Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Benhua Zeng is active.

Publication


Featured researches published by Benhua Zeng.


EBioMedicine | 2015

Dietary Modulation of Gut Microbiota Contributes to Alleviation of Both Genetic and Simple Obesity in Children

Chenhong Zhang; Aihua Yin; Hongde Li; Ruirui Wang; Guojun Wu; Jian Shen; Menghui Zhang; Linghua Wang; Yaping Hou; Haimei Ouyang; Yan Zhang; Yinan Zheng; Jicheng Wang; Xiaofei Lv; Yulan Wang; Feng Zhang; Benhua Zeng; Wenxia Li; Feiyan Yan; Yufeng Zhao; Xiaoyan Pang; Xiaojun Zhang; Huaqing Fu; Feng Chen; Naisi Zhao; Bruce R. Hamaker; Laura C. Bridgewater; David Weinkove; Karine Clément; Joël Doré

Gut microbiota has been implicated as a pivotal contributing factor in diet-related obesity; however, its role in development of disease phenotypes in human genetic obesity such as Prader–Willi syndrome (PWS) remains elusive. In this hospitalized intervention trial with PWS (n = 17) and simple obesity (n = 21) children, a diet rich in non-digestible carbohydrates induced significant weight loss and concomitant structural changes of the gut microbiota together with reduction of serum antigen load and alleviation of inflammation. Co-abundance network analysis of 161 prevalent bacterial draft genomes assembled directly from metagenomic datasets showed relative increase of functional genome groups for acetate production from carbohydrates fermentation. NMR-based metabolomic profiling of urine showed diet-induced overall changes of host metabotypes and identified significantly reduced trimethylamine N-oxide and indoxyl sulfate, host-bacteria co-metabolites known to induce metabolic deteriorations. Specific bacterial genomes that were correlated with urine levels of these detrimental co-metabolites were found to encode enzyme genes for production of their precursors by fermentation of choline or tryptophan in the gut. When transplanted into germ-free mice, the pre-intervention gut microbiota induced higher inflammation and larger adipocytes compared with the post-intervention microbiota from the same volunteer. Our multi-omics-based systems analysis indicates a significant etiological contribution of dysbiotic gut microbiota to both genetic and simple obesity in children, implicating a potentially effective target for alleviation. Research in context Poorly managed diet and genetic mutations are the two primary driving forces behind the devastating epidemic of obesity-related diseases. Lack of understanding of the molecular chain of causation between the driving forces and the disease endpoints retards progress in prevention and treatment of the diseases. We found that children genetically obese with Prader–Willi syndrome shared a similar dysbiosis in their gut microbiota with those having diet-related obesity. A diet rich in non-digestible but fermentable carbohydrates significantly promoted beneficial groups of bacteria and reduced toxin-producers, which contributes to the alleviation of metabolic deteriorations in obesity regardless of the primary driving forces.


Nature Immunology | 2015

Commensal bacteria direct selective cargo sorting to promote symbiosis

Qin Zhang; Ying Pan; Ruiqing Yan; Benhua Zeng; Haifang Wang; Xinwen Zhang; Wenxia Li; Hong Wei; Zhihua Liu

Mucosal immunity protects a host from intestinal inflammation and infection and is profoundly influenced by symbiotic bacteria. Here we report that in mice symbiotic bacteria directed selective cargo sorting in Paneth cells to promote symbiosis through Nod2, a cytosolic bacterial sensor, and the multifunctional protein kinase LRRK2, both encoded by inflammatory bowel disease (IBD)-associated genes. Commensals recruited Nod2 onto lysozyme-containing dense core vesicles (DCVs), which was required for DCV localization of LRRK2 and a small GTPase, Rab2a. Deficiency of Nod2, LRRK2 or Rab2a or depletion of commensals resulted in lysosomal degradation of lysozyme. Thus, commensal bacteria and host factors orchestrate the lysozyme-sorting process to protect the host from enteric infection, implicating Paneth cell dysfunction in IBD pathogenesis.


Science | 2018

Gut bacteria selectively promoted by dietary fibers alleviate type 2 diabetes

Liping Zhao; Feng Zhang; Xiaoying Ding; Guojun Wu; Yan Y. Lam; Xuejiao Wang; Huaqing Fu; Xinhe Xue; Chunhua Lu; Jilin Ma; Lihua Yu; Chengmei Xu; Zhongying Ren; Ying Xu; Songmei Xu; Hongli Shen; Xiuli Zhu; Yu Shi; Qingyun Shen; Weiping Dong; Rui Liu; Yunxia Ling; Yue Zeng; Xingpeng Wang; Qianpeng Zhang; Jing Wang; Linghua Wang; Yanqiu Wu; Benhua Zeng; Hong Wei

Microbial modulation of diabetes Short-chain fatty acids (SCFAs) are produced by various human gut microbes. SCFAs act as an energy source to the colonic epithelium and are also sensed by host signaling pathways that modulate appetite and inflammation. Deficiency of gut SCFAs is associated with type 2 diabetes. Zhao et al. found that adopting a high-fiber diet promoted the growth of SCFA-producing organisms in diabetic humans. The high-fiber diet induced changes in the entire gut microbe community and correlated with elevated levels of glucagon-like peptide-1, a decline in acetylated hemoglobin levels, and improved blood-glucose regulation. Science, this issue p. 1151 Increasing dietary fiber intake increases the abundance of short-chain fatty acid–producing gut microbes and relieves diabetes. The gut microbiota benefits humans via short-chain fatty acid (SCFA) production from carbohydrate fermentation, and deficiency in SCFA production is associated with type 2 diabetes mellitus (T2DM). We conducted a randomized clinical study of specifically designed isoenergetic diets, together with fecal shotgun metagenomics, to show that a select group of SCFA-producing strains was promoted by dietary fibers and that most other potential producers were either diminished or unchanged in patients with T2DM. When the fiber-promoted SCFA producers were present in greater diversity and abundance, participants had better improvement in hemoglobin A1c levels, partly via increased glucagon-like peptide-1 production. Promotion of these positive responders diminished producers of metabolically detrimental compounds such as indole and hydrogen sulfide. Targeted restoration of these SCFA producers may present a novel ecological approach for managing T2DM.


Scientific Reports | 2016

Role of the Gut Microbiome in Modulating Arthritis Progression in Mice

Xiaofei Liu; Benhua Zeng; Juan Zhang; Wenxia Li; Fangxiang Mou; Heng Wang; Qinghua Zou; Bing Zhong; Like Wu; Hong Wei; Yongfei Fang

Genetics alone cannot explain most cases of rheumatoid arthritis (RA). Thus, investigating environmental factors such as the gut microbiota may provide new insights into the initiation and progression of RA. In this study, we performed 16S rRNA sequencing to characterise the gut microbiota of DBA1 mice that did or did not develop arthritis after induction with collagen. We found that divergence in the distribution of microbiota after induction was pronounced and significant. Mice susceptible to collagen-induced arthritis (CIA) showed enriched operational taxonomic units (OTUs) affiliated with the genus Lactobacillus as the dominant genus prior to arthritis onset. With disease development, the abundance of OTUs affiliated with the families Bacteroidaceae, Lachnospiraceae, and S24-7 increased significantly in CIA-susceptible mice. Notably, germ-free mice conventionalized with the microbiota from CIA-susceptible mice showed a higher frequency of arthritis induction than those conventionalized with the microbiota from CIA-resistant mice. Consistently, the concentration of the cytokine interleukin-17 in serum and the proportions of CD8+T cells and Th17 lymphocytes in the spleen were significantly higher in the former group, whereas the abundances of dendritic cells, B cells, and Treg cells in the spleen were significantly lower. Our results suggest that the gut microbiome influences arthritis susceptibility.


Scientific Reports | 2015

Microbiota prevents cholesterol loss from the body by regulating host gene expression in mice

Chun-Yan Zhong; Wei-Wei Sun; Yinyan Ma; Hongling Zhu; Pan Yang; Hong Wei; Benhua Zeng; Qian Zhang; Yu Liu; Wenxia Li; Yixin Chen; Liqing Yu; Zhi-Yuan Song

We have previously observed that knockout of Niemann-Pick C1-Like 1 (NPC1L1), a cholesterol transporter essential for intestinal cholesterol absorption, reduces the output of dry stool in mice. As the food intake remains unaltered in NPC1L1-knockout (L1-KO) mice, we hypothesized that NPC1L1 deficiency may alter the gut microbiome to reduce stool output. Consistently, here we demonstrate that the phyla of fecal microbiota differ substantially between L1-KO mice and their wild-type controls. Germ-free (GF) mice have reduced stool output. Inhibition of NPC1L1 by its inhibitor ezetimibe reduces stool output in specific pathogen-free (SPF), but not GF mice. In addition, we show that GF versus SPF mice have reduced intestinal absorption and increased fecal excretion of cholesterol, particularly after treatment with ezetimibe. This negative balance of cholesterol in GF mice is associated with reduced plasma and hepatic cholesterol, and likely caused by reduced expression of NPC1L1 and increased expression of ABCG5 and ABCG8 in small intestine. Expression levels of other genes in intestine and liver largely reflect a state of cholesterol depletion and a decrease in intestinal sensing of bile acids. Altogether, our findings reveal a broad role of microbiota in regulating whole-body cholesterol homeostasis and its response to a cholesterol-lowering drug, ezetimibe.


Scientific Reports | 2016

Gut microbiota can transfer fiber characteristics and lipid metabolic profiles of skeletal muscle from pigs to germ-free mice.

Honglin Yan; Hui Diao; Yi Xiao; Wenxia Li; Bing Yu; Jun He; Jie Yu; Ping Zheng; Xiangbing Mao; Yuheng Luo; Benhua Zeng; Hong Wei; Daiwen Chen

Obesity causes changes in microbiota composition, and an altered gut microbiota can transfer obesity-associated phenotypes from donors to recipients. Obese Rongchang pigs (RP) exhibited distinct fiber characteristics and lipid metabolic profiles in their muscle compared with lean Yorkshire pigs (YP). However, whether RP have a different gut microbiota than YP and whether there is a relationship between the microbiota and muscle properties are poorly understood. The present study was conducted to test whether the muscle properties can be transferred from pigs to germ-free (GF) mice. High-throughput pyrosequencing confirms the presence of distinct core microbiota between pig breeds, with alterations in taxonomic distribution and modulations in β diversity. RP displayed a significant higher Firmicutes/Bacteroidetes ratio and apparent genera differences compared with YP. Transplanting the porcine microbiota into GF mice replicated the phenotypes of the donors. RP and their GF mouse recipients exhibited a higher body fat mass, a higher slow-contracting fiber proportion, a decreased fiber size and fast IIb fiber percentage, and enhanced lipogenesis in the gastrocnemius muscle. Furthermore, the gut microbiota composition of colonized mice shared high similarity with their donor pigs. Taken together, the gut microbiota of obese pigs intrinsically influences skeletal muscle development and the lipid metabolic profiles.


Nature Communications | 2017

Remodelling of the gut microbiota by hyperactive NLRP3 induces regulatory T cells to maintain homeostasis

Xiaomin Yao; Chenhong Zhang; Yue Xing; Guang Xue; Qianpeng Zhang; Fengwei Pan; Guojun Wu; Yingxin Hu; Qiuhong Guo; Ailing Lu; Xiaoming Zhang; Rongbin Zhou; Zhigang Tian; Benhua Zeng; Hong Wei; Warren Strober; Liping Zhao; Guangxun Meng

Inflammasomes are involved in gut homeostasis and inflammatory pathologies, but the role of NLRP3 inflammasome in these processes is not well understood. Cryopyrin-associated periodic syndrome (CAPS) patients with NLRP3 mutations have autoinflammation in skin, joints, and eyes, but not in the intestine. Here we show that the intestines of CAPS model mice carrying an Nlrp3R258W mutation maintain homeostasis in the gut. Additionally, such mice are strongly resistant to experimental colitis and colorectal cancer; this is mainly through a remodelled gut microbiota with enhanced anti-inflammatory capacity due to increased induction of regulatory T cells (Tregs). Mechanistically, NLRP3R258W functions exclusively in the lamina propria mononuclear phagocytes to directly enhance IL-1β but not IL-18 secretion. Increased IL-1β boosts local antimicrobial peptides to facilitate microbiota remodelling. Our data show that NLRP3R258W-induced remodelling of the gut microbiota, induces local Tregs to maintain homeostasis and compensate for otherwise-detrimental intestinal inflammation.Inflammasomes are involved in gut homeostasis and inflammatory pathologies. The authors show that a hyperactive NLRP3 inflammasome maintains gut homeostasis through remodelling of the gut microbiota and induction of regulatory T cells.


Scientific Reports | 2016

Microbiota Modulates Behavior and Protein Kinase C mediated cAMP response element-binding protein Signaling.

Li Zeng; Benhua Zeng; Haiyang Wang; Bo Li; Ran Huo; Peng Zheng; Xiaotong Zhang; Xiangyu Du; Meiling Liu; Zheng Fang; Xue-Jiao Xu; Chanjuan Zhou; Jianjun Chen; Wenxia Li; Jing Guo; Hong Wei; Peng Xie

Evolutionary pressure drives gut microbiota–host coevolution and results in complex interactions between gut microbiota and neural development; however, the molecular mechanisms by which the microbiota governs host behavior remain obscure. Here, we report that colonization early in life is crucial for the microbiota to modulate brain development and behavior; later colonization or deletion of microbiota cannot completely reverse the behaviors. Microarray analysis revealed an association between absence of gut microbiota and expression in cAMP responding element-binding protein (CREB) regulated genes in the hippocampus. The absence of gut microbiota from birth was shown to be associated with decreased CREB expression, followed by decreases of protein kinase C beta (PRKCB) and AMPA receptors expression, and an increase of phosphorylation CREB (pCREB) expression. Microbiota colonization in adolescence restored CREB and pCREB expression, but did not alter PRKCB and AMPARs expression. The removal of the gut microbiota from SPF mice using antibiotics only reduced pCREB expression. These findings suggest that (i) colonization of the gut microbiota early in life might facilitate neurodevelopment via PKC–CREB signaling and (ii) although GF mice and ABX mice display reduced anxiety-related behaviors, the molecular mechanisms behind this might differ.


Journal of Immunology | 2017

Rip2 Is Required for Nod2-Mediated Lysozyme Sorting in Paneth Cells

Haifang Wang; Xinwen Zhang; Zhanguang Zuo; Qin Zhang; Ying Pan; Benhua Zeng; Wenxia Li; Hong Wei; Zhihua Liu

Paneth cells play an important role in maintaining intestinal homeostasis by secreting a large number of antimicrobial peptides into the intestinal lumen. In this study, we found that Rip2 is required for lysozyme sorting in Paneth cells in a manner that is dependent on Nod2, LRRK2, and Rab2a. Rip2 deficiency in mouse led to lysosomal degradation of lysozyme in Paneth cells and prevented the recruitment of Rab2a onto dense core vesicles (DCVs). Like Nod2 and LRRK2, Rip2 localizes to DCVs in Paneth cells, and its DCV localization depends on Nod2 and LRRK2. Thus, we delineated a genetic pathway, consisting of Nod2–LRRK2–Rip2–Rab2a, which is required for lysozyme sorting. Taken together, our results indicate that the lysozyme-sorting process in Paneth cells is orchestrated by a number of host factors and highlight the importance of Paneth cell function in intestinal homeostasis.


Cell Reports | 2016

The Gut Epithelial Receptor LRRC19 Promotes the Recruitment of Immune Cells and Gut Inflammation

Shuisong Cao; Xiaomin Su; Benhua Zeng; Hui Yan; Yugang Huang; Enlin Wang; Huan Yun; Yuan Zhang; Feifei Liu; Wenxia Li; Hong Wei; Yongzhe Che; Rongcun Yang

Summary Commensal microbes are necessary for a healthy gut immune system. However, the mechanism involving these microbes that establish and maintain gut immune responses is largely unknown. Here, we have found that the gut immune receptor leucine-rich repeat (LRR) C19 is involved in host-microbiota interactions. LRRC19 deficiency not only impairs the gut immune system but also reduces inflammatory responses in gut tissues. We demonstrate that the LRRC19-associated chemokines CCL6, CCL9, CXCL9, and CXCL10 play a critical role in immune cell recruitment and intestinal inflammation. The expression of these chemokines is associated with regenerating islet-derived (REG) protein-mediated microbiotas. We also found that the expression of REGs may be regulated by gut Lactobacillus through LRRC19-mediated activation of NF-κB. Therefore, our study establishes a regulatory axis of LRRC19, REGs, altered microbiotas, and chemokines for the recruitment of immune cells and the regulation of intestinal inflammation.

Collaboration


Dive into the Benhua Zeng's collaboration.

Top Co-Authors

Avatar

Hong Wei

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Wenxia Li

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Bing Yu

Chinese Ministry of Education

View shared research outputs
Top Co-Authors

Avatar

Chenhong Zhang

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Daiwen Chen

Chinese Ministry of Education

View shared research outputs
Top Co-Authors

Avatar

Guojun Wu

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Jie Yu

Chinese Ministry of Education

View shared research outputs
Top Co-Authors

Avatar

Jun He

Chinese Ministry of Education

View shared research outputs
Top Co-Authors

Avatar

Liping Zhao

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Xiangbing Mao

Chinese Ministry of Education

View shared research outputs
Researchain Logo
Decentralizing Knowledge