Network


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

Hotspot


Dive into the research topics where Jingwei Cai is active.

Publication


Featured researches published by Jingwei Cai.


Journal of Clinical Investigation | 2015

Intestinal farnesoid X receptor signaling promotes nonalcoholic fatty liver disease

Changtao Jiang; Cen Xie; Fei Li; Limin Zhang; Robert G. Nichols; Kristopher W. Krausz; Jingwei Cai; Yunpeng Qi; Zhong-Ze Fang; Shogo Takahashi; Naoki Tanaka; Dhimant Desai; Shantu Amin; Istvan Albert; Andrew D. Patterson; Frank J. Gonzalez

Nonalcoholic fatty liver disease (NAFLD) is a major worldwide health problem. Recent studies suggest that the gut microbiota influences NAFLD pathogenesis. Here, a murine model of high-fat diet-induced (HFD-induced) NAFLD was used, and the effects of alterations in the gut microbiota on NAFLD were determined. Mice treated with antibiotics or tempol exhibited altered bile acid composition, with a notable increase in conjugated bile acid metabolites that inhibited intestinal farnesoid X receptor (FXR) signaling. Compared with control mice, animals with intestine-specific Fxr disruption had reduced hepatic triglyceride accumulation in response to a HFD. The decrease in hepatic triglyceride accumulation was mainly due to fewer circulating ceramides, which was in part the result of lower expression of ceramide synthesis genes. The reduction of ceramide levels in the ileum and serum in tempol- or antibiotic-treated mice fed a HFD resulted in downregulation of hepatic SREBP1C and decreased de novo lipogenesis. Administration of C16:0 ceramide to antibiotic-treated mice fed a HFD reversed hepatic steatosis. These studies demonstrate that inhibition of an intestinal FXR/ceramide axis mediates gut microbiota-associated NAFLD development, linking the microbiome, nuclear receptor signaling, and NAFLD. This work suggests that inhibition of intestinal FXR is a potential therapeutic target for NAFLD treatment.


mSystems | 2016

Farnesoid X Receptor Signaling Shapes the Gut Microbiota and Controls Hepatic Lipid Metabolism

Limin Zhang; Cen Xie; Robert G. Nichols; Siu Hung Joshua Chan; Changtao Jiang; Ruixin Hao; Philip B. Smith; Jingwei Cai; Margaret Simons; Emmanuel Hatzakis; Costas D. Maranas; Frank J. Gonzalez; Andrew D. Patterson

The farnesoid X receptor (FXR) plays an important role in mediating the dialog between the host and gut microbiota, particularly through modulation of enterohepatic circulation of bile acids. Mounting evidence suggests that genetic ablation of Fxr in the gut or gut-restricted chemical antagonism of the FXR promotes beneficial health effects, including the prevention of nonalcoholic fatty liver disease in rodent models. However, questions remain unanswered, including whether modulation of FXR activity plays a role in shaping the gut microbiota community structure and function and what metabolic pathways of the gut microbiota contribute in an FXR-dependent manner to the host phenotype. In this report, new insights are gained into the metabolic contribution of the gut microbiota to the metabolic phenotypes, including establishing a link between FXR antagonism, bacterial bile salt hydrolase activity, and fermentation. Multiple approaches, including unique mouse models as well as metabolomics and genome-scale metabolic models, were employed to confirm these results. ABSTRACT The gut microbiota modulates obesity and associated metabolic phenotypes in part through intestinal farnesoid X receptor (FXR) signaling. Glycine-β-muricholic acid (Gly-MCA), an intestinal FXR antagonist, has been reported to prevent or reverse high-fat diet (HFD)-induced and genetic obesity, insulin resistance, and fatty liver; however, the mechanism by which these phenotypes are improved is not fully understood. The current study investigated the influence of FXR activity on the gut microbiota community structure and function and its impact on hepatic lipid metabolism. Predictions about the metabolic contribution of the gut microbiota to the host were made using 16S rRNA-based PICRUSt (phylogenetic investigation of communities by reconstruction of unobserved states), then validated using 1H nuclear magnetic resonance-based metabolomics, and results were summarized by using genome-scale metabolic models. Oral Gly-MCA administration altered the gut microbial community structure, notably reducing the ratio of Firmicutes to Bacteroidetes and its PICRUSt-predicted metabolic function, including reduced production of short-chain fatty acids (substrates for hepatic gluconeogenesis and de novo lipogenesis) in the ceca of HFD-fed mice. Metabolic improvement was intestinal FXR dependent, as revealed by the lack of changes in HFD-fed intestine-specific Fxr-null (FxrΔIE) mice treated with Gly-MCA. Integrative analyses based on genome-scale metabolic models demonstrated an important link between Lactobacillus and Clostridia bile salt hydrolase activity and bacterial fermentation. Hepatic metabolite levels after Gly-MCA treatment correlated with altered levels of gut bacterial species. In conclusion, modulation of the gut microbiota by inhibition of intestinal FXR signaling alters host liver lipid metabolism and improves obesity-related metabolic dysfunction. IMPORTANCE The farnesoid X receptor (FXR) plays an important role in mediating the dialog between the host and gut microbiota, particularly through modulation of enterohepatic circulation of bile acids. Mounting evidence suggests that genetic ablation of Fxr in the gut or gut-restricted chemical antagonism of the FXR promotes beneficial health effects, including the prevention of nonalcoholic fatty liver disease in rodent models. However, questions remain unanswered, including whether modulation of FXR activity plays a role in shaping the gut microbiota community structure and function and what metabolic pathways of the gut microbiota contribute in an FXR-dependent manner to the host phenotype. In this report, new insights are gained into the metabolic contribution of the gut microbiota to the metabolic phenotypes, including establishing a link between FXR antagonism, bacterial bile salt hydrolase activity, and fermentation. Multiple approaches, including unique mouse models as well as metabolomics and genome-scale metabolic models, were employed to confirm these results.


Journal of Proteome Research | 2016

Antioxidant Drug Tempol Promotes Functional Metabolic Changes in the Gut Microbiota.

Jingwei Cai; Limin Zhang; Richard A. Jones; Jared B. Correll; Emmanuel Hatzakis; Philip B. Smith; Frank J. Gonzalez; Andrew D. Patterson

Recent studies have identified the important role of the gut microbiota in the pathogenesis and progression of obesity and related metabolic disorders. The antioxidant tempol was shown to prevent or reduce weight gain and modulate the gut microbiota community in mice; however, the mechanism by which tempol modulates weight gain/loss with respect to the host and gut microbiota has not been clearly established. Here we show that tempol (0, 1, 10, and 50 mg/kg p.o. for 5 days) decreased cecal bacterial fermentation and increased fecal energy excretion in a dose-dependent manner. Liver (1)H NMR-based metabolomics identified a dose-dependent decrease in glycogen and glucose, enhanced glucogenic and ketogenic activity (tyrosine and phenylalanine), and increased activation of the glycolysis pathway. Serum (1)H NMR-based metabolomics indicated that tempol promotes enhanced glucose catabolism. Hepatic gene expression was significantly altered as demonstrated by an increase in Pepck and G6pase and a decrease in Hnf4a, ChREBP, Fabp1, and Cd36 mRNAs. No significant change in the liver and serum metabolomic profiles was observed in germ-free mice, thus establishing a significant role for the gut microbiota in mediating the beneficial metabolic effects of tempol. These results demonstrate that tempol modulates the gut microbial community and its function, resulting in reduced host energy availability and a significant shift in liver metabolism toward a more catabolic state.


Analytical Chemistry | 2017

Orthogonal Comparison of GC–MS and 1H NMR Spectroscopy for Short Chain Fatty Acid Quantitation

Jingwei Cai; Jingtao Zhang; Yuan Tian; Limin Zhang; Emmanuel Hatzakis; Kristopher W. Krausz; Philip B. Smith; Frank J. Gonzalez; Andrew D. Patterson

Short chain fatty acids (SCFAs) are important regulators of host physiology and metabolism and may contribute to obesity and associated metabolic diseases. Interest in SCFAs has increased in part due to the recognized importance of how production of SCFAs by the microbiota may signal to the host. Therefore, reliable, reproducible, and affordable methods for SCFA profiling are required for accurate identification and quantitation. In the current study, four different methods for SCFA (acetic acid, propionic acid, and butyric acid) extraction and quantitation were compared using two independent platforms including gas chromatography coupled with mass spectrometry (GC-MS) and 1H nuclear magnetic resonance (NMR) spectroscopy. Sensitivity, recovery, repeatability, matrix effect, and validation using mouse fecal samples were determined across all methods. The GC-MS propyl esterification method exhibited superior sensitivity for acetic acid and butyric acid measurement (LOD < 0.01 μg mL-1, LOQ < 0.1 μg mL-1) and recovery accuracy (99.4%-108.3% recovery rate for 100 μg mL-1 SCFA mixed standard spike in and 97.8%-101.8% recovery rate for 250 μg mL-1 SCFAs mixed standard spike in). NMR methods by either quantitation relative to an internal standard or quantitation using a calibration curve yielded better repeatability and minimal matrix effects compared to GC-MS methods. All methods generated good calibration curve linearity (R2 > 0.99) and comparable measurement of fecal SCFA concentration. Lastly, these methods were used to quantitate fecal SCFAs obtained from conventionally raised (CONV-R) and germ free (GF) mice. Results from global metabolomic analysis of feces generated by 1H NMR and bomb calorimetry were used to further validate these approaches.


Journal of Nutritional Biochemistry | 2018

Vitamin A deficiency in mice alters host and gut microbial metabolism leading to altered energy homeostasis

Yuan Tian; Robert G. Nichols; Jingwei Cai; Andrew D. Patterson; Margherita T. Cantorna

Vitamin A deficiency (A-) is a worldwide public health problem. To better understand how vitamin A status influences gut microbiota and host metabolism, we systematically analyzed urine, cecum, serum and liver samples from vitamin A sufficient (A+) and deficient (A-) mice using 1H NMR-based metabolomics, quantitative (q)PCR and 16S rRNA gene sequencing coupled with multivariate data analysis. The microbiota in the cecum of A- mice showed compositional as well as functional shifts compared to the microbiota from A+ mice. Targeted 1H NMR analyses revealed significant changes in microbial metabolite concentrations including higher butyrate and hippurate and decreased acetate and 4-hydroxyphenylacetate in A+ relative to A- mice. Bacterial butyrate-producing genes including butyryl-CoA:acetate CoA-transferase and butyrate kinase were significantly higher in bacteria from A+ versus bacteria from A- mice. A- mice had disturbances in multiple metabolic pathways including alterations in energy (hyperglycemia, glycogenesis, TCA cycle and lipoprotein biosynthesis), amino acid and nucleic acid metabolism. A- mice had hyperglycemia, liver dysfunction, changes in bacterial metabolism and altered gut microbial communities. Moreover, integrative analyses indicated a strong correlation between gut microbiota and host energy metabolism pathways in the liver. Vitamin A regulates host and bacterial metabolism, and the result includes alterations in energy homeostasis.


Nature Medicine | 2018

Gut microbiota and intestinal FXR mediate the clinical benefits of metformin

Lulu Sun; Cen Xie; Guang Wang; Yue Wu; Qing Wu; Xuemei Wang; Jia Liu; Yangyang Deng; Jialin Xia; Bo Chen; Songyang Zhang; Chuyu Yun; Guan Lian; Xiujuan Zhang; Heng Zhang; William H. Bisson; Jingmin Shi; Xiaoxia Gao; Pupu Ge; Cuihua Liu; Kristopher W. Krausz; Robert G. Nichols; Jingwei Cai; Bipin Rimal; Andrew D. Patterson; Xian Wang; Frank J. Gonzalez; Changtao Jiang

The anti-hyperglycemic effect of metformin is believed to be caused by its direct action on signaling processes in hepatocytes, leading to lower hepatic gluconeogenesis. Recently, metformin was reported to alter the gut microbiota community in humans, suggesting that the hyperglycemia-lowering action of the drug could be the result of modulating the population of gut microbiota. However, the critical microbial signaling metabolites and the host targets associated with the metabolic benefits of metformin remained elusive. Here, we performed metagenomic and metabolomic analysis of samples from individuals with newly diagnosed type 2 diabetes (T2D) naively treated with metformin for 3 d, which revealed that Bacteroides fragilis was decreased and the bile acid glycoursodeoxycholic acid (GUDCA) was increased in the gut. These changes were accompanied by inhibition of intestinal farnesoid X receptor (FXR) signaling. We further found that high-fat-diet (HFD)-fed mice colonized with B. fragilis were predisposed to more severe glucose intolerance, and the metabolic benefits of metformin treatment on glucose intolerance were abrogated. GUDCA was further identified as an intestinal FXR antagonist that improved various metabolic endpoints in mice with established obesity. Thus, we conclude that metformin acts in part through a B. fragilis–GUDCA–intestinal FXR axis to improve metabolic dysfunction, including hyperglycemia.Metformin decreases the levels of Bacteroides fragilis while increasing the bile acid GUDCA to antagonize intestinal FXR and improves the metabolic health of humans and mice.


Current protocols in immunology | 2018

Structural and Functional Analysis of the Gut Microbiome for Toxicologists

Robert G. Nichols; Jingwei Cai; Iain A. Murray; Imhoi Koo; Philip B. Smith; Gary H. Perdew; Andrew D. Patterson

Characterizing the reciprocal interactions between toxicants, the gut microbiota, and the host, holds great promise for improving our mechanistic understanding of toxic endpoints. Advances in culture‐independent sequencing analysis (e.g., 16S rRNA gene amplicon sequencing) combined with quantitative metabolite profiling (i.e., metabolomics) have provided new ways of studying the gut microbiome and have begun to illuminate how toxicants influence the structure and function of the gut microbiome. Developing a standardized protocol is important for establishing robust, reproducible, and importantly, comparative data. This protocol can be used as a foundation for examining the gut microbiome via sequencing‐based analysis and metabolomics. Two main units follow: (1) analysis of the gut microbiome via sequencing‐based approaches; and (2) functional analysis of the gut microbiome via metabolomics.


Cell Metabolism | 2015

Microbiota-Dependent Hepatic Lipogenesis Mediated by Stearoyl CoA Desaturase 1 (SCD1) Promotes Metabolic Syndrome in TLR5-Deficient Mice

Vishal Singh; Benoit Chassaing; Limin Zhang; Beng San Yeoh; Xia Xiao; Manish Kumar; Mark T. Baker; Jingwei Cai; Rachel Walker; Kamil Borkowski; K.J. Harvatine; Nagendra Singh; Gregory C. Shearer; James M. Ntambi; Bina Joe; Andrew D. Patterson; Andrew T. Gewirtz; Matam Vijay-Kumar


Journal of Functional Foods | 2018

Prebiotic effects of white button mushroom (Agaricus bisporus) feeding on succinate and intestinal gluconeogenesis in C57BL/6 mice

Yuan Tian; Robert G. Nichols; Pratiti Roy; Wei Gui; Philip B. Smith; Jingtao Zhang; Yang-Ding Lin; Veronika Weaver; Jingwei Cai; Andrew D. Patterson; Margherita T. Cantorna


Hypertension | 2018

Attenuation of Microbiotal Dysbiosis and Hypertension in a CRISPR/Cas9 Gene Ablation Rat Model of GPER1

Harshal Waghulde; Xi Cheng; Sarah Galla; Blair Mell; Jingwei Cai; Shondra M. Pruett-Miller; Guillermo Vazquez; Andrew D. Patterson; Matam Vijay Kumar; Bina Joe

Collaboration


Dive into the Jingwei Cai's collaboration.

Top Co-Authors

Avatar

Andrew D. Patterson

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

Robert G. Nichols

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

Frank J. Gonzalez

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Philip B. Smith

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Cen Xie

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Kristopher W. Krausz

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Yuan Tian

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

Limin Zhang

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

Emmanuel Hatzakis

Pennsylvania State University

View shared research outputs
Researchain Logo
Decentralizing Knowledge