Effect of β 1,3/1,6 glucan on gut microbiota of yellow feathered broilers

Similar documents
Symptomspesifikke probiotikum. Linda Mulder, MSc. Vårseminaret 2014

FEEDING THE DAIRY COW DURING LACTATION

Probiotics for the Treatment of Adult Gastrointestinal Disorders

NUTRITION OF THE BODY

LARGE BREED COMPLETE DOG FOOD

The impact of antibiotics on the gut microbiota

Effect of feeding Bacillus-based probiotic on growth performance and health of broilers. Dr. Girish Channarayapatna Evonik Industries, Singapore

National Food Safety Standard Standard for nutrition labelling of prepackaged foods

Introduction. Introduction Nutritional Requirements. Six Major Classes of Nutrients. Water 12/1/2011. Regional Hay School -- Bolivar, MO 1

THE DIGESTIVE SYSTEM

Nutrition Education Competencies Aligned with the California Health Education Content Standards

Endocrine System: Practice Questions #1

PROCESSING OF WHEAT FOR GROWING-FINISHING SWINE

Give a NOD to diabetes:

Bio-chemical. researches concerning Aloe arborescens and. cultivated with organic protocols from Azienda Agricola G. Dester.

Overview of the Cattle Immune System 1

Introduction. Pathogenesis of type 2 diabetes

Green Fluorescent Protein (GFP): Genetic Transformation, Synthesis and Purification of the Recombinant Protein

OMEGA 3 REPORT. Source: and

Medical Microbiology Culture Media :

10.1 The function of Digestion pg. 402

AmphoraNet: Taxonomic Composition Analysis of Metagenomic Shotgun Sequencing Data

The Polyphenol Study. Effect of Dietary Polyphenols on Intestinal Microbiota, Intestinal Inflammation and Metabolic Syndrome

Functional properties of fructans and their potential role in management of intestinal inflammatory pathologies

Definition: What is the Microbiome- A Big Picture

UTILIZATION of PLASMA ACTIVATED WATER in Biotechnology, Pharmacology and Medicine. JSC TECHNOSYSTEM-ECO Moscow, Russia April, 2009

Getting Enough Fiber In Your Diet Does Not Have To Be Like This!

UTI CAT FOOD COMPARISON CHART

Restore and Maintain treatment protocol

Controlling intestinal microflora with BioHealth, MicroHealth and LactoHealth. Below illustration shows clearly the effects of unbalanced microflora

Hill s Evidence-Based Clinical Nutrition for Dermatology Specialists

CHAPTER 6: RECOMBINANT DNA TECHNOLOGY YEAR III PHARM.D DR. V. CHITRA

Nutrition Guide For Bobwhite Quail Production

Dietary emulsifiers impact the mouse gut microbiota promoting colitis and metabolic syndrome

PATIENT INFORMATION LEAFLET: CENTRUM. Read the contents of this leaflet carefully before you start using CENTRUM, because it

BEC Feed Solutions. Steve Blake BEC Feed Solutions

Course Curriculum for Master Degree in Food Science and Technology/ Department of Nutrition and Food Technology

Chapter 18: Applications of Immunology

Metabolism Dr.kareema Amine Al-Khafaji Assistant professor in microbiology, and dermatologist Babylon University, College of Medicine, Department of

U.S. Meat Animal Research Center Clay Center, NE

WHY IS THIS IMPORTANT?

Determination of Specific Nutrients in Various Foods. Abstract. Humans need to consume food compounds such as carbohydrates, proteins, fats,

Accelerate genomic breakthroughs in microbiology. Gain deeper insights with powerful bioinformatic tools.

EUROPEAN COMMISSION HEALTH & CONSUMER PROTECTION DIRECTORATE-GENERAL

Nutrition and Toxicants in Autoimmune Disease: Implications for Prevention and Treatment

3.0 Treatment of Infection

Land O Lakes Feed DDGS. Nutrients Concentrate: United States Ethanol Outlook. A Growing Opportunity

Product Information: Jevity 1.2 Cal

How To Get Healthy

Carbohydrates, proteins and lipids

Carbon-organic Compounds

R. Landstorfer et al. BMC Genomics, Audrey Segura

FACULTY OF MEDICAL SCIENCE

Bacterial Transformation with Green Fluorescent Protein. Table of Contents Fall 2012

Water Quality For Poultry

Payback News. Beef Cows-The Cheapest Mineral Isn t

Product Information: Jevity 1.5 Cal

An Essential Part of a Healthy Diet. Soluble vs Insoluble Fibre. Are You Getting Enough Fibre? Health Benefits of a High-Fibre Diet 4

Production of prebiotics from PATRICK ADLERCREUTZ, DIV. OF BIOTECHNOLOGY

Consequences of 100% organic diets for pigs and poultry

Complex Microbial Communities. Single-Stage Chemostat Model

Chapter 43: The Immune System

Product Information: Jevity 1.5 Cal

NUTRIENTS: THEIR INTERACTIONS

Detailed Course Descriptions for the Human Nutrition Program

Objectives. What is undernutrition? What is undernutrition? What does undernutrition look like?

Without intrinsic factor, which is a unique protein made in the stomach, vitamin B12 cannot gain access to the rest of the body where it is needed.

TOTAL PROTEIN FIBRINOGEN

GUIDELINES FOR THE REGISTRATION OF BIOLOGICAL PEST CONTROL AGENTS FOOD AND AGRICULTURE ORGANIZATION OF THE UNITED NATIONS

How To Understand The Human Body

Growth & Feeding Puppies Karen Hedberg BVSc Growth

Introduction to Microbiology The Microbial World and You (Chapter 1) Lecture Materials for Amy Warenda Czura, Ph.D. Suffolk County Community College

Dietary Fiber and Alcohol. Nana Gletsu Miller, PhD Spring 2014

Methyl groups, like vitamins, are

Food Allergy Gluten & Diabetes Dr Gary Deed Mediwell 314 Old Cleveland Road Coorparoo

Level 3. Applying the Principles of Nutrition to a Physical Activity Programme Level 3

Name: Hour: Elements & Macromolecules in Organisms

Week 6: Digestive Health

Beneficial Microflora in Honey Bee Colonies

NUTRIENT SPECIFICATIONS OF TURKEY WASTE MATERIAL

HIGH FIBER DIET. (Article - Web Site) August 20, 2003

ROLLED VERSUS WHOLE CORN: EFFECTS ON RUMINAL FERMENTATION OF FEEDLOT STEERS

Overview. Nutritional Aspects of Primary Biliary Cirrhosis. How does the liver affect nutritional status?

Disc Diffusion Susceptibility Methods

Migrene og mikroflora. Linda Mulder, MSc. Vårseminaret 2014

14/11/2014. Copper supply affects growth performance and health in growing pigs. Outline. Copper as essential trace elements

Pay-Per-Click Suggested Words

Effects of Antibiotics on Bacterial Growth and Protein Synthesis: Student Laboratory Manual

25-hydroxyvitamin D: from bone and mineral to general health marker

Focus on Preventing Disease. keeping an eye on a healthy bottom line. Cattle Industry

Digestion of feeds in the milk-fed calf

Grouping to Increase Milk Yield and Decrease Feed Costs

CORN BY-PRODUCTS IN DAIRY COW RATIONS

CODEX STANDARD FOR FOLLOW-UP FORMULA CODEX STAN This standard applies to the composition and labelling of follow-up formula.

Chapter 2. The Chemistry of Life Worksheets

Heat of combustion (gross energy)

What is Geriatric? Geriatric Nutrition of Companion Animals. Age Chart. Diseases Associated with Older Pets

1. The diagram below represents a biological process

Pediatrics. Specialty Courses for Medical Assistants

Forage Crises? Extending Forages and Use of Non-forage Fiber Sources. Introduction

Transcription:

https://doi.org/10.1186/s13568-022-01458-y ORIGINAL ARICLE Effect of β 1,3/1,6 glucan on gut microbiota of yellow feathered broilers Open Access Jingge Wang 1,2, Zibin Zheng 1,3, Hua ang 1, Jie Chen 4, ingping Xiao 1, Xiaofeng Ji 1, Zhenming Zhang 2, Hailian He 2, Baoan Ding 2* and Biao ang 1* Abstract β-1,3/1,6-glucan as a prebiotic improves immune performance in animals. hese functions are closely related to the effect of β-1,3/1,6-glucan on gut microbiota structure. However, the effect of β-1,3/1,6-glucan on the gut microbiota structure of broilers is unclear. he aim of this study was to confirm the effects of β-1,3/1,6-glucan on the cecal microflora structure of yellow-feathered broilers. his study monitored the antimicrobial resistance (AMR) level of Escherichia coli in feces of yellow-feathered broilers by standard broth dilution method and mastered the AMR level of chickens selected. he effects of β-1,3/1,6-glucan on gut microbiota were investigated by 16S rrna sequencing. he results showed that the number of isolated multidrug-resistant E. coli strains accounted for 98.41%. At 14, 21, and 28 days of age, supplemented of 0.2%, 0.1%, and 0.1% β-1,3/1,6-glucan in yellow-feathered broiler diets significantly altered gut microbial composition, and beneficial bacteria Alistipes, Bacteroides and Faecalibacterium were significantly increased. hese findings provide guidance and recommendations for β-1,3/1,6-glucan as a broiler feed additive to improve the growth of broilers. Keywords: β-1,3/1,6-glucan, ellow-feathered broilers, Gut microbiota, 16S rrna Introduction Antibiotics used in animal breeding are one of the important sources of environmental antibiotics pollution. he increasing concentration of antibiotics in the environment makes bacteria evolve more extensive antimicrobial resistance (AMR) and further changes the composition of the microbial community (Danner et al. 2019). Antibiotics in livestock and poultry feeding processes can cause gut microbiota disorder and hinder animals average growth and metabolism (Lillehoj et al. 2018). he European Union, the United States and China imposed Jingge Wang and Zibin Zheng have equally contributed to this work *Correspondence: dingbaoan1967@163.com; tangbiao@zaas.ac.cn 1 State Key Laboratory for Managing Biotic and Chemical hreats to the Quality and Safety of Agro Products & Institute of Agro Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China 2 College of Agriculture and Animal Husbandry, Qinghai University, Xining 810016, China Full list of author information is available at the end of the article a complete ban of all anti-biotics in animal feed to promote growth in January 2006, January 2017 and October 2020, respectively (Salim et al. 2018). In promoting nonantibiotic breeding, finding suitable antibiotic substitutes is an essential link in the feeding mode of reducing the use of antibiotics. Currently, a number of possible alternatives to AGP are used, including acidifiers, probiotics, enzymes, algae and herbal products, microflora enhancers, and immuno-modulators (Salim et al. 2018; Seidavi et al. 2021). β-glucan is considered a natural prebiotic with various biological functions, such as antioxidant, free radical scavenging, anti-tumor, anti-cancer, and immune activation, competing with pathogenic microorganisms for binding sites in intestinal epithelial cells to prevent inflammation (Baldassano et al. 2017; De Marco et al. 2021; Mo et al. 2017; Virginio et al. 2021; Xu et al. 2009). he main chain of β-1,3/1,6-glucan is a linear skeleton with β-1,3 bonds, and the side chain is a highly branched β-1,6 bond (Baldassano et al. 2017). he Author(s) 2022. Open Access his article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. he images or other third party material in this article are included in the article s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. o view a copy of this licence, visit http:// creat iveco mmons. org/ licen ses/ by/4. 0/.

Page 2 of 13 It is commonly found in the cell walls of yeast, bacteria, fungi, algae, and plants (Baldassano et al. 2017). β-1,3/1,6-glucan was proved to be a source of substances with immune-stimulating properties in yeast cells in 1955 (Louis et al. 1955). It is a green, healthy, safe, and effective immune adjuvant (Bobadilla et al. 2013; Wu et al. 2016). β-1,3/1,6-glucan can reduce the colonization of Salmonella in the intestinal tract, relieve the level of intestinal and visceral organ injury caused by Salmonella infection, promote the number of probiotics such as Bifidobacterium and Lactobacillus, and also have a significant immune effect against parasitic and viral diseases (Horst et al. 2019; Shao et al. 2016). β-1,3/1,6-glucan is an essential component of a prebiotic-rich diet that promotes the growth and metabolism of the gastrointestinal microbiota (Horst et al. 2019; Shao et al. 2016). Gut microbiota is a vast microbial ecosystem and complex ecosystem, easily affected by many factors, such as the environment, age, diet, and feed additive (Chen et al. 2020; Fassarella et al. 2021). Gut microbiota participate in vital physiological processes, such as energy homeostasis, metabolism, intestinal epithelial health, immune activity and neural development (Barko et al. 2018). Jayachandran et al. reported that the gut microbiota might realize the immunomodulatory effect of β-glucan as the mediator of the immune response (Jayachandran et al. 2018). At present, the research on β-glucan and gut microbiota mainly include rats (Aoe et al. 2019), mice (Shi et al. 2020), fish (Harris et al. 2020), dogs (Van et al. 2020), weaned piglets (Metzler-Zebeli et al. 2011), and calves (Virginio et al. 2021), which have not been reported in broilers. Among them, β-glucan distinctly raised the number of Lactobacilli and Bifidobacteria in the colon of weaned pigs, and also increased the concentration of butyrate in the stomach, cecum and colon, which may be beneficial to the intestinal development of weaned pigs (Metzler-Zebeli et al. 2011). β-glucan significantly increased acetic acid and butyrate concentrations in cecum of rats. he abundance of Bacteroidetes in cecum was significantly increased, and the abundance of Firmicutes in cecum was significantly decreased, which was helpful to induce secreted IgA to neutralize the toxins produced by microorganisms (Aoe et al. 2019). In this study, the AMR of yellow-feathered broilers was monitored. he effects of β-1,3/1,6-glucan on the gut microbiota of broilers from hatching to 28 days of age were studied. his experiment analyzed the effect of β-1,3/1,6-glucan as a substitute antibody product in improving the gut microbial composition by 16S rrna. Materials and methods Animals and sampling his study was conducted on a farm in Huangzhong County, Qinghai Province. 240 1-day-old yellow-feathered broilers (male) with similar genetic and growth status were randomly divided into four groups with four replicates per group and 15 broilers per replicate. he broilers were housed in 16 cages with a size of 150 cm 80 cm 38 cm (15 broilers each). he negative control group () was fed a basal diet. In the antibiotic group (), 0.02% tylosin (Ringpu, China, with a purity of ) was added to the basal diet. β-1,3/1,6-glucan groups 1 and 2 (, ) were supplemented with 0.1% and 0.2% β-1,3/1,6-glucan (Xingzhongcheng, China, from yeast) in basal diet, respectively. he composition and nutritional level of the basal diet are shown in able 1. he broilers had free access to feed and water during the experiment. We used incandescent lamps, which were slightly higher at 40 lx for the first week. After the second week, the light intensity gradually decreased and was 25 lx. he light was 24 h a day in the first week, then decreased to 0.5 h a day until the 26th day, and then kept for 17 h a day. he room temperature is controlled to be 35 33 on the 1st 7th day, 32.5 29.5 on the 8th 14th day, and 29 24 on the 15th 28th day. he humidity of the chicken house shall be kept between 60 7, and the chicken house able 1 Ingredient composition and analysed nutrient contents of the basal diet Composition Proportion% Nutritional level Corn 56.81 Dry matter 84.36% Soybean meal 28.00 Metabolic energy 11.84 (MJ/kg) Soybean oil 2.00 Crude protein 19.55% Wheat bran 5.30 Crude fat 4.47% east powder 3.00 Crude fiber 3.81% Stone powder 1.96 Ash content 2.8 Calcium hydrogen phosphate 1.10 Calcium 1.09% Sodium Chloride 0.30 Phosphorus 0.64% Methionine 0.23 Methionine 0.55% Lysine 0.35 Lysine 1.45% hreonine 0.08 hreonine 0.86% Sodium sulphate 0.10 Broiler multi mineral 0.12 5 choline chloride 0.10 Baking soda 0.05 Premix 0.50 otal 100.00 Multi mineral content per kilogram of broiler: Copper 8 mg, Iron 60 mg, zinc 60 mg, manganese 60 mg, Selenium 0.15 mg and Iodine 0.35 mg. Nutritional levels are calculated values

Page 3 of 13 shall be disinfected and cleaned regularly according to the routine immunization procedure. Use 0.1% bromogeramine solution to sterilize chickens, once a week, spray steriliza-tion. One yellow-feathered broiler was randomly selected from each replicate of each group for fresh feces collection at 7, 14, 21, and 28 days of age, respectively. One yellow-feathered broiler was randomly selected from each replicate of each group at 14, 21, and 28 days of age. After the chicks were dissected, the cecal contents were collected with sterile cotton swabs and put into a 5 ml sterile centrifuge tube. Refrigerate in a liquid nitrogen tank after sealing. Identification of Escherichia coli isolates and antimicrobial susceptibility testing A total of 64 fresh fecal samples were collected at 7, 14, 21, and 28 days of age using Cary-Blair transport medium to isolate of Escherichia coli. Matrix-assisted laser desorption ionization-time-of-flight mass spectrometry (Bruker MALDI Biotyper System, Germany) was used for strain identification. AMR of E. coli strains was detected by the broth dilution method. he isolation and identification of E. coli strains and AMR detection methods were described in previous studies (ang et al. 2021). DNA extraction, sequencing, and data analysis According to the manufacturer s instructions, the microbial genomic DNA was extracted from cecum contents of yellow-feathered broilers using the QIAamp DNA Stool Mini Kit (QIAGEN, US). he concentration of DNA in the extracted samples was detected with a NanoDrop 2000 spectrophotometer (hermofisher, US). he extracted DNA samples above were submitted to Shanghai Majorbio Bio-pharm echnology Co, Ltd for sequence analysis. he V3 V4 region of the bacterial 16S rrna gene was amplified from each genomic DNA sample by using the primers 338F (5 -AC CC ACG GGA GCA GCA-3 ) 806R (5 -GGA CA CHV GGG W CA A-3 ). Sequencing libraries were then constructed using ruseqm DNA Sample Prep Kit and sequenced on an Illumina MiSeq 300 platform. After sample splitting of PE reads obtained by MiSeq sequencing, double-ended reads were firstly controlled and filtered according to sequencing quality and, at the same time, spliced according to the overlapping relationship between double-ended reads to obtain optimized data after quality control spliced. hen, sequence denoising methods (DADA2/Deblur) were used to process the optimized data, and Amplicon Sequence Variant (ASV) was used to represent the sequence and abundance information. Statistical analysis and visualization All statistical analyses were performed by SPSS 23.0 (IBM, US) using an unpaired two-tailed Student s t-test. Data are presented as the mean ± SEM. Results were considered significant when P < 0.05. Based on the representative sequences and abundance information of ASV, taxonomic analysis, community diversity analysis, species difference analysis, correlation analysis, and a series of statistical or visual analyses can be carried out. Alpha diversity indices (Chao and Shannon) via QIIME software (Version 1.7.0) and shown with R software (Version 2.15.3) (Lawley & annock 2017). Principal coordinate analysis (PCoA) was performed to analyze the beta diversity. Circos and Pie charts were generated to show taxa distribution at the phylum and genus levels. Results Isolation of E. coli and antimicrobial susceptibility 63 strains of E. coli were isolated and identified from 64 stool samples collected in this research, with an isolation rate of 98.44%. 63 of the E. coli strains to 13 antibiotics are shown in Fig. 1a and Fig. 1b. In terms of the MIC distribution (Fig. 1c), the MIC values of the antibiotics ampicillin (AMP), amoxicillin/clavulanate potassium (AMC), cefotaxime (CX), ceftiofur (CEF), gentamicin (GEN), tetracycline (E), ciprofloxacin (CIP), sulfamethoxazole (/S) and florfenicol (FFC) were highly resistant to these antibiotics. AMP, AMC, CEF, E, and CIP showed the highest AMR rates of over, followed by CX, /S, and FFC, at 85.71%, 85.71%, 88.89%, while GEN exhibited AMR rates greater than 7. Among the isolates, strains with multidrug resistance (MDR) accounted for 98.41% of the total isolates. In addition, their AMR rates for amikacin (AMK), colistin (CS) and tigecycline (IG) were 11.11%, 4.76% and 1.59%, respectively. All tested E. coli strains were sensitive to meropenem (MEM). hese results indicate that the AMR of chicken-derived E. coli in this region is profound. As shown in Fig. 2 and able 2, there were 18 types of AMR spectrum of 63 strains. AMP-AMC-CX-GEN- CEF-CIP-/S-E-FFC was the most AMR phenotype of 31 strains (49.21%). here were 3 strains (4.76%) of 8 classes of antibiotics, 38 strains (60.32%) of 7 classes of antibiotics, 12 strains (19.05%) of 6 classes of antibiotics, 7 strains (11.11%) of 5 classes of antibiotics. It shows that the MDR of E. coli in this chicken farm is numerous, and there are many kinds of AMR profiles. 16S amplicon sequencing 2,500,002 optimized 16S amplicon sequences with 1,034,571,892 bp and an average length of 414 bp were obtained from 48 cecal content samples. As shown in

Page 4 of 13 Fig. 1 he susceptibility test results of 63 strains of E. coli to 13 antibiotics. a AMR rates of Escherichia coli. b he distribution of multidrug-resistant strains. c MIC distributions of 13 antibiotics in isolated E. coli strains. MIC minimum inhibitory concentration, MDR multidrug resistance, AMP ampicillin, AMC amoxicillin/clavulanate potassium, CX cefotaxime, CEF ceftiofur, AMK amikacin, GEN gentamicin, E tetracycline, IG tigecycline, MEM meropenem, CS colistin, CIP ciprofloxacin, /S sulfamethoxazole, FFC florfenicol Fig. 3a, gentle rank-abundance curves at the ASV level indicate that species are evenly distributed in the sample community. As shown in Fig. 3b, the dilution curve under the sobs index of ASV level reflects that the amount of sequencing data of the submitted samples are reasonable. he coverage index of all sequencing samples was more significant than 0.99, indicating that the sequencing results were reliable. As shown in Venn Fig. 3c, d, e, the number of

Page 5 of 13 Sensitive Intermediate Resistant CEF 7d 14d 21d 28d Fig. 2 AMR profiles of 63 E. coli strains. AMP ampicillin, AMC amoxicillin/clavulanate potassium, CX cefotaxime, CEF ceftiofur, AMK amikacin, GEN gentamicin, E tetracycline, IG tigecycline, MEM meropenem, CS colistin, CIP ciprofloxacin, /S sulfamethoxazole, FFC florfenicol CX GEN AMC AMP E CIP FFC /S IG MEM CS AMK able 2 he AMR patterns of 63 of E. coli strains ID Antibiotic-resistant pattern Number Percentage 1 AMP-AMC-CX-GEN-CEF-CIP-/S-E-FFC 31 49.21 2 AMP-AMC-CX-CEF-CIP-/S-E-FFC 4 6.35 3 AMP-AMC-CX-GEN-CEF-CIP-E-FFC 4 6.35 4 AMP-AMC-CX-CEF-CIP-E-FFC 3 4.76 5 AMP-CX-GEN-CEF-CIP-/S-E-FFC 3 4.76 6 AMP-AMC-GEN-CEF-CIP-/S-E-FFC 2 3.17 7 AMP-AMC-CX-AMK-GEN-CEF-CIP-/S-E 2 3.17 8 AMP-AMC-CX-AMK-GEN-CEF-CIP-/S-E-FFC 2 3.17 9 AMP-AMC-CX-AMK-GEN-CS-CEF-CIP-/S-E-FFC 2 3.17 10 AMP-AMC-CIP-/S-E-FFC 2 3.17 11 AMP-AMC-CX-CEF-CIP-/S-E-IG-FFC 1 1.59 12 AMP-AMC-CX-GEN-CS-CEF-CIP-/S-E-FFC 1 1.59 13 AMP-AMC-GEN-CEF-CIP-FFC 1 1.59 14 AMC-CX-AMK-GEN-CIP-/S-E 1 1.59 15 AMP-AMC-CEF-CIP-/S-E 1 1.59 16 AMP-AMC-CIP-/S-E 1 1.59 17 AMP-AMC-/S-E 1 1.59 18 AMP-AMC-E 1 1.59 ASVs shared by the four groups at 14 days of age, 21 days of age, and 28 days of age were 109, 119, and 133, respectively, indicating that the bacterial species similarity in cecal contents of yellow-feathered broilers increased, possibly due to the gradual stability of gut microbiota with the increase of age. β 1,3/1,6 glucan administration altered broilers gut microbiota he alpha-diversity indicated that the Chao index had a downward trend from the control group to the group in 14 days (P < 0.05) (Fig. 4a, b), and there was an upward trend from 21 days (P < 0.05) (Fig. 4c, d) in

Page 6 of 13 a b 400 Rarefaction curves Relative Abundance 1% 0.1% 0.01% Rank-Abundance curves 14d 21d 28d Sobs index 350 300 250 200 150 100 50 0.001% 100 200 300 0 0 1000 2000 3000 4000 5000 6000 7000 8000 9000 ASV Level Rank Number of Reads Sampled c d e 100 51 5 38 14 110 131 62 26 17 165 92 49 65 15 104 23 21 8 8 109 62 36 27 113 86 42 53 16 46 119 19 9 11 49 107 33 55 8 27 133 39 22 34 91 14d 21d 28d Fig. 3 Analysis of ASV level data by amplification sequencing. ASV level Rank-Abundance curves a, Sobs exponential dilution curves b, and Ven diagram c, d, e of samples. he richness of species in the horizontal direction of the Rank-Abundance curve is reflected by the width of the curve, and the smoothness of the curve reflects the evenness of species in the sample. Different colors represent different grouped samples. he sobs index rarefaction curve was constructed to reflect the microbial diversity of each sample at different sequencing quantities. Different colors rep-resent different samples. Venn diagram can intuitively represent the similarity and overlap of ASV components of samples. Different colors represent different grouped samples yellow-feathered broilers cecum. he beta-diversity showed significant differences in cecal contents microbiota community at 14, 21, and 28 days of age (Fig. 5a). he beta-diversity showed significant changes in the microbial community members of cecal contents from the control group to,, and groups (Fig. 5b, c, d). Especially in the group, its clustering is far from all other groups. In broilers cecum, groups were closer to the control group. In 21 days, the group was as far away from the control group as the group. Next, we analyzed broilers microbiota composition in the cecum of broilers in the,,, and groups. axonomic analysis showed that the dominant bacteria phyla were Firmicutes and Bacteriodetes, accounting for more than 93.43% of the total sequences in all samples (Fig. 6). Compared to the control group, the relative abundance of Firmicutes in the group and group were increased by 13.64% (P < 0.05) and 12.6 (P < 0.05) in 14 days of broilers, respectively (Fig. 6a). However, the relative abundance of Bacteriodetes in the group and group was decreased by 14.47% (P < 0.05) and 16.94% (P < 0.05) in 21 days samples relative to the control group, respectively (Fig. 6b). Firmicutes were less by 20.17%, and Bacteriodetes increased by 18.65%, abundant in 28 days samples of the group compared to the control group (P < 0.05) (Fig. 6c). At the genus level, cecal contents samples were dominated by Alistipes (13.07%), Bacteroides (10.66%), Ruminococcus_torques_group (10.46%), unclassified_f Lachnospiraceae (7.86%), Faecalibacterium (6.13%), and Lactobacillus (5.31%) (Fig. 7). Among the top 5 taxa, at 14 days of age, compared with the control group, Faecalibacterium had a downward trend in the group, and it significantly rose in the and groups (P < 0.05). unclassified_f Lachnospiraceae had an extremely significant decline in the group compared with the control group at 14 days of age (P < 0.05). At the 21 days of age, the group had a significant increase

Page 7 of 13 a 300 250 200 b 14 d 14 d 4.50 4.25 4.00 3.75 Group c 400 350 300 250 200 150 e 400 350 300 250 200 3.50 d 21 d 5.00 21 d 4.50 4.00 f 28 d 5.00 28 d 4.50 4.00 3.50 Chao Index Shannon Index Fig. 4 Alpha diversity, including Chao index a, c, e and Shannon index b, d, f in the control group (), antibiotic group (), 0.1% β-1,3/1,6-glucan (), and 0.2% β-1,3/1,6-glucan () groups in the cecum. Data was processed through log10, expressed as mean ± SEM (n = 8), and analyzed by one-way ANOVA analysis. he different superscript letters on the boxplot represent a significant difference (P < 0.05) in Bacteroides and unclassified_f Lachnospiraceae (P < 0.05), and the latter significantly rose in and group (P < 0.05) compared with the control group. At the 21 days of age, the group had a significant decline in Alistipes (P < 0.05), Ruminococcus_torques_group (P < 0.05), and Faecalibacterium (P < 0.05), and Ruminococcus_torques_group significantly decline in group (P < 0.05) compared with the control group. At 28 days of age, compared with the control group, the contents of Alistipes and Bacteroides in the group were significantly increased and decreased (P < 0.05). At 28 days of age, compared with the control group, the group had a significant increase in Ruminococcus_torques_group and unclassified_f Lachnospiraceae and it significantly declines in Bacteroides (P < 0.05). Bacterial taxa differentially represented in broilers cecum microbiota Broiler cecum bacterial features were analyzed by using lefse. Hierarchically clustered heat map for the significantly different bacterial genera in cecal contents of 14 day old broiler, 21 day old broilers and 28 day old broilers (Fig. 8). he abundance of these significantly different features was shown on the heat map. At 14 days of age (Fig. 8a), Bifidobacterium and Lactobacillus significantly increased in the group compared with other groups (,, ). At 21 days of age (Fig. 8b), Megamonas and Enorma had a significant increase in the group compared with other groups (,, ), Bacteroides, and Helicobacter had a significant increase in the group compared with other groups (,, ). At 28 days of age (Fig. 8c), Ruminococcus_torques_ group, Desulfovibrio, Defluviitaleaceae_UCG_011, and unclassified_f Lachnospiraceae in the group were significantly higher than other groups (,, ). Discussion β-1,3/1,6-glucan can improve the immune level of the body, increase the expression of immune factors in intestinal and other damaged parts, and alleviate the damage

Page 8 of 13 a PCoA2 (10.81%) 0.2 0.1 0.0-0.1-0.2 age Group 14d 21d 28d b PCoA2 (21.4) 0.4 0.2 0.0-0.2 14d Group -0.3 c PCoA2 (18.77%) 0.4 0.2 0.0-0.3-0.2-0.1 0.0 0.1 0.2 PCoA1 (19.12%) 21d Group d PCoA2 (19.86%) 0.4 0.2 0.0-0.2-0.3-0.2-0.1 0.0 0.1 0.2 PCoA1 (38.19%) 28d Group -0.2-0.1 0.0 0.1 0.2 0.3 PCoA1 (36.33%) -0.3-0.2-0.1 0.0 0.1 0.2 0.3 PCoA1 (23.43%) Fig. 5 PCoA of the broilers gut microbial community composition of different age a, and the (control group), (antibiotic group), (0.1% β-1,3/1,6-glucan), and (0.2% β-1,3/1,6-glucan) group of broilers aged 14 days b, 21 days c and 28 days d based on the Bray Curtis distances showed distinct clusters. he cecum microbial community structure between the 14, 21, and 28 days of age were differentiated by colors (red, green, blue, respectively). he cecum microbial community structure between the,,, and groups were differentiated by colors (purple, blue, green, red, respectively) to the body after pathogen infection (Bobadilla et al. 2013; Wu et al. 2016). he β-glucan regulating gut microbiota is a potential method to reduce disease susceptibility and improve growth performance in piglets (Luo et al. 2019). In this study, we detected the AMR of yellowfeathered broilers to 13 antibiotics from birth to day 28 and found that broilers in this study were highly resistant to multiple antibiotics. here was no difference in AMR among different groups. he structure of gut microbiota is critical to intestinal function (David et al. 2014). he composition of gut microbes is influenced by factors such as antibiotic treatment and diet (Clemente et al. 2018; Dixit et al. 2021). At 14 and 21 days, 0.1% and 0.2% β-1,3/1,6-glucan supplementation increased cecal microflora diversity, respectively. At 28 days, supplementation of 0.1% and 0.2% β-1,3/1,6-glucan increased cecal microflora diversity, but no significant difference. he results showed that different glucan contents had different effects on the gut microbiota diversity of broilers at different ages. Fassarella et al. found that β-glucan had no significant difference in the alpha diversity of gut microbiota in early pigs, and a PCoA analysis showed that β-glucan had a significant effect on feces samples before weaning (Hugo et al. 2020). Velikonja et al. showed that different daily intakes of glucan (6 g vs. 3 g per day) had different effects on gut microbiota diversity (Velikonja et al. 2019). hese results are similar to our findings. he addition of different glucan levels (0.1% and 0.2%) has different effects on the gut microbiota diversity of chicks. However, our study discovers that adding different levels of β-1,3/1,6- glucan may have the opposite effect on the diversity of gut microbiota. he interaction between diet-microorganism-host is closely related to body health and disease (Clemente et al. 2018). Dysbiosis of the gut microbiota can lead to diseases as diverse as inflammatory bowel disease (IBD), systemic inflammatory arthritis, and multiple sclerosis (Clemente et al. 2018; Velikonja et al. 2019). In this study, the dominant bacteria in the cecal contents of chicks were Alistipes, Bacteroides, Ruminococcus_torques_group, unclassified_f Lachnospiraceae,

Page 9 of 13 a Firmicutes 2 8 3 7 6 5 4 3 4 2 5 7 6 8 5 6 4 3 2 7 14d 8 8 7 6 5 4 3 2 2 3 4 5 6 7 8 2 3 4 5 6 7 8 Bacteroidota Campilobacterota Actinobacteriota Proteobacteria Cyanobacteria Desulfobacterota unclassified_k norank_d Bacteria b Firmicutes 3 2 8 7 6 4 5 4 3 2 5 6 7 6 8 7 5 4 3 8 2 21d 8 7 6 5 2 4 3 3 2 4 5 6 7 Bacteroidota 8 2 3 4 5 6 7 8 Campilobacterota Actinobacteriota Proteobacteria Desulfobacterota Cyanobacteria Deferribacterota unclassified_k norank_d Bacteria c Firmicutes 3 2 8 7 6 4 5 4 3 5 2 6 7 6 8 7 5 4 8 3 2 % 28d 9 00 7 8 % 6 5 2 4 3 3 2 4 5 6 Bacteroidota 7 8 2 3 4 5 6 7 8 Actinobacteriota Proteobacteria Cyanobacteria Campilobacterota Deferribacterota Desulfobacterota Synergistota Verrucomicrobiota unclassified_k norank_d Bacteria Fig. 6 Bacterial communities in the cecum of broilers aged 14 days a, 21 days b and 28 days c at phylum level in groups (control group), (antibiotic group), (0.1% β-1,3/1,6-glucan), and (0.2% β-1,3/1,6-glucan). he cecum microbial community structure between the,, and, and groups were differentiated by colors (green, red, blue, orange, respectively)

Page 10 of 13 100 Sequence number percent (%) 75 50 25 0 14 d 21 d 28 d other unclassified_f Ruminococcaceae Helicobacter norank_f_muribaculaceae norank_f_eubacterium _coprostanoligenes_group Bifidobacterium Butyricicoccus norank_f norank_o RF39 UCG-005 Subdoligranulum Sellimonas Blautia Enorma Fournierella Megamonas norank_f Barnesiellaceae norank_f norank_o Clostridia_vadinBB60_group Rikenella norank_f Ruminococcaceae Erysipelatoclostridium Lachnoclostridium unclassified_f Oscillospiraceae Barnesiella Phascolarctobacterium norank_f norank_o Clostridia_UCG-014 Lactobacillus Faecalibacterium unclassified_f Lachnospiraceae Ruminococcus_torques_group Bacteroides Alistipes Fig. 7 he top 30 features in (control group), (antibiotic group), (0.1% β-1,3/1,6-glucan), and (0.2% β-1,3/1,6-glucan) groups of cecum microbiome in broilers. Each color indicates the relative abundance of a bacterial taxon on the bar chart Faecalibacterium and Lactobacillus. Studies have shown that Alistipes produce short-chain fatty acids and reduce intestinal inflammation, which may have a protective effect on many diseases, including liver fibrosis, colitis, cancer immunotherapy, and cardiovascular disease (Parker et al. 2020). Bacteroidetes and Faecalibacterium have been positively correlated with human health and are considered health-promoting gut microbiota (Wang et al. 2020). Bacteroides encodes polysaccharide binding proteins in the outer membrane through polysaccharide utilization sites (PUL) to capture polysaccharides and decompose polysaccharides into oligosaccharides to promote the utilization and uptake of dietary polysaccharides by Bacteroides (amura et al. 2017). Faecalibacterium facilitates the utilization of acetate in the intestine (Virginio Junior et al. 2021). Acetate is used as an energy source for the liver and peripheral tissues and as a signal molecule in gluconeogenesis and lipogenesis (Zambell et al. 2003). Ruminococcus_torques_group and unclassified_f Lachnospiraceae belong to Firmicutes

Page 11 of 13 a c 14d Category1 norank_f Ruminococcaceae Escherichia_Shigella norank_f norank_o RF39 Negativibacillus Coprobacter norank_f UCG_010 yzzerella Barnesiella Fournierella unclassified_f Oscillospiraceae unclassified_f Ruminococcaceae Intestinimonas Faecalibacterium Erysipelatoclostridium Subdoligranulum Bifidobacterium Shuttleworthia Lactobacillus Blautia norank_f Flavobacteriaceae Butyricimonas Anaerofilum norank_f Eubacterium_coprostanoligenes_group Eubacterium_hallii_group unclassified_c Bacilli 28d Category1 norank_f Barnesiellaceae unclassified_f Christensenellaceae CAG_352 Parasutterella Akkermansia NK4A214_group unclassified_o Erysipelotrichales norank_f norank_o Gastranaerophilales Lachnoclostridium norank_f norank_o Clostridia_vadinBB60_group unclassified_f Hungateiclostridiaceae V9D2013_group Mucispirillum Synergistes Parabacteroides Sutterella Colidextribacter Pseudoflavonifractor UCG_005 Ruminococcus_torques_group Desulfovibrio GCA_900066575 Defluviitaleaceae_UCG_011 unclassified_f Lachnospiraceae Ruminococcus_gauvreauii_group Bacteroides norank_f Muribaculaceae DU089 unclassified_o Bacteroidales Caproiciproducens Category1 3 2 1 0-1 21d Category1 uzzerella Megamonas norank_f norank_o norank_c Clostridia Enorma Marvinbryantia norank_f Oscillospiraceae CHKCI001 Candidatus_Arthromitus Collinsella Campylobacter Helicobacter Butyricicoccus Enterococcus Sellimonas Flavonifractor Bilophila Ruminococcus Oscillibacter Christensenellaceae_R_7_group Anaeroplasma norank_f norank_o Clostridia_UCG_014 Alistipes norank_f Peptococcaceae Phascolarctobacterium Rikenella Monoglobus Bacillus Frisingicoccus Fig. 8 Hierarchically clustered heat map for the significantly different bacterial genera in cecal contents of 14 day old broiler a, 21 day old broilers b and 28 day old broilers c. Heat map indicated 83 bacterial taxa were identified by LEfSe (LDA > 3) in broilers at 14 days of age (n = 25) a, 21 days of age (n = 28) b and 28 days of age (n = 30) c cecum microbiome. he top 1000 features were used for lefse analysis. Heat map shows the average relative abundances on a Z-score b (Bassanini et al. 2019). Ruminococcus_torques aggravates the symptoms of neurodegenerative disease amyotrophic lateral sclerosis (Blacher et al. 2019). Mucosa-associated bacteria Ruminococcus_torques are significantly increased in the intestinal epithelial cells of IBD, such as ulcerative colitis and Crohn s disease (Png et al. 2010). Although members of Lachnospiraceae are among the primary producers of short-chain fatty acids, the different taxonomic groups of Lachnospiraceae have also been associated with different enteral and parenteral diseases (Vacca et al. 2020). Metabolic syndrome, obesity, diabetes, liver disease, and IBD are all inflammatory diseases

Page 12 of 13 associated with the Lachnospiraceae family or specific taxonomic groups of Lachnospiraceae (Vacca et al. 2020; Zeng et al. 2016). In addition, they appear to be associated with major depressive disorder and multiple sclerosis syndrome (Cheung et al. 2019; Vacca et al. 2020). Meanwhile, the increased abundance of Lachnospiraceae was associated with aging (Odamaki et al. 2016). his study found that adding 0.2% β-1,3/1,6-glucan to the diet of 14 days of age yellow-feathered broilers can significantly improve the abundance of Faecalibacterium that promote intestinal health and significantly reduce the abundance of unclassified_f Lachnospiraceae associated with host aging and many diseases. Supplementation of 0.1% β-1,3/1,6-glucan in diets of yellow-feathered broilers at 21 days of age significantly reduced the abundance of Ruminococcus_torques_group, which is associated with host enteritis and neurological diseases. Supplementation of 0.1% β-1,3/1,6-glucan at 28 days of age significantly increased the abundance of Alistipes, which promotes intestinal nutrition and protection. Huali et al. reported that β-glucan significantly increased the abundance of Bacteroidetes and Faecalibacterium. On the contrary, the abundance of Lachnospiraceae and Ruminococcus significantly decreased, similar to the results in this study (Wang et al. 2020). Hugo et al. found significant differences in Ruminococcus in feces when β-glucan dietary intervention was administered at the pre-weaning stage, highlighting the potential regulatory role of the microbiota on dietary fiber (Hugo et al. 2020). Angelis et al. reported that after the intervention of a barley glucan diet on the human body, the abundance of Ruminococcus increased, and the abundance of other Firmicutes such as Faecalibacterium decreased (De Angelis et al. 2015). hese results indicate that dietary supplementation of β-glucan can regulate the gut microbiota structure, and the source and amount of β-glucan can significantly affect sensitive bacteria. In this study, the supplementation of 0.1% and 0.2% β-1,3/1,6-glucan in diets of yellow-feathered broilers had different effects on the cecal microflora structure of broilers at different ages. Supplementation of 0.2%, 0.1%, and 0.1% β-1,3/1,6- glucan in the diets of yellow-feathered broilers at 14, 21, and 28 days of age had significant effects on the maintenance of gut microbiota structure that was more conducive to intestinal nutrient absorption and immune resistance. In this study, we found that yellow-feathered broilers from hatching to the 28th day were highly AMR to 9 commonly used antibiotics. At 14, 21, and 28 days of age, yellow-feathered broilers supplemented with 0.2%, 0.1%, and 0.1% β-1,3/1,6-glucan significantly changed the gut microbiota composition and beneficial bacteria such a beneficial bacteria Alistipes, Bacteroides and Faecalibacterium significantly increased. hese results demonstrate that β-1,3/1,6-glucan as an antibiotic substitute can improve gut microbiota composition, which is helpful for the promotion and application of alternative antibiotic products. Acknowledgements Not applicable. Author contributions Conceptualization, B and BD; investigation, B, ZZ, JC, X, XJ, HH and H; methodology, JW, ZZ and BD; supervision, B and H; visualization, ZZ, JW and B; writing original draft B, JW and ZZ. All authors have read and approved the final manuscript. Funding his work was supported by the Key Research and Development Program of Zhejiang Province (2020C02031), the State Key Laboratory for Managing Biotic and Chemical hreats to the Quality and Safety of Agro-products (2010DS700124-ZZ2008, ZZ2102), and the Collaborative Extension Plan of Major Agricultural echnologies in Zhejiang Province (2021XGXM03). Availability of data and materials he data presented in this study are deposited in the (NCBI SRA) repository (accession number: PRJNA820361). Declarations Ethics approval and consent to participate All procedures outlined herein were in compliance with the national laws and regulations for animal experimentation and were performed in accordance with the oversight principles mandated by the Qinghai University Animal Care Committee for the care and use of experimental animals (Approval no. IACUC, SL-2022009). Written informed consent was obtained from the owners for the participation of their animals in this study. Consent for publication All authors have reviewed the final version of the manuscript and approve it for publication. Competing interests he authors have declared that there are no conflicts of interests. Author details 1 State Key Laboratory for Managing Biotic and Chemical hreats to the Quality and Safety of Agro Products & Institute of Agro Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China. 2 College of Agriculture and Animal Husbandry, Qinghai University, Xining 810016, China. 3 Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China. 4 Zhejiang Provincial Center for Animal Disease Prevention and Control, Hangzhou 310020, China. Received: 5 July 2022 Accepted: 27 August 2022 References Aoe S, amanaka C, Fuwa M, amiya, Nakayama, Miyoshi, Kitazono E (2019) Effects of BARLEmax and high-beta-glucan barley line on short-chain fatty acids production and microbiota from the cecum to the distal colon in rats. PLoS ONE 14(6):e0218118 Baldassano S, Accardi G, Vasto S (2017) Beta-glucans and cancer: the influence of inflammation and gut peptide. Eur J Med Chem 142:486 492 Barko PC, McMichael MA, Swanson KS, Williams DA (2018) he Gastrointestinal microbiome: a review. J Vet Intern Med 32(1):9 25

Page 13 of 13 Bassanini G, Ceccarani C, Borgo F, Severgnini M, Rovelli V, Morace G, Verduci E, Borghi E (2019) Phenylketonuria diet promotes shifts in Firmicutes populations. Front Cell Infect Microbiol 9:101 Blacher E, Bashiardes S, Shapiro H, Rothschild D, Mor U, Dori-Bachash M, Kleimeyer C, Moresi C, Harnik, Zur M, Zabari M, Brik RB, Kviatcovsky D, Zmora N, Cohen, Bar N, Levi I, Amar N, Mehlman, Brandis A, Biton I, Kuperman, soory M, Alfahel L, Harmelin A, Schwartz M, Israelson A, Arike L, Johansson MEV, Hansson GC, Gotkine M, Segal E, Elinav E (2019) Potential roles of gut microbiome and metabolites in modulating ALS in mice. Nature 572(7770):474 480 Bobadilla F, Rodriguez-irado C, Imarai M, Galotto MJ, Andersson R (2013) Soluble beta-1,3/1,6-glucan in seaweed from the southern hemisphere and its immunomodulatory effect. Carbohydr Polym 92(1):241 248 Chen X, Zhang, Ma W, Wang Z (2020) Effects of Ligustrum lucidum on egg production, egg quality, and caecal microbiota of hens during the late laying period. Ital J Anim Sci 19(1):687 696 Cheung SG, Goldenthal AR, Uhlemann AC, Mann JJ, Miller JM, Sublette ME (2019) Systematic review of gut microbiota and major depression. Front Psychiatry 10:34 Clemente JC, Manasson J, Scher JU (2018) he role of the gut microbiome in systemic inflammatory disease. BMJ 360:j5145 Danner MC, Robertson A, Behrends V, Reiss J (2019) Antibiotic pollution in surface fresh waters: occurrence and effects. Sci otal Environ 664:793 804 David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, Ling AV, Devlin AS, Varma, Fischbach MA, Biddinger SB, Dutton RJ, urnbaugh PJ (2014) Diet rapidly and reproducibly alters the human gut microbiome. Nature 505(7484):559 563 De Angelis M, Montemurno E, Vannini L, Cosola C, Cavallo N, Gozzi G, Maranzano V, Di Cagno R, Gobbetti M, Gesualdo L (2015) Effect of whole-grain barley on the human fecal microbiota and metabolome. Appl Environ Microbiol 81(22):7945 7956 Dixit K, Chaudhari D, Dhotre D, Shouche, Saroj S (2021) Restoration of dysbiotic human gut microbiome for homeostasis. Life Sci 278:119622 Fassarella M, Blaak EE, Penders J, Nauta A, Smidt H, Zoetendal EG (2021) Gut microbiome stability and resilience: elucidating the response to perturbations in order to modulate gut health. Gut 70(3):595 605 Harris SJ, Bray DP, Adamek M, Hulse DR, Steinhagen D, Hoole D (2020) Effect of beta-1/3,1/6-glucan upon immune responses and bacteria in the gut of healthy common carp (Cyprinus carpio). J Fish Biol 96(2):444 455 Horst G, Levine R, Chick R, Hofacre C (2019) Effects of beta-1,3-glucan (AletaM) on vaccination response in broiler chickens. Poult Sci 98(4):1643 1647 de Hugo V, Geervliet M, Jansen CA, Rutten V, van Hees H, Groothuis N, Wells JM, Savelkoul HFJ, ijhaar E, Smidt H (2020) Impact of yeast-derived betaglucans on the porcine gut microbiota and immune system in early life. Microorganisms 8(10):1573 Jayachandran M, Chen J, Chung SSM, Xu B (2018) A critical review on the impacts of beta-glucans on gut microbiota and human health. J Nutr Biochem 61:101 110 Lawley B, annock GW (2017) Analysis of 16S rrna Gene Amplicon sequences using the QIIME software package. Methods Mol Biol 1537:153 163 Lillehoj H, Liu, Calsamiglia S, Fernandez-Miyakawa ME, Chi F, Cravens RL, Oh S, Gay CG (2018) Phytochemicals as antibiotic alternatives to promote growth and enhance host health. Vet Res 49(1):76 Luo J, Chen D, Mao X, He J, u B, Cheng L, Zeng D (2019) Purified beta-glucans of different molecular weights enhance growth performance of LPSchallenged piglets via improved gut barrier function and microbiota. Animals 9(9):602 De Marco Castro E, Calder PC, Roche HM (2021) β-1,3/1,6-glucans and immunity: state of the art and future directions. Mol Nutr Food Res 65(1):e1901071 Metzler-Zebeli BU, Zijlstra R, Mosenthin R, Ganzle MG (2011) Dietary calcium phosphate content and oat beta-glucan influence gastrointestinal microbiota, butyrate-producing bacteria and butyrate fermentation in weaned pigs. FEMS Microbiol Ecol 75(3):402 413 Mo L, Chen, Li W, Guo S, Wang X, An H, Zhan (2017) Anti-tumor effects of (1 >3)-beta-d-glucan from Saccharomyces cerevisiae in S180 tumorbearing mice. Int J Biol Macromol 95:385 392 Odamaki, Kato K, Sugahara H, Hashikura N, akahashi S, Xiao JZ, Abe F, Osawa R (2016) Age-related changes in gut microbiota composition from newborn to centenarian: a cross-sectional study. BMC Microbiol 16:90 Parker BJ, Wearsch PA, Veloo ACM, Rodriguez-Palacios A (2020) he genus Alistipes: Gut bacteria with emerging implications to inflammation, cancer, and mental health. Front Immunol 11:906 Pillemer L, Schoenberg MD, Blum L, Wurz L (1955) Properdin system and immunity.ii. Interaction of the properdin system with polysaccharides. Science 122(3169):545 549 Png CW, Linden SK, Gilshenan KS, Zoetendal EG, McSweeney CS, Sly LI, McGuckin MA, Florin H (2010) Mucolytic bacteria with increased prevalence in IBD mucosa augment in vitro utilization of mucin by other bacteria. Am J Gastroenterol 105(11):2420 2428 Salim HM, Huque KS, Kamaruddin KM, Beg M (2018) Global restriction of using antibiotic growth promoters and alternative strategies in poultry production. Sci Prog 101(1):52 75 Seidavi A, avakoli M, Slozhenkina M, Gorlov I, Hashem NM, Asroosh F, aha AE, Abd El-Hack ME, Swelum AA (2021) he use of some plant-derived products as effective alternatives to antibiotic growth promoters in organic poultry production: a review. Environ Sci Pollut Res Int 28(35):47856 47868 Shao, Wang Z, ian X, Guo, Zhang H (2016) east beta-d-glucans induced antimicrobial peptide expressions against Salmonella infection in broiler chickens. Int J Biol Macromol 85:573 584 Shi H, u, Lin D, Zheng P, Zhang P, Hu M, Wang Q, Pan W, ang X, Hu et al (2020) β-glucan attenuates cognitive impairment via the gut-brain axis in diet-induced obese mice. Microbiome 8(1):143 amura K, Hemsworth GR, Dejean G, Rogers E, Pudlo NA, Urs K, Jain N, Davies GJ, Martens EC, Brumer H (2017) Molecular mechanism by which prominent human gut Bacteroidetes utilize mixed-linkage beta-glucans, major health-promoting cereal polysaccharides. Cell Rep 21(2):417 430 ang B, Ma, He X, Zhou Q, Chang J, Qian M, Xia X, ang H (2021) Similar antimicrobial resistance of Escherichia coli strains isolated from retail chickens and poultry farms. Foodborne Pathog Dis 18(7):489 496 Vacca M, Celano G, Calabrese FM, Portincasa P, Gobbetti M, De Angelis M (2020) he controversial role of human gut Lachnospiraceae. Microorganisms 8(4):573 Van den Abbeele P, Duysburgh C, Rakebrandt M, Marzorati M (2020) Dried yeast cell walls high in beta-glucan and mannan-oligosaccharides positively affect microbial composition and activity in the canine gastrointestinal tract in vitro. J Anim Sci 98(6):1 10 Velikonja A, Lipoglavsek L, Zorec M, Orel R, Avgustin G (2019) Alterations in gut microbiota composition and metabolic parameters after dietary intervention with barley beta glucans in patients with high risk for metabolic syndrome development. Anaerobe 55:67 77 Virginio Junior GF, Reis ME, da Silva AP, de oledo AF, Cezar AM, Mendes LW, Greco L, Montenegro H, Coutinho LL, Bittar CMM (2021) Does algae beta-glucan affect the fecal bacteriome in dairy calves? PLoS ONE 16(9):e0258069 Wang H, Chen G, Li X, Zheng F, Zeng X (2020) east beta-glucan, a potential prebiotic, showed a similar probiotic activity to inulin. Food Funct 11(12):10386 10396 Wu S, seng, Nan FH (2016) β-1,3 1,6-glucan modulate the non-specific immune response to enhance the survival in the Vibrio alginolyticus infection of aiwan abalone (Haliotis diversicolor supertexta). Fish Shellfish Immunol 54:556 563 Xu S, Huo J, Lee KG, Kurosaki, Lam KP (2009) Phospholipase Cγ2 is critical for Dectin-1-mediated Ca 2+ flux and cytokine production in dendritic cells. J Biol Chem 284(11):7038 7046 Zambell KL, Fitch MD, Fleming SE (2003) Acetate and butyrate are the major substrates for de novo lipogenesis in rat colonic epithelial cells. J Nutr 133(11):3509 3515 Zeng H, Ishaq SL, Zhao FQ, Wright AG (2016) Colonic inflammation accompanies an increase of beta-catenin signaling and Lachnospiraceae/Streptoco ccaceae bacteria in the hind gut of high-fat diet-fed mice. J Nutr Biochem 35:30 36 Publisher s Note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.