medium containing 0.1, 1, 10, and 20 mg/mL acarbose in the proper order and handled following the same steps as above described. These final enrichment cultures were diluted to different concentrations and streaked onto mGAM agar plates. After 24 h of incubation at 37 ℃, a certain number of colonies appeared on agar plates and were then picked separately and inoculated into the liquid mGAM medium containing 10 mg/mL acarbose. After 24 h of incubation at 37 ℃, the cultures were centrifuged at 14,000 g for 10 min at 4 ℃. The obtained supernatanta were used for HPLC analysis to determine acarbose concentrations.
Once acarbose in the medium was found to be consumed, the corresponding colonies that displayed activity were picked out for sequencing (accession number: PRJNA801205). Here, we identified Klebsiella grimontii TD1 as an active acarbose-degrading strain in the human gut microbiome. This strain was inoculated into liquid mGAM medium (containing 1.5 mg/mL of acarbose) to ensure its acarbose degradation ability. Simultaneously, the grown K. grimontii TD1 cells were harvested by centrifugation at 4,000 g for 10 min at 4 ℃ and then stored at ‒20 ℃ for the following experiments.
Animal studies
Four-week-old male C57BL/6J mice were purchased from Charles River Laboratories China (Beijing, China). The animals were maintained under controlled temperature, humidity, and lighting conditions (12-hour light: 12-hour darkness cycle) with free access to food and water. All experiments were performed according to the ethical guidelines of the Institute of Neuroscience, Chinese Academy of Sciences (ethical review approval number: NA-041-2021-R1). The establishment of high-fat diet (HFD)/Streptozotocin (STZ)-induced T2DM mice was performed based on the previous method21. After one-week acclimatization period, the mice were randomly divided into two groups: (i) NC (n = 9), the normal control fed a standard chow diet; (ii) HFD (n = 60), the high-fat diet control fed a diet with 60 kcal% Fat (Research diet, New Brunswick, NJ, USA; D12492). After 12 weeks of treatment, the mice of the HFD group were fasted overnight and intraperitoneally injected with a single dose (40 mg/kg body weight) of freshly prepared STZ (Cayman Chemical Co., USA) solution for five consecutive days. After two weeks, the fasting blood glucose (FBG) was assayed by tail vein blood sampling using a blood glucose Accu-Chek Performa (Roche Diagnostics). The mice with FBG > 14 mM were selected as T2DM and divided into four groups: PBS, acarbose, K. grimontii TD1, and K. grimontii TD1 + acarbose groups. The PBS group (T2DM + PBS, n = 11) was gavaged with PBS solutions; the acarbose group (T2DM + acarbose, n = 11) was treated with 50 mg/kg/day acarbose (Sinopharm Chemical Reagent Beijing Co. Ltd) by gavage; the K. grimontii TD1 group (T2DM + K. grimontii TD1, n = 10) was gavaged with 1×1011 CFU viable cell of K. grimontii TD1; the acarbose + K. grimontii TD1 group (T2DM + acarbose + K. grimontii TD1, n = 10) was gavaged with 50 mg/kg/day acarbose as well as 1×1011 CFU K. grimontii TD1. All the mice were gavaged once per day for two weeks. On the fourteenth day, all the mice in the four groups were fasted overnight and gavaged as abovementioned in the next morning. Additionally, a starch solution (1 g/kg) was gavaged to all the mice at the same time. The tail vein blood glucose at 0, 60, 120, 180, and 240-min post-gavage was assayed with the blood glucose Accu-Chek Performa (Roche Diagnostics). Then, the incremental glucose levels at different time points were calculated. The areas under the glucose response curve (AUCs) were calculated using GraphPad Software (GraphPad Prism version 7.0).
RNA-Seq and data analysis
The K. grimontii TD1 strain was grown in MS medium (34.0 g/L Na2HPO4, 15.0 g/L KH2PO4, 2.5 g/L NaCl, 5.0 g/L NH4Cl, 2 ml 1M MgSO4, 20 ml 20% starch, 0.1 ml 1M CaCl2) with (2 mg/mL) or without the supplementation of acarbose. The cells at the logarithmic phase of growth were harvested by the centrifugation at 4,000 g for 10 min at 4 ℃ and then frozen immediately in liquid nitrogen.
Total RNA was extracted using the TRIzol® Reagent kit (R0016, Beyotime, Shanghai) according to the manufacturer’s protocol and the residual DNA was eliminated using DNase I (Takara). RNA concentration was determined using a NanoDrop spectrophotometer (Thermo Fisher Scientific, Waltham, MA). High-quality RNA samples were used to construct sequencing libraries.
RNA-seq transcriptome library was established using the TruSeq™ RNA sample preparation Kit from Illumina (San Diego, CA) according to the manufacturer’s instructions using 2 µg of total RNA. Briefly, ribosomal RNA (rRNA) depletion instead of poly adenylation selection is adopted to enrich RNA suing a Ribo-Zero Magnetic kit (epicenter). The obtained mRNAs were broken into short fragments (200 nt). Then, cDNA was synthesized via reverse transcriptase using a SuperScript double-stranded cDNA synthesis kit (Invitrogen, CA) with random hexamer primers (Illumina). The second strand of cDNA was generated by incorporating deoxyuridine triphosphate (dUTP) in place of deoxythymidine triphosphate (dTTP), aiming to create blunt-ended cDNA. The yielding double-stranded cDNA was subjected to end-repair, phosphorylation, and 3′ adenylation and adapter ligation in sequential. The second strand of cDNA with dUTP was degraded using UNG enzyme (Uracil-N-Glycosylase). The cDNA fragments of 200 bp on 2% Low Range Ultra Agarose were purified and then used for PCR amplification using Phusion DNA polymerase (NEB) for 15 reaction cycles. After quantified using a micro fluorometer (TBS-380, TurnerBioSystems, USA), the library was sequenced on the Illumina HiSeq × Ten sequencer using paired-end sequencing. Processing of original images for sequences base-calling and calculation of quality value were performed using the Illumina data processing pipeline (version 1.6), thereby generating 150-bp paired-end reads. A Perl program was written to filter low-quality sequences and reads with more than 5% of N bases (unknown bases) or containing adaptor sequences.
The generated raw data were used for bioinformatics analysis. All the analyses were performed using the free online platform of Majorbio Cloud Platform (www.majorbio.com) from Shanghai Majorbio Bio-pharm Technology Co., Ltd. The software and parameters are the same as previously described22.
qRT-PCR was conducted to verify RNA-Seq data,. Briefly, grown cells after 24 h cultivation were harvested, frozen quickly in liquid nitrogen, and then ground into powder. RNA samples were extracted with the Ultrapure RNA Kit (SparkJade, Shanghai, China), followed by digestion with DNase I (SparkJade, Shanghai, China) to remove residual chromosomal DNA. qRT-PCR was carried out in a MyiQ2 thermal cycler (Bio-Rad, USA) using a SYBR Green PCR premix kit (SparkJade, Shanghai, China). The relative transcript levels of target genes were normalized to gapA (L5654_11535, an internal control), and the relative fold changes in transcription (tested strains versus control strains) were determined as previously described23. The experiments were conducted with three independent RNA samples (biological replicates). The primers used are listed in Supplementary Table 5.
Fast protein liquid chromatography (FPLC) and liquid chromatography–tandem mass spectrometry (LC-MS/MS) analyses
FPLC was used to purify target enzymes. First, 250 µL of the frozen stock of K. grimontii TD1 was inoculated into 300 mL of liquid mGAM medium and grown at 37 ℃ for 12 h. The grown cells were harvested by centrifugation at 4,000 g for 5 min at 4 ℃ and resuspended in 10 mL of buffer (50 mM Tris-HCl, 100 mM NaCl, 10% glycerol, pH 7.9). Resuspended cells were lysed through a cell disruptor (French Press, Constant Systems Limited, UK). The lysates were then clarified by centrifugation (14,000 g for 1 h at 4 ℃) and the supernatant (400 µL) was subjected to protein purification using FPLC (AKTA PURE,GE healthcare, UK) with an anion column (Hitrap™ Q HP, GE healthcare, UK). Solution A (50 mM Tris-HCl, 50 mM NaCl, 1% glycerol, pH 7.9) and solution B (50 mM Tris-HCl, 1 M NaCl, 1% glycerol, pH 7.9) were used as the mobile phases to form a gradient as follows: 0–100% solution B for 0–30 min at a flow rate of 3 mL/min. Samples were collected every two minutes for the measurement of acarbose-degrading activity.
The fractions (Q16-Q18) with acarbose-degrading activities were subjected to FPLC for another round of purification using a cation column (Hitrap™ SP HP, GE healthcare, UK) via the same procedure as above described, yielding further subdivided active fractions (H27-H31).
Finally, the fractions H27-H31 were concentrated 5-fold using a spin concentrator with a 10 kDa cutoff (4, 000 g for 1 h at 4 ℃) and then subjected to FPLC for size-exclusion chromatography using a molecular-sieve column (Hiload 16/600 Superdex 200, GE healthcare, UK). The fraction was eluted using a mobile phase (pH 8.0, 50 mM Tris-HCl, 100 mM NaCl, and 1% glycerol) and a flow rate of 0.3 mL/min. Samples were collected every 0.5 mL outflow and subjected to the measurement of acarbose-degrading activity. Next, all the samples were run on SDS-PAGE, and the gel bands that only occurred in the samples with acarbose-degrading activity were cut out and purified for LC-MS/MS analysis, aiming to confirm protein identities within the bands and assess the presence of the target enzymes for acarbose degradation (Supplementary Table 6). The analyses were carried out by Applied Protein Technology (Shanghai, China) on a high pH reverse-phase high-pressure liquid chromatography (HPLC) (Thermo Scientific, Vilnius, Lithuania) followed by a Q Exactive mass spectrometer (Thermo Scientific, Vilnius, Lithuania) as previously described24. The UniProt database of Klebsiella grimontii (https://www.ncbi.nlm.nih.gov/bioproject/PRJNA605147) was searched using Mascot (v.2.3.02, Matrix Science).
Construction of plasmids
All the primers, plasmids, and strains used in this study are listed in Supplementary Tables 5 and 6, respectively.
The vector expressing the gene (L5654_01665) encoding the acarbose hydrolase from K. grimontii TD1 in E. coli DH5α was constructed as follows. The DNA fragment of the gene was obtained by PCR amplification using the K. grimontii genomic DNA as the template and the primers L5654_01665-f/ L5654_01665-r. The PCR product was analyzed by agarose gel electrophoresis and the target band was recovered using a DNA gel recovery kit (Axygen Biotechnology Company Limited, Hangzhou, China). The yielding DNA fragment was linked with the linear plasmid pET28a vector (digested with NdeI and XhoI) using a Hieff Clone Plus One Cloning Kit (10912ES10, Yeasen, China), generating the target plasmid pET28a- L5654_01665. The constructed plasmid was checked by DNA sequencing.
The plasmids expressing the other genes (Supplementary Table 6) from K. grimontii TD1 were constructed with the same steps except for the primers used in PCR amplification.
Protein production and purification
The plasmids harboring the coding sequences of the target proteins were transformed into E. coli BL21 strain (DE3) for protein production. All the manipulations were carried out in an anaerobic chamber (Thermo Forma, Inc., Waltham, MA, USA). Briefly, transformants were grown on agar plates (LB medium containing 50 µg/mL of kanamycin) at 37 ℃. Colonies occurred on agar plates were then picked out and inoculated into liquid LB media (containing 50 µg/mL kanamycin) for cultivation at 37 ℃ for 12 h. Grown cells (2 mL) were inoculated into 200 mL of liquid LB medium (containing 50 µg/mL of kanamycin) for further cultivation at 37 ℃. When their biomass (OD600) reached ~ 0.8, protein expression was induced at 16 ℃ with the supplementation of 1 mM IPTG. After 16 h of incubation, E. coli cells were harvested by centrifugation at 5,000 g for 10 min at 4 ℃, and then resuspended in buffer (50 mM Tris-HCl, 100 mM NaCl, 10% glycerol, pH 7.9). Cells were lysed using a cell disruptor (French Press, Constant Systems Limited, UK), and the lysate was clarified by centrifugation at 12,000 g for 1 h at 4 ℃. The supernatant was loaded onto a Ni2+ Sepharose™ 6 fast flow agarose column (GE Healthcare, Waukesha, WI, USA) for purification. The column was then washed with 150 mL protein solution A (20 mM Tris-HCl, pH 7.9, 10% glycerol, 500 mM KCl, 10 mM imidazole) followed by 150 mL protein solution B (20 mM Tris-HCl, pH 7.9, 10% glycerol, 500 mM KCl, 25 mM imidazole) and 300 mL protein solution C (20 mM Tris-HCl, pH 7.9, 10% glycerol, 500 mM KCl, 50 mM imidazole). The elute fractions from solution C (containing target proteins) were applied to an Amicon Ultra 15 Centrifugal Filter (Milipore Billerica MA) for desalting and imidazole removal using a buffer containing 50 mM Tris-HCl (pH 7.9), 100 mM NaCl, 10% (v/v) glycerol, and 3 mM DTT. The purified protein was stored in 50% (w/v) glycerol at ‒80 ℃.
Identification of degradation products from acarbose
The identities of the degradation products from acarbose were determined as described below by using a LC-MS spectrometer detector (Q Exactive, Thermo Fisher Scientific, USA). Briefly, a mixture that contained 0.00087 µM of purified Apg (acarbose hydrolase), 2 mg/mL acarbose, and 1 mL of buffer (50 mM Tris-HCl, 100 mM NaCl, 10% glycerol, pH 7.9) was incubated at 37 ℃ for 30 min. The control was the same mixture except that the heat-inactivated Apg protein. The reaction was stopped by heating at 100 ℃ for 5 min.
Next, samples were analyzed by a Q Exactive quadrupole orbitrap high resolution mass spectrometry coupled with a Dionex Ultimate 3000 RSLC (HPG) ultra-performance liquid chromatography (UPLC-Q-Orbitrap-HRMS) system (Thermo Fisher Scientific), with a HESI ionization source. In brief, samples (the injection volume was 2 µL) were firstly separated with an ACQUITY UPLC HSS T3 column (100 mm × 2.1 mm, 1.8-µm particle size; Waters). The mobile phase consisted of solution A (2 mM ammonium formate and 0.1% formic acid in water) and solution B (acetonitrile and methanol, 1:1). The gradient elution was set as follows: 0–0.5 min, 1% B; 0.5–15 min, 1–30% B; 15–20 min, 30%‒90% B; 20–25 min, 90% B; 25.1–30 min, 1% B. The flow rate was 0.3 mL/min and column temperature was 45 ℃. All MS experiments were performed in positive and negative ion modes using a heated ESI source. The source and ion transfer parameters applied were as followed: spray voltage 3.5 kV (positive) and 2.8 kV (negative). For the ionization mode, the sheath gas, aux gas, capillary temperature, and heater temperature were maintained at 40, 10 (arbitrary units), 275 ℃ and 350 ℃, respectively. The S-Lens RF level was set at 50. The Orbitrap mass analyzer was operated at a resolving power of 70,000 in full-scan mode (scan range: 200‒1200 m/z; automatic gain control (AGC) target: 1e6) and of 175.00 in the Top 3 data-dependent MS2 mode (stepped normalized collision energy: 10, 20, and 30; injection time: 50 ms; isolation window: 4 m/z; AGC target: 1e5) with a dynamic exclusion setting of 5.0 seconds.
Determination of the inhibition of acarbose, acarviosine-glucose, and acarviosine on α-amylase activity
The inhibitory effect of acarbose on α-amylase was determined as previously described25. Briefly, a mixture (50 µL) containing 5 µL porcine pancreatic α-amylase (50 U/ml) (10080-25G, Sigma Aldrich, USA), 100 mM acarbose, 100 mM acarviosine-glucose, and 100 mM acarviosine was pre-incubated at 20 ℃ for 10 min, followed by addition of 100 µL of 0.2% soluble starch (dissolved in 100 mM phosphate buffer, pH 6.9) and incubation at 20 ℃ for 30 min. The reaction was terminated by adding 20 µL of 1 M HCl. The residual starch in the reaction mixture was determined as follows: 100 µL of the reaction mixture was mixed with 25 µL of Lugol’s iodine solution (G1069, solarbio, China). The absorption of the mixture at 630 nm was measured using a microplate reader (CLS3922-100EA, Corning, USA). The concentration of starch was calculated by comparing the absorption of the samples to the standard curve for soluble starch. Each assay was repeated in triplicate.
Apg enzyme assays and kinetics
All the assays were performed in an anaerobic chamber (Thermo Forma, Inc., Waltham, MA, USA). Briefly, the buffer contained 50 mM Tris-HCl (pH 7.9), 100 mM NaCl, 10% (v/v) glycerol, and 3 mM DTT. The reaction mixture (200 µL) containing 0.00087 µM Apg, 2 mg/mL of substrates (acarbose, maltopentaose, maltotriose, maltotetraose, starch, maltose, or isomaltotriose), and buffer was incubated at 37 ℃ for 10 min. The reaction was terminated by heating at 100 ℃ for 5 min. The reaction mixture was then centrifuged at 14,000 g for 10 min at 4 ℃, and the supernatant was used to determine glucose concentration. Each assay was repeated in triplicate.
The concentration of the purified Apg protein was determined using the Bio-Rad assay reagent (Bio-Rad Laboratories, Hercules, CA, USA) with bovine serum albumin as a standard. One unit of Apg activity (U) was defined as the amount of enzyme that catalyzes the formation of 1.0 µmol of glucose per minute under the given experimental conditions.
To determine the Km and kcat values of Apg for acarbose under anaerobic conditions, this enzyme was tested at a concentration ([E]) of 0.00087 µM with various concentrations ([S]) of the substrates (acarbose) in the reaction system. The reaction mixture (200 µL) that contained 50 mM Tris-HCl (pH 7.9), 100 mM NaCl, 10% (v/v) glycerol, 3 mM DTT, 0.00087 µM purified Apg, and different concentrations of acarbose (0.09, 0.15, 0.22, 0.31. 0.46, 0.93, 1.55, 2.79, 3.72, 4.65, 5.27, and 6.20 µM) was incubated at 37 ℃ for 5 min. The reaction was terminated by heating at 100 ℃ for 5 min. The reaction mixture was then centrifuged at 14,000 g for 10 min at 4 ℃. The supernatant was used to determine acarbose consumption by HPLC. Initial rates of substrate consumption (V) were obtained by linear regression of the data points within the initial reaction time (0–5 min) (the substrate consumption versus time). The observed rate constant (kobs) was calculated according to the equation (kobs = V/[E]), and then fit to the equation, kobs = kcat* [S]/(Km + [S]), in Graphpad Prism, yielding Km and kcat values.
Phylogenetic analysis of Apg homologs
The protein sequence of Apg was used as the query for a BLASTp search against the non-redundant protein database at NCBI. The resulting Apg homologues with identity no less than 80% were selected with the removal of duplicates and unclassified entries according to their descriptions. For a better visualization, at most five Apg homologues within a genus were adopted. The chosen protein sequences were aligned using the MUSCLE algorithm in MEGA1126 with the default setting. A maximum likelihood phylogenetic tree was generated from this alignment in MEGA11 with the default setting and tested by bootstrapping 100 times. At last, the taxonomic information was highlighted with different colors manually.
Assessment of the prevalence and abundance of Apg+ bacteria in human gut microbiome
The protein sequence of Apg was used as the query for a BLASTp search against the non-redundant protein database at NCBI to obtain its homologues. Taxa containing Apg homologue genes (identity ≥ 85%, 42 in total) were selected as Apg+. Meanwhile, the taxonomic abundances of healthy human and type 2 diabetes patient intestinal bacterial metagenome samples and their meta-information were downloaded from GMrepo (https://gmrepo.humangut.info/home) with the phenotype ID of D006262 and D003924. We only kept the samples generated by metagenomics but not amplicon, QC passed, and not using antibiotic recently. For the metagenomic samples generated in this study, we annotated and summarized the abundances of all the genes belonging to a specific species. The summarized abundance of Apg+ bacteria was calculated as the summation of all Apg+ taxa abundances in a sample. Samples with a summarized abundance greater than 95% of all the samples were considered as technical outliers and excluded. A sample presenting more than 0.01% of the summarized abundance was defined as Apg+.
DNA extraction, library construction, and metagenomic sequencing
The E.Z.N.A.® Soil DNA Kit (Omega Bio-Tek, Norcross, GA, U.S.) was adopted to extract genomic DNA from patient stool samples according to the manufacturer’s method. The concentration and purity of the extracted DNA were evaluated using a micro fluorometer (TBS-380, TurnerBioSystems, USA) and NanoDrop spectrophotometer (NanoDrop 2000, Thermo Fisher Scientific, Waltham, MA), respectively. The quality of the extracted DNA was examined by visualizing the DNA using agarose gel electrophoresis (1% agarose gel).
Then, the extracted DNA was fragmented to an average size of ~ 400 bp by sonication with a Covaris M220 sonicator (Gene Company Limited, China). The paired-end library was constructed by using the NEXTflexTM Rapid DNA-Seq Library Prep Kit (Bioo Scientific, Austin, TX, USA). Adapters containing the full complement of sequencing primer hybridization sites were ligated to blunt-ends of fragments. Paired-end sequencing was performed on Illumina NovaSeq/Hiseq Xten (Illumina Inc., San Diego, CA, USA) at Majorbio Bio-Pharm Technology Co., Ltd. (Shanghai, China) using NovaSeq Reagent Kits/HiSeq X Reagent Kits according to the manufacturer’s instructions (www.illumina.com).
Raw data processing and bioinformatics analysis
The raw reads from metagenome sequencing were cleaned by trimming adaptor sequences and removing low-quality sequences using the fastp27 (https://github.com/OpenGene/fastp, version 0.20.0) on the Majorbio Cloud Platform (cloud.majorbio.com). The remaining clean reads were mapped to the human hg38 reference genome using BWA28 (http://bio-bwa.sourceforge.net, version 0.7.9a) to identify and remove the human host originated reads. These high-quality reads were then assembled to contigs using MEGAHIT29. Contigs with the length being or over 300 bp were selected as the final assembling result.
Bioinformatics analysis of metagenomic data was performed as previously described30–32, including the construction of non-redundant gene sets, functional enrichment analysis, and species annotation under different taxa33. Briefly, the clean reads after quality control were mapped to representative gene sequences (95% identity) using SOAPaligner. Based on the NCBI NR database, we annotated gene sets for viruses, bacteria, fungi, protozoa, and archaea using Diamond (version 0.8.35). In the generated unified database, each gene is assigned to a highest-scoring taxonomy, which facilitates simultaneous assessment of these microbial species in the gut ecosystem of patients with T2DM. One-way analysis of variance (ANOVA) was used to analyze the diversity parameters (ACE and Simpson) of the gut microbiota of diabetes at different response acarbose-treatment (high-response group and low-response group). T-test was performed to analyze the bacteria with the highest abundance (phylum and species) in the gut of different response acarbose-treatment.
Molecular modeling of Apg
The enzyme-ligand complex 3D structure was processed using Protein Preparation in Schrödinger/2020-4 (Schrödinger, LLC, New York, NY), including assigning bond orders using CCD database, adding hydrogens, creating zero-order bonds to metals, creating disulfide bonds, optimizing H-bond assignment, and restrained minimization. Then, a docking grid was defined around the centroid of the ligand with a similar size of the ligand using Glide in Schrödinger/2020-4. Meanwhile, 3D structures of the small molecules of interest (i.e., acarbose and its derivatives, and other potential substrates of Apg) were processed by LigPrep in Schrödinger/2020-4, retaining the neutralized form and specified chiralities. Finally, each small molecule was docked into the catalytic site of the enzyme using Glide in Schrödinger/2020-4 and scored by the XP mode with expanded sampling, except that the conformation of acarbose binding to human α-amylase was extracted from PDB 1XCX. The docking scores in the result files were taken as estimations of ΔG (kcal/mol)
Data availability
Data supporting the findings of this study are available within the paper and the Supplementary Information files. K. grimontii TD1 genome sequencing raw data is available through NCBI-Genome associated with NCBI-Bioproject accession PRJNA801205. All RNA-seq raw data is available through NCBI-SRA associated with NCBI-Bioproject accession PRJNA801096. Datasets and strains generated and analyzed in the study are available from the corresponding author upon reasonable requests. A reporting summary for this Article is available as a Supplementary Information file.
Code availability
The amino acid sequences of enzymes (containing EC numbers) for SSN analysis were downloaded from KEGG. The generated source code is available at https://github.com/lovingstudy/Apg_gut.