Network


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

Hotspot


Dive into the research topics where Lars Eckmann is active.

Publication


Featured researches published by Lars Eckmann.


Cell | 2004

IKKβ Links Inflammation and Tumorigenesis in a Mouse Model of Colitis-Associated Cancer

Florian R. Greten; Lars Eckmann; Tim F. Greten; Jin Mo Park; Zhi-Wei Li; Laurence J. Egan; Martin F. Kagnoff; Michael Karin

A link between inflammation and cancer has long been suspected, but its molecular nature remained ill defined. A key player in inflammation is transcription factor NF-kappaB whose activity is triggered in response to infectious agents and proinflammatory cytokines via the IkappaB kinase (IKK) complex. Using a colitis-associated cancer model, we show that although deletion of IKKbeta in intestinal epithelial cells does not decrease inflammation, it leads to a dramatic decrease in tumor incidence without affecting tumor size. This is linked to increased epithelial apoptosis during tumor promotion. Deleting IKKbeta in myeloid cells, however, results in a significant decrease in tumor size. This deletion diminishes expression of proinflammatory cytokines that may serve as tumor growth factors, without affecting apoptosis. Thus, specific inactivation of the IKK/NF-kappaB pathway in two different cell types can attenuate formation of inflammation-associated tumors. In addition to suppressing apoptosis in advanced tumors, IKKbeta may link inflammation to cancer.


Cancer Cell | 2009

IL-6 and Stat3 Are Required for Survival of Intestinal Epithelial Cells and Development of Colitis-Associated Cancer

Sergei I. Grivennikov; Eliad Karin; Janoš Terzić; Daniel Mucida; Guann-Yi Yu; Sivakumar Vallabhapurapu; Jürgen Scheller; Stefan Rose-John; Hilde Cheroutre; Lars Eckmann; Michael Karin

Colitis-associated cancer (CAC) is the most serious complication of inflammatory bowel disease. Proinflammatory cytokines have been suggested to regulate preneoplastic growth during CAC tumorigenesis. Interleukin 6 (IL-6) is a multifunctional NF-kappaB-regulated cytokine that acts on epithelial and immune cells. Using genetic tools, we now demonstrate that IL-6 is a critical tumor promoter during early CAC tumorigenesis. In addition to enhancing proliferation of tumor-initiating cells, IL-6 produced by lamina propria myeloid cells protects normal and premalignant intestinal epithelial cells (IECs) from apoptosis. The proliferative and survival effects of IL-6 are largely mediated by the transcription factor Stat3, whose IEC-specific ablation has profound impact on CAC tumorigenesis. Thus, the NF-kappaB-IL-6-Stat3 cascade is an important regulator of the proliferation and survival of tumor-initiating IECs.


Journal of Clinical Investigation | 1995

A distinct array of proinflammatory cytokines is expressed in human colon epithelial cells in response to bacterial invasion.

Hyun C. Jung; Lars Eckmann; Suk-Kyun Yang; Asit Panja; Joshua Fierer; E. Morzycka-Wroblewska; Martin F. Kagnoff

Pathogenic bacteria that penetrate the intestinal epithelial barrier stimulate an inflammatory response in the adjacent intestinal mucosa. The present studies asked whether colon epithelial cells can provide signals that are important for the initiation and amplification of an acute mucosal inflammatory response. Infection of monolayers of human colon epithelial cell lines (T84, HT29, Caco-2) with invasive strains of bacteria (Salmonella dublin, Shigella dysenteriae, Yersinia enterocolitica, Listeria monocytogenes, enteroinvasive Escherichia coli) resulted in the coordinate expression and upregulation of a specific array of four proinflammatory cytokines, IL-8, monocyte chemotactic protein-1, GM-CSF, and TNF alpha, as assessed by mRNA levels and cytokine secretion. Expression of the same cytokines was upregulated after TNF alpha or IL-1 stimulation of these cells. In contrast, cytokine gene expression was not altered after infection of colon epithelial cells with noninvasive bacteria or the noninvasive protozoan parasite, G. lamblia. Notably, none of the cell lines expressed mRNA for IL-2, IL-4, IL-5, IL-6, IL-12p40, IFN-gamma, or significant levels of IL-1 or IL-10 in response to the identical stimuli. The coordinate expression of IL-8, MCP-1, GM-CSF and TNF alpha appears to be a general property of human colon epithelial cells since an identical array of cytokines, as well as IL-6, also was expressed by freshly isolated human colon epithelial cells. Since the cytokines expressed in response to bacterial invasion or other proinflammatory agonists have a well documented role in chemotaxis and activation of inflammatory cells, colon epithelial cells appear to be programmed to provide a set of signals for the activation of the mucosal inflammatory response in the earliest phases after microbial invasion.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Paneth cells directly sense gut commensals and maintain homeostasis at the intestinal host-microbial interface.

Shipra Vaishnava; Cassie L. Behrendt; Anisa S. Ismail; Lars Eckmann; Lora V. Hooper

The intestinal epithelium is in direct contact with a vast microbiota, yet little is known about how epithelial cells defend the host against the heavy bacterial load. To address this question we studied Paneth cells, a key small intestinal epithelial lineage. We found that Paneth cells directly sense enteric bacteria through cell-autonomous MyD88-dependent toll-like receptor (TLR) activation, triggering expression of multiple antimicrobial factors. Paneth cells were essential for controlling intestinal barrier penetration by commensal and pathogenic bacteria. Furthermore, Paneth cell-intrinsic MyD88 signaling limited bacterial penetration of host tissues, revealing a role for epithelial MyD88 in maintaining intestinal homeostasis. Our findings establish that gut epithelia actively sense enteric bacteria and play an essential role in maintaining host-microbial homeostasis at the mucosal interface.


Nature | 2012

Adenoma-linked barrier defects and microbial products drive IL-23/IL-17-mediated tumour growth.

Sergei I. Grivennikov; Kepeng Wang; Daniel Mucida; C. Andrew Stewart; Bernd Schnabl; Dominik Jauch; Koji Taniguchi; Guann Yi Yu; Christoph H. Österreicher; Kenneth E. Hung; Christian Datz; Ying Feng; Eric R. Fearon; Mohamed Oukka; Lino Tessarollo; Vincenzo Coppola; Felix Yarovinsky; Hilde Cheroutre; Lars Eckmann; Giorgio Trinchieri; Michael Karin

Approximately 2% of colorectal cancer is linked to pre-existing inflammation known as colitis-associated cancer, but most develops in patients without underlying inflammatory bowel disease. Colorectal cancer often follows a genetic pathway whereby loss of the adenomatous polyposis coli (APC) tumour suppressor and activation of β-catenin are followed by mutations in K-Ras, PIK3CA and TP53, as the tumour emerges and progresses. Curiously, however, ‘inflammatory signature’ genes characteristic of colitis-associated cancer are also upregulated in colorectal cancer. Further, like most solid tumours, colorectal cancer exhibits immune/inflammatory infiltrates, referred to as ‘tumour-elicited inflammation’. Although infiltrating CD4+ TH1 cells and CD8+ cytotoxic T cells constitute a positive prognostic sign in colorectal cancer, myeloid cells and T-helper interleukin (IL)-17-producing (TH17) cells promote tumorigenesis, and a ‘TH17 expression signature’ in stage I/II colorectal cancer is associated with a drastic decrease in disease-free survival. Despite its pathogenic importance, the mechanisms responsible for the appearance of tumour-elicited inflammation are poorly understood. Many epithelial cancers develop proximally to microbial communities, which are physically separated from immune cells by an epithelial barrier. We investigated mechanisms responsible for tumour-elicited inflammation in a mouse model of colorectal tumorigenesis, which, like human colorectal cancer, exhibits upregulation of IL-23 and IL-17. Here we show that IL-23 signalling promotes tumour growth and progression, and development of a tumoural IL-17 response. IL-23 is mainly produced by tumour-associated myeloid cells that are likely to be activated by microbial products, which penetrate the tumours but not adjacent tissue. Both early and late colorectal neoplasms exhibit defective expression of several barrier proteins. We propose that barrier deterioration induced by colorectal-cancer-initiating genetic lesions results in adenoma invasion by microbial products that trigger tumour-elicited inflammation, which in turn drives tumour growth.


Gastroenterology | 1993

Differential cytokine expression by human intestinal epithelial cell lines: Regulated expression of interleukin 8

Lars Eckmann; Hyun C. Jung; Cornelia Schürer-Maly; Asit Panja; E. Morzycka-Wroblewska; Martin F. Kagnoff

BACKGROUND To characterize the role of intestinal epithelial cells in mucosal host defense, we have examined constitutive cytokine expression and regulated expression of interleukin (IL)-8 by human colonic epithelial cells. METHODS Cytokine expression by the human colonic epithelial cell lines, T84, Caco-2, SW620, and HT29 was assessed by using polymerase chain reaction amplification of reverse-transcribed RNA. Regulated IL-8 expression was analyzed by nuclear run-off assays, Northern blot analysis, and enzyme-linked immunosorbent assay. RESULTS The cell lines constitutively expressed messenger RNA (mRNA) for IL-8 and transforming growth factor beta 1. In addition, some cell lines expressed mRNA for IL-1 alpha, IL-1 beta, IL-10 and tumor necrosis factor alpha (TNF alpha). None of the cell lines expressed mRNA for IL-2, IL-4, IL-5, IL-6, or interferon gamma. Cell lines secreted IL-8 either constitutively or after stimulation with the physiological agonists TNF alpha, IL-1 beta, or lipopolysaccharide. Increased IL-8 secretion after TNF alpha stimulation of T84 cells was accompanied by increased IL-8 mRNA levels and an increased transcription rate of the IL-8 gene. IL-8 was preferentially secreted at the basolateral surface of polarized T84 cells. In further studies, freshly isolated human colon epithelial cells also secreted IL-8. CONCLUSIONS These results support the notion of bidirectional communication between intestinal epithelial cells and mucosal immune and inflammatory cells.


Nature Cell Biology | 2006

Maintenance of colonic homeostasis by distinctive apical TLR9 signalling in intestinal epithelial cells

Jongdae Lee; Ji-Hun Mo; Kyoko Katakura; Irit Alkalay; Adam N. Rucker; Yu-Tsueng Liu; Hyun-Ku Lee; Carol Shen; Gady Cojocaru; Steve Shenouda; Martin F. Kagnoff; Lars Eckmann; Yinon Ben-Neriah; Eyal Raz

The mechanisms by which commensal bacteria suppress inflammatory signalling in the gut are still unclear. Here, we present a cellular mechanism whereby the polarity of intestinal epithelial cells (IECs) has a major role in colonic homeostasis. TLR9 activation through apical and basolateral surface domains have distinct transcriptional responses, evident by NF-κB activation and cDNA microarray analysis. Whereas basolateral TLR9 signals IκBα degradation and activation of the NF-κB pathway, apical TLR9 stimulation invokes a unique response in which ubiquitinated IκB accumulates in the cytoplasm preventing NF-κB activation. Furthermore, apical TLR9 stimulation confers intracellular tolerance to subsequent TLR challenges. IECs in TLR9-deficient mice, when compared with wild-type and TLR2-deficient mice, display a lower NF-κB activation threshold and these mice are highly susceptible to experimental colitis. Our data provide a case for organ-specific innate immunity in which TLR expression in polarized IECs has uniquely evolved to maintain colonic homeostasis and regulate tolerance and inflammation.


Journal of Clinical Investigation | 1997

Secretion of proinflammatory cytokines by epithelial cells in response to Chlamydia infection suggests a central role for epithelial cells in chlamydial pathogenesis.

Stephanie Rasmussen; Lars Eckmann; Alison J. Quayle; Li Shen; You-Xun Zhang; Deborah J. Anderson; Joshua Fierer; Richard S. Stephens; Martin F. Kagnoff

Chlamydia species infect epithelial cells at mucosal surfaces, and are major causes of sexually transmitted diseases. Infection is characterized by inflammation which is exacerbated upon reinfection, ultimately leading to tissue damage and scarring. Although central for the development of disease manifestations, little is known about the mechanisms that initiate and sustain the inflammatory response to Chlamydia. Infection of cervical and colonic epithelial cells with Chlamydia trachomatis and Chlamydia psittaci is shown in the present studies to upregulate mRNA expression and secretion of the proinflammatory cytokines IL-8, GRO alpha, GM-CSF, and IL-6. In contrast to the rapid, but transient, cytokine induction following infection with other invasive bacteria, the epithelial cytokine response to Chlamydia was delayed until 20-24 h after infection, persisted throughout the chlamydial growth cycle (2-4 d), and required bacterial protein synthesis. Moreover, epithelial cell lines and primary endocervical epithelial cells released IL-1alpha after Chlamydia infection, and increased secretion of the proinflammatory cytokines could be inhibited by anti-IL-1alpha. This suggests that IL-1alpha, released following lysis of infected epithelial cells, may amplify the inflammatory response by stimulating additional cytokine production by noninfected neighboring cells. These findings suggest a novel pathophysiologic concept wherein the acute host response to Chlamydia at mucosal surfaces is primarily initiated and sustained by epithelial cells, the first and major targets of chlamydial infection.


Cell | 2007

NF-kappaB is a negative regulator of IL-1beta secretion as revealed by genetic and pharmacological inhibition of IKKbeta.

Florian R. Greten; Melek C. Arkan; Julia Bollrath; Li-Chung Hsu; Jason Goode; Cornelius Miething; Serkan Göktuna; Michael Neuenhahn; Joshua Fierer; Stephan Paxian; Nico van Rooijen; Yajun Xu; Timothy D. Ocain; Bruce Jaffee; Dirk H. Busch; Justus Duyster; Roland M. Schmid; Lars Eckmann; Michael Karin

IKKbeta-dependent NF-kappaB activation plays a key role in innate immunity and inflammation, and inhibition of IKKbeta has been considered as a likely anti-inflammatory therapy. Surprisingly, however, mice with a targeted IKKbeta deletion in myeloid cells are more susceptible to endotoxin-induced shock than control mice. Increased endotoxin susceptibility is associated with elevated plasma IL-1beta as a result of increased pro-IL-1beta processing, which was also seen upon bacterial infection. In macrophages enhanced pro-IL-1beta processing depends on caspase-1, whose activation is inhibited by NF-kappaB-dependent gene products. In neutrophils, however, IL-1beta secretion is caspase-1 independent and depends on serine proteases, whose activity is also inhibited by NF-kappaB gene products. Prolonged pharmacologic inhibition of IKKbeta also augments IL-1beta secretion upon endotoxin challenge. These results unravel an unanticipated role for IKKbeta-dependent NF-kappaB signaling in the negative control of IL-1beta production and highlight potential complications of long-term IKKbeta inhibition.


Journal of Clinical Investigation | 2005

Toll-like receptor 9–induced type I IFN protects mice from experimental colitis

Kyoko Katakura; Jongdae Lee; Daniel Rachmilewitz; Gloria C. Li; Lars Eckmann; Eyal Raz

Experimental colitis is mediated by inflammatory or dysregulated immune responses to microbial factors of the gastrointestinal tract. In this study we observed that administration of Toll-like receptor 9 (TLR9) agonists suppressed the severity of experimental colitis in RAG1-/- but not in SCID mice. This differential responsiveness between phenotypically similar but genetically distinct animals was related to a partial blockade in TLR9 signaling and defective production of type I IFN (i.e., IFN-alpha/beta) in SCID mice upon TLR9 stimulation. The addition of neutralization antibodies against type I IFN abolished the antiinflammatory effects induced by TLR9 agonists, whereas the administration of recombinant IFN-beta mimicked the antiinflammatory effects induced by TLR9 agonists in this model. Furthermore, mice deficient in the IFN-alpha/beta receptor exhibited more severe colitis than wild-type mice did upon induction of experimental colitis. These results indicate that TLR9-triggered type I IFN has antiinflammatory functions in colitis. They also underscore the important protective role of type I IFN in intestinal homeostasis and suggest that strategies to modulate innate immunity may be of therapeutic value for the treatment of intestinal inflammatory conditions.

Collaboration


Dive into the Lars Eckmann's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael Karin

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joshua Fierer

University of California

View shared research outputs
Top Co-Authors

Avatar

Sara M. Dann

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael B. Dwinell

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Sharon L. Reed

University of California

View shared research outputs
Top Co-Authors

Avatar

Christine Le

University of California

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge