Network


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

Hotspot


Dive into the research topics where Marcus Mühlbauer is active.

Publication


Featured researches published by Marcus Mühlbauer.


Science | 2012

Intestinal Inflammation Targets Cancer-Inducing Activity of the Microbiota

Janelle C. Arthur; Ernesto Perez-Chanona; Marcus Mühlbauer; Sarah Tomkovich; Joshua M. Uronis; Ting Jia Fan; Barry J. Campbell; Turki Abujamel; Belgin Dogan; Arlin B. Rogers; Jonathan Rhodes; Alain Stintzi; Kenneth W. Simpson; Jonathan J. Hansen; Temitope O. Keku; Anthony A. Fodor; Christian Jobin

Of Microbes and Cancer Inflammation is a well-established driver of tumorigenesis. For example, patients with inflammatory bowel disease have an elevated risk of developing colorectal cancer (CRC). Whether the gut microbiota also contributes to the development of CRC is less well understood. Arthur et al. (p. 120, published online 16 August; see the Perspective by Schwabe and Wang) now show that the microbiota does indeed promote tumorigenesis in an inflammation-driven model of CRC in mice. Although germ-free mice were protected against developing cancer, colonization of mice with Escherichia coli was sufficient to drive tumorigenesis. Microbes resident in the gut can promote colorectal cancer in mice in an inflammation-independent manner. Inflammation alters host physiology to promote cancer, as seen in colitis-associated colorectal cancer (CRC). Here, we identify the intestinal microbiota as a target of inflammation that affects the progression of CRC. High-throughput sequencing revealed that inflammation modifies gut microbial composition in colitis-susceptible interleukin-10–deficient (Il10−/−) mice. Monocolonization with the commensal Escherichia coli NC101 promoted invasive carcinoma in azoxymethane (AOM)–treated Il10−/− mice. Deletion of the polyketide synthase (pks) genotoxic island from E. coli NC101 decreased tumor multiplicity and invasion in AOM/Il10−/− mice, without altering intestinal inflammation. Mucosa-associated pks+ E. coli were found in a significantly high percentage of inflammatory bowel disease and CRC patients. This suggests that in mice, colitis can promote tumorigenesis by altering microbial composition and inducing the expansion of microorganisms with genotoxic capabilities.


PLOS ONE | 2009

Modulation of the Intestinal Microbiota Alters Colitis-Associated Colorectal Cancer Susceptibility

Joshua M. Uronis; Marcus Mühlbauer; Hans H. Herfarth; Tara C. Rubinas; Gieira S. Jones; Christian Jobin

It is well established that the intestinal microbiota plays a key role in the pathogenesis of Crohns disease (CD) and ulcerative colitis (UC) collectively referred to as inflammatory bowel disease (IBD). Epidemiological studies have provided strong evidence that IBD patients bear increased risk for the development of colorectal cancer (CRC). However, the impact of the microbiota on the development of colitis-associated cancer (CAC) remains largely unknown. In this study, we established a new model of CAC using azoxymethane (AOM)-exposed, conventionalized-Il10−/− mice and have explored the contribution of the host intestinal microbiota and MyD88 signaling to the development of CAC. We show that 8/13 (62%) of AOM-Il10−/− mice developed colon tumors compared to only 3/15 (20%) of AOM- wild-type (WT) mice. Conventionalized AOM-Il10−/− mice developed spontaneous colitis and colorectal carcinomas while AOM-WT mice were colitis-free and developed only rare adenomas. Importantly, tumor multiplicity directly correlated with the presence of colitis. Il10−/− mice mono-associated with the mildly colitogenic bacterium Bacteroides vulgatus displayed significantly reduced colitis and colorectal tumor multiplicity compared to Il10−/− mice. Germ-free AOM-treated Il10−/− mice showed normal colon histology and were devoid of tumors. Il10−/−; Myd88−/− mice treated with AOM displayed reduced expression of Il12p40 and Tnfα mRNA and showed no signs of tumor development. We present the first direct demonstration that manipulation of the intestinal microbiota alters the development of CAC. The TLR/MyD88 pathway is essential for microbiota-induced development of CAC. Unlike findings obtained using the AOM/DSS model, we demonstrate that the severity of chronic colitis directly correlates to colorectal tumor development and that bacterial-induced inflammation drives progression from adenoma to invasive carcinoma.


Journal of Immunology | 2007

Gnotobiotic IL-10−/−;NF-κBEGFP Mice Reveal the Critical Role of TLR/NF-κB Signaling in Commensal Bacteria-Induced Colitis

Thomas Karrasch; Joo Sung Kim; Marcus Mühlbauer; Scott T. Magness; Christian Jobin

Commensal bacteria and TLR signaling have been associated with the maintenance of intestinal homeostasis in dextran sodium sulfate-induced intestinal injury. The aim of this study was to determine the in vivo role of TLR/NF-κB activation in a model of commensal bacteria-induced T cell-mediated colitis. A NF-κB reporter gene mouse (NF-κBEGFP) (EGFP, enhanced GFP) was crossed to the colitogenic susceptible strain IL-10−/− and derived into germfree conditions using embryo-transfer technology. Germfree IL-10wt/wt;NF-κBEGFP and IL-10−/−;NF-κBEGFP mice (wt, wild type) were dual associated with the nonpathogenic commensal bacteria strains Enterococcus faecalis and Escherichia coli. EGFP was detected using macroimaging, confocal microscopy, and flow cytometry. IL-10−/−;MyD88−/− mice were used to assess E. faecalis/E. coli-induced TLR-dependent signaling and IL-23 gene expression. Dual-associated IL-10−/−;NF-κBEGFP mice developed severe inflammation by 7 wk. Macroscopic analysis showed elevated EGFP expression throughout the colon of bacteria-associated IL-10−/−;NF-κBEGFP mice. Confocal microscopy analysis revealed EGFP-positive enterocytes during the early phase of bacterial colonization (1 wk) in both IL-10wt/wt and IL-10−/− mice, while the signal shifted toward lamina propria T cells, dendritic cells, neutrophils, and macrophages in IL-10−/− mice during colitis (7 wk). The NF-κB inhibitor BAY 11-7085 attenuated E. faecalis/E. coli-induced EGFP expression and development of colitis. Additionally, E. faecalis/E. coli-induced NF-κB signaling and IL-23 gene expression were blocked in bone marrow-derived dendritic cells derived from IL-10−/−;MyD88−/− mice. We conclude that bacteria-induced experimental colitis involves the activation of TLR-induced NF-κB signaling derived mostly from mucosal immune cells. Blocking TLR-induced NF-κB activity may represent an attractive strategy to treat immune-mediated intestinal inflammation.


Nature Communications | 2014

Microbial genomic analysis reveals the essential role of inflammation in bacteria-induced colorectal cancer

Janelle C. Arthur; Raad Z. Gharaibeh; Marcus Mühlbauer; Ernesto Perez-Chanona; Joshua M. Uronis; Jonathan McCafferty; Anthony A. Fodor; Christian Jobin

Enterobacteria, especially Escherichia coli, are abundant in patients with inflammatory bowel disease or colorectal cancer (CRC). However, it is unclear whether cancer is promoted by inflammation-induced expansion of E. coli and/or changes in expression of specific microbial genes. Here we use longitudinal (2, 12 and 20 weeks) 16S rRNA sequencing of luminal microbiota from ex-germ free mice to show that inflamed Il10−/− mice maintain a higher abundance of Enterobacteriaceae than healthy wild-type mice. Experiments with mono-colonized Il10−/− mice reveal that host inflammation is necessary for E. coli cancer-promoting activity. RNA-sequence analysis indicates significant changes in E. coli gene catalogue in Il10−/− mice, with changes mostly driven by adaptation to the intestinal environment. Expression of specific genes present in the tumor-promoting E. coli pks island are modulated by inflammation/CRC development. Thus, progression of inflammation in Il10−/− mice supports Enterobacteriaceae and alters a small subset of microbial genes important for tumor development.


Gastroenterology | 2011

Microbial Colonization Induces Dynamic Temporal and Spatial Patterns of NF-κB Activation in the Zebrafish Digestive Tract

Michelle Kanther; Xiaolun Sun; Marcus Mühlbauer; Lantz C. Mackey; Edward J. Flynn; Michel Bagnat; Christian Jobin; John F. Rawls

BACKGROUND & AIMS The nuclear factor κ-light-chain enhancer of activated B cells (NF-κB) transcription factor pathway is activated in response to diverse microbial stimuli to regulate expression of genes involved in immune responses and tissue homeostasis. However, the temporal and spatial activation of NF-κB in response to microbial signals have not been determined in whole living organisms, and the molecular and cellular details of these responses are not well understood. We used in vivo imaging and molecular approaches to analyze NF-κB activation in response to the commensal microbiota in transparent gnotobiotic zebrafish. METHODS We used DNA microarrays, in situ hybridization, and quantitative reverse transcription polymerase chain reaction analyses to study the effects of the commensal microbiota on gene expression in gnotobiotic zebrafish. Zebrafish PAC2 and ZFL cells were used to study the NF-κB signaling pathway in response to bacterial stimuli. We generated transgenic zebrafish that express enhanced green fluorescent protein under transcriptional control of NF-κB, and used them to study patterns of NF-κB activation during development and microbial colonization. RESULTS Bacterial stimulation induced canonical activation of the NF-κB pathway in zebrafish cells. Colonization of germ-free transgenic zebrafish with a commensal microbiota activated NF-κB and led to up-regulation of its target genes in intestinal and extraintestinal tissues of the digestive tract. Colonization with the bacterium Pseudomonas aeruginosa was sufficient to activate NF-κB, and this activation required a functional flagellar apparatus. CONCLUSIONS In zebrafish, transcriptional activity of NF-κB is spatially and temporally regulated by specific microbial factors. The observed patterns of NF-κB-dependent responses to microbial colonization indicate that cells in the gastrointestinal tract respond robustly to the microbial environment.


The ISME Journal | 2013

Stochastic changes over time and not founder effects drive cage effects in microbial community assembly in a mouse model

Jonathan McCafferty; Marcus Mühlbauer; Raad Z. Gharaibeh; Janelle C. Arthur; Ernesto Perez-Chanona; Wei Sha; Christian Jobin; Anthony A. Fodor

Maternal transmission and cage effects are powerful confounding factors in microbiome studies. To assess the consequences of cage microenvironment on the mouse gut microbiome, two groups of germ-free (GF) wild-type (WT) mice, one gavaged with a microbiota harvested from adult WT mice and another allowed to acquire the microbiome from the cage microenvironment, were monitored using Illumina 16S rRNA sequencing over a period of 8 weeks. Our results revealed that cage effects in WT mice moved from GF to specific pathogen free (SPF) conditions take several weeks to develop and are not eliminated by the initial gavage treatment. Initial gavage influenced, but did not eliminate a successional pattern in which Proteobacteria became less abundant over time. An analysis in which 16S rRNA sequences are mapped to the closest sequenced whole genome suggests that the functional potential of microbial genomes changes significantly over time shifting from an emphasis on pathogenesis and motility early in community assembly to metabolic processes at later time points. Functionally, mice allowed to naturally acquire a microbial community from their cage, but not mice gavaged with a common biome, exhibit a cage effect in Dextran Sulfate Sodium-induced inflammation. Our results argue that while there are long-term effects of the founding community, these effects are mitigated by cage microenvironment and successional community assembly over time, which must both be explicitly considered in the interpretation of microbiome mouse experiments.


Nature Medicine | 2015

Inflammasome-independent role of AIM2 in suppressing colon tumorigenesis via DNA-PK and Akt.

Justin E. Wilson; Alex Petrucelli; Liang Chen; A. Alicia Koblansky; Agnieszka D. Truax; Yoshitaka Oyama; Arlin B. Rogers; W. June Brickey; Yuli Wang; Monika Schneider; Marcus Mühlbauer; Wei Chun Chou; Brianne R. Barker; Christian Jobin; Nancy L. Allbritton; Dale A. Ramsden; Beckley K. Davis; Jenny P.Y. Ting

The inflammasome activates caspase-1 and the release of interleukin-1β (IL-1β) and IL-18, and several inflammasomes protect against intestinal inflammation and colitis-associated colon cancer (CAC) in animal models. The absent in melanoma 2 (AIM2) inflammasome is activated by double-stranded DNA, and AIM2 expression is reduced in several types of cancer, but the mechanism by which AIM2 restricts tumor growth remains unclear. We found that Aim2-deficient mice had greater tumor load than Asc-deficient mice in the azoxymethane/dextran sodium sulfate (AOM/DSS) model of colorectal cancer. Tumor burden was also higher in Aim2−/−/ApcMin/+ than in APCMin/+ mice. The effects of AIM2 on CAC were independent of inflammasome activation and IL-1β and were primarily mediated by a non–bone marrow source of AIM2. In resting cells, AIM2 physically interacted with and limited activation of DNA-dependent protein kinase (DNA-PK), a PI3K-related family member that promotes Akt phosphorylation, whereas loss of AIM2 promoted DNA-PK–mediated Akt activation. AIM2 reduced Akt activation and tumor burden in colorectal cancer models, while an Akt inhibitor reduced tumor load in Aim2−/− mice. These findings suggest that Akt inhibitors could be used to treat AIM2-deficient human cancers.


Gastroenterology | 2010

Tumor Necrosis Factor and Interferon-γ Down-regulate Klotho in Mice With Colitis

Robert D. Thurston; Claire B. Larmonier; Pawel Majewski; Rajalakshmy Ramalingam; Monica T. Midura-Kiela; Daniel Laubitz; Alain Vandewalle; David G. Besselsen; Marcus Mühlbauer; Christian Jobin; Pawel R. Kiela; Fayez K. Ghishan

BACKGROUND & AIMS Klotho (KL) is an anti-inflammatory protein that protects the endothelium from nitric oxide (NO)-induced dysfunction, reduces the expression of endothelial adhesion molecules, and potentially regulates T-cell functions. KL deficiency leads to premature senescence and impaired Ca2+/Pi homeostasis, which can lead to inflammatory bowel disease (IBD)-associated osteopenia/osteoporosis. We investigated the changes in renal expression of Kl as a consequence of colitis. METHODS We studied 3 mouse models of IBD: colitis induced by trinitrobenzene sulfonic acid, colitis induced by microflora (in gnotobiotic interleukin-10(-/-)), and colitis induced by adoptive transfer of CD4(+)CD45RB(high) T cells. Effects of the tumor necrosis factor (TNF) and interferon (IFN)-gamma on Kl expression and the activity of its promoter were examined in renal epithelial cells (mpkDCT4 and mIMCD3). RESULTS Renal expression of Kl messenger RNA (mRNA) and protein was reduced in all 3 models of IBD. Reduced level of KL correlated with the severity of colitis; the effect was reversed by neutralizing antibodies against TNF. In vitro, TNF inhibited Kl expression, an effect potentiated by IFN-gamma. The combination of TNF and IFN-gamma increased expression of inducible nitric oxide synthase (iNOS) and increased NO production. The effect of IFN-gamma was reproduced by exposure to an NO donor and reversed by the iNOS inhibitor. In cells incubated with TNF and/or IFN-gamma, Kl mRNA stability was unaffected, whereas Kl promoter activity was reduced, indicating that these cytokines regulate Kl at the transcriptional level. CONCLUSIONS The down-regulation of KL that occurs during inflammation might account for the extraintestinal complications such as abnormalities in bone homeostasis that occur in patients with IBD.


Molecular Pharmacology | 2008

Interleukin-8 expression is regulated by histone deacetylases through the nuclear factor-kappaB pathway in breast cancer.

Carine Chavey; Marcus Mühlbauer; Carine Bossard; Ariane Freund; Sébastien Durand; Christian Jorgensen; Christian Jobin; Gwendal Lazennec

We have reported recently that the chemokine interleukin 8 (IL-8)/CXCL8 was overexpressed in invasive estrogen receptor (ERα)-negative breast cancer cells compared with ERα-positive breast cancer cells. We now demonstrate that histone deacetylases (HDACs) play an essential role in the regulation of IL-8 gene expression in ERα-positive MCF-7 breast cancer cells. Treatment of MCF-7 cells with the HDAC inhibitor trichostatin A (TSA) led to a strong up-regulation of IL-8 protein and RNA levels in MCF-7 cells. The up-regulation of IL-8 in MCF-7 cells was time- and concentration-dependent. Moreover, run-on and transfection experiments demonstrated that IL-8 induction by HDAC inhibitors was transcriptional and involved mainly the nuclear factor-κB (NF-κB) site of the IL-8 promoter. These observations are corroborated by an up-regulation of NF-κB activity in MCF-7 cells in the presence of TSA. In addition, blocking NF-κB pathway by adenoviral delivery of a dominant-negative IκBorIκB kinase complex 2 (IKK2) mutant abolished IL-8 gene induction by histone deacetylase inhibitors. HDAC inhibitors triggered IKK phosphorylation and up-regulated p65 nuclear translocation, although they decreased the protein levels of IκBα, which accounts for NF-κB activation. TSA increased binding of acetylated histone 3 to the IL-8 gene promoter. In summary, our results demonstrate that NF-κB pathway repression by HDAC is responsible for the low expression of IL-8 in ERα-positive breast cancer cells.


Nature Communications | 2016

Altered intestinal microbiota–host mitochondria crosstalk in new onset Crohn’s disease

Walid Mottawea; Cheng-Kang Chiang; Marcus Mühlbauer; Amanda E. Starr; James Butcher; Turki Abujamel; Shelley A. Deeke; Annette Brandel; Hu Zhou; Shadi Shokralla; Mehrdad Hajibabaei; Ruth Singleton; Eric I. Benchimol; Christian Jobin; David R. Mack; Daniel Figeys; Alain Stintzi

Intestinal microbial dysbiosis is associated with Crohns disease (CD). However, the mechanisms leading to the chronic mucosal inflammation that characterizes this disease remain unclear. In this report, we use systems-level approaches to study the interactions between the gut microbiota and host in new-onset paediatric patients to evaluate causality and mechanisms of disease. We report an altered host proteome in CD patients indicative of impaired mitochondrial functions. In particular, mitochondrial proteins implicated in H2S detoxification are downregulated, while the relative abundance of H2S microbial producers is increased. Network correlation analysis reveals that Atopobium parvulum controls the central hub of H2S producers. A. parvulum induces pancolitis in colitis-susceptible interleukin-10-deficient mice and this phenotype requires the presence of the intestinal microbiota. Administrating the H2S scavenger bismuth mitigates A. parvulum-induced colitis in vivo. This study reveals that host–microbiota interactions are disturbed in CD and thus provides mechanistic insights into CD pathogenesis.

Collaboration


Dive into the Marcus Mühlbauer's collaboration.

Top Co-Authors

Avatar

Christian Jobin

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Ernesto Perez-Chanona

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Anthony A. Fodor

University of North Carolina at Charlotte

View shared research outputs
Top Co-Authors

Avatar

Janelle C. Arthur

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Joshua M. Uronis

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Raad Z. Gharaibeh

University of North Carolina at Charlotte

View shared research outputs
Top Co-Authors

Avatar

Sarah Tomkovich

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

A. Alicia Koblansky

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Agnieszka D. Truax

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge