dietary exposure to antibiotic residues facilitates

34
Dietary Exposure to Antibiotic Residues Facilitates Metabolic Disorder by Altering the Gut Microbiota and Bile Acid Composition Rou-An Chen Institute of Food Science and Technology, National Taiwan University Wei-Kai Wu Department of Medical Research, National Taiwan University Hospital https://orcid.org/0000-0002- 9476-8998 Suraphan Panyod Institute of Food Science and Technology, National Taiwan University Po-Yu Liu National Taiwan University https://orcid.org/0000-0003-1290-0850 Hsiao-Li Chuang National Laboratory Animal Center, National Applied Research Laboratories Yi-Hsun Chen Department of Internal Medicine, College of Medicine, National Taiwan University Qiang Lyu Department of Chemistry, National Taiwan University Hsiu-Ching Hsu School of Pharmacy, College of Medicine, National Taiwan University Tzu-Lung Lin Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University Ting-Chin Shen Division of Gastroenterology, Perelman School of Medicine, University of Pennsylvania Yu-Tang Yang Department of Internal Medicine, College of Medicine, National Taiwan University Hsin-Bai Zou Department of Chemistry, National Taiwan University Huai-Syuan Huang Institute of Food Science and Technology, National Taiwan University Yu-En Lin Institute of Food Science and Technology, National Taiwan University Chieh-Chang Chen

Upload: others

Post on 04-Apr-2022

2 views

Category:

Documents


0 download

TRANSCRIPT

Dietary Exposure to Antibiotic Residues FacilitatesMetabolic Disorder by Altering the Gut Microbiotaand Bile Acid CompositionRou-An Chen 

Institute of Food Science and Technology, National Taiwan UniversityWei-Kai Wu 

Department of Medical Research, National Taiwan University Hospital https://orcid.org/0000-0002-9476-8998Suraphan Panyod 

Institute of Food Science and Technology, National Taiwan UniversityPo-Yu Liu 

National Taiwan University https://orcid.org/0000-0003-1290-0850Hsiao-Li Chuang 

National Laboratory Animal Center, National Applied Research LaboratoriesYi-Hsun Chen 

Department of Internal Medicine, College of Medicine, National Taiwan UniversityQiang Lyu 

Department of Chemistry, National Taiwan UniversityHsiu-Ching Hsu 

School of Pharmacy, College of Medicine, National Taiwan UniversityTzu-Lung Lin 

Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang GungUniversityTing-Chin Shen 

Division of Gastroenterology, Perelman School of Medicine, University of PennsylvaniaYu-Tang Yang 

Department of Internal Medicine, College of Medicine, National Taiwan UniversityHsin-Bai Zou 

Department of Chemistry, National Taiwan UniversityHuai-Syuan Huang 

Institute of Food Science and Technology, National Taiwan UniversityYu-En Lin 

Institute of Food Science and Technology, National Taiwan UniversityChieh-Chang Chen 

Department of Internal Medicine, National Taiwan University Hospital https://orcid.org/0000-0003-2953-210XChi-Tang Ho 

Department of Food Science, Rutgers UniversityHsin-Chih Lai 

Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang GungUniversityMing-Shiang Wu 

National Taiwan University Hospital https://orcid.org/0000-0002-1940-6428Cheng-Chih Hsu 

Department of Chemistry, National Taiwan University https://orcid.org/0000-0002-2892-5326Lee-Yan Sheen  ( [email protected] )

National Taiwan University

Letter

Keywords: antibiotic residue, metabolic dysfunction, tylosin, gut microbiota

Posted Date: November 18th, 2021

DOI: https://doi.org/10.21203/rs.3.rs-1066621/v1

License: This work is licensed under a Creative Commons Attribution 4.0 International License.  Read Full License

1

Dietary Exposure to Antibiotic Residues Facilitates Metabolic Disorder by 1

Altering the Gut Microbiota and Bile Acid Composition 2

Rou-An Chena†, Wei-Kai Wub†, Suraphan Panyoda, Po-Yu Liuc, Hsiao-Li Chuangd, Yi-3

Hsun Chenc, Qiang Lyue, Hsiu-Ching Hsuf, Tzu-Lung Ling, Ting-Chin David Shenh, 4

Yu-Tang Yangc, Hsin-Bai Zoue, Huai-Syuan Huanga, Yu-En Lina, Chieh-Chang Cheni, 5

Chi-Tang Hoj, Hsin-Chih Laig, Ming-Shiang Wuc,i, Cheng-Chih Hsue* and Lee-Yan 6

Sheena,k,l* 7

aInstitute of Food Science and Technology, National Taiwan University, Taipei, 8

Taiwan; bDepartment of Medical Research, National Taiwan University Hospital, 9

Taipei, Taiwan; cDepartment of Internal Medicine, College of Medicine, National 10

Taiwan University, Taipei, Taiwan; dNational Laboratory Animal Center, National 11

Applied Research Laboratories, Taipei, Taiwan; eDepartment of Chemistry, National 12

Taiwan University, Taipei, Taiwan; fThe Metabolomics Core Laboratory, Center of 13

Genomic Medicine, National Taiwan University, Taipei, Taiwan; gDepartment of 14

Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung 15

University, Taoyuan, Taiwan; hDivision of Gastroenterology, Perelman School of 16

Medicine, University of Pennsylvania, Pennsylvania, USA; iDepartment of Internal 17

Medicine, National Taiwan University Hospital, Taipei, Taiwan; jDepartment of Food 18

Science, Rutgers University, New Brunswick, New Jersey, USA; kCenter for Food and 19

Biomolecules, National Taiwan University, Taipei, Taiwan; lNational Center for Food 20

Safety Education and Research, National Taiwan University, Taipei, Taiwan 21

22

†There authors contributed equally for the study. 23

*Corresponding authors: 24

Lee-Yan Sheen, No. 1, Sec. 4, Roosevelt Rd., Taipei, Taiwan 10617; Tel: 886-2-25

33661572; E-mail: [email protected] 26

Cheng-Chih Hsu, No. 1, Sec. 4, Roosevelt Rd., Taipei, Taiwan 10617; Tel.: 886-27

33661681; E-mail: [email protected] 28

29

2

Dietary Exposure to Antibiotic Residues Facilitates Metabolic Disorder by 30

Altering the Gut Microbiota and Bile Acid Composition 31

Low dose antibiotic residues in food potentially contribute to obesity and metabolic 32

dysfunction1. However, the effect of chronic exposure to very low-dose antibiotic 33

residue (~1000-fold lower than the therapeutic dose) on gut microbiota and host 34

metabolism is poorly understood. Herein the effect of exposure to a residual dose 35

of tylosin—an antibiotic growth promoter—on host metabolism and gut 36

microbiota is explored in a mouse model. Theoretical maximal daily intake 37

(TMDI) dose of tylosin facilitates high-fat diet-induced obesity, induces insulin 38

resistance, and perturbs gut microbiota composition. Moreover, obesity-related 39

phenotypes are transferrable to germ-free recipient mice following fecal 40

microbiota transplantation. Tylosin TMDI exposure restricted to early life is 41

sufficient to induce metabolic complications, alter the abundance of specific 42

bacteria related to host metabolic homeostasis later in life, and modify the 43

composition of short-chain fatty acids and bile acids. Finally, tylosin TMDI 44

exposure induces lasting metabolic consequences via elevating the ratio of primary 45

to secondary bile acids and its downstream FGF15 signaling pathway. Hence, 46

exposure to very low doses of antibiotic residues, whether continuously or in early 47

life, can exert long-lasting effects on host metabolism by altering gut microbiota 48

and their metabolites. 49

Antibiotics used as growth promoters in livestock and animal husbandry can be 50

detected in animal-derived food2-5. Although the maximum residue limits (MRLs) of 51

these veterinary antibiotics has been evaluated, these estimated levels do not consider 52

the effects of antibiotic residues on gut microbiota and microbial metabolites, or the 53

effects of altered gut microbiota on host metabolism6. Recent studies have established 54

3

an association between exposure to veterinary antibiotics and children obesity1,7. 55

Additionally, animal studies have indicated that exposure to sub-therapeutic doses of 56

antibiotics leads to obesity and NAFLD8-10. In this study, we aimed to further elucidate 57

whether chronic exposure to acceptable residual amounts of antibiotic, which doses are 58

hundred-fold lower than the sub-therapeutic doses used in previous studies, could cause 59

obesity complications. Thus, C57BL6J mice were fed an acceptable daily intake (ADI, 60

0.37 mg/kg/day) and theoretical maximum daily intake (TMDI, 0.047 mg/kg/day) dose 61

of tylosin (Fig. 1a). The ADI dose is the maximum daily dose with no adverse effect6, 62

while the TMDI dose is an estimate of maximum dietary intake level obtained for 63

MRLs and the sum of average daily per capita consumption for each food product6, 64

which was used for simulating the ingestion of antibiotic residues through food 65

consumption in the present study. Additionally, a normal-chow diet and high-fat diet 66

were provided to assess the potential synergistic effects of antibiotics and diet on host 67

metabolism. 68

Compared with NCD-CON (normal chow diet/control) mice, NCD-ADI and 69

NCD-TMDI mice showed significantly greater weight gain from weaning to 5 weeks of 70

age. NCD-ADI mice also exhibited higher weight gain from weeks 13 to 15 (Fig. 1b) 71

with no significant changes detected among the NCD groups in relative fat or lean mass 72

(Extended Data Fig. 1a, b). In contrast, high-fat diet (HFD)-ADI and HFD-TMDI 73

mice exhibited significantly increased weight gain compared with HFD-CON mice 74

from weaning to 17 weeks of age (Fig. 1c). HFD-ADI and HFD-TMDI mice had 75

increased relative fat mass at weeks 8, 12, and 17, compared with HFD-CON mice (Fig. 76

1d), whereas HFD-TMDI mice had decreased relative lean mass (Fig. 1e). Likewise, 77

HFD-TMDI mice had increased visceral fat, including epididymal and perinephric 78

adipose tissues (Fig. 1f, g and Extended Data Fig. 1 c, d). HFD-ADI and HFD-TMDI 79

4

mice also exhibited adipocyte hypertrophy (Fig. 1h) and elevated adipocyte size (Fig. 80

1j) compared with HFD-CON mice. Hence, tylosin-induced adiposity is evident early in 81

life for NCD and HFD mice; however, the continuous effects of tylosin-induced 82

adiposity were preferentially observed in HFD-fed mice. 83

We next sought to investigate whether exposure to residual tylosin dose is 84

causally associated with metabolic complications, including hepatic abnormalities and 85

glucose homeostasis. Given that tylosin increased the fat mass in HFD-fed mice, not 86

NCD-fed mice, additional investigations were conducted in the HFD-fed mice. 87

Histological examination of the liver revealed that tylosin-treated mice exhibited 88

increased lipid droplet formation (Fig. 1i), more inflammatory foci, and higher fatty 89

liver scores (Fig. 1k), suggesting that the presence of residual tylosin caused more 90

severe NAFLD. Both HFD-ADI and HFD-TMDI mice exhibited a trend toward higher 91

plasma glucose during the oral glucose tolerance test (OGTT) (Extended Data Fig. 2a) 92

and fasting glucose test (Extended Data Fig. 2b), with an increased OGTTAUC found in 93

HFD-TMDI mice (Fig 2l). The fasting insulin level and homeostasis model assessment 94

of insulin resistance (HOMA-IR) index were also elevated in HFD-ADI mice, 95

compared with HFD-CON mice (Fig. 2m, n). Hence, a residual dose of tylosin can 96

exacerbate HFD-induced adverse effects on metabolic complications. Remarkably, 97

although the tylosin ADI dose is defined as the maximum antibiotic dose that can be 98

ingested daily without health risk6, it facilitated obesity-related metabolic disorders in 99

our mice model. As such, the tolerance level of antibiotics may need to be re-evaluated 100

in consideration of chronic metabolic diseases which were emerging in western 101

lifestyle. 102

As sub-therapeutic doses of antibiotic treatments have been shown to disrupt the 103

development and maturation of gut microbiota with metabolic consequences8,11, we next 104

5

asked whether ADI and TMDI doses of tylosin alter gut microbiota composition. To 105

this end, fecal 16S rRNA sequencing was performed in mice at 3 (weaning), 8, and 17 106

weeks of age to elucidate changes in the gut microbiota. Compared with HFD-TMDI 107

and HFD-CON mice, HFD-ADI mice had significantly reduced Shannon indices at 108

week 3, which markedly increased during weeks 3–17 (Fig. 2a). Principal coordinate 109

analysis (PCoA) based on Bray-Curtis dissimilarity further revealed that the 110

microbiomes of NCD and HFD mice clustered separately (Extended Data Fig. 3a), 111

indicating that diet may represent the most critical factor influencing the composition of 112

the gut microbiota, while residual tylosin dose exhibited a secondary effect in shifting 113

the gut microbiota in a dose-dependent manner according to the PCoA2 axis (Extended 114

Data Fig. 3a). 115

Further, we analyzed the subsets of NCD (Extended Data Fig. 3b) and HFD-116

fed mice (Fig. 2b) separately to observe the effect of tylosin on age-related microbiota 117

development. In both groups, ADI dose of tylosin substantially influenced the gut 118

microbiota composition, while TMDI dose of tylosin showed a lesser impact. Notably, 119

the gut microbiota of tylosin-treated mice was obviously changed at the early-life stage 120

(week 3) in both NCD and HFD groups, with gradually shifted toward the control group 121

at weeks 8 and 17, respectively. Bray-Curtis distances from HFD-CON mice to HFD-122

ADI or HFD-TMDI mice were also shortened as the mice aged (Fig. 2c). These 123

findings suggest that gut microbiota has increased susceptibility to the perturbation 124

caused by residual tylosin in early life, yet susceptibility may decrease at later stages, 125

possibly due to the establishment and maturation of gut microbiota. 126

To further investigate differentially abundant bacteria modified by tylosin TMDI 127

dose, we identified significantly-altered bacteria at the amplicon sequence variants 128

(ASV) level (defined as log2-fold change >1, p values < 0.05; Fig. 2d, Extended Data 129

6

Fig. 4). Oscillibacter, Blautia and Parasutterella, which are associated with obesity and 130

inflammatory bowel disease12-14, were significantly enriched in HFD-TMDI mice, 131

whereas Bifidobacterium was significantly reduced (Fig. 2d). TMDI dose of tylosin 132

may decrease beneficial bacteria while enriching pathogenic bacteria. 133

We next assessed the association between obesity-related features of 17-week-134

old HFD-fed mice and their gut microbiota composition by the envfit function in the 135

vegan R package, the result is displayed in PCoA (Fig. 2e). The results showed that 136

vectors indicating both obesity-related biomarkers (the blue arrows) and antibiotic 137

exposure (the red arrow) were arrowed toward the mice treated with tylosin in the same 138

direction, indicating the obesity outcomes were associated with the tylosin-shifted gut 139

microbiome. We further performed Spearman's correlation between PCoA1 and PCoA2 140

of gut microbiota against individual obesity-related parameters (Supplementary Table. 141

1) and found that the PCoA2 was significantly correlated with obesogenic phenotypes 142

(fat gain, fat mass, and relative fat mass), fatty liver score, and insulin resistance 143

parameters (fasting insulin and HOMA-IR). 144

To understand the overall obesity phenotype and its relationship with tylosin 145

altered-microbiome, we performed a dimension reduction of the obesity and metabolic 146

disorders related biomarkers by using the principal component analysis (PCA). The 147

result showed that the obesity phenotypes in tylosin-treated mice were significantly 148

different from control mice (p < 0.0001; Extended data Fig. 5) We subsequently 149

calculated a correlation between PC1obesity biomarkers and the PCoA2microbiota which 150

displaying a positive correlation (r = 0.71, p = 0.003). 151

Given the above association between the metabolic disorder phenotypes and 152

tylosin-altered gut microbiota, we conducted a fecal microbiota transplantation (FMT) 153

study to investigate their causative role. Since a TMDI dose can adequately simulate 154

7

human exposure to antibiotics in food, feces from HFD-TMDI mice were transplanted 155

to germ-free mice. Compared with germ-free mice that received feces from HFD-CON 156

mice (FMT-CON), the HFD-TMDI recipient mice (FMT-TMDI) showed higher body 157

weight at 11, 12, 13, 14 weeks of age (Fig. 2g), increased weight gain 2 weeks post-158

FMT (Fig. 2h), and slightly elevated fat mass at week 20 (Fig. 2i), implying that the 159

microbiome from HFD-TMDI mice increased the adiposity of the recipient mice. 160

Additionally, HFD-TMDI mice exhibited increased plasma glucose levels in OGTTAUC, 161

and a higher HOMA-IR index (Fig. 2j, k). Thus, a TMDI dose of tylosin-altered 162

microbiota induced obesity and insulin resistance in germ-free recipients, indicating that 163

residual tylosin exposure facilitates metabolic disorders through the alternation of gut 164

microbiota. 165

Growing evidence indicates that antibiotic exposure during infancy, which has 166

been considered the critical window of gut microbiota development8, is associated with 167

increased risk of being overweight and obese15,16. More specifically, antibiotic exposure 168

during early life, even at sub-therapeutic levels, disturbs the colonization and maturation 169

of the intestinal microbiota, leading to lasting effects on the metabolism of the host8,17. 170

Accordingly, an early-exposure experiment was conducted to investigate the influence 171

of early-life exposure to a TMDI dose of tylosin on obesity-related phenotypes and the 172

gut microbiota (Fig. 3a). 173

Cont-TMDI mice, which were continuously exposed to TMDI dose of tylosin 174

throughout the experimental period, displayed continuously elevated body weight, 175

relative fat mass, visceral fat mass, and OGTTAUC, compared with HFD-CON mice 176

(Fig. 3b-e). Interestingly, Early-TMDI mice, which were exposed to tylosin TMDI 177

during pregnancy and the nursing period, also exhibited consistently elevated body 178

weight, relative fat mass, fasting insulin, and HOMA-IR index values after cessation of 179

8

tylosin exposure (Fig. 3b-g). These findings suggest that exposure to antibiotic residue 180

in food products during early life is sufficient to induce long-lasting effects on 181

metabolism and lead to obesity. Compared with previous studies showing that antibiotic 182

exposure limited to early life induces elevated adiposity later in life8, our findings 183

further demonstrate that metabolic complications can emerge later in life even with a 184

food-grade low dose of antibiotic residue. 185

The overall gut microbiota composition based on PCoA of Bray-Curtis distances 186

showed that tylosin influenced the gut microbiota composition at weeks 5 and 20 (p < 187

0.05; Fig. 3h). Compared with CON mice, Cont-TMDI mice showed a greater 188

difference than Early-TMDI mice, suggesting that continuous tylosin exposure has a 189

more significant effect compared to exposure restricted to early life (Fig. 3h). 190

Furthermore, the Early-TMDI and CON mice exhibited greater differences at week 5 191

than week 20, possibly due to establishment of the gut microbiota (Fig. 3h). 192

Interestingly, despite Early-TMDI mice exhibited a more similarly resembled microbial 193

community to that of CON mice at week 20 than at week 5 (Fig. 3h), their fat mass at 194

week 20 was more significantly increased then at week 5 (Fig. 3c). This finding 195

suggests that the impaired metabolic phenotype can persist despite resilience of the 196

microbiota. That is, minor disruption of the microbiota in early life caused by antibiotic 197

residue appears to be sufficient for inducing significant adiposity18,19. 198

Studies have indicated that microbial colonization can be perturbed by exposure 199

to antibiotic early in life, which, in turn, contributes to metabolic disorders in 200

adulthood20,21. Thus, we proposed that although the overall gut microbiota composition 201

can restore in later life, the abundance of specific bacteria associated with metabolic 202

homeostasis of the host are persistently depleted. To investigate this hypothesis, we 203

identified 32 bacterial genera significantly altered by early or continuous exposure to 204

9

TMDI dose of tylosin (Fig. 3i), then examined the association between these bacterial 205

genera and obesity-related phenotypes (Fig. 3j). Bacterial genera increased in both 206

Early-TMDI and Cont-TMDI mice, including Anaerofustis, and demonstrated a 207

significant positive correlation with obesity-related phenotypes (Fig. 3i). In contrast, 208

genera that were depleted in Early-TMDI and Cont-TMDI mice, including bacteria 209

belonging to the Lachnospiraceae and Ruminococcaceae families, exhibited a 210

significantly negative correlation with obesity-related phenotypes (Fig. 3i). Previous 211

studies have also reported that these bacteria are related to obesity. Anaerofustis, the 212

tylosin-enriched bacterium is increased in obese humans22, while tylosin-depleted 213

Ruminococcaceae and Lachnospiraceae are associated with lower long-term weight 214

gain23. Notably, despite the discontinuation of tylosin exposure in Early-TMDI mice at 215

3 weeks of age, several bacterial genera that are negatively correlated with obesity-216

related phenotypes remained diminished at week 20. Consistent with our hypothesis, 217

although early exposure to residual dose of antibiotics may not cause a significant 218

impact on the overall microbial composition in later life, early-depletion of certain 219

bacteria, that continued to be lifelong lost, may contribute to metabolic dysfunctions in 220

later life. 221

Given that antibiotics reportedly alter short-chain fatty acid (SCFA), bile acids 222

composition and their signaling pathways, thereby leading to metabolic 223

consequences24,25, we next investigated the effect of TMDI dose of tylosin on these two 224

major metabolites. SCFA analysis showed that propionic acid and butyric acid, two 225

main SCFAs associated with enhanced intestinal barrier function and insulin 226

sensitivity26,27, exhibited decreasing trends in Cont-TMDI mice (Fig. 4a). Isovaleric 227

acid, a branched SCFA reported to improve insulin-stimulated glucose uptake and 228

enhance insulin sensitivity28, was significantly decreased in Cont-TMDI mice (Fig. 4a). 229

10

Reduction of SCFAs could be attributed to the reduction of Lachnospiraceae and 230

Ruminococcaceae in Early-TMDI and Cont-TMDI mice (Fig. 3i)29,30. 231

The total amount of bile acids and the ratio of conjugated to unconjugated bile 232

acids were not significantly altered by tylosin exposure (Extended Data Fig. 6a, b). 233

Notably, the ratio of primary bile acids (PBA) to secondary bile acids (SBA) was 234

significantly increased in Cont-TMDI and Early-TMDI mice (Fig. 4b). Indeed, a 235

decreased PBA/SBA ratio in NAFLD patients who have undergone bariatric surgery is 236

associated with improved insulin sensitivity, indicating that an increased PBA/SBA 237

ratio tends to induce metabolic disorder31,32. To further investigate the effect of tylosin 238

on the conversion of PBA to SBA, the detected bile acids were classified into non-12-239

OH bile acids (Fig. 4c), muricholic acids (MCA; Fig. 4d), and 12-OH bile acids (Fig. 240

4e) based on their metabolic pathway33. The levels of PBAs, namely, chenodeoxycholic 241

acid, α-MCA, 𝛽-MCA, and cholic acid (Fig. 4c-e) were significantly increased, whereas 242

those of the SBAs, including ursodeoxycholic acid (UDCA) and ω-MCA (Fig. 4c, d), 243

were decreased in tylosin-treated mice, possibly due to inhibition of bacteria associated 244

with epimerization and dehydroxylation of PBAs, such as Clostridia, 245

Peptostreptococcus34 (Extended Data Fig. 6c, d). 246

Antibiotics have also been found to increase the PBA/SBA ratio with a 247

subsequent decrease in plasma fibroblast growth factor 19 (FGF19; human orthologue 248

of FGF15). FGF19 is an insulin-like hormone secreted by intestinal epithelium to 249

regulate hepatic lipid and glucose metabolism by binding to FGFR4 in the liver, thereby 250

decreasing peripheral insulin sensitivity24,35,36. Moreover, Parasutterella, the genus 251

enriched in tylosin-treated mice (Fig. 2e and Extended Data Fig. 4a), reportedly 252

increases 𝛽-MCA and decreases ileal Fgf15 gene expression13. Hence, the 253

FGF15/FGFR4 signaling pathway was further explored. Tylosin reduced ileal FGF15 254

11

expression (Fig. 4g) and portal vein FGF15 levels (Fig. 4f) with subsequent reduction in 255

hepatic FGFR4 levels (Fig. 4h). Collectively, tylosin TMDI-treated mice showed an 256

increased PBA/SBA ratio, as well as lower FGF15 levels in the ileum and portal vein, 257

and decreased expression of hepatic FGFR4, which may cause metabolic disorders by 258

affecting metabolism-related signaling pathways in the liver35-37. 259

This study has certain limitations. First, we only investigated the in vivo effects 260

of one antibiotic (tylosin), whereas exposure to other antibiotics may lead to different 261

outcomes owing to the specific antimicrobial action and spectrum of each antibiotic. 262

Second, the human dietary pattern is dynamic, with daily exposure to multiple types of 263

antibiotics, and even pesticides. Future research is warranted to investigate the effects of 264

other antibiotics at residual amounts, as well as the combination of different antibiotics 265

to better reflect real-life conditions. 266

In conclusion (Fig. 4i), tylosin at ADI and TMDI doses, which are generally 267

regarded as harmless, facilitated HFD-induced metabolic complications and perturbated 268

the gut microbiota composition. Moreover, the altered gut microbiota was critical for 269

tylosin TMDI-induced metabolic consequences, suggesting that continuous exposure to 270

very low doses of antibiotic residues in food can affect human metabolism by altering 271

gut microbiota. Since the gut microbial community is dynamic and susceptible to 272

environmental shifts in early life, early exposure to residual doses of tylosin was 273

sufficient to persistently deplete specific bacteria involved in the metabolic homeostasis 274

of the host, which may induce lasting metabolic consequences. Finally, we 275

demonstrated that residual tylosin altered the conversion of bile acids with downstream 276

effects on the FGF15 signaling pathway, a possible mechanism that participates in the 277

lasting metabolic disorders. Taken together, these findings indicate that permissible 278

12

exposure levels of antibiotic residues should be re-established while considering its 279

impact on the gut microbiota, for which this study provides valuable insights. 280

281

13

Materials and Methods 282

Antibiotic selection and dose calculation 283

Tylosin was selected as a model antibiotic growth promoter due to its high annual 284

consumption as a veterinary antibiotic (Bureau of Animal and Plant Health Inspection 285

and Quarantine, 2014) and its presence in animal-derived food38-40. The ADI and TMDI 286

doses were obtained from the World Health Organization Technical Report Series41. 287

Experimental design of the animal studies 288

Animal experiments were performed with permission from the Institutional Animal 289

Care and Use Committee of National Taiwan University (approval number: NTU-106-290

EL-051and NALC 107-0-006-R2). All mice were purchased from the National 291

Laboratory Animal Center (Taipei City, Taiwan). For animal studies Ⅰ and Ⅲ, each 292

experimental group comprised 3 C57BL/6J mother mice. The number of mice in each 293

experimental group varied according to the number of offspring of the mother mice. 294

Ⅰ. Antibiotic residue exposure model 295

C57BL/6J mother mice received tylosin at ADI (0.37 mg/kg) and TMDI (0.047 mg/kg) 296

doses 10 days before giving birth, and their offspring were continuously administered 297

the antibiotic. Tylosin was administered through drinking water. Control mice (CON) 298

did not receive antibiotics. To investigate the syngenetic effect of tylosin and diet, the 299

offspring were randomly divided into NCD (MFG, Oriental Yeast Co., Ltd., Tokyo, 300

Japan) and HFD (60% kcal from fat) groups (D12492, Research Diets, New Brunswick, 301

NJ, USA). Body composition was measured at 8, 12, and 17 weeks. The metabolic 302

measurements and OGTT were performed at 20 weeks prior to sacrifice. After 303

euthanasia by carbon dioxide inhalation, blood and tissue samples were collected and 304

stored at −80 ℃. 305

Ⅱ. Fecal microbial transplantation study 306

14

Eight-week-old C57BL/6 germ-free mice were randomly divided into fecal microbiota 307

transplantation-control (FMT-CON) and FMT-TMDI groups, which were transplanted 308

with fecal microbiota from HFD-CON and HFD-TMDI mice, respectively. After FMT, 309

the recipient mice were housed in two independent isolators and fed with irradiated 310

HFD until 20 weeks of age. Before the recipient mice were euthanized, the body 311

composition analysis and OGTT were performed. 312

Ⅲ. Early-life exposure model 313

The mice were divided into three groups: CON, feeding conditions were the same as 314

those of HFD-CON; Early-TMDI, exposure duration was limited to the gestation and 315

lactation period; Cont-TMDI, feeding conditions were the same as those of HFD-TMDI. 316

TMDI administration of tylosin started on day 10 of gestation. Before the mice were 317

weaned, both Early-TMDI and Cont-TMDI mice were exposed to a TMDI dose of 318

tylosin. After weaning, only the Cont-TMDI mice were continuously exposed to tylosin 319

through drinking water. Body composition was measured at 5, 10, 15, and 20 weeks. 320

The OGTT was performed before euthanizing the mice at 20 weeks. 321

Body composition analysis 322

Body composition was determined using Minispec LF50 TD-NMR Body Composition 323

Analyzer (Bruker, Billerica, MA, USA), which provides the measurement of body 324

weight and lean and fat mass. The relative fat mass was calculated as the fat mass 325

(g)/body weight (g) ratio. 326

Glucose and insulin sensitivity 327

For the OGTT, mice were deprived of food for 5 h. Blood was collected from the 328

submandibular vein, and the glucose levels were measured using a glucometer (Roche, 329

Basel, Switzerland) at 0, 15, 30, 60, 90, and 120 min after oral administration with 2 330

g/kg glucose. The fasting insulin levels were detected by an enzyme-linked 331

15

immunosorbent assay (ELISA) kit (Mercodia, Uppsala, Sweden). The HOMA-IR index 332

was calculated using the formula: fasting glucose (nmol/L) × fasting insulin 333

(µU/mL)/22.5 42. 334

Histopathological analysis of liver and adipose tissue 335

Liver and adipose tissue sections were collected and fixed in 10% formalin solution. 336

Histopathological analysis was performed by the formalin-fixed, paraffin-embedded, 337

and hematoxylin and eosin (H&E)-stained slide method. The fatty liver score was 338

estimated by a pathologist as previously described43. The fatty liver score included the 339

evaluation of steatosis (macrovesicular, microvesicular, and hypertrophy) and 340

inflammation (number of inflammatory foci). The visceral adipocyte quantification was 341

performed by HCImage Live software (HCImage, Sewickley, PA, USA). 342

Quantification of plasma lipopolysaccharides 343

Plasma samples were centrifuged and added to HEK-Blue™ mTLR4 cell lines in the 344

HEK-Blue™ Detection medium (InvivoGen, San Diego, CA, USA). After 24 h of 345

incubation, the color of secreted embryonic alkaline phosphatase (SEAP) released by 346

the reporter cells was measured by a spectrophotometer at 620 nm44. 347

Metabolic measurements 348

Metabolic measurements (food intake, locomotor activity, VO2 consumption, and VCO2 349

production) were made using the Promethion metabolic phenotyping system (Sable 350

Systems, Las Vegas, NV, USA). Monitoring was performed for 24–48 h with ad libitum 351

access to food and water after mice had been acclimatized to cages for 6–12 h. 352

Fecal microbiota extraction and 16S rRNA gene sequencing 353

The mice in each group were randomly selected for fecal microbiota analysis. Mice 354

feces were collected and frozen immediately at −80 °C. Fecal DNA extraction and 355

library preparation and sequencing were performed according to the protocol provided 356

16

by QIAGEN (QIAGEN, MD, USA). Briefly, fecal microbial DNA was isolated by 357

using QIAamp 96 PowerFecal QIAcube HT Kit (QIAGEN, Germantown, MD, USA). 358

The 16S V4 region was amplified by the forward primer (F515, 5ʹ-GTGCCAGCMG 359

CCGCGGTAA-3ʹ) and the reverse primer (R806, 5ʹ-GGACTACHVGGGTWTCTAAT-360

3ʹ). The reaction conditions were as follows: 3 min at 95 °C, followed by 25 cycles of 361

95 °C for 30 s, 55 °C for 30 s, 72 °C for 30 s, and 5 min at 72 °C for a final extension. 362

For library construction, the amplified PCR product was attached with Illumina 363

sequencing adapters by Nextera XT Index Kit then purified using AMPure XP beads. 364

The library quantification was conducted using the DNA 1000 kit and 2100 Bioanalyzer 365

Instrument (Agilent Technologies, Santa Clara, CA, USA). The sequencing (paired-end 366

reads, 2 × 150 bp) was performed by Illumina NextSeq (Illumina, San Diego, CA, 367

USA). 368

Bioinformatic analysis for microbial taxonomic profiling 369

The amplicon sequences were processed by a QIIME 2 pipeline (version 2019.10, 370

https://qiime2.org). The primer sequences of raw reads were trimmed using the cutadapt 371

plugin. The trimmed single-end (forward) sequences were subsequently denoised with 372

the DADA2 plugin of QIIME245. To obtain qualified data, we truncated reads to 130 bp 373

from the 3ʹ end based on the quality score. The DADA2 outputs of high confident ASVs 374

went through quality filtering, denoising, and chimeric-read removing. The classify-375

consensus-vsearch plugin was applied for taxonomy assignment by aligning against 376

SILVA 132 99% 16S rDNA sequences, and identity cutoff was set at ≥80% sequence 377

similarity by default. The vegan package in R was used to calculate Shannon index and 378

perform principal coordinate analysis (PCoA) based on the Bray–Curtis distance46. The 379

permutation multivariate analysis of variance (ANOVA) using distance matrixes 380

(adonis) was performed to calculate the significance of microbiota composition among 381

17

groups. The PCoA plot of gut microbiota with an association between obesity-related 382

features of 17-week-old HFD-fed mice was performed by the envfit function in the 383

vegan R package, displaying as the vectors (p < 0.05). 384

Correlation analysis 385

The Spearman’s correlation analysis between PCoA1 and PCoA2 of gut microbiota 386

against individual obesity-related parameters was performed with FDR-adjusted p-387

value. The correlation coefficient and p-value of Spearman’s correlation between 388

PC1obesity biomarkers (representing the overall obesity phenotype) and PCoA2microbiota 389

(representing the gut microbiota composition) was calculated. 390

Principal component analysis (PCA) of obesity biomarkers 391

PCA was used for dimension reduction of obesity biomarkers10, including body weight 392

at 17 weeks of age, fat mass at 17 weeks of age, relative fat mass at 17 weeks of age, 393

weight gain between 3 to 17 weeks of age, fat mass gain between 8 to 17 weeks of age, 394

fasting glucose, OGTTAUC, fasting insulin, HOMA-IR, weight of eWAT, and fatty liver 395

score. 396

Fecal short-chain fatty acid analysis 397

The SCFAs we analyzed included acetic acid (C2), propionic acid (C3), butyric acid 398

(C4), isobutyric acid (C4), valeric acid (C5), and isovaleric acid (C5). Briefly, 20 mg of 399

the raw feces was dissolved in 500 μL of a 0.5% H3PO4 aqueous solution and 400

homogenized with Geno/Grinder® at 1,000 rpm for 2 min. The sample solutions were 401

then centrifuged at 18,000 × g for 10 min at 4 °C to separate depositions. Afterwards, 402

285 µL of the supernatant was collected in a clean centrifuge tube, and 15 µL of 403

acetate-d3 was added. For the liquid–liquid extraction of SCFAs, 300 μL of butanol and 404

the final solution were mixed and centrifuged. Last, 20 μL of internal standard 405

propionate-d5 was added to 180 μL of the upper organic layer. GC–MS analysis was 406

18

performed using an Agilent 7890A gas chromatograph (Agilent Technologies) coupled 407

with a Pegasus 4D GC × GC–TOF–MS system (Leco Corporation, St. Joseph, MI, 408

USA) using an VF-WAXms capillary column47. 409

Fecal bile acid analysis 410

The bile acid quantification included four primary bile acids (𝛼-MCA, 𝛽-MCA, CA, 411

and UDCA), four secondary bile acids (𝜔-MCA, CDCA, DCA, and LCA), and seven 412

conjugated bile acids (T𝛽-MCA, TUDCA, TCA, GCA, TCDCA, TDCA, TLCA). 413

Briefly, 20 mg of raw feces was dissolved in 200 μL of 70% d3-cholic acid aqueous 414

solution containing 2-ppm d4-cholic acid as internal standard, and it was homogenized 415

using an ultrasonicator for 30 min. The sample solutions were then centrifuged at 416

18,000 × g for 5 min. LC–MS analysis was performed using UltiMate 3000 liquid 417

chromatography system (Thermo Fisher Scientific, Dreieich, Germany) coupled with a 418

high-resolution Q Exactive Plus instrument equipped with the ESI source (Thermo 419

Fisher Scientific), and an Acquity HSS T3 (2.1 × 100 mm, 1.7 µm) column (Waters, 420

Milford, MA, USA). 421

Biochemical analysis of the FGF15–FGFR4 pathway 422

For western blotting, frozen tissues were lysed in lysis buffer containing 7-M urea, 2-M 423

thiourea, 2% CHAPS, 0.002% bromophenol blue, 60-mM DTT, and a protease and 424

phosphatase inhibitor cocktail. The cell lysates were sonicated for 5 min and centrifuged 425

at 17,500 × g for 30 min at 4 °C. Total protein content was measured based on the 426

Bradford assay with a Bradford reagent (Bioshop Canada Inc., Burlington, Canada) and 427

an ELISA reader. For the loading buffer, 62.5-mM Tris-HCl, 10% glycerol, 2% SDS, 428

and 0.01% bromophenol blue were mixed with the protein samples then heated at 95 °C 429

for 10 min for protein denaturation. Proteins were separated by 10% or 12% 430

SDS−polyacrylamide gel electrophoresis, they were then transferred onto 431

19

polyvinylidene difluoride membranes (Millipore, Burlington, MA, USA). After 432

blocking with 5% bovine serum albumin in tris-buffered saline with Tween 20, the 433

membranes were incubated with rabbit monoclonal anti–GAPDH antibody (1:5000; 434

Catalog No. 5174; Cell Signaling, Danvers, MA, USA), or rabbit polyclonal anti–435

FGF15 antibody (1:500; Catalog No. ab229630; Abcam, Cambridge, UK), or rabbit 436

polyclonal anti–FGFR4 antibody (1:500; Catalog No. ab119378; Abcam) at 4 °C for 16 437

h and subsequently incubated with HRP-linked anti-rabbit IgG antibody (1:3000; 438

Catalog No. 7074; Cell Signaling). The protein expression signal was captured and 439

quantified using the BioSpectrum AC imaging system (UVP, Upland, CA, USA) and 440

ImageJ (version 1.53; National Institutes of Health, Bethesda, MD, USA), 441

respectively48. Portal FGF15 levels were quantified using an ELISA kit (Mercodia, 442

Uppsala, Sweden). 443

Statistical analysis 444

Data were represented as mean ± standard deviation (SD) or mean ± standard error of 445

the mean (SEM). One-way ANOVA and Tukey’s range test or Student’s t-test were 446

applied for intergroup comparisons. Statistical assessment of the gut microbiome, 447

SCFAs, and bile acids was performed using Kruskal–Wallis with/without false 448

discovery rate or one-way ANOVA with Tukey’s range test. All statistical data were 449

analyzed using GraphPad Prism software (version 9.2.0; GraphPad Software, San 450

Diego, CA, USA) or RStudio (version 1.2.5001, RStudio, Boston, MA, USA). 451

452

20

Acknowledgments: This study was funded by the Ministry of Science and Technology 453

(MOST), Taiwan (MOST 106-3114-B-002-003, MOST 107-2321-B-002-039, MOST 454

108-2321-B-002-051, MOST 109-2327-B-002-005, and MOST 109-2314-B-002-064-455

MY3). We would like to acknowledge the technical support provided by the following 456

institutes: Fecal microbiota transplantation, isolator service and metabolic measurement 457

were conducted by National Laboratory Animal Center, National Applied Research 458

Laboratories, Taiwan. Analysis of bile acids was supported by the National Taiwan 459

University Consortia of Key Technologies. Analysis of short-chain fatty acid was 460

supported by the ‘Center of Precision Medicine’ from The Featured Areas Research 461

Center Program by the MOST. Body composition analysis was performed by Taiwan 462

Mouse Clinic, Academia Sinica and Taiwan Animal Consortium. We thank 463

BIOTOOLS for bioinformatics technique support. 464

Author contributions: R.A.C. designed and performed the animal study, performed 465

metabolomic, bioinformatics, and statistical analysis, and drafted the manuscript; 466

W.K.W. proposed, designed, and instructed the study; S.P. assisted the experiments and 467

bioinformatics analysis; P.Y.L. instructed the bioinformatics analysis; H.L.C. performed 468

the germ-free mice study and assisted the metabolic measurement; Y.H.C. assisted the 469

germ-free animal study and performed the LPS analysis; Q.L. instructed the bile acid 470

analysis; H.C.H. and H.B.Z. performed SCFA analysis and assisted with mass 471

spectrometry analysis; T.L.L. instructed the LPS analysis; Y.T.Y. conducted the PCR 472

and library preparation for 16S rRNA sequencing; H.S.H. and Y.E.L. instructed the 473

western blotting; S.P., T.C.D.S., W.K.W., P.Y.L. and L.Y.S. critically revised the 474

manuscript; W.K.W., L.Y.S., C.C.H., M.S.W., H.C.L., C.C.C. and C.T.H., provided 475

professional insights, techniques, and relevant resources for the study. 476

Data availability: The raw 16s rRNA sequencing data are accessible at the National 477

Center for Biotechnology Information Short Read Archive (BioProject: 478

PRJNA715326). 479

Competing interests: The authors declare that the research was conducted in the 480

absence of any commercial or financial relationships that could be construed as a 481

potential conflict of interest. 482

483

21

References 484

1 Wang, H. et al. Antibiotics detected in urines and adipogenesis in school 485

children. Environ Int 89-90, 204–211 (2016). 486

2 Brussow, H. Growth promotion and gut microbiota: insights from antibiotic use. 487

Environ Microbiol 17, 2216–2227 (2015). 488

3 Ji, K. et al. Influence of water and food consumption on inadvertent antibiotics 489

intake among general population. Environ Res 110, 641–649 (2010). 490

4 Li, N., Ho, K. W. K., Ying, G. G. & Deng, W. J. Veterinary antibiotics in food, 491

drinking water, and the urine of preschool children in Hong Kong. Environ Int 492

108, 246–252 (2017). 493

5 Wang, H. et al. Antibiotic residues in meat, milk and aquatic products in 494

Shanghai and human exposure assessment. Food Control 80, 217–225 (2017). 495

6 WHO. Principles and methods for the risk assessment of chemicals in food. 496

Environmental Health Criteria 70. 240 (2009). 497

7 Riley, L. W., Raphael, E. & Faerstein, E. Obesity in the United States - 498

dysbiosis from exposure to low-dose antibiotics? Front Public Health 1, 69 499

(2013). 500

8 Cox, L. M. et al. Altering the intestinal microbiota during a critical 501

developmental window has lasting metabolic consequences. Cell 158, 705–721 502

(2014). 503

9 Cho, I. et al. Antibiotics in early life alter the murine colonic microbiome and 504

adiposity. Nature 488, 621–626 (2012). 505

10 Mahana, D. et al. Antibiotic perturbation of the murine gut microbiome 506

enhances the adiposity, insulin resistance, and liver disease associated with high-507

fat diet. Genome Med 8, 48 (2016). 508

11 Livanos, A. E. et al. Antibiotic-mediated gut microbiome perturbation 509

accelerates development of type 1 diabetes in mice. Nat Microbiol 1, 16140 510

(2016). 511

12 Egshatyan, L. et al. Gut microbiota and diet in patients with different glucose 512

tolerance. Endocr Connect 5, 1–9 (2016). 513

13 Ju, T., Kong, J. Y., Stothard, P. & Willing, B. P. Defining the role of 514

Parasutterella, a previously uncharacterized member of the core gut microbiota. 515

ISME J 13, 1520–1534 (2019). 516

14 Lam, Y. Y. et al. Increased gut permeability and microbiota change associate 517

with mesenteric fat inflammation and metabolic dysfunction in diet-induced 518

obese mice. PLoS One 7, e34233 (2012). 519

15 Chen, L. W. et al. Implication of gut microbiota in the association between 520

infant antibiotic exposure and childhood obesity and adiposity accumulation. Int 521

J Obes (Lond) 44, 1508–1520 (2020). 522

16 Mbakwa, C. A. et al. Early Life Antibiotic Exposure and Weight Development 523

in Children. J Pediatr 176, 105–113 e102 (2016). 524

22

17 Schulfer, A. F. et al. The impact of early-life sub-therapeutic antibiotic treatment 525

(STAT) on excessive weight is robust despite transfer of intestinal microbes. 526

ISME J 13, 1280–1292 (2019). 527

18 Cox, L. M. & Blaser, M. J. Antibiotics in early life and obesity. Nat Rev 528

Endocrinol 11, 182–190 (2015). 529

19 Nobel, Y. R. et al. Metabolic and metagenomic outcomes from early-life pulsed 530

antibiotic treatment. Nat Commun 6, 7486 (2015). 531

20 Matamoros, S., Gras-Leguen, C., Le Vacon, F., Potel, G. & de La Cochetiere, 532

M. F. Development of intestinal microbiota in infants and its impact on health. 533

Trends Microbiol 21, 167–173 (2013). 534

21 Zeissig, S. & Blumberg, R. S. Life at the beginning: perturbation of the 535

microbiota by antibiotics in early life and its role in health and disease. Nat 536

Immunol 15, 307–310 (2014). 537

22 Insenser, M. et al. Gut Microbiota and the Polycystic Ovary Syndrome: 538

Influence of Sex, Sex Hormones, and Obesity. J Clin Endocrinol Metab 103, 539

2552–2562 (2018). 540

23 Menni, C. et al. Gut microbiome diversity and high-fibre intake are related to 541

lower long-term weight gain. Int J Obes (Lond) 41, 1099–1105 (2017). 542

24 Vrieze, A. et al. Impact of oral vancomycin on gut microbiota, bile acid 543

metabolism, and insulin sensitivity. J Hepatol 60, 824–831 (2014). 544

25 Reijnders, D. et al. Effects of Gut Microbiota Manipulation by Antibiotics on 545

Host Metabolism in Obese Humans: A Randomized Double-Blind Placebo-546

Controlled Trial. Cell Metab 24, 341 (2016). 547

26 Canfora, E. E., Jocken, J. W. & Blaak, E. E. Short-chain fatty acids in control of 548

body weight and insulin sensitivity. Nat Rev Endocrinol 11, 577–591 (2015). 549

27 Cani, P. D. et al. Microbial regulation of organismal energy homeostasis. Nat 550

Metab 1, 34–46 (2019). 551

28 Heimann, E., Nyman, M., Palbrink, A. K., Lindkvist-Petersson, K. & Degerman, 552

E. Branched short-chain fatty acids modulate glucose and lipid metabolism in 553

primary adipocytes. Adipocyte 5, 359–368 (2016). 554

29 Louis, P. & Flint, H. J. Formation of propionate and butyrate by the human 555

colonic microbiota. Environ Microbiol 19, 29–41 (2017). 556

30 Vacca, M. et al. The Controversial Role of Human Gut Lachnospiraceae. 557

Microorganisms 8 (2020). 558

31 Belgaumkar, A. P. et al. Changes in Bile Acid Profile After Laparoscopic Sleeve 559

Gastrectomy are Associated with Improvements in Metabolic Profile and Fatty 560

Liver Disease. Obes Surg 26, 1195–1202 (2016). 561

32 Talavera-Urquijo, E., Beisani, M., Balibrea, J. M. & Alverdy, J. C. Is bariatric 562

surgery resolving NAFLD via microbiota-mediated bile acid ratio reversal? A 563

comprehensive review. Surg Obes Relat Dis 16, 1361–1369 (2020).. 564

33 Jia, W., Wei, M., Rajani, C. & Zheng, X. Targeting the alternative bile acid 565

synthetic pathway for metabolic diseases. Protein Cell (2020). 566

23

34 Jia, W., Xie, G. & Jia, W. Bile acid-microbiota crosstalk in gastrointestinal 567

inflammation and carcinogenesis. Nat Rev Gastroenterol Hepatol 15, 111–128 568

(2018). 569

35 Ge, H. et al. Fibroblast growth factor receptor 4 (FGFR4) deficiency improves 570

insulin resistance and glucose metabolism under diet-induced obesity conditions. 571

J Biol Chem 289, 30470–30480 (2014). 572

36 Potthoff, M. J. et al. FGF15/19 regulates hepatic glucose metabolism by 573

inhibiting the CREB-PGC-1alpha pathway. Cell Metab 13, 729–738 (2011). 574

37 Shin, D. J. & Osborne, T. F. FGF15/FGFR4 integrates growth factor signaling 575

with hepatic bile acid metabolism and insulin action. J Biol Chem 284, 11110–576

11120 (2009). 577

38 Onyinye Stella, O. I., Vivienne Ezenduka, E. & John Anaelom, N. Screening for 578

tylosin and other antimicrobial residues in fresh and fermented (nono) cow milk 579

in Delta state, South-South, Nigeria. Vet World 13, 458–464 (2020). 580

39 USDA. FY2015 Food Safety and Inspection Service (FSIS) National Residue 581

Program Data. (2015). 582

40 USDA. 2016 Food Safety and Inspection Service (FSIS) National Residue 583

Program Data. (2016). 584

41 WHO. Evaluation of certain veterinary drug residues in food. WHO Technical 585

Report Series 954 (2008). 586

42 Matthews, D. R. et al. Homeostasis model assessment: insulin resistance and 587

beta-cell function from fasting plasma glucose and insulin concentrations in 588

man. Diabetologia 28, 412–419 (1985). 589

43 Liang, W. et al. Establishment of a general NAFLD scoring system for rodent 590

models and comparison to human liver pathology. PLoS One 9, e115922 (2014). 591

44 Lai, H. C. et al. Gut microbiota modulates COPD pathogenesis: role of anti-592

inflammatory Parabacteroides goldsteinii lipopolysaccharide. Gut, gutjnl-2020-593

322599 (2021). 594

45 Suzuki, T. A., Martins, F. M. & Nachman, M. W. Altitudinal variation of the gut 595

microbiota in wild house mice. Mol Ecol 28, 2378–2390, 596

doi:10.1111/mec.14905 (2019). 597

46 Oksanen, J. et al. vegan: Community Ecology Package. (2020). 598

47 Hsu, Y. L. et al. Evaluation and Optimization of Sample Handling Methods for 599

Quantification of Short-Chain Fatty Acids in Human Fecal Samples by GC-MS. 600

J Proteome Res 18, 1948–1957 (2019). 601

48 Panyod, S. et al. Diet Supplementation with Allicin Protects against Alcoholic 602

Fatty Liver Disease in Mice by Improving Anti-inflammation and Antioxidative 603

Functions. J Agric Food Chem 64, 7104–7113 (2016). 604

605

24

Figures 606

607

Fig. 1 | Residual dose of tylosin facilitates HFD-induced obesity and metabolic 608

disorders. 609

a, Experimental design of antibiotic residue exposure model. b, Body weight changes in 610

NCD and c, HFD mice (n = 8–12/group). d, Relative fat mass and e, relative lean mass 611

changes in HFD mice (n = 8–12/group). f, Weight of epididymal adipose tissue and g, 612

perinephric adipose tissue (n = 8–12/group). h-i, histological features of H&E stained in 613

epididymal adipose tissue (h) and liver (i) (n = 8–12/group). j, Adipocyte diameter of 614

epididymal adipose tissue (n = 6/group). k, Fatty liver score including steatosis 615

(macrovesicular, microvesicular, and hypertrophy) and inflammation (number of 616

inflammatory foci) (n = 8–12/group). l, Area under the curve (AUC) derived from the 617

OGTT (n = 7–8/group). m, Plasma insulin level after overnight fasting (n = 7–8/group). 618

n, HOMA-IR index represented as an indicator of insulin resistance (n = 7–8/group). 619

Data are expressed as mean ± SD. For b-e, and j, Statistical analyses were performed 620

by one-way ANOVA with Tukey’s range test within diet groups (NCD or HFD) as 621

40

50 HFD-CON

HFD-ADI

HFD-TMDI*

#

20

-30

30

-40

40

-50

50

-60

60

-70

70

-80

80

-90

90

-10

0

10

0-1

10

11

0-1

20

12

0-1

30

13

0-1

40

14

0-1

50

15

0-1

60

16

0-1

70

17

0-1

80

18

0-1

90

19

0-2

00

-10

0

10

20

30

40

Adipocyte size (µm)

Perc

cent of adip

ocyte

s (

%)

##

**

#

#

***

* *

##

**

##

**

#

**##

*

**

***

*

40

50 HFD-CON

HFD-ADI

HFD-TMDI*

#HFD-ConHFD-ADIHFD-TMDI

HFD-CON

HFD-ADI

HFD-TMDI

b c

d e f g h

j k l m n

HFD-CON

HFD-CON

HFD-ADI

HFD-ADI

HFD-TMDI

HFD-TMDI

100 μm

100 μm

100 μm

100 μm

100 μm

100 μm

i

8 12 17

0

10

20

30

40

Age (week)

Rela

tive fat m

ass (

%)

*

#

*

#

*

8 12 17

50

55

60

65

70

75

80

Age (week)

Rela

tive le

an m

ass (

%)

**

#

**

#

*

3 4 5 6 7 8 9 10 11 12 13 14 15 16 17

0

5

10

15

20

25

30

35

40

45

50

Age (week)

Body w

eig

ht (g

)

HFD-CON

HFD-ADI

HFD-TMDI

**

##

***

*****

*****

******* ***

**

*** **

#

####

## ####

## ### #####

# # # #

*#

3 4 5 6 7 8 9 10 11 12 13 14 15 16 17

0

5

10

15

20

25

30

35

40

45

50

Age (week)

Body w

eig

ht (g

)

NCD-CON

NCD-ADI

NCD-TMDI

**

***

#

#####

#*

*#

Normal chow diet (NCD)

or high-fat diet (HFD)NursingPregnancy

NCD-CON

NCD-ADI

NCD-TMDI

HFD-CON

HFD-ADI

HFD-TMDI

-10 d 0 d 3 wks 20 wks8 wks 12 wks 17 wks

NCD group

HFD group

Microbiome analysis

Body composition analysis

OGTT and metabolic measurements

a

CON ADI TMDI

0.0

0.5

1.0

1.5

Perinephric a

dip

ose tis

sue (

g)

HFD

*

CON ADI TMDI

0

1

2

3

Epid

idym

al a

dip

ose tis

sue (

g)

HFD

*

CON ADI TMDI

30000

40000

50000

60000

70000O

GT

TA

UC (

mg/d

L x

min

)

HFD

p=0.05

CON ADI TMDI

0

5000

10000

15000

Insulin

(pg/m

l)HFD

**

CON ADI TMDI

0

50

100

150

200

250

HO

MA

-IR

(m

mol/L

x µ

U/L

)

HFD

**

p=0.051

40

50 HFD-CON

HFD-ADI

HFD-TMDI*

#

Mice

12

12

8

12

12

12

25

follows: CON vs ADI (#p < 0.05; ##p < 0.01); CON vs TMDI (*p < 0.05; **p < 0.01; 622

***p < 0.001). For f-g, and k-n, one-way ANOVA with Tukey’s range test (*p < 0.05; 623

**p < 0.01; ***p < 0.001). Abbreviations: ADI, acceptable daily intake; AUC, area 624

under curve; CON, control; HFD, high-fat diet; HOMA-IR, homeostatic model 625

assessment of insulin resistance; NCD, normal chow diet; OGTT, oral glucose tolerance 626

test; TMDI, theoretical maximum daily intake. 627

628

26

629

Fig. 2 | Residual dose of tylosin remodels the gut microbiota composition, and their 630

microbiota shift verifies its pathogenesis on generating the obesogenic and 631

metabolic phenotype in germ-free mice. 632

a, Change in Shannon diversity index in 3-, 8-, 17-week-old HFD-fed mice (n = 633

6/group). b, Gut microbiota composition as represented by principal coordinate analysis 634

(PCoA) of Bray-Curtis distances for 3-, 8-, and 17-week-old HFD-fed mice (n = 635

6/group). c, Bray-Curtis dissimilarity index comparing distances of tylosin-treated 636

groups to CON at different time points (n = 6/group). d, Volcano plot showing the 637

enriched or decreased bacteria based on log2 fold change >1 and significant difference 638

(p < 0.05) using the Wilcoxon signed-rank test (n = 6/group). e, Bray-Curtis distances-639

based PCoA of 17-week-old HFD-fed mice's gut microbiota and the fitted antibiotic 640

exposure/obesity-related variables which significantly correlated to the shifted 641

microbiome (p < 0.05) by using the envfit package in R (n = 5-6/group). f, Spearman's 642

a c

fe

g h i j k

b

40

50 HFD-CON

HFD-ADI

HFD-TMDI*

#

3 8 17

2.5

3.0

3.5

4.0

Age (week)

Shannon in

dex **

##

0.2

0.4

0.6

0.8****

**

3 8 17 3 8 17

0.0

0.2

0.4

0.6

0.8

1.0

Bra

y-C

urt

is d

ista

nce

CON to ADI

HFD

CON to TMDI

***

****

****

****

**

8 9 10 11 12 13 14 15 16 17 18 19 20

20

30

40

50

60

Age (week)

Body w

eig

ht (g

)

FMT-CON

FMT-TMDI

**

*

*

●●

●●

●●

●●

● ●

●●

●●●

● ●

0.4

0.2

0.0

0.2

●●

3

3

3

8

8

8

17

17

17

HFD-CON-3wk

HFD-CON-8wk

HFD-CON-17wk

HFD-ADI-3wk

HFD-ADI-8wk

HFD-ADI-17wk

HFD-TMDI-3wk

HFD-TMDI-8wk

HFD-TMDI-17wk

3

8

17

3

8

17

3

8

17

PCoA1 (23.89%)

-0.2 0.0 0.2 0.4

PC

oA

2 (

15

.91

%)

-0.4

-0.2

0.2

0.0

d

CON TMDI

-2

0

2

4

6

8

Body w

eig

ht change

in 2

weeks a

fter

FM

T (

g)

FMT

*

CON TMDI

20

30

40

50

Rela

tive fat m

ass (

%)

FMT

p=0.086

CON TMDI

1500

2000

2500

3000

OG

TT

AU

C (

mg/d

L x

min

)

FMT

****

CON TMDI

0

10

20

30

HO

MA

-IR

(m

mol/L

x µ

U/L

)

FMT

*

FMT

Adonis: p < 0.0001

1.0

1.5

2.0

0.5

0.0

-lo

g1

0 (p

.valu

e)

log2 fold change

Bifidobacterium

Oscillibacter

Blautia

Parasutterella

0 5 10-5-10

Decreased genus

Enriched genus

-0.6 -0.4 -0.2 0.0 0.2 0.4

-0.4

-0.2

0.0

0.2

0.4

0.6

PCoA1 (32.4%)

PC

oA

2 (

25.2

%)

Body weight

Relative fat mass

Fatty liver score

Fasting glucose

OGTTAUC

Antibiotic exposure

Adonis: p < 0.0001

r=−0.71

p=0.003HFD-ADI

HFD-CON

HFD-TMDI

●●

●●

●●

●●

● ● ●

r=−0.71

p=0.003

-4 -2 0 2

-0.4

0.4

-0.2

0.0

0.2

r = 0.71 p = 0.003

HFD-ADI

HFD-CON

HFD-TMDI

r=−0.71

p=0.003HFD-ADI

HFD-CON

HFD-TMDI

PC1obesity biomarkers (84.4%)

PC

oA

2m

icro

bio

ta (

25

.2%

)

r = 0.71 p = 0.003

27

correlation of PCoA2microbiota and PC1obesity biomarkers (n = 5-6/group). g, Body weight 643

change in mice transplanted with feces of HFD-CON and HFD-TMDI mice (n = 644

10/group). h, Body weight change at 2 weeks after transplantation (ΔBW(W10-W8)) (n = 645

10/group). i, Relative fat mass at 20 weeks of age (n = 10/group). j, AUC derived from 646

the OGTT (n = 9/group). k, HOMA-IR index (n = 9/group). Data are expressed as mean 647

± SD. For a, Statistical analyses were performed by Wilcoxon signed-rank test with 648

FDR-adjust as follow: HFD-CON vs HFD-ADI (##p < 0.01); HFD-CON vs HFD-TMDI 649

(**p < 0.01). For c, One-way ANOVA with Tukey’s range test (**p < 0.01; ***p < 650

0.001; ****p < 0.0001). For e, Wilcoxon signed-rank test with FDR-adjust. For g-k, un-651

paired t-test (*p < 0.05; ****p < 0.0001). Abbreviations: AUC, area under curve; CON, 652

control; FMT, fecal microbiota transplantation; HOMA-IR, homeostatic model 653

assessment of insulin resistance; OGTT, oral glucose tolerance test; TMDI, theoretical 654

maximum daily intake. 655

656

28

657

Fig. 3 | Early-life exposure of tylosin residue sufficiently induces obesity and 658

metabolic disorder disease and modifies gut microbiota composition by enhancing 659

the re-structuring of obesity-related genera with deep-rooted change. 660

a, Experimental design of early-life exposure model. b, Body weight change (n = 8-661

11/group). c, Relative fat mass change (n = 8-11/group). d, Weight of visceral adipose 662

tissue (n = 8-11/group). e, AUC derived from the OGTT (n = 7-8/group). f, Insulin level 663

after overnight fasting (n = 7-8/group). g, HOMA-IR index (n = 7-8/group). h, PCoA 664

based on Bray-Curtis distances of gut microbiota at 5 and 20 weeks of age (n = 665

8/group). i, Heatmap showing 32 bacteria with significant differences (q < 0.05) among 666

groups and j, their Spearman's correlation with obesity-related variables (n = 8/group). 667

Data are expressed as mean ± SD. For b-c, Statistical analyses were performed by one-668

way ANOVA with Tukey’s range test as follow: CON vs Early-TMDI (#p < 0.05 and 669

##p < 0.01), CON vs Cont-TMDI (*p < 0.05; **p < 0.01; ***p < 0.001), or Cont-TMDI 670

CON

Cont-TMDI

Early-TMDI

-10 d 0 d 3 wks 20 wks5 wks 10 wks 15 wks

High-fat diet (HFD)NursingPregnancy

Microbiome analysis

Body composition analysis

SCFAs and BAs analysis

OGTT and metabolic measurements

a b c

d

g

fe i

5 10 15 20

0

10

20

30

40

50

Age (week)

Rela

tive fat m

ass (

%)

CONCont-TMDI

Early-TMDI

*

*

*

##

* #

4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20

0

10

20

30

40

50

Age (week)

Body w

eig

ht (g

)

CON

Cont-TMDI

Early-TMDI

* #

***

****

*****

** **** *

* * **

** **

#

###

## # # #

##

CON ContTMDI

EarlyTMDI

0

2

4

6

8

Vecera

l adip

ose tis

sue (

g)

*

CON ContTMDI

EarlyTMDI

1600

2000

2400

2800

OG

TT

AU

C (

mg/d

L x

min

)

*

CON ContTMDI

EarlyTMDI

0

1000

2000

3000

4000

Insulin

g/L

) *

CON ContTMDI

EarlyTMDI

0

10

20

30

40

HO

MA

-IR

(m

mol/L

x µ

U/L

)

**

p=0.09

●● ●

●●

● ●

●●

PCoA1 ( 42.85 %)

PC

oA

2 (

19.0

4 %

)

−0.4 −0.2 0.0 0.2 0.4

−0.3

−0.2

−0.1

0.0

0.1

0.2

0.3

5

20

5

20

5

20

CON-5wk

CON-20wk

5

20

5

20

5

20

Cont-TMDI-5wk

Cont-TMDI-20wk

Early-TMDI-5wk

Early-TMDI-20wk

h

Mice

8

7

11

j

Adonis: p < 0.0001

29

vs Early-TMDI (†p < 0.05). For d-g, One-way ANOVA with Tukey’s range test (*p < 671

0.05; **p < 0.01). For h, Adonis was performed to test the difference among groups. 672

For i, Kruskal-Wallis test with FDR-adjusted p-value (q < 0.05). For j, Spearman’s 673

correlation with FDR-adjusted p-value (* q < 0.1; **q < 0.05). Abbreviations: AUC, 674

area under curve; CON, control; HFD, high-fat diet; HOMA-IR, homeostatic model 675

assessment of insulin resistance; OGTT, oral glucose tolerance test; TMDI, theoretical 676

maximum daily intake. 677

30

678

Fig. 4 | The modification of fecal primary-secondary bile acids ratio by gut 679

microbiota and downregulates FGF15 signaling pathway is involved in the 680

consequences of obesogenic and metabolic dysfunctions caused by tylosin residue 681

exposure at TMDI dose. 682

a, Fecal short-chain fatty acids levels (n = 7/group). b, Ratio of fecal primary bile acids 683

to secondary bile acids (n = 7-11/group). c, Levels of non-12-OH bile acids, d, 684

muricholic acids and e, 12-OH bile acids (n = 7-11/group). f, Portal FGF15 levels (n = 685

6-11/group). g, Western blotting of ileal FGF15 expression normalized to GAPDH (n = 686

4/group). h, Western blotting of hepatic FGFR4 level normalized to GAPDH. Data are 687

presented by mean ± SD (n = 4/group). For a-h, Statistical analyses were performed by 688

one-way ANOVA with Tukey’s range test (*p < 0.05; **p < 0.01; ***p < 0.001). 689

Abbreviations: AGP, antibiotic growth promoter; α-MCA, α-muricholic acid; β-MCA, 690

β-muricholic acid; ω-MCA, ω- muricholic acid; CON, control; CDCA, 691

a b c

d

g

fe

h

CON ContTMDI

EarlyTMDI

0.0

0.5

1.0

1.5

2.0

PB

A / S

BA

*

*

T!MCA "MCA !MCA #MCA

0.0

0.2

0.4

0.6

0.8

5

10

15

20

Bile

acid

s (

%)

**

**

******

*

Primary Secondary

TCDCA CDCA TUDCA UDCA LCA TLCA

0

2

4

20

40

60

80

100

Bile

acid

s (

%)

CON

Cont-TMDI

Early-TMDI

Primary Secondary

**

**

* *

Acetic acid

Propionicacid

Butyricacid

Valericacid

Isobutyricacid

Isovalericacid

0

100

200

300

4001000

2000

3000

4000

Short

chain

fatty a

cid

s (

µg/g

)

CON

Cont-TMDI

Early-TMDI

p=0.066

p=0.068

p=0.059** *

CON ContTMDI

EarlyTMDI

0

10

20

30

Port

al F

GF

15 (

pg/m

L) *

TCA CA TDCA DCA

0.0

0.2

0.4

0.6

0.810

20

30

40

Bile

acid

s (

%)

Primary Secondary

*

CON

Cont-TMDI

Early-TMDI

CON

Cont-TMDI

Early-TMDI

iFGF15

GAPDH

FGFR4

GAPDH

CON ContTMDI

EarlyTMDI

0.0

0.4

0.8

1.2

Rela

tive e

xpre

ssio

n le

vel

(FG

FR

4 / G

AP

DH

) ***

*

CON ContTMDI

EarlyTMDI

0.0

0.4

0.8

1.2

Rela

tive e

xpre

ssio

n le

vel

(FG

F15 / G

AP

DH

)

**

**

31

chenodeoxycholic acid; FGF15, fibroblast growth factor 15; FGFR4, fibroblast growth 692

factor receptor 4; GAPDH, glyceraldehyde 3-phosphate dehydrogenase; LCA, 693

lithocholic acid; PBA, primary bile acid; SBA, secondary bile acid; T-β-MCA, tauro-694

beta-muricholic acid; TCDCA, taurochenodeoxycholic acid; TLCA, taurolithocholic 695

acid; TMDI, theoretical maximum daily intake; TUDCA, tauroursodeoxycholic acid; 696

UDCA, ursodeoxycholic acid. 697

Supplementary Files

This is a list of supplementary �les associated with this preprint. Click to download.

ExtendedDataFigures20211109NatMetab.pdf

Supplementarytable20211109NatMetabver1.pdf