Network


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

Hotspot


Dive into the research topics where Lillian Chau is active.

Publication


Featured researches published by Lillian Chau.


Journal of Clinical Investigation | 2015

Engineering the gut microbiota to treat hyperammonemia

Ting-Chin David Shen; Lindsey Albenberg; Kyle Bittinger; Christel Chehoud; Ying-Yu Chen; Colleen A. Judge; Lillian Chau; Josephine Ni; Michael Sheng; Andrew Lin; Benjamin J. Wilkins; Elizabeth L. Buza; James D. Lewis; Yevgeny Daikhin; Ilana Nissim; Marc Yudkoff; Frederic D. Bushman; Gary D. Wu

Increasing evidence indicates that the gut microbiota can be altered to ameliorate or prevent disease states, and engineering the gut microbiota to therapeutically modulate host metabolism is an emerging goal of microbiome research. In the intestine, bacterial urease converts host-derived urea to ammonia and carbon dioxide, contributing to hyperammonemia-associated neurotoxicity and encephalopathy in patients with liver disease. Here, we engineered murine gut microbiota to reduce urease activity. Animals were depleted of their preexisting gut microbiota and then inoculated with altered Schaedler flora (ASF), a defined consortium of 8 bacteria with minimal urease gene content. This protocol resulted in establishment of a persistent new community that promoted a long-term reduction in fecal urease activity and ammonia production. Moreover, in a murine model of hepatic injury, ASF transplantation was associated with decreased morbidity and mortality. These results provide proof of concept that inoculation of a prepared host with a defined gut microbiota can lead to durable metabolic changes with therapeutic utility.


Science Translational Medicine | 2017

A role for bacterial urease in gut dysbiosis and Crohn’s disease

Josephine Ni; Ting-Chin David Shen; Eric Z. Chen; Kyle Bittinger; Aubrey Bailey; Manuela Roggiani; Alexandra Sirota-Madi; Elliot S. Friedman; Lillian Chau; Andrew Lin; Ilana Nissim; Justin Scott; Abigail Lauder; Christopher Hoffmann; Gloriany Rivas; Lindsey Albenberg; Robert N. Baldassano; Jonathan Braun; Ramnik J. Xavier; Clary B. Clish; Marc Yudkoff; Hongzhe Li; Mark Goulian; Frederic D. Bushman; James D. Lewis; Gary D. Wu

Bacterial urease activity of the gut microbiota alters nitrogen flux, leading to gut dysbiosis and worsening of colitis in mice. Nitrogen flux and gut dysbiosis Ni et al. used shotgun metagenomic and metabolomic analysis of fecal samples from pediatric patients with Crohn’s disease. They demonstrated an association between disease severity, gut dysbiosis, and free amino acids. A heavy isotope–labeled nitrogen flux analysis showed that bacterial urease activity led to the transfer of host-derived nitrogen to the gut microbiota, boosting amino acid synthesis. Inoculation of a murine host with Escherichia coli engineered to express urease led to dysbiosis associated with worsened immune-mediated colitis and increased amino acid production. A potential role for nitrogen flux in the development of gut dysbiosis suggests that urease may be a potential target for developing treatments for inflammatory bowel diseases. Gut dysbiosis during inflammatory bowel disease involves alterations in the gut microbiota associated with inflammation of the host gut. We used a combination of shotgun metagenomic sequencing and metabolomics to analyze fecal samples from pediatric patients with Crohn’s disease and found an association between disease severity, gut dysbiosis, and bacterial production of free amino acids. Nitrogen flux studies using 15N in mice showed that activity of bacterial urease, an enzyme that releases ammonia by hydrolysis of host urea, led to the transfer of murine host-derived nitrogen to the gut microbiota where it was used for amino acid synthesis. Inoculation of a conventional murine host (pretreated with antibiotics and polyethylene glycol) with commensal Escherichia coli engineered to express urease led to dysbiosis of the gut microbiota, resulting in a predominance of Proteobacteria species. This was associated with a worsening of immune-mediated colitis in these animals. A potential role for altered urease expression and nitrogen flux in the development of gut dysbiosis suggests that bacterial urease may be a potential therapeutic target for inflammatory bowel diseases.


Proceedings of the National Academy of Sciences of the United States of America | 2018

Microbes vs. chemistry in the origin of the anaerobic gut lumen

Elliot S. Friedman; Kyle Bittinger; Tatiana V. Esipova; Likai Hou; Lillian Chau; Jack Jiang; Clementina Mesaros; Peder J. Lund; Xue Liang; Garret A. FitzGerald; Mark Goulian; Daeyeon Lee; Benjamin A. Garcia; Ian A. Blair; Sergei A. Vinogradov; Gary D. Wu

Significance It is generally thought that the gut microbes consume oxygen and maintain the lumen in a deeply anaerobic state. However, we found that the gut of germ-free mice is also deeply anaerobic, suggesting that there exist other mechanisms responsible for oxygen consumption in addition to the microbes’ respiration. These mechanisms comprise oxidative reactions, such as the oxidation of lipids and other organic substrates. Both the microbiota and the oxidative chemistry regulate luminal oxygen levels that in turn influence the composition of the microbial communities throughout the intestinal tract. The succession from aerobic and facultative anaerobic bacteria to obligate anaerobes in the infant gut along with the differences between the compositions of the mucosally adherent vs. luminal microbiota suggests that the gut microbes consume oxygen, which diffuses into the lumen from the intestinal tissue, maintaining the lumen in a deeply anaerobic state. Remarkably, measurements of luminal oxygen levels show nearly identical pO2 (partial pressure of oxygen) profiles in conventional and germ-free mice, pointing to the existence of oxygen consumption mechanisms other than microbial respiration. In vitro experiments confirmed that the luminal contents of germ-free mice are able to chemically consume oxygen (e.g., via lipid oxidation reactions), although at rates significantly lower than those observed in the case of conventionally housed mice. For conventional mice, we also show that the taxonomic composition of the gut microbiota adherent to the gut mucosa and in the lumen throughout the length of the gut correlates with oxygen levels. At the same time, an increase in the biomass of the gut microbiota provides an explanation for the reduction of luminal oxygen in the distal vs. proximal gut. These results demonstrate how oxygen from the mammalian host is used by the gut microbiota, while both the microbes and the oxidative chemical reactions regulate luminal oxygen levels, shaping the composition of the microbial community throughout different regions of the gut.


bioRxiv | 2018

Posttranscriptional regulation of intestinal epithelial cell repair by RNA binding protein IMP1

Priya Chatterji; Kelly A. Whelan; Sarah F. Andres; Fernando Cid Samper; Lauren A Simon; Rei Mizuno; Emma T. Lundsmith; David S.M. Lee; Shun Liang; H.R. Sagara Wijeratne; Stefanie Marti; Lillian Chau; Patrick A Williams; Veronique Giroux; Benjamin J. Wilkins; Gary D. Wu; Premal Shah; Gian Gaetano Tartaglia; Kathryn E Hamilton

RNA binding proteins, such as IMP1, are emerging as essential regulators of intestinal development and cancer. IMP1 hypomorphic mice exhibit severe intestinal growth defects, yet it’s role in adult intestinal epithelium is unclear. We employed ribosome profiling to test the effect of IMP1 loss on the “translatome” in colon cancer cell lines. In parallel, we evaluated mice with intestinal epithelial-specific Imp1 deletion (Imp1ΔIEC) following irradiation or colitis models. Ribosome-profiling revealed translation efficiency changes for multiple pathways important for intestinal homeostasis, including autophagy, in IMP1 knockout cells. We found increased autophagy flux in Imp1ΔIEC mice, reinforced through in silico and biochemical analyses revealing direct binding of IMP1 to autophagy transcripts MAP1LC3B and ATG3. We found that Imp1ΔIEC mice exhibit enhanced recovery following irradiation, which is attenuated with genetic deletion of autophagy gene Atg7. Finally, we demonstrated that IMP1 is upregulated in Crohn’s disease patients and Imp1 loss lessened colitis severity in mice. These studies demonstrate that IMP1 acts as a posttranscriptional regulator of gut epithelial repair post-irradiation and colitis, in part through modulation of autophagy.


Gastroenterology | 2018

FXR-Dependent Modulation of the Human Small Intestinal Microbiome by the Bile Acid Derivative Obeticholic Acid

Elliot S. Friedman; Yun Li; Ting-Chin David Shen; Jack Jiang; Lillian Chau; Luciano Adorini; Farah Babakhani; Jeffrey E. Edwards; D. Shapiro; Chunyu Zhao; Rotonya M. Carr; Kyle Bittinger; Hongzhe Li; Gary D. Wu

BACKGROUND & AIMS Intestinal bacteria can modify the composition of bile acids and bile acids, which are regulated by the farnesoid X receptor, affect the survival and growth of gut bacteria. We studied the effects of obeticholic acid (OCA), a bile acid analogue and farnesoid X receptor agonist, on the intestinal microbiomes of humans and mice. METHODS We performed a phase I study in 24 healthy volunteers given OCA (5, 10, or 25 mg/d for 17 days). Fecal and plasma specimens were collected at baseline (day 0) and on days 17 (end of dosing) and 37 (end of study). The fecal specimens were analyzed by shotgun meta-genomic sequencing. A Uniref90 high-stringency genomic analysis was used to assign specific genes to the taxonomic signature of bacteria whose abundance was associated with OCA. Male C57BL/6 mice were gavage fed daily with water, vehicle, or OCA (10 mg/kg) for 2 weeks. Small intestine luminal contents were collected by flushing with saline and fecal pellets were collected at baseline and day 14. Mouse samples were analyzed by 16S-tagged sequencing. Culture experiments were performed to determine the taxonomic-specific effects of bile acids and OCA on bacterial growth. RESULTS Suppression of endogenous bile acid synthesis by OCA in subjects led to a reversible induction of gram-positive bacteria that are found in the small intestine and are components of the diet and oral microbiota. We found that bile acids decreased proliferation of these bacteria in minimum inhibitory concentration assays. In these organisms, there was an increase in the representation of microbial genomic pathways involved in DNA synthesis and amino acid metabolism with OCA treatment of subjects. Consistent with these findings, mice fed OCA had lower endogenous bile acid levels and an increased proportion of Firmicutes, specifically in the small intestine, compared with mice fed water or vehicle. CONCLUSIONS In studying the effects of OCA in humans and mice, we found evidence for interactions between bile acids and features of the small intestinal microbiome. These findings indicate that farnesoid X receptor activation alters the intestinal microbiota and could provide opportunities for microbiome biomarker discovery or new approaches to engineering the human microbiome. ClinicalTrials.gov, NCT01933503.


Immunity | 2013

Dendritic Cell Expression of the Signaling Molecule TRAF6 Is Critical for Gut Microbiota-Dependent Immune Tolerance

Daehee Han; Matthew C. Walsh; Pedro J. Cejas; Nicholas N. Dang; Young Mi Kim; Jihyun Kim; Laetitia Charrier-Hisamuddin; Lillian Chau; Qin Zhang; Kyle Bittinger; Frederic D. Bushman; Laurence A. Turka; Hao Shen; Boris Reizis; Gary D. Wu; Yongwon Choi


Gastroenterology | 2018

824 - Inflammation-Induced Alterations in Colonic Epithelial Mitochondria Function Revealed by Fatty Acid Oxidation

Lillian Chau; Sarah A. Smith; Sayaka A. Ogawa; Kelly A. Whelan; Kathryn E. Hamilton; Jie Chen; Sue A. Keilbaugh; Franz Fogt; Meenakshi Bewtra; Barry Slaff; Aalim M. Weljie; Michael Bennett; Hiroshi Nakagawa; Gary D. Wu


Gastroenterology | 2018

299 - FXR-Dependent Modification of the Human Small Intestinal Microbiome

Elliot S. Friedman; Ting-Chin David Shen; Yun Li; Jack Jiang; Lillian Chau; Luciano Adorini; Farah Babakhani; Jeffrey E. Edwards; Chunyu Zhao; Kyle Bittinger; Hongzhe Li; Gary D. Wu


Gastroenterology | 2018

Su1056 - Unraveling Post-Transcriptional Regulation of Intestinal Response to Acute Challenge by the RNA Binding Protein IMP1

Priya Chatterji; Lauren Simon; Sarah F. Andres; Rei Mizuno; Emma Lundsmith; Lillian Chau; Veronique Giroux; Amanda Mah; Laurianne Van Landeghem; Gary D. Wu; Kelly A. Whelan; Anil K. Rustgi; Kathryn E. Hamilton


Gastroenterology | 2017

A Role for Bacterial Urease in Crohn's Disease and Gut Dysbiosis

Josephine Ni; Ting-Chin David Shen; Eric Z. Chen; Aubrey Bailey; Manuela Roggiani; Alexandra Sirota-Madi; Elliot S. Friedman; Lillian Chau; Lindsey Albenberg; Robert N. Baldassano; Jonathan Braun; Ramnik J. Xavier; Clary B. Clish; Marc Yudkoff; Hongzhe Li; Mark Goulian; Frederic D. Bushman; James D. Lewis; Gary D. Wu

Collaboration


Dive into the Lillian Chau's collaboration.

Top Co-Authors

Avatar

Gary D. Wu

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Kyle Bittinger

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hongzhe Li

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Jack Jiang

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

James D. Lewis

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lindsey Albenberg

Children's Hospital of Philadelphia

View shared research outputs
Researchain Logo
Decentralizing Knowledge