201
|
From mice to men: Murine models of colorectal cancer for use in translational research. Crit Rev Oncol Hematol 2015; 98:94-105. [PMID: 26558688 DOI: 10.1016/j.critrevonc.2015.10.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 08/28/2015] [Accepted: 10/27/2015] [Indexed: 12/18/2022] Open
Abstract
Colorectal cancer (CRC) is the third most common carcinoma worldwide and despite advances in treatment, survival for patients with metastatic disease remains poor. With nearly 50% of patients developing metastases, in vivo investigation is essential to improve outcomes for these patients and numerous murine models of CRC have been developed to allow the study of chemoprevention and chemotherapy, in addition to improving our understanding of the pathogenesis of CRC. Selecting the most appropriate murine model for a specific application will maximize the conversion of potential therapies from the laboratory to clinical practice and requires an understanding of the various models available. This review will provide an overview of the murine models currently used in CRC research, discussing the limitations and merits of each and their most relevant application. It is aimed at the developing researcher, acting as a guide to prompt further reading in planning a specific study.
Collapse
|
202
|
Whiting N, Hu J, Shah JV, Cassidy MC, Cressman E, Zacharias Millward N, Menter DG, Marcus CM, Bhattacharya PK. Real-Time MRI-Guided Catheter Tracking Using Hyperpolarized Silicon Particles. Sci Rep 2015; 5:12842. [PMID: 26239953 PMCID: PMC4523869 DOI: 10.1038/srep12842] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 07/13/2015] [Indexed: 11/25/2022] Open
Abstract
Visualizing the movement of angiocatheters during endovascular interventions is typically accomplished using x-ray fluoroscopy. There are many potential advantages to developing magnetic resonance imaging-based approaches that will allow three-dimensional imaging of the tissue/vasculature interface while monitoring other physiologically-relevant criteria, without exposing the patient or clinician team to ionizing radiation. Here we introduce a proof-of-concept development of a magnetic resonance imaging-guided catheter tracking method that utilizes hyperpolarized silicon particles. The increased signal of the silicon particles is generated via low-temperature, solid-state dynamic nuclear polarization, and the particles retain their enhanced signal for ≥ 40 minutes--allowing imaging experiments over extended time durations. The particles are affixed to the tip of standard medical-grade catheters and are used to track passage under set distal and temporal points in phantoms and live mouse models. With continued development, this method has the potential to supplement x-ray fluoroscopy and other MRI-guided catheter tracking methods as a zero-background, positive contrast agent that does not require ionizing radiation.
Collapse
Affiliation(s)
- Nicholas Whiting
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Jingzhe Hu
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
- Department of Bioengineering, Rice University, Houston, TX 77030
| | - Jay V. Shah
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712
| | - Maja C. Cassidy
- Kavli Institute of NanoScience, Delft University of Technology, Delft, Netherlands
| | - Erik Cressman
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston TX 77030
| | - Niki Zacharias Millward
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - David G. Menter
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston TX, 77030
| | | | - Pratip K. Bhattacharya
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| |
Collapse
|
203
|
PTPN2 controls differentiation of CD4⁺ T cells and limits intestinal inflammation and intestinal dysbiosis. Mucosal Immunol 2015; 8:918-29. [PMID: 25492475 DOI: 10.1038/mi.2014.122] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 10/30/2014] [Indexed: 02/07/2023]
Abstract
Loss-of-function variants within the gene locus encoding protein tyrosine phosphatase non-receptor type 2 (PTPN2) are associated with increased risk for Crohn's disease (CD). A disturbed regulation of T helper (Th) cell responses causing loss of tolerance against self- or commensal-derived antigens and an altered intestinal microbiota plays a pivotal role in CD pathogenesis. Loss of PTPN2 in the T-cell compartment causes enhanced induction of Th1 and Th17 cells, but impaired induction of regulatory T cells (Tregs) in several mouse colitis models, namely acute and chronic dextran sodium sulfate colitis, and T-cell transfer colitis models. This results in increased susceptibility to intestinal inflammation and intestinal dysbiosis which is comparable with that observed in CD patients. We detected inflammatory infiltrates in liver, kidney, and skin and elevated autoantibody levels indicating systemic loss of tolerance in PTPN2-deficient animals. CD patients featuring a loss-of-function PTPN2 variant exhibit enhanced Th1 and Th17 cell, but reduced Treg markers when compared with PTPN2 wild-type patients in serum and intestinal tissue samples. Our data demonstrate that dysfunction of PTPN2 results in aberrant T-cell differentiation and intestinal dysbiosis similar to those observed in human CD. Our findings indicate a novel and crucial role for PTPN2 in chronic intestinal inflammation.
Collapse
|
204
|
Eschborn M, Weigmann B, Reissig S, Waisman A, Saloga J, Bellinghausen I. Activated glycoprotein A repetitions predominant (GARP)–expressing regulatory T cells inhibit allergen-induced intestinal inflammation in humanized mice. J Allergy Clin Immunol 2015; 136:159-68. [DOI: 10.1016/j.jaci.2015.04.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 03/31/2015] [Accepted: 04/03/2015] [Indexed: 12/25/2022]
|
205
|
Barrett CW, Reddy VK, Short SP, Motley AK, Lintel MK, Bradley AM, Freeman T, Vallance J, Ning W, Parang B, Poindexter SV, Fingleton B, Chen X, Washington MK, Wilson KT, Shroyer NF, Hill KE, Burk RF, Williams CS. Selenoprotein P influences colitis-induced tumorigenesis by mediating stemness and oxidative damage. J Clin Invest 2015; 125:2646-60. [PMID: 26053663 DOI: 10.1172/jci76099] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 04/30/2015] [Indexed: 12/19/2022] Open
Abstract
Patients with inflammatory bowel disease are at increased risk for colon cancer due to augmented oxidative stress. These patients also have compromised antioxidant defenses as the result of nutritional deficiencies. The micronutrient selenium is essential for selenoprotein production and is transported from the liver to target tissues via selenoprotein P (SEPP1). Target tissues also produce SEPP1, which is thought to possess an endogenous antioxidant function. Here, we have shown that mice with Sepp1 haploinsufficiency or mutations that disrupt either the selenium transport or the enzymatic domain of SEPP1 exhibit increased colitis-associated carcinogenesis as the result of increased genomic instability and promotion of a protumorigenic microenvironment. Reduced SEPP1 function markedly increased M2-polarized macrophages, indicating a role for SEPP1 in macrophage polarization and immune function. Furthermore, compared with partial loss, complete loss of SEPP1 substantially reduced tumor burden, in part due to increased apoptosis. Using intestinal organoid cultures, we found that, compared with those from WT animals, Sepp1-null cultures display increased stem cell characteristics that are coupled with increased ROS production, DNA damage, proliferation, decreased cell survival, and modulation of WNT signaling in response to H2O2-mediated oxidative stress. Together, these data demonstrate that SEPP1 influences inflammatory tumorigenesis by affecting genomic stability, the inflammatory microenvironment, and epithelial stem cell functions.
Collapse
|
206
|
Walldorf J, Hermann M, Porzner M, Pohl S, Metz H, Mäder K, Zipprich A, Christ B, Seufferlein T. In-vivo monitoring of acute DSS-Colitis using Colonoscopy, high resolution Ultrasound and bench-top Magnetic Resonance Imaging in Mice. Eur Radiol 2015; 25:2984-91. [PMID: 25981216 DOI: 10.1007/s00330-015-3714-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Revised: 01/07/2015] [Accepted: 03/11/2015] [Indexed: 12/13/2022]
Abstract
OBJECTIVE The aim of this study was to establish and evaluate (colour Doppler-) high-resolution-ultrasound (hrUS) and bench-top magnetic resonance imaging (btMRI) as new methods to monitor experimental colitis. MATERIALS AND METHODS hrUS, btMRI and endoscopy were performed in mice without colitis (n = 15), in mice with acute colitis (n = 14) and in mice with acute colitis and simultaneous treatment with infliximab (n = 19). RESULTS Determination of colon wall thickness using hrUS (32 MHz) and measurement of the cross-sectional colonic areas by btMRI allowed discrimination between the treatment groups (mean a vs. b vs. c - btMRI: 922 vs. 2051 vs. 1472 pixel, hrUS: 0.26 vs. 0.45 vs. 0.31 mm). btMRI, endoscopy, hrUS and colour Doppler-hrUS correlated to histological scoring (p < 0.05), while endoscopy and btMRI correlated to post-mortem colon length (p < 0.05). CONCLUSIONS The innovative in vivo techniques btMRI and hrUS are safe and technically feasible. They differentiate between distinct grades of colitis in an experimental setting, and correlate with established post-mortem parameters. In addition to endoscopic procedures, these techniques provide information regarding colon wall thickness and perfusion. Depending on the availability of these techniques, their application increases the value of in vivo monitoring in experimental acute colitis in small rodents. KEY POINTS • Improved in vivo monitoring might balance interindividual differences in murine colitis. • In monitoring murine colitis, btMRI and hrUS are safe and technically feasible. • Very short examination times underline the usefulness especially of hrUS. • Results of btMRI and hrUS correlate with endoscopic and post-mortem findings.
Collapse
Affiliation(s)
- J Walldorf
- Department of Internal Medicine I, Martin Luther University Halle-Wittenberg, Ernst-Grube-Strasse 40, 06120, Halle, Germany,
| | | | | | | | | | | | | | | | | |
Collapse
|
207
|
Mielke L, Preaudet A, Belz G, Putoczki T. Confocal laser endomicroscopy to monitor the colonic mucosa of mice. J Immunol Methods 2015; 421:81-88. [PMID: 25960174 PMCID: PMC5803490 DOI: 10.1016/j.jim.2015.04.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 03/27/2015] [Accepted: 04/15/2015] [Indexed: 12/13/2022]
Abstract
The gastrointestinal tract is a unique organ system that provides an epithelial barrier between our underlying immune system and luminal pathogens. Disruption of gastrointestinal homeostasis, as a result of impaired barrier function, is associated with numerous pathologies including inflammatory bowel disease and colorectal cancer. In parallel to the clinical development of endoscopy technologies to monitor and diagnose these pathologies in humans, advanced mouse colonoscopy techniques are being developed. When these technologies are coupled with model systems of human disease, which are essential to our understanding of the pathophysiology of gastrointestinal diseases, the requirement for euthanasia of multiple cohorts of mice is eliminated. Here we highlight the suitability of white light endoscopy to monitor the progression of colitis in mice. We further outline the experimental power of combined standard endoscopy with confocal microendoscopy, which permits visualization of fluorescent markers in a single animal in real-time. Together, these technologies will enhance our understanding of the interplay between components of the gastrointestinal microenvironment and their role in disease. Monitoring of mucosal damage using white light endoscopy Monitoring of the epithelial barrier using confocal endomicroscopy Monitoring of the vasculature architecture using confocal endomicroscopy
Collapse
Affiliation(s)
- Lisa Mielke
- The Walter and Eliza Hall Institute for Medical Research, Melbourne, Australia; The Department of Medical Biology, University of Melbourne, Australia
| | - Adele Preaudet
- The Walter and Eliza Hall Institute for Medical Research, Melbourne, Australia
| | - Gabrielle Belz
- The Walter and Eliza Hall Institute for Medical Research, Melbourne, Australia; The Department of Medical Biology, University of Melbourne, Australia
| | - Tracy Putoczki
- The Walter and Eliza Hall Institute for Medical Research, Melbourne, Australia; The Department of Medical Biology, University of Melbourne, Australia.
| |
Collapse
|
208
|
KAG-308, a newly-identified EP4-selective agonist shows efficacy for treating ulcerative colitis and can bring about lower risk of colorectal carcinogenesis by oral administration. Eur J Pharmacol 2015; 754:179-89. [DOI: 10.1016/j.ejphar.2015.02.021] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 02/10/2015] [Accepted: 02/11/2015] [Indexed: 12/21/2022]
|
209
|
β-Arrestin2 encourages inflammation-induced epithelial apoptosis through ER stress/PUMA in colitis. Mucosal Immunol 2015; 8:683-95. [PMID: 25354317 DOI: 10.1038/mi.2014.104] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 09/23/2014] [Indexed: 02/04/2023]
Abstract
β-Arrestins (β-arrs) are regulators and mediators of G protein-coupled receptor signaling, and accumulating evidence suggests that they are functionally involved in inflammation and autoimmune diseases. However, the effect of β-arrs is unclear in inflammatory bowel disease (IBD), and the role of β-arr2 is unknown in ulcerative colitis (UC) and Crohn's disease (CD). The aim of this study is to investigate whether β-arr2 encourages inflammation-induced epithelial apoptosis through endoplasmic reticulum (ER) stress/p53-upregulated modulator of apoptosis (PUMA) in colitis. In the present study, the results showed that β-arr2 was increased in specimens from patients with UC or CD. Furthermore, a β-arr2 deficiency significantly repressed intestinal inflammation, ameliorated colitis, and alleviated mucosal apoptosis in mice. In addition, the targeted deletion of β-arr2 depressed ER stress, inhibited PUMA, and downregulated PUMA-mediated mitochondrial apoptotic signaling in colitis. β-Arr2, an important modulator of G protein-coupled receptor function, binds eIF2α to activate ER stress signaling. Furthermore, the knockdown of PUMA dramatically prevented β-arr2-induced apoptosis via alleviating ER stress in vitro. The results suggest that β-arr2 encourages inflammation-induced epithelial apoptosis through ER stress/PUMA in colitis and that β-arr2 is a potential therapeutic target for colitis.
Collapse
|
210
|
Vong LB, Yoshitomi T, Matsui H, Nagasaki Y. Development of an oral nanotherapeutics using redox nanoparticles for treatment of colitis-associated colon cancer. Biomaterials 2015; 55:54-63. [PMID: 25934452 DOI: 10.1016/j.biomaterials.2015.03.037] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 03/19/2015] [Accepted: 03/23/2015] [Indexed: 12/12/2022]
Abstract
Oral chemotherapy is the preferred treatment for colon cancer. However, this strategy faces many challenges, including instability in the gastrointestinal (GI) tract, insufficient bioavailability, low tumor targeting, and severe adverse effects. In this study, we designed a novel redox nanoparticle (RNP(O)) that is an ideal oral therapeutics for colitis-associated colon cancer treatment. RNP(O) possesses nitroxide radicals in the core, which act as reactive oxygen species (ROS) scavengers. Orally administered RNP(O) highly accumulated in colonic mucosa, and specifically internalized in cancer tissues, but less in normal tissues. Despite of long-term oral administration of RNP(O), no noticeable toxicities were observed in major organs of mice. Because RNP(O) effectively scavenged ROS, it significantly suppressed tumor growth after accumulation at tumor sites. Combination of RNP(O) with the conventional chemotherapy, irinotecan, led to remarkably improved therapeutic efficacy and effectively suppressed its adverse effects on GI tract. Therefore, RNP(O) is promising oral nanotherapeutics for cancer therapies.
Collapse
Affiliation(s)
- Long Binh Vong
- Department of Materials Science, Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8573, Japan
| | - Toru Yoshitomi
- Department of Materials Science, Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8573, Japan
| | - Hirofumi Matsui
- Master's School of Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8575, Japan; Division of Gastroenterology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8575, Japan
| | - Yukio Nagasaki
- Department of Materials Science, Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8573, Japan; Master's School of Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8575, Japan; Satellite Laboratory, International Center for Materials Nanoarchitectonics (WPI-MANA), National Institute for Materials Science (NIMS), University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8573, Japan.
| |
Collapse
|
211
|
Choi JW, Kim P, Kim JK, Kim YR, Fukumura D, Yun SH. Longitudinal Tracing of Spontaneous Regression and Anti-angiogenic Response of Individual Microadenomas during Colon Tumorigenesis. Am J Cancer Res 2015; 5:724-32. [PMID: 25897337 PMCID: PMC4402496 DOI: 10.7150/thno.10734] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 02/11/2015] [Indexed: 12/19/2022] Open
Abstract
Angiogenesis is essential for the progression of cancer, but its involvement in the initial phase of colon tumorigenesis is not well understood. Using intravital endomicroscopy, we visualized the natural history of early pre-tumorous lesions and adenomas in the colon of conditional Apc-knockout and Apc/Kras double mutant mouse models. Early lesions emerged about 4 weeks after the onset of somatic mutations, accompanying vascular dilation when the size of lesions reached about 200 μm, but most lesions regressed spontaneously and cleared within 10 weeks after their emergence. Anti-angiogenic treatments with vascular endothelial growth factor receptor (VEGFR) antagonists reduced the size of the early lesions and the number of polyps. We found surprisingly that anti-angiogenic treatments delayed the natural clearance of transient lesions by up to several weeks in both genetic models. The results represent the previously unexpected role of early angiogenesis on the spontaneous regression of early-stage colon tumors.
Collapse
|
212
|
Helicobacter pylori-specific protection against inflammatory bowel disease requires the NLRP3 inflammasome and IL-18. Inflamm Bowel Dis 2015; 21:854-61. [PMID: 25742401 DOI: 10.1097/mib.0000000000000318] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND The Gram-negative bacterium Helicobacter pylori is a constituent of the human gastric microbiota. Chronic infection with H. pylori causes gastritis and predisposes to gastric carcinoma but has also been inversely linked to various allergic and chronic inflammatory conditions. In particular, large meta-analyses have documented an inverse association between H. pylori infection and the risk of developing ulcerative colitis and Crohn's disease. METHODS We investigated possible protective effects of experimental H. pylori infection and of regular treatment with H. pylori extract in 2 mouse models of colitis and in mouse models of type I diabetes and multiple sclerosis. The mechanism of protection was examined in mouse strains lacking specific innate immune recognition pathways and cytokines. RESULTS We show here that experimental infection with H. pylori and administration of regular doses of H. pylori extract both alleviate the clinical and histopathological features of dextran sodium sulfate-induced chronic colitis and of T-cell transfer-induced colitis. High resolution endoscopy of the protected animals revealed the accumulation of large amounts of colonic mucus upon H. pylori exposure, which could be attributed to transcriptional activation of the mucin 2 gene. The protection against dextran sodium sulfate-induced colitis was dependent on the NLRP3 inflammasome and interleukin-18 signaling. Other autoimmune diseases, i.e., experimental autoimmune encephalomyelitis and type I diabetes, were not controlled by H. pylori. CONCLUSIONS In summary, we propose here that the immunomodulatory activity of an ancient constituent of the gut microbiota, H. pylori, may be exploited for the prevention and/or treatment of inflammatory bowel diseases.
Collapse
|
213
|
Ernst M, Preaudet A, Putoczki T. Non-invasive assessment of the efficacy of new therapeutics for intestinal pathologies using serial endoscopic imaging of live mice. J Vis Exp 2015:52383. [PMID: 25867916 PMCID: PMC4401233 DOI: 10.3791/52383] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Animal models of inflammatory bowel disease (IBD) and colorectal cancer (CRC) have provided significant insight into the cell intrinsic and extrinsic mechanisms that contribute to the onset and progression of intestinal diseases. The identification of new molecules that promote these pathologies has led to a flurry of activity focused on the development of potential new therapies to inhibit their function. As a result, various pre-clinical mouse models with an intact immune system and stromal microenvironment are now heavily used. Here we describe three experimental protocols to test the efficacy of new therapeutics in pre-clinical models of (1) acute mucosal damage, (2) chronic colitis and/or colitis-associated colon cancer, and (3) sporadic colorectal cancer. We also outline procedures for serial endoscopic examination that can be used to document the therapeutic response of an individual tumor and to monitor the health of individual mice. These protocols provide complementary experimental platforms to test the effectiveness of therapeutic compounds shown to be well tolerated by mice.
Collapse
Affiliation(s)
- Matthias Ernst
- The Walter and Eliza Hall Institute for Medical Research; The Department of Medical Biology, University of Melbourne; Olivia Newton-John Cancer Research Institute
| | - Adele Preaudet
- The Walter and Eliza Hall Institute for Medical Research
| | - Tracy Putoczki
- The Walter and Eliza Hall Institute for Medical Research; The Department of Medical Biology, University of Melbourne;
| |
Collapse
|
214
|
Leoni G, Neumann PA, Kamaly N, Quiros M, Nishio H, Jones HR, Sumagin R, Hilgarth RS, Alam A, Fredman G, Argyris I, Rijcken E, Kusters D, Reutelingsperger C, Perretti M, Parkos CA, Farokhzad OC, Neish AS, Nusrat A. Annexin A1-containing extracellular vesicles and polymeric nanoparticles promote epithelial wound repair. J Clin Invest 2015; 125:1215-27. [PMID: 25664854 DOI: 10.1172/jci76693] [Citation(s) in RCA: 262] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Accepted: 01/02/2015] [Indexed: 12/13/2022] Open
Abstract
Epithelial restitution is an essential process that is required to repair barrier function at mucosal surfaces following injury. Prolonged breaches in epithelial barrier function result in inflammation and further damage; therefore, a better understanding of the epithelial restitution process has potential for improving the development of therapeutics. In this work, we demonstrate that endogenous annexin A1 (ANXA1) is released as a component of extracellular vesicles (EVs) derived from intestinal epithelial cells, and these ANXA1-containing EVs activate wound repair circuits. Compared with healthy controls, patients with active inflammatory bowel disease had elevated levels of secreted ANXA1-containing EVs in sera, indicating that ANXA1-containing EVs are systemically distributed in response to the inflammatory process and could potentially serve as a biomarker of intestinal mucosal inflammation. Local intestinal delivery of an exogenous ANXA1 mimetic peptide (Ac2-26) encapsulated within targeted polymeric nanoparticles (Ac2-26 Col IV NPs) accelerated healing of murine colonic wounds after biopsy-induced injury. Moreover, one-time systemic administration of Ac2-26 Col IV NPs accelerated recovery following experimentally induced colitis. Together, our results suggest that local delivery of proresolving peptides encapsulated within nanoparticles may represent a potential therapeutic strategy for clinical situations characterized by chronic mucosal injury, such as is seen in patients with IBD.
Collapse
|
215
|
Meinhardt K, Kroeger I, Bauer R, Ganss F, Ovsiy I, Rothamer J, Büttner M, Atreya I, Waldner M, Bittrich M, Lehmann CH, Rieger MA, Beilhack A, Zeiser R, Edinger M, Dudziak D, Mackensen A, Rehli M, Ullrich E. Identification and characterization of the specific murine NK cell subset supporting graft- versus-leukemia- and reducing graft- versus-host-effects. Oncoimmunology 2015; 4:e981483. [PMID: 25949862 PMCID: PMC4368119 DOI: 10.4161/2162402x.2014.981483] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2014] [Accepted: 10/23/2014] [Indexed: 11/19/2022] Open
Abstract
Clinical studies investigating the impact of natural killer (NK) cells in allogeneic hematopoietic stem cell transplantation settings have yielded promising results. However, NK cells are a functionally and phenotypically heterogeneous population. Therefore, we addressed the functional relevance of specific NK cell subsets distinguished by expression of CD117, CD27 and CD11b surface markers in graft-versus-leukemia (GVL)-reaction and graft-versus-host-disease (GVHD). Our results clearly demonstrate that the subset of c-Kit−CD27−CD11b+ NK cells expressed multiple cytotoxic pathway genes and provided optimal graft-versus-leukemia-effects, while significantly reducing T cell proliferation induced by allogeneic dendritic cells. Furthermore, these NK cells migrated to inflamed intestinal tissues where graft-versus-host-colitis was efficiently mitigated. For the first time, we identified the c-Kit−CD27−CD11b+ NK cell population as the specific effector NK cell subset capable of significantly diminishing GVHD in fully mismatched bone marrow transplantation settings. In conclusion, the subset of c-Kit−CD27−CD11b+ NK cells not only supports GVL, but also plays a unique role in the protection against GVHD by migrating to the peripheral GVHD target organs where they exert efficient immunoregulatory activities. These new insights demonstrate the importance of selecting the optimal NK cell subset for cellular immunotherapy following allogeneic hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Kathrin Meinhardt
- Department of Internal Medicine 5; Hematology and Oncology; Friedrich-Alexander Universität Erlangen-Nürnberg; University Hospital Erlangen ; Erlangen, Germany ; Childrens Hospital; Department of Pediatric Hematology and Oncology; Johann Wolfgang Goethe University ; Frankfurt, Germany ; LOEWE Center for Cell and Gene Therapy; Johann Wolfgang Goethe University ; Frankfurt, Germany
| | - Irena Kroeger
- Department of Internal Medicine 5; Hematology and Oncology; Friedrich-Alexander Universität Erlangen-Nürnberg; University Hospital Erlangen ; Erlangen, Germany
| | - Ruth Bauer
- Department of Internal Medicine 5; Hematology and Oncology; Friedrich-Alexander Universität Erlangen-Nürnberg; University Hospital Erlangen ; Erlangen, Germany ; Childrens Hospital; Department of Pediatric Hematology and Oncology; Johann Wolfgang Goethe University ; Frankfurt, Germany ; LOEWE Center for Cell and Gene Therapy; Johann Wolfgang Goethe University ; Frankfurt, Germany
| | - Franziska Ganss
- Department of Internal Medicine 5; Hematology and Oncology; Friedrich-Alexander Universität Erlangen-Nürnberg; University Hospital Erlangen ; Erlangen, Germany ; Childrens Hospital; Department of Pediatric Hematology and Oncology; Johann Wolfgang Goethe University ; Frankfurt, Germany ; LOEWE Center for Cell and Gene Therapy; Johann Wolfgang Goethe University ; Frankfurt, Germany
| | - Ilja Ovsiy
- Childrens Hospital; Department of Pediatric Hematology and Oncology; Johann Wolfgang Goethe University ; Frankfurt, Germany ; LOEWE Center for Cell and Gene Therapy; Johann Wolfgang Goethe University ; Frankfurt, Germany
| | - Johanna Rothamer
- Department of Internal Medicine 5; Hematology and Oncology; Friedrich-Alexander Universität Erlangen-Nürnberg; University Hospital Erlangen ; Erlangen, Germany ; Childrens Hospital; Department of Pediatric Hematology and Oncology; Johann Wolfgang Goethe University ; Frankfurt, Germany ; LOEWE Center for Cell and Gene Therapy; Johann Wolfgang Goethe University ; Frankfurt, Germany
| | - Maike Büttner
- Department of Nephropathology; Friedrich-Alexander Universität Erlangen-Nürnberg; University Hospital Erlangen ; Erlangen, Germany
| | - Imke Atreya
- Department of Internal Medicine 1; Friedrich-Alexander Universität Erlangen-Nürnberg; University Hospital Erlangen ; Erlangen, Germany
| | - Maximilian Waldner
- Department of Internal Medicine 1; Friedrich-Alexander Universität Erlangen-Nürnberg; University Hospital Erlangen ; Erlangen, Germany
| | - Max Bittrich
- Department of Internal Medicine 5; Hematology and Oncology; Friedrich-Alexander Universität Erlangen-Nürnberg; University Hospital Erlangen ; Erlangen, Germany
| | - Christian Hk Lehmann
- Department of Dermatology; Laboratory of Dendritic Cell Biology; Friedrich-Alexander Universität Erlangen-Nürnberg; University Hospital Erlangen ; Erlangen, Germany
| | - Michael A Rieger
- LOEWE Center for Cell and Gene Therapy; Johann Wolfgang Goethe University ; Frankfurt, Germany ; Department of Hematology/Oncology; Johann Wolfgang Goethe University ; Frankfurt, Germany
| | - Andreas Beilhack
- IZKF Research Group for Experimental Stem Cell Transplantation; Department of Medicine II; University Hospital ; Würzburg, Germany
| | - Robert Zeiser
- Division of Hematology and Oncology; Department of Medicine; Freiburg University Medical Center; Albert-Ludwigs-University ; Freiburg, Germany
| | - Matthias Edinger
- Department of Internal Medicine 3; University Hospital Regensburg ; Regensburg; Germany ; Regensburg Center for Interventional Immunology (RCI); University Regensburg ; Regensburg, Germany
| | - Diana Dudziak
- Department of Dermatology; Laboratory of Dendritic Cell Biology; Friedrich-Alexander Universität Erlangen-Nürnberg; University Hospital Erlangen ; Erlangen, Germany
| | - Andreas Mackensen
- Department of Internal Medicine 5; Hematology and Oncology; Friedrich-Alexander Universität Erlangen-Nürnberg; University Hospital Erlangen ; Erlangen, Germany
| | - Michael Rehli
- Department of Internal Medicine 3; University Hospital Regensburg ; Regensburg; Germany ; Regensburg Center for Interventional Immunology (RCI); University Regensburg ; Regensburg, Germany
| | - Evelyn Ullrich
- Department of Internal Medicine 5; Hematology and Oncology; Friedrich-Alexander Universität Erlangen-Nürnberg; University Hospital Erlangen ; Erlangen, Germany ; Childrens Hospital; Department of Pediatric Hematology and Oncology; Johann Wolfgang Goethe University ; Frankfurt, Germany ; LOEWE Center for Cell and Gene Therapy; Johann Wolfgang Goethe University ; Frankfurt, Germany
| |
Collapse
|
216
|
Kreiser S, Eckhardt J, Kuhnt C, Stein M, Krzyzak L, Seitz C, Tucher C, Knippertz I, Becker C, Günther C, Steinkasserer A, Lechmann M. Murine CD83-positive T cells mediate suppressor functions in vitro and in vivo. Immunobiology 2015; 220:270-9. [PMID: 25151500 DOI: 10.1016/j.imbio.2014.08.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 07/22/2014] [Accepted: 08/04/2014] [Indexed: 10/24/2022]
Abstract
The CD83 molecule (CD83) is a well-known surface marker present on mature dendritic cells (mDC). In this study, we show that CD83 is also expressed on a subset of T cells which mediate regulatory T cell (Treg)-like suppressor functions in vitro and in vivo. Treg-associated molecules including CD25, cytotoxic T lymphocyte antigen-4 (CTLA-4), glucocorticoid-induced TNFR family-related gene (GITR), Helios and neuropilin-1 (NRP-1) as well as forkhead box protein 3 (FOXP3) were specifically expressed by these CD83(+) T cells. In contrast, CD83(-) T cells showed a naive T cell phenotype with effector T cell properties upon activation. Noteworthy, CD83(-) T cells were not able to upregulate CD83 despite activation. Furthermore, CD83(+) T cells suppressed the proliferation and inflammatory cytokine release of CD83(-) T cells in vitro. Strikingly, stimulated CD83(+) T cells released soluble CD83 (sCD83), which has been reported to possess immunosuppressive properties. In vivo, using the murine transfer colitis model we could show that CD83(+) T cells were able to suppress colitis symptoms while CD83(-) T cells possessed effector functions. In addition, this CD83 expression is also conserved on expanded human Treg. Thus, from these studies we conclude that CD83(+) T cells share important features with regulatory T cells, identifying CD83 as a novel lineage marker to discriminate between different T cell populations.
Collapse
Affiliation(s)
- Simon Kreiser
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen D-91052, Germany
| | - Jenny Eckhardt
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen D-91052, Germany
| | - Christine Kuhnt
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen D-91052, Germany
| | - Marcello Stein
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen D-91052, Germany
| | - Lena Krzyzak
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen D-91052, Germany
| | - Christine Seitz
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen D-91052, Germany
| | - Christine Tucher
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen D-91052, Germany
| | - Ilka Knippertz
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen D-91052, Germany
| | - Christoph Becker
- Department of Medicine 1, University Hospital Erlangen, Erlangen D-91052, Germany
| | - Claudia Günther
- Department of Medicine 1, University Hospital Erlangen, Erlangen D-91052, Germany
| | - Alexander Steinkasserer
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen D-91052, Germany
| | - Matthias Lechmann
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen D-91052, Germany; Department of Medicine 1, University Hospital Erlangen, Erlangen D-91052, Germany.
| |
Collapse
|
217
|
Near-Infrared Confocal Laser Endomicroscopy Detects Colorectal Cancer via an Integrin αvβ3 Optical Probe. Mol Imaging Biol 2015; 17:450-60. [DOI: 10.1007/s11307-015-0825-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
218
|
Keenan MR, Leung SJ, Rice PS, Wall RA, Barton JK. Dual optical modality endoscopic imaging of cancer development in the mouse colon. Lasers Surg Med 2015; 47:30-9. [PMID: 25449147 PMCID: PMC4304897 DOI: 10.1002/lsm.22307] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/16/2014] [Indexed: 01/07/2023]
Abstract
BACKGROUND AND OBJECTIVE We utilize a miniature, dual-modality endoscope that combines fluorescence-based surface magnifying chromoendoscopy (SMC) and optical coherence tomography (OCT) to follow the anatomical changes that occur during adenoma development in the mouse colon. MATERIALS AND METHODS Twenty-five mice were treated with the carcinogen azoxymethane (AOM) to induce tumor development in the distal colon, or were treated with saline as control, and were imaged over six months. OCT detects adenoma number with high sensitivity and specificity and can measure lesion size. In methylene blue-lavaged colons, SMC detects changes in the colonic crypts. SMC images of control mouse colons exhibit reticulated patterns of crypts of equal size, forming either a dot or honeycomb pattern. RESULTS Images of AOM-treated colons show mild crypt irregularities even in grossly normal tissue. Images of small to medium adenoma exhibit larger crypts, more intense signal, and irregular spacing whereas those of large adenoma have heterogeneous, intense signal and loss of crypt structure. CONCLUSIONS The combination of OCT and SMC permits the detection of neoplastic events from the earliest stages of crypt irregularities before gross tissue changes are noted, through to measuring the growth of protruding adenoma.
Collapse
Affiliation(s)
- Molly R Keenan
- The University of Arizona, Department of Biomedical Engineering, 1657 E. Helen Street, 210240, Tucson, Arizona, 85721
| | | | | | | | | |
Collapse
|
219
|
Gerster R, Eloranta JJ, Hausmann M, Ruiz PA, Cosin-Roger J, Terhalle A, Ziegler U, Kullak-Ublick GA, von Eckardstein A, Rogler G. Anti-inflammatory Function of High-Density Lipoproteins via Autophagy of IκB Kinase. Cell Mol Gastroenterol Hepatol 2014; 1:171-187.e1. [PMID: 28247863 PMCID: PMC5301135 DOI: 10.1016/j.jcmgh.2014.12.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 12/12/2014] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Plasma levels of high-density lipoprotein (HDL) cholesterol are frequently found decreased in patients with inflammatory bowel disease (IBD). Therefore, and because HDL exerts anti-inflammatory activities, we investigated whether HDL and its major protein component apolipoprotein A-I (apoA-I) modulate mucosal inflammatory responses in vitro and in vivo. METHODS The human intestinal epithelial cell line T84 was used as the in vitro model for measuring the effects of HDL on the expression and secretion of tumor necrosis factor (TNF), interleukin-8 (IL-8), and intracellular adhesion molecule (ICAM). Nuclear factor-κB (NF-κB)-responsive promoter activity was studied by dual luciferase reporter assays. Mucosal damage from colitis induced by dextran sodium sulphate (DSS) and 2,4,6-trinitrobenzenesulfonic acid (TNBS) was scored by colonoscopy and histology in apoA-I transgenic (Tg) and apoA-I knockout (KO) and wild-type (WT) mice. Myeloperoxidase (MPO) activity and TNF and ICAM expression were determined in intestinal tissue samples. Autophagy was studied by Western blot analysis, immunofluorescence, and electron microscopy. RESULTS HDL and apoA-I down-regulated TNF-induced mRNA expression of TNF, IL-8, and ICAM, as well as TNF-induced NF-κB-responsive promoter activity. DSS/TNBS-treated apoA-I KO mice displayed increased mucosal damage upon both colonoscopy and histology, increased intestinal MPO activity and mRNA expression of TNF and ICAM as compared with WT and apoA-I Tg mice. In contrast, apoA-I Tg mice showed less severe symptoms monitored by colonoscopy and MPO activity in both the DSS and TNBS colitis models. In addition, HDL induced autophagy, leading to recruitment of phosphorylated IκB kinase to the autophagosome compartment, thereby preventing NF-κB activation and induction of cytokine expression. CONCLUSIONS Taken together, the in vitro and in vivo findings suggest that HDL and apoA-I suppress intestinal inflammation via autophagy and are potential therapeutic targets for the treatment of IBD.
Collapse
Key Words
- 3-MA, 3-methyl adenine
- ApoA-I, apolipoprotein A-I
- Apolipoprotein A-I
- Autophagy
- CD, Crohn’s disease
- DAPI, 4′,6-diamidino-2-phenylindole
- DSS, dextran sodium sulphate
- EMSA, electrophoretic mobility shift assay
- HDL, high-density lipoprotein
- IBD, inflammatory bowel disease
- ICAM, intracellular adhesion molecule
- IL, interleukin
- Inflammatory Bowel Disease
- KO, knockout
- LC3II, light chain 3 II
- MEICS, murine endoscopic index of colitis severity
- MPO, myeloperoxidase
- NF-κB
- NF-κB, nuclear factor κB
- PBS, phosphate-buffered saline
- PFA, paraformaldehyde
- PI-3, phosphatidylinositol-3
- RT-PCR, real-time polymerase chain reaction
- TNBS, 2,4,6-trinitrobenzenesulfonic acid
- TNF, tumor necrosis factor
- Tg, transgenic
- WT, wild type
- mTOR, the mammalian target of rapamycin
- p-IKK, phosphorylated IκB kinase
- siRNA, small interfering RNA
Collapse
Affiliation(s)
- Ragam Gerster
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, Schlieren, Switzerland
- Zurich Center of Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Jyrki J. Eloranta
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, Schlieren, Switzerland
| | - Martin Hausmann
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Pedro A. Ruiz
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Jesus Cosin-Roger
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
- Departamento de Farmacología and CIBERehd, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | - Anne Terhalle
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Urs Ziegler
- Centre for Microscopy and Image Analysis, University Hospital Zurich, Zurich, Switzerland
| | - Gerd A. Kullak-Ublick
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, Schlieren, Switzerland
- Zurich Center of Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Arnold von Eckardstein
- Zurich Center of Integrative Human Physiology, University of Zurich, Zurich, Switzerland
- Institute of Clinical Chemistry, University Hospital Zurich, Zurich, Switzerland
| | - Gerhard Rogler
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
- Zurich Center of Integrative Human Physiology, University of Zurich, Zurich, Switzerland
- Correspondence Address correspondence to: Gerhard Rogler, MD, PhD, Division of Gastroenterology and Hepatology, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland. fax: +41-0-44-255-9497.
| |
Collapse
|
220
|
Charanya T, York T, Bloch S, Sudlow G, Liang K, Garcia M, Akers WJ, Rubin D, Gruev V, Achilefu S. Trimodal color-fluorescence-polarization endoscopy aided by a tumor selective molecular probe accurately detects flat lesions in colitis-associated cancer. JOURNAL OF BIOMEDICAL OPTICS 2014; 19:126002. [PMID: 25473883 PMCID: PMC4255434 DOI: 10.1117/1.jbo.19.12.126002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 10/24/2014] [Indexed: 05/10/2023]
Abstract
Colitis-associated cancer (CAC) arises from premalignant flat lesions of the colon, which are difficult to detect with current endoscopic screening approaches. We have developed a complementary fluorescence and polarization reporting strategy that combines the unique biochemical and physical properties of dysplasia and cancer for real-time detection of these lesions. Using azoxymethane-dextran sodium sulfate (AOM-DSS) treated mice, which recapitulates human CAC and dysplasia, we show that an octapeptide labeled with a near-infrared (NIR) fluorescent dye selectively identified all precancerous and cancerous lesions. A new thermoresponsive sol-gel formulation allowed topical application of the molecular probe during endoscopy. This method yielded high contrast-to-noise ratios (CNR) between adenomatous tumors (20.6 ± 1.65) and flat lesions (12.1 ± 1.03) and surrounding uninvolved colon tissue versus CNR of inflamed tissues (1.62±0.42) Incorporation of nanowire-filtered polarization imaging into NIR fluorescence endoscopy shows a high depolarization contrast in both adenomatous tumors and flat lesions in CAC, reflecting compromised structural integrity of these tissues. Together, the real-time polarization imaging provides real-time validation of suspicious colon tissue highlighted by molecular fluorescence endoscopy.
Collapse
Affiliation(s)
- Tauseef Charanya
- Washington University in St. Louis, Department of Radiology, 4525 Scott Avenue, East Building, St. Louis, Missouri 63110, United States
- Washington University in St. Louis, Department of Biomedical Engineering, 1 Brookings Drive, St. Louis, Missouri 63110, United States
| | - Timothy York
- Washington University in St. Louis, Department of Computer Science and Engineering, 1 Brookings Drive, St. Louis, Missouri 63110, United States
| | - Sharon Bloch
- Washington University in St. Louis, Department of Radiology, 4525 Scott Avenue, East Building, St. Louis, Missouri 63110, United States
| | - Gail Sudlow
- Washington University in St. Louis, Department of Radiology, 4525 Scott Avenue, East Building, St. Louis, Missouri 63110, United States
| | - Kexian Liang
- Washington University in St. Louis, Department of Radiology, 4525 Scott Avenue, East Building, St. Louis, Missouri 63110, United States
| | - Missael Garcia
- Washington University in St. Louis, Department of Computer Science and Engineering, 1 Brookings Drive, St. Louis, Missouri 63110, United States
| | - Walter J. Akers
- Washington University in St. Louis, Department of Radiology, 4525 Scott Avenue, East Building, St. Louis, Missouri 63110, United States
| | - Deborah Rubin
- Washington University in St. Louis, Department of Medicine, 660 South Euclid Avenue, St. Louis, Missouri 63110, United States
| | - Viktor Gruev
- Washington University in St. Louis, Department of Computer Science and Engineering, 1 Brookings Drive, St. Louis, Missouri 63110, United States
| | - Samuel Achilefu
- Washington University in St. Louis, Department of Radiology, 4525 Scott Avenue, East Building, St. Louis, Missouri 63110, United States
- Washington University in St. Louis, Department of Biomedical Engineering, 1 Brookings Drive, St. Louis, Missouri 63110, United States
- Washington University in St. Louis, Department of Biochemistry and Molecular Biophysics, 660 South Euclid Avenue, St. Louis, Missouri 63110, United States
- Address all correspondence to: Samuel Achilefu, E-mail:
| |
Collapse
|
221
|
Ileva LV, Bernardo M, Young MR, Riffle LA, Tatum JL, Kalen JD, Choyke PL. In vivo MRI virtual colonography in a mouse model of colon cancer. Nat Protoc 2014; 9:2682-92. [PMID: 25340441 PMCID: PMC8381262 DOI: 10.1038/nprot.2014.178] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
We have developed a reliable noninvasive method for monitoring colonic tumors and mucosal inflammation in a mouse model of colon cancer using magnetic resonance colonography (MRC). After a mild cleansing enema, the colon is filled with Fluorinert, a perfluorinated liquid that does not produce a proton MR signal. The mouse is placed in a dedicated volume MR receiver coil, and high-resolution images are acquired in three planes. The Fluorinert enema distends the mouse colon, creating an artifact-free black homogeneous background, allowing clear delineation of the inflamed colonic wall and visualization of luminal tumors in various stages of development. A gadolinium-based contrast agent can be administered i.v. to the animal to detect mural inflammation or tumor vascularity. This technique is useful for serial monitoring of the effects of preventive or therapeutic strategies on tumor development without killing the animal or requiring invasive endoscopies. The animal preparation and imaging can be completed in ∼1.5 h.
Collapse
Affiliation(s)
- Lilia V Ileva
- Small Animal Imaging Program, Laboratory Animal Sciences Program, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Marcelino Bernardo
- Molecular Imaging Program, Laboratory Animal Sciences Program, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Matthew R Young
- Laboratory of Cancer Prevention, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland, USA
| | - Lisa A Riffle
- Small Animal Imaging Program, Laboratory Animal Sciences Program, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - James L Tatum
- Cancer Imaging Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Joseph D Kalen
- Small Animal Imaging Program, Laboratory Animal Sciences Program, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Peter L Choyke
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
222
|
Saud SM, Li W, Morris NL, Matter MS, Colburn NH, Kim YS, Young MR. Resveratrol prevents tumorigenesis in mouse model of Kras activated sporadic colorectal cancer by suppressing oncogenic Kras expression. Carcinogenesis 2014; 35:2778-86. [PMID: 25280562 DOI: 10.1093/carcin/bgu209] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Sporadic and non-hereditary mutations account for the majority of colorectal cancers (CRC). After the loss of adenomatous polyposis coli (APC) function and activation of the β-catenin/LEF signaling pathway, activating mutations in Kras are major drivers of sporadic CRC. Preventing the outgrowth of cells that develop sporadic mutations will decrease CRC. Resveratrol, a naturally occurring polyphenolic compound has anti-inflammatory, anti-oxidant and anti-cancer activities. We used a genetically engineered mouse model for sporadic CRC where the APC locus is knocked out and Kras is activated specifically in the distal colon to determine the effects of resveratrol on preventing and treating CRC. Feeding mice a diet supplemented with 150 or 300 ppm resveratrol (105 and 210mg daily human equivalent dose, respectively) before tumors were visible by colonoscopy resulted in a 60% inhibition of tumor production. In the 40% of mice that did develop tumors Kras expression was lost in the tumors. In a therapeutic assay where tumors were allowed to develop prior to treatment, feeding tumor bearing mice with resveratrol resulted in a complete remission in 33% of the mice and a 97% decrease in tumor size in the remaining mice. Analysis of miRNA expression in non-tumoral and tumoral colonic tissue of resveratrol treated mice showed an increased expression of miR-96, a miRNA previously shown to regulate Kras translation. These data indicate that resveratrol can prevent the formation and growth of colorectal tumors by downregulating Kras expression.
Collapse
Affiliation(s)
- Shakir M Saud
- Nutritional Science Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, MD, USA, Laboratory of Cancer Prevention, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Weidong Li
- Laboratory of Cancer Prevention, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA, Oncology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Nicole L Morris
- Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick, MD, USA and
| | - Matthias S Matter
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Nancy H Colburn
- Laboratory of Cancer Prevention, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Young S Kim
- Nutritional Science Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, MD, USA
| | - Matthew R Young
- Laboratory of Cancer Prevention, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA,
| |
Collapse
|
223
|
Takahashi N, Vereecke L, Bertrand MJM, Duprez L, Berger SB, Divert T, Gonçalves A, Sze M, Gilbert B, Kourula S, Goossens V, Lefebvre S, Günther C, Becker C, Bertin J, Gough PJ, Declercq W, van Loo G, Vandenabeele P. RIPK1 ensures intestinal homeostasis by protecting the epithelium against apoptosis. Nature 2014; 513:95-9. [PMID: 25186904 DOI: 10.1038/nature13706] [Citation(s) in RCA: 363] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Accepted: 07/23/2014] [Indexed: 11/09/2022]
Abstract
Receptor interacting protein kinase 1 (RIPK1) has an essential role in the signalling triggered by death receptors and pattern recognition receptors. RIPK1 is believed to function as a node driving NF-κB-mediated cell survival and inflammation as well as caspase-8 (CASP8)-dependent apoptotic or RIPK3/MLKL-dependent necroptotic cell death. The physiological relevance of this dual function has remained elusive because of the perinatal death of RIPK1 full knockout mice. To circumvent this problem, we generated RIPK1 conditional knockout mice, and show that mice lacking RIPK1 in intestinal epithelial cells (IECs) spontaneously develop severe intestinal inflammation associated with IEC apoptosis leading to early death. This early lethality was rescued by antibiotic treatment, MYD88 deficiency or tumour-necrosis factor (TNF) receptor 1 deficiency, demonstrating the importance of commensal bacteria and TNF in the IEC Ripk1 knockout phenotype. CASP8 deficiency, but not RIPK3 deficiency, rescued the inflammatory phenotype completely, indicating the indispensable role of RIPK1 in suppressing CASP8-dependent apoptosis but not RIPK3-dependent necroptosis in the intestine. RIPK1 kinase-dead knock-in mice did not exhibit any sign of inflammation, suggesting that RIPK1-mediated protection resides in its kinase-independent platform function. Depletion of RIPK1 in intestinal organoid cultures sensitized them to TNF-induced apoptosis, confirming the in vivo observations. Unexpectedly, TNF-mediated NF-κB activation remained intact in these organoids. Our results demonstrate that RIPK1 is essential for survival of IECs, ensuring epithelial homeostasis by protecting the epithelium from CASP8-mediated IEC apoptosis independently of its kinase activity and NF-κB activation.
Collapse
Affiliation(s)
- Nozomi Takahashi
- 1] VIB Inflammation Research Center, Technologiepark 927, B-9052 Ghent, Belgium [2] Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, B-9052 Ghent, Belgium
| | - Lars Vereecke
- 1] VIB Inflammation Research Center, Technologiepark 927, B-9052 Ghent, Belgium [2] Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, B-9052 Ghent, Belgium
| | - Mathieu J M Bertrand
- 1] VIB Inflammation Research Center, Technologiepark 927, B-9052 Ghent, Belgium [2] Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, B-9052 Ghent, Belgium
| | - Linde Duprez
- 1] VIB Inflammation Research Center, Technologiepark 927, B-9052 Ghent, Belgium [2] Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, B-9052 Ghent, Belgium
| | - Scott B Berger
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, Pennsylvania 19426, USA
| | - Tatyana Divert
- 1] VIB Inflammation Research Center, Technologiepark 927, B-9052 Ghent, Belgium [2] Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, B-9052 Ghent, Belgium
| | - Amanda Gonçalves
- 1] VIB Inflammation Research Center, Technologiepark 927, B-9052 Ghent, Belgium [2] Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, B-9052 Ghent, Belgium [3] VIB Bio Imaging Core Gent, Technologiepark 927, B-9052 Ghent, Belgium
| | - Mozes Sze
- 1] VIB Inflammation Research Center, Technologiepark 927, B-9052 Ghent, Belgium [2] Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, B-9052 Ghent, Belgium
| | - Barbara Gilbert
- 1] VIB Inflammation Research Center, Technologiepark 927, B-9052 Ghent, Belgium [2] Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, B-9052 Ghent, Belgium
| | - Stephanie Kourula
- 1] VIB Inflammation Research Center, Technologiepark 927, B-9052 Ghent, Belgium [2] Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, B-9052 Ghent, Belgium
| | - Vera Goossens
- 1] VIB Inflammation Research Center, Technologiepark 927, B-9052 Ghent, Belgium [2] Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, B-9052 Ghent, Belgium
| | - Sylvie Lefebvre
- 1] VIB Inflammation Research Center, Technologiepark 927, B-9052 Ghent, Belgium [2] Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, B-9052 Ghent, Belgium
| | - Claudia Günther
- Department of Medicine 1, Friedrich-Alexander-University, D-91054 Erlangen, Germany
| | - Christoph Becker
- Department of Medicine 1, Friedrich-Alexander-University, D-91054 Erlangen, Germany
| | - John Bertin
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, Pennsylvania 19426, USA
| | - Peter J Gough
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, Pennsylvania 19426, USA
| | - Wim Declercq
- 1] VIB Inflammation Research Center, Technologiepark 927, B-9052 Ghent, Belgium [2] Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, B-9052 Ghent, Belgium
| | - Geert van Loo
- 1] VIB Inflammation Research Center, Technologiepark 927, B-9052 Ghent, Belgium [2] Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, B-9052 Ghent, Belgium
| | - Peter Vandenabeele
- 1] VIB Inflammation Research Center, Technologiepark 927, B-9052 Ghent, Belgium [2] Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, B-9052 Ghent, Belgium [3] Methusalem program, Ghent University, Technologiepark 927, B-9052 Ghent, Belgium
| |
Collapse
|
224
|
Weber DA, Sumagin R, McCall IC, Leoni G, Neumann PA, Andargachew R, Brazil JC, Medina-Contreras O, Denning TL, Nusrat A, Parkos CA. Neutrophil-derived JAML inhibits repair of intestinal epithelial injury during acute inflammation. Mucosal Immunol 2014; 7:1221-32. [PMID: 24621992 PMCID: PMC4340686 DOI: 10.1038/mi.2014.12] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Revised: 01/20/2014] [Accepted: 02/04/2014] [Indexed: 02/04/2023]
Abstract
Neutrophil transepithelial migration (TEM) during acute inflammation is associated with mucosal injury. Using models of acute mucosal injury in vitro and in vivo, we describe a new mechanism by which neutrophils infiltrating the intestinal mucosa disrupt epithelial homeostasis. We report that junctional adhesion molecule-like protein (JAML) is cleaved from neutrophil surface by zinc metalloproteases during TEM. Neutrophil-derived soluble JAML binds to the epithelial tight junction protein coxsackie-adenovirus receptor (CAR) resulting in compromised barrier and inhibition of wound repair, through decreased epithelial proliferation. The deleterious effects of JAML on barrier and wound repair are reversed with an anti-JAML monoclonal antibody that inhibits JAML-CAR binding. JAML released from transmigrating neutrophils across inflamed epithelia may thus promote recruitment of leukocytes and aid in clearance of invading microorganisms. However, sustained release of JAML under pathologic conditions associated with persistence of large numbers of infiltrated neutrophils would compromise intestinal barrier and inhibit mucosal healing. Thus, targeting JAML-CAR interactions may improve mucosal healing responses under conditions of dysregulated neutrophil recruitment.
Collapse
Affiliation(s)
- Dominique A. Weber
- Epithelial Pathobiology and Mucosal Inflammation Unit, Department of Pathology and Laboratory Medicine
| | - Ronen Sumagin
- Epithelial Pathobiology and Mucosal Inflammation Unit, Department of Pathology and Laboratory Medicine
| | - Ingrid C. McCall
- Epithelial Pathobiology and Mucosal Inflammation Unit, Department of Pathology and Laboratory Medicine
| | - Giovanna Leoni
- Epithelial Pathobiology and Mucosal Inflammation Unit, Department of Pathology and Laboratory Medicine
| | - Philipp A. Neumann
- Epithelial Pathobiology and Mucosal Inflammation Unit, Department of Pathology and Laboratory Medicine
| | - Rakieb Andargachew
- Epithelial Pathobiology and Mucosal Inflammation Unit, Department of Pathology and Laboratory Medicine
| | - Jennifer C. Brazil
- Epithelial Pathobiology and Mucosal Inflammation Unit, Department of Pathology and Laboratory Medicine,Department of Pediatrics, Emory University, Atlanta, Georgia 30322
| | - Oscar Medina-Contreras
- Epithelial Pathobiology and Mucosal Inflammation Unit, Department of Pathology and Laboratory Medicine,Department of Pediatrics, Emory University, Atlanta, Georgia 30322
| | - Timothy L. Denning
- Epithelial Pathobiology and Mucosal Inflammation Unit, Department of Pathology and Laboratory Medicine,Department of Pediatrics, Emory University, Atlanta, Georgia 30322
| | - Asma Nusrat
- Epithelial Pathobiology and Mucosal Inflammation Unit, Department of Pathology and Laboratory Medicine
| | - Charles A. Parkos
- Epithelial Pathobiology and Mucosal Inflammation Unit, Department of Pathology and Laboratory Medicine,Department of Pediatrics, Emory University, Atlanta, Georgia 30322
| |
Collapse
|
225
|
Brückner M, Lenz P, Nowacki TM, Pott F, Foell D, Bettenworth D. Murine endoscopy for in vivo multimodal imaging of carcinogenesis and assessment of intestinal wound healing and inflammation. J Vis Exp 2014:51875. [PMID: 25226434 PMCID: PMC4828016 DOI: 10.3791/51875] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Mouse models are widely used to study pathogenesis of human diseases and to evaluate diagnostic procedures as well as therapeutic interventions preclinically. However, valid assessment of pathological alterations often requires histological analysis, and when performed ex vivo, necessitates death of the animal. Therefore in conventional experimental settings, intra-individual follow-up examinations are rarely possible. Thus, development of murine endoscopy in live mice enables investigators for the first time to both directly visualize the gastrointestinal mucosa and also repeat the procedure to monitor for alterations. Numerous applications for in vivo murine endoscopy exist, including studying intestinal inflammation or wound healing, obtaining mucosal biopsies repeatedly, and to locally administer diagnostic or therapeutic agents using miniature injection catheters. Most recently, molecular imaging has extended diagnostic imaging modalities allowing specific detection of distinct target molecules using specific photoprobes. In conclusion, murine endoscopy has emerged as a novel cutting-edge technology for diagnostic experimental in vivo imaging and may significantly impact on preclinical research in various fields.
Collapse
Affiliation(s)
| | - Philipp Lenz
- Department of Medicine B, University Hospital Münster
| | | | | | - Dirk Foell
- Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster
| | | |
Collapse
|
226
|
Zigmond E, Varol C, Kaplan M, Shapira O, Melzer E. Low-Level Light Therapy Induces Mucosal Healing in a Murine Model of Dextran-Sodium-Sulfate Induced Colitis. Photomed Laser Surg 2014; 32:450-7. [DOI: 10.1089/pho.2013.3626] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Affiliation(s)
- Ehud Zigmond
- Research Center for Digestive Tract and Liver Diseases, Sourasky Medical Center & the Sackler Faculty of Medicine, Tel-Aviv, Israel
| | - Chen Varol
- Research Center for Digestive Tract and Liver Diseases, Sourasky Medical Center & the Sackler Faculty of Medicine, Tel-Aviv, Israel
| | - Michail Kaplan
- Research Center for Digestive Tract and Liver Diseases, Sourasky Medical Center & the Sackler Faculty of Medicine, Tel-Aviv, Israel
| | - Oz Shapira
- Department of Cardiothoracic Surgery, Hebrew University, Hadassah Medical Center, Jerusalem, Israel
| | - Ehud Melzer
- Gastroenterology and Hepatology, Kaplan Medical Center, Rehovot & Hebrew University Jerusalem, Israel
| |
Collapse
|
227
|
Bär F, Föh B, Pagel R, Schröder T, Schlichting H, Hirose M, Lemcke S, Klinger A, König P, Karsten CM, Büning J, Lehnert H, Fellermann K, Ibrahim SM, Sina C. Carboxypeptidase E modulates intestinal immune homeostasis and protects against experimental colitis in mice. PLoS One 2014; 9:e102347. [PMID: 25051500 PMCID: PMC4106776 DOI: 10.1371/journal.pone.0102347] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2014] [Accepted: 06/18/2014] [Indexed: 12/24/2022] Open
Abstract
Enteroendocrine cells (EEC) produce neuropeptides, which are crucially involved in the maintenance of the intestinal barrier. Hence, EEC dysfunction is suggested to be involved in the complex pathophysiology of inflammatory bowel disease (IBD), which is characterized by decreased intestinal barrier function. However, the underlying mechanisms for EEC dysfunction are not clear and suitable models for a better understanding are lacking. Here, we demonstrate that Carboxypeptidase E (CPE) is specifically expressed in EEC of the murine colon and ileum and that its deficiency is associated with reduced intestinal levels of Neuropeptide Y (NPY) and Peptide YY (PYY), which are both produced by EEC. Moreover, cpe−/− mice exhibit an aggravated course of DSS-induced chronic colitis compared to wildtype littermates. In addition, we observed elevated mucosal IL-6 and KC transcript levels already at baseline conditions in cpe−/− mice. Moreover, supernatants obtained from isolated intestinal crypts of cpe−/− mice lead to increased IL-6 and KC expression in MODE-K cells in the presence of LPS. This effect was reversible by co-administration of recombinant NPY, suggesting a CPE mediated immunosuppressive effect in the intestines by influencing the processing of specific neuropeptides. In this context, the chemotaxis of bone marrow derived macrophages towards respective supernatants was enhanced. In conclusion, our data point to an anti-inflammatory role of CPE in the intestine by influencing local cytokine levels and thus regulating the migration of myeloid immune cells into the mucosa. These findings highlight the importance of EEC for intestinal homeostasis and propose EEC as potential therapeutic targets in IBD.
Collapse
Affiliation(s)
- Florian Bär
- Medical Department I, University Hospital Schleswig-Holstein, Lübeck, Germany
- Institute of Anatomy, University of Lübeck, Lübeck, Germany
| | - Bandik Föh
- Medical Department I, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - René Pagel
- Medical Department I, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Torsten Schröder
- Medical Department I, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Heidi Schlichting
- Medical Department I, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Misa Hirose
- Department of Dermatology, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Susanne Lemcke
- Department of Dermatology, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Antje Klinger
- Institute of Anatomy, University of Lübeck, Lübeck, Germany
| | - Peter König
- Institute of Anatomy, University of Lübeck, Lübeck, Germany
| | - Christian M. Karsten
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Jürgen Büning
- Medical Department I, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Hendrik Lehnert
- Medical Department I, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Klaus Fellermann
- Medical Department I, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Saleh M. Ibrahim
- Department of Dermatology, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Christian Sina
- Medical Department I, University Hospital Schleswig-Holstein, Lübeck, Germany
- * E-mail:
| |
Collapse
|
228
|
Eckhardt J, Kreiser S, Döbbeler M, Nicolette C, DeBenedette MA, Tcherepanova IY, Ostalecki C, Pommer AJ, Becker C, Günther C, Zinser E, Mak TW, Steinkasserer A, Lechmann M. Soluble CD83 ameliorates experimental colitis in mice. Mucosal Immunol 2014; 7:1006-18. [PMID: 24424524 DOI: 10.1038/mi.2013.119] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 12/02/2013] [Indexed: 02/04/2023]
Abstract
The physiological balance between pro- and anti-inflammatory processes is dysregulated in inflammatory bowel diseases (IBD) as in Crohn's disease and ulcerative colitis. Conventional therapy uses anti-inflammatory and immunosuppressive corticosteroids to treat acute-phase symptoms. However, low remission rate and strong side effects of these therapies are not satisfying. Thus, there is a high medical need for new therapeutic strategies. Soluble CD83, the extracellular domain of the transmembrane CD83 molecule, has been reported to have interesting therapeutic and immunosuppressive properties by suppressing dendritic cell (DC)-mediated T-cell activation and inducing tolerogenic DCs. However, the expression and function of CD83 in IBD is still unknown. Here, we show that CD83 expression is upregulated by different leukocyte populations in a chemical-induced murine colitis model. Furthermore, in this study the potential of sCD83 to modulate colitis using an experimental murine colitis model was investigated. Strikingly, sCD83 ameliorated the clinical disease symptoms, drastically reduced mortality, and strongly decreased inflammatory cytokine expression in mesenteric lymph nodes and colon. The infiltration of macrophages and granulocytes into colonic tissues was vigorously inhibited. Mechanistically, we could show that sCD83-induced expression of indolamine 2,3-dioxygenase is essential for its protective effects.
Collapse
Affiliation(s)
- J Eckhardt
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - S Kreiser
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - M Döbbeler
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - C Nicolette
- Argos Therapeutics, Durham, North Carolina, USA
| | | | | | - C Ostalecki
- Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - A J Pommer
- Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - C Becker
- Department of Medicine 1, University Hospital Erlangen, Erlangen, Germany
| | - C Günther
- Department of Medicine 1, University Hospital Erlangen, Erlangen, Germany
| | - E Zinser
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - T W Mak
- The Campbell Family Institute for Breast Cancer Research at Princess Margaret Hospital, University Health Network, Toronto, Ontario, Canada
| | - A Steinkasserer
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - M Lechmann
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| |
Collapse
|
229
|
Pastille E, Bardini K, Fleissner D, Adamczyk A, Frede A, Wadwa M, von Smolinski D, Kasper S, Sparwasser T, Gruber AD, Schuler M, Sakaguchi S, Roers A, Müller W, Hansen W, Buer J, Westendorf AM. Transient ablation of regulatory T cells improves antitumor immunity in colitis-associated colon cancer. Cancer Res 2014; 74:4258-69. [PMID: 24906621 DOI: 10.1158/0008-5472.can-13-3065] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Regulatory T cells (Treg) are supportive to cancer development in most tissues, but their role in colitis-associated colon cancer (CAC) remains unclear. In this study, we investigated the role of CD4(+)Foxp3(+) Treg in a mouse model of CAC and in patients with colon cancer. These Treg were increased strongly in number in a mouse model of CAC and in the peripheral blood of patients with colon cancer, exhibiting an activated phenotype as defined by elevated expression of GARP, CD103, CTLA-4, and IL10, along with an increased suppressive effect on the proliferation and Th1 cytokine expression of CD4(+)CD25(-) responder T cells ex vivo. Transient ablation of CD4(+)Foxp3(+) Treg during tumor development in the CAC model suppressed tumor outgrowth and distribution, accompanied by an increased number of CD8(+)IFNγ/granzyme B-producing effector T cells. Conversely, inactivation of IL10 in Treg did not elevate the antitumor response but instead further boosted tumor development. Our results establish a tumor-promoting function for Treg during CAC formation, but they also suggest that a selective, transient ablation of Treg can evoke antitumor responses, with implications for immunotherapeutic interventions in patients with CAC.
Collapse
Affiliation(s)
- Eva Pastille
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Katrin Bardini
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Diana Fleissner
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Alexandra Adamczyk
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Annika Frede
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Munisch Wadwa
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | | | - Stefan Kasper
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Tim Sparwasser
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Medicine, Hannover, Germany
| | - Achim D Gruber
- Institute of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Martin Schuler
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany. German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Shimon Sakaguchi
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Axel Roers
- Institute for Immunology, Technische Universität Dresden, Medical Faculty Carl Gustav Carus, Dresden, Germany
| | - Werner Müller
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Wiebke Hansen
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Jan Buer
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Astrid M Westendorf
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany.
| |
Collapse
|
230
|
Wang T, Li Q, Xiao P, Ahn J, Kim YE, Park Y, Kim M, Song M, Chung E, Chung WK, Ahn GO, Kim S, Kim P, Myung SJ, Kim KH. Gradient index lens based combined two-photon microscopy and optical coherence tomography. OPTICS EXPRESS 2014; 22:12962-70. [PMID: 24921493 DOI: 10.1364/oe.22.012962] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
We report a miniaturized probe-based combined two-photon microscopy (TPM) and optical coherence tomography (OCT) system. This system is to study the colorectal cancer in mouse models by visualizing both cellular and structural information of the colon in 3D with TPM and OCT respectively. The probe consisted of gradient index (GRIN) lenses and a 90° reflecting prism at its distal end for side-viewing, and it was added onto an objective lens-based TPM and OCT system. The probe was 2.2 mm in diameter and 60 mm in length. TPM imaging was performed by raster scanning of the excitation focus at the imaging speed of 15.4 frames/s. OCT imaging was performed by combining the linear sample translation and probe rotation along its axis. This miniaturized probe based dual-modal system was characterized with tissue phantoms containing fluorescent microspheres, and applied to image mouse colonic tissues ex vivo as a demonstration. As OCT and TPM provided structural and cellular information of the tissues respectively, this probe based multi-modal imaging system can be helpful for in vivo studies of preclinical animal models such as mouse colonic tumorigenesis.
Collapse
|
231
|
Aluminum enhances inflammation and decreases mucosal healing in experimental colitis in mice. Mucosal Immunol 2014; 7:589-601. [PMID: 24129165 PMCID: PMC3998638 DOI: 10.1038/mi.2013.78] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 08/12/2013] [Accepted: 08/14/2013] [Indexed: 02/04/2023]
Abstract
The increasing incidence of inflammatory bowel diseases (IBDs) in developing countries has highlighted the critical role of environmental pollutants as causative factors in their pathophysiology. Despite its ubiquity and immune toxicity, the impact of aluminum in the gut is not known. This study aimed to evaluate the effects of environmentally relevant intoxication with aluminum in murine models of colitis and to explore the underlying mechanisms. Oral administration of aluminum worsened intestinal inflammation in mice with 2,4,6-trinitrobenzene sulfonic acid- and dextran sodium sulfate-induced colitis and chronic colitis in interleukin 10-negative (IL10(-/-)) mice. Aluminum increased the intensity and duration of macroscopic and histologic inflammation, colonic myeloperoxidase activity, inflammatory cytokines expression, and decreased the epithelial cell renewal compared with control animals. Under basal conditions, aluminum impaired intestinal barrier function. In vitro, aluminum induced granuloma formation and synergized with lipopolysaccharide to stimulate inflammatory cytokines expression by epithelial cells. Deleterious effects of aluminum on intestinal inflammation and mucosal repair strongly suggest that aluminum might be an environmental IBD risk factor.
Collapse
|
232
|
Naumov I, Zilberberg A, Shapira S, Avivi D, Kazanov D, Rosin-Arbesfeld R, Arber N, Kraus S. CD24 knockout prevents colorectal cancer in chemically induced colon carcinogenesis and in APC(Min)/CD24 double knockout transgenic mice. Int J Cancer 2014; 135:1048-59. [PMID: 24500912 DOI: 10.1002/ijc.28762] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2013] [Accepted: 01/20/2014] [Indexed: 02/06/2023]
Abstract
Increased expression of CD24 is seen in a large variety of solid tumors, including up to 90% of gastrointestinal (GI) tumors. Stable derivatives of SW480 colorectal cancer (CRC) cells that overexpress CD24 proliferate faster, and increase cell motility, saturation density, plating efficiency, and growth in soft agar. They also produce larger tumors in nude mice as compared to the parental SW480 cells. Most significantly, even depletion of one copy of the CD24 allele in the APC(Min/+) mice of a transgenic mouse model led to a dramatic reduction in tumor burden in all sections of the small intestine. Homozygous deletion of both CD24 alleles resulted in complete abolishment of tumor formation. Moreover, CD24 knockout mice exhibited resistance to chemically induced inflammation-associated CRC. Finally, a new signal transduction pathway is suggested: namely, CD24 expression downstream to COX2 and PGE2 synthesis, which is directly regulated by β-catenin. CD24 is shown in vitro and in vivo as being an important oncogene in the gut, and one that plays a critical role in the initiation and progression of carcinogenesis.
Collapse
Affiliation(s)
- Inna Naumov
- The Integrated Cancer Prevention Center Tel Aviv Sourasky Medical Center Tel Aviv Israel Affiliated to the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | | | | | | | | | | | | |
Collapse
|
233
|
Adachi T, Hinoi T, Sasaki Y, Niitsu H, Saito Y, Miguchi M, Shimomura M, Ohdan H. Colonoscopy as a tool for evaluating colorectal tumor development in a mouse model. Int J Colorectal Dis 2014; 29:217-23. [PMID: 24212401 DOI: 10.1007/s00384-013-1791-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/18/2013] [Indexed: 02/04/2023]
Abstract
PURPOSE A sporadic colon cancer mouse model with conditional mutations in adenomatous polyposis coli (Apc) is biologically relevant for human colorectal cancer (CRC). This study aimed to determine the utility and limitations of colonoscopy for evaluating colon tumors in this mouse model. METHODS We compared the estimates of location, size, and miss rate of tumors detected during colonoscopy with those determined by necropsy. Sixty-six CPC-Apc mice originating from Apc (F/wt) mice harbor a Cdx2-Cre transgene in which colorectal tumorigenesis was driven by Apc allelic loss. The sensitivity and specificity of colonoscopy for detecting tumors in a mouse CRC model were investigated. RESULTS A strong positive correlation was found between tumor location as measured by colonoscopy and the location determined by necropsy (p < 0.001). A total of 120 tumors were graded during colonoscopy (grades 1-5: 0, 8, 20, 27, and 65 lesions, respectively), and a strong positive correlation was found between the tumor grade determined by colonoscopy and size measured by necropsy (grades 2-5: 2.08, 2.98, 4.02, and 5.09 mm, respectively; p < 0.005). Although the miss rate was 47.1 %, most of the missed tumors (96 %) were in close proximity (within 5 mm) of another tumor. CONCLUSIONS A colonoscopic method for the reliable measurement of colorectal tumors in vivo has been established. The application of this technique to mouse models of colon carcinogenesis will provide a better understanding of the dynamics of tumor growth.
Collapse
Affiliation(s)
- Tomohiro Adachi
- Department of Surgery, Division of Frontier Medical Sciences, Programs for Biomedical Research, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | | | | | | | | | | | | | | |
Collapse
|
234
|
Characterisation of enterocolitis in the piroxicam-accelerated interleukin-10 knock out mouse--a model mimicking inflammatory bowel disease. J Crohns Colitis 2014; 8:147-60. [PMID: 23994255 DOI: 10.1016/j.crohns.2013.08.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 07/30/2013] [Accepted: 08/06/2013] [Indexed: 02/08/2023]
Abstract
BACKGROUND In inflammatory bowel disease a defective mucosal barrier, a dysregulated immune response and an excessive reactivity against the gut microbiota are assumed to cause a breakdown of the intestinal homeostasis and lead to chronic inflammation. Piroxicam treatment is a method for induction of colitis in IL-10 k.o. mice, which integrates a dysfunction of both the intestinal barrier and the immune system. However, the translational value of this model has not been thoroughly clarified. AIM To characterise the piroxicam-accelerated colitis (PAC) IL-10 k.o. model with respect to clinical features, pathogenic mechanisms and its ability to respond to existing therapies. METHODS The PAC IL-10k.o. model was established on a C57BL/6J background and the clinical manifestations, immunological mechanisms and efficacy of ampicillin and anti-IL-12/23p40 treatment were assessed. RESULTS The PAC IL-10 k.o. mice developed weight loss and diarrhoea, and colonoscopy revealed a thickened granulomatous mucosa. Histological evaluation of ileum and colon showed Crohn's disease-like changes with pronounced hyperplasia and focal transmural inflammation. Ileitis was also observed in piroxicam treated wild type mice. The total number of neutrophils, monocytes and natural killer cells was elevated in the blood compared to IL-10 k.o. and wild type mice, indicating a role of the innate immune system in the pathogenesis. These findings were supported by analyses of the intestinal cytokine profile. Ampicillin and anti-IL-12/23p40 treatment significantly suppressed disease in the model. CONCLUSION The PAC IL-10 k.o. model resembles several features of Crohn's disease and could be a useful in vivo model in preclinical research.
Collapse
|
235
|
Stuart E, Buchert M, Putoczki T, Thiem S, Farid R, Elzer J, Huszar D, Waring PM, Phesse TJ, Ernst M. Therapeutic Inhibition of Jak Activity Inhibits Progression of Gastrointestinal Tumors in Mice. Mol Cancer Ther 2014; 13:468-74. [DOI: 10.1158/1535-7163.mct-13-0583-t] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
236
|
Marques CC, Castelo-Branco MT, Pacheco RG, Buongusto F, do Rosário A, Schanaider A, Coutinho-Silva R, de Souza HSP. Prophylactic systemic P2X7 receptor blockade prevents experimental colitis. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1842:65-78. [PMID: 24184714 DOI: 10.1016/j.bbadis.2013.10.012] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Revised: 10/16/2013] [Accepted: 10/22/2013] [Indexed: 12/31/2022]
Abstract
BACKGROUND The P2X7 receptor (P2X7-R) is a non-selective adenosine triphosphate-gated cation channel present in epithelial and immune cells, and involved in inflammatory response. Extracellular nucleotides released in conditions of cell stress or inflammation may function as a danger signal alerting the immune system from inflammation. We investigated the therapeutic action of P2X7-R blockade in a model of inflammatory bowel disease. METHODS Rats with trinitrobenzene sulfonic (TNBS) acid-induced colitis were treated with the P2X7-R antagonists A740003 or brilliant blue G (BBG) through intra-peritoneal (IP) or intra-colonic (IC) injection prior to colitis induction. Clinical and endoscopic follow-up, histological scores, myeloperoxidase activity, densities of collagen fibers and goblet cells were evaluated. P2X7-R expression, NF-kappa B and Erk activities, and densities of T-cells and macrophages were analyzed by immunoperoxidase. The inflammatory response was determined by measuring inflammatory cytokines in cultures of colon explants, by enzyme-linked immunosorbent assay. Colonic apoptosis was determined by the TUNEL assay. RESULTS IP-BBG significantly attenuated the severity of colitis, myeloperoxidase activity, collagen deposition, densities of lamina propria T-cells and macrophages, while maintaining goblet cell densities. IP-BBG inhibited the increase in P2X7-R expression in parallel with apoptotic rates. TNF-α and interleukin-1β stabilized in low levels, while TGF-β and interleukin-10 did not change following IP-BBG-therapy. Colonic NF-kappa-B and Erk activation were significantly lower in IP-BBG-treated animals. Prophylactic IP-A740003 also protected rats against the development of TNBS-colitis. CONCLUSIONS Prophylactic systemic P2X7-R blockade is effective in the prevention of experimental colitis, probably due to a systemic anti-inflammatory action, interfering with a stress-inflammation amplification loop mediated by P2X7-R.
Collapse
Affiliation(s)
- Carla Caldas Marques
- Departamento de Clínica Médica, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-913, Brazil; Laboratório de Imunologia Celular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-590, Brazil; Programa de Imunobiologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-590, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
237
|
de Britto MAP, Soletti RC, Schanaider A, Madi K, de Souza HSP, Machado JC. Endoluminal ultrasound biomicroscopy as a reliable tool for in vivo assessment of colonic inflammation in rats. Int J Colorectal Dis 2013; 28:1613-1620. [PMID: 23925435 DOI: 10.1007/s00384-013-1755-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/23/2013] [Indexed: 02/04/2023]
Abstract
PURPOSE Endoscopic ultrasound (EUS) imaging of the colon is an important diagnostic tool for early neoplasia, although usually restricted to the rectum in inflammatory bowel disease (IBD). This study aimed to evaluate the ability of an endoluminal ultrasound biomicroscopic (eUBM) system to detect and characterize lesions simulating Crohn's disease in the colon of rats in vivo. METHODS Colitis was induced with trinitrobenzene sulfonic acid instillated in the distal colon. Eighteen Wistar rats were submitted to eUBM in three time points: week 1 group (18 animals examined on day 3 after colitis induction), week 2 group (12 animals on days 3 and 10), and week 3 group (7 animals on days 3, 10, and 17). This design yielded distinct inflammation intensities. Three untreated rats were used for acquisition of control images. Scores were used for comparison with histology. RESULTS Scores for eUBM and histology in the different moments of examination achieved a Spearman's rank correlation coefficient of 0.87 (p < 0.001). Findings of wall thickening presented positive predictive value (PPV) and sensitivity of 94 and of 100 %, respectively. Superficial and deep ulcers presented a PPV of 89 and 80 %, respectively, and negative predictive values of 100 and 85 %, respectively. CONCLUSION Accurate detection and analysis of the lesions was achieved. The model is essential for the clinical development of the technique and a reproducible method for the evaluation of experimental colitis. eUBM might be applicable in different segments of the gut, developing into a novel adjunct method for IBD evaluation.
Collapse
Affiliation(s)
- Marcelo Alexandre Pinto de Britto
- Post-Graduation Program in Surgical Sciences, Department of Surgery, School of Medicine, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | | | | | | | | |
Collapse
|
238
|
Paul Olson TJ, Hadac JN, Sievers CK, Leystra AA, Deming DA, Zahm CD, Albrecht DM, Nomura A, Nettekoven LA, Plesh LK, Clipson L, Sullivan R, Newton MA, Schelman WR, Halberg RB. Dynamic tumor growth patterns in a novel murine model of colorectal cancer. Cancer Prev Res (Phila) 2013; 7:105-13. [PMID: 24196829 DOI: 10.1158/1940-6207.capr-13-0163] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Colorectal cancer often arises from adenomatous colonic polyps. Polyps can grow and progress to cancer, but may also remain static in size, regress, or resolve. Predicting which polyps progress and which remain benign is difficult. We developed a novel long-lived murine model of colorectal cancer with tumors that can be followed by colonoscopy. Our aim was to assess whether these tumors have similar growth patterns and histologic fates to human colorectal polyps to identify features to aid in risk stratification of colonic tumors. Long-lived Apc(Min/+) mice were treated with dextran sodium sulfate to promote colonic tumorigenesis. Tumor growth patterns were characterized by serial colonoscopy with biopsies obtained for immunohistochemistry and gene expression profiling. Tumors grew, remained static, regressed, or resolved over time with different relative frequencies. Newly developed tumors demonstrated higher rates of growth and resolution than more established tumors that tended to remain static in size. Colonic tumors were hyperplastic lesions (3%), adenomas (73%), intramucosal carcinomas (20%), or adenocarcinomas (3%). Interestingly, the level of β-catenin was higher in adenomas that became intratumoral carcinomas than those that failed to progress. In addition, differentially expressed genes between adenomas and intramucosal carcinomas were identified. This novel murine model of intestinal tumorigenesis develops colonic tumors that can be monitored by serial colonoscopy, mirror growth patterns seen in human colorectal polyps, and progress to colorectal cancer. Further characterization of cellular and molecular features is needed to determine which features can be used to risk-stratify polyps for progression to colorectal cancer and potentially guide prevention strategies.
Collapse
Affiliation(s)
- Terrah J Paul Olson
- University of Wisconsin-Madison K4/532 Clinical Science Center, 600 Highland Avenue, Madison, WI 53792.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
239
|
Chua ACG, Klopcic BRS, Ho DS, Fu SK, Forrest CH, Croft KD, Olynyk JK, Lawrance IC, Trinder D. Dietary iron enhances colonic inflammation and IL-6/IL-11-Stat3 signaling promoting colonic tumor development in mice. PLoS One 2013; 8:e78850. [PMID: 24223168 PMCID: PMC3819375 DOI: 10.1371/journal.pone.0078850] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Accepted: 09/16/2013] [Indexed: 12/18/2022] Open
Abstract
Chronic intestinal inflammation and high dietary iron are associated with colorectal cancer development. The role of Stat3 activation in iron-induced colonic inflammation and tumorigenesis was investigated in a mouse model of inflammation-associated colorectal cancer. Mice, fed either an iron-supplemented or control diet, were treated with azoxymethane and dextran sodium sulfate (DSS). Intestinal inflammation and tumor development were assessed by endoscopy and histology, gene expression by real-time PCR, Stat3 phosphorylation by immunoblot, cytokines by ELISA and apoptosis by TUNEL assay. Colonic inflammation was more severe in mice fed an iron-supplemented compared with a control diet one week post-DSS treatment, with enhanced colonic IL-6 and IL-11 release and Stat3 phosphorylation. Both IL-6 and ferritin, the iron storage protein, co-localized with macrophages suggesting iron may act directly on IL-6 producing-macrophages. Iron increased DSS-induced colonic epithelial cell proliferation and apoptosis consistent with enhanced mucosal damage. DSS-treated mice developed anemia that was not alleviated by dietary iron supplementation. Six weeks post-DSS treatment, iron-supplemented mice developed more and larger colonic tumors compared with control mice. Intratumoral IL-6 and IL-11 expression increased in DSS-treated mice and IL-6, and possibly IL-11, were enhanced by dietary iron. Gene expression of iron importers, divalent metal transporter 1 and transferrin receptor 1, increased and iron exporter, ferroportin, decreased in colonic tumors suggesting increased iron uptake. Dietary iron and colonic inflammation synergistically activated colonic IL-6/IL-11-Stat3 signaling promoting tumorigenesis. Oral iron therapy may be detrimental in inflammatory bowel disease since it may exacerbate colonic inflammation and increase colorectal cancer risk.
Collapse
Affiliation(s)
- Anita C. G. Chua
- School of Medicine and Pharmacology, Fremantle Hospital, University of Western Australia, Fremantle, Western Australia, Australia
- Western Australian Institute for Medical Research, Perth, Western Australia, Australia
- * E-mail:
| | - Borut R. S. Klopcic
- School of Medicine and Pharmacology, Fremantle Hospital, University of Western Australia, Fremantle, Western Australia, Australia
- Centre for Inflammatory Bowel Diseases, Fremantle Hospital, Fremantle, Western Australia, Australia
| | - Desiree S. Ho
- School of Medicine and Pharmacology, Fremantle Hospital, University of Western Australia, Fremantle, Western Australia, Australia
- Western Australian Institute for Medical Research, Perth, Western Australia, Australia
| | - S. Kristine Fu
- School of Medicine and Pharmacology, Fremantle Hospital, University of Western Australia, Fremantle, Western Australia, Australia
- Centre for Inflammatory Bowel Diseases, Fremantle Hospital, Fremantle, Western Australia, Australia
| | - Cynthia H. Forrest
- Department of Histopathology, PathWest, Fremantle Hospital, Fremantle, Western Australia, Australia
- School of Pathology and Laboratory Medicine, University of Western Australia, Perth, Western Australia, Australia
| | - Kevin D. Croft
- School of Medicine and Pharmacology, Royal Perth Hospital, University of Western Australia, Perth, Western Australia, Australia
| | - John K. Olynyk
- Department of Gastroenterology, Fremantle Hospital, Fremantle, Western Australia, Australia
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Western Australia, Australia
- Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia
| | - Ian C. Lawrance
- School of Medicine and Pharmacology, Fremantle Hospital, University of Western Australia, Fremantle, Western Australia, Australia
- Centre for Inflammatory Bowel Diseases, Fremantle Hospital, Fremantle, Western Australia, Australia
| | - Debbie Trinder
- School of Medicine and Pharmacology, Fremantle Hospital, University of Western Australia, Fremantle, Western Australia, Australia
- Western Australian Institute for Medical Research, Perth, Western Australia, Australia
| |
Collapse
|
240
|
Scher JU, Sczesnak A, Longman RS, Segata N, Ubeda C, Bielski C, Rostron T, Cerundolo V, Pamer EG, Abramson SB, Huttenhower C, Littman DR. Expansion of intestinal Prevotella copri correlates with enhanced susceptibility to arthritis. eLife 2013; 2:e01202. [PMID: 24192039 PMCID: PMC3816614 DOI: 10.7554/elife.01202 10.7554/elife.01202] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Rheumatoid arthritis (RA) is a prevalent systemic autoimmune disease, caused by a combination of genetic and environmental factors. Animal models suggest a role for intestinal bacteria in supporting the systemic immune response required for joint inflammation. Here we performed 16S sequencing on 114 stool samples from rheumatoid arthritis patients and controls, and shotgun sequencing on a subset of 44 such samples. We identified the presence of Prevotella copri as strongly correlated with disease in new-onset untreated rheumatoid arthritis (NORA) patients. Increases in Prevotella abundance correlated with a reduction in Bacteroides and a loss of reportedly beneficial microbes in NORA subjects. We also identified unique Prevotella genes that correlated with disease. Further, colonization of mice revealed the ability of P. copri to dominate the intestinal microbiota and resulted in an increased sensitivity to chemically induced colitis. This work identifies a potential role for P. copri in the pathogenesis of RA. DOI: http://dx.doi.org/10.7554/eLife.01202.001.
Collapse
Affiliation(s)
- Jose U Scher
- Department of Medicine, New York University School of Medicine and Hospital for Joint Diseases, New York, United States
| | - Andrew Sczesnak
- Molecular Pathogenesis Program, The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, United States,Graduate Program in Bioinformatics and Computational Biology, University of California, San Francisco, San Francisco, United States
| | - Randy S Longman
- Molecular Pathogenesis Program, The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, United States,Jill Roberts IBD Center, Department of Medicine, Weill Cornell Medical College, New York, United States
| | - Nicola Segata
- Centre for Integrative Biology, University of Trento, Trento, Italy,Department of Biostatistics, Harvard School of Public Health, Boston, United States
| | - Carles Ubeda
- Immunology Program, Infectious Diseases Service, and The Lucille Castori Center for Microbes, Inflammation, and Cancer, Memorial Sloan-Kettering Cancer Center, New York, United States,Centro Superior de Investigacion en Salud Publica, University of Valencia, Valencia, Spain
| | - Craig Bielski
- Department of Biostatistics, Harvard School of Public Health, Boston, United States
| | - Tim Rostron
- Department of Medicine, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Vincenzo Cerundolo
- Department of Medicine, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Eric G Pamer
- Immunology Program, Infectious Diseases Service, and The Lucille Castori Center for Microbes, Inflammation, and Cancer, Memorial Sloan-Kettering Cancer Center, New York, United States
| | - Steven B Abramson
- Department of Medicine, New York University School of Medicine and Hospital for Joint Diseases, New York, United States
| | - Curtis Huttenhower
- Department of Biostatistics, Harvard School of Public Health, Boston, United States
| | - Dan R Littman
- Molecular Pathogenesis Program, The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, United States,Howard Hughes Medical Institute, New York University School of Medicine, New York, United States,For correspondence:
| |
Collapse
|
241
|
Scher JU, Sczesnak A, Longman RS, Segata N, Ubeda C, Bielski C, Rostron T, Cerundolo V, Pamer EG, Abramson SB, Huttenhower C, Littman DR. Expansion of intestinal Prevotella copri correlates with enhanced susceptibility to arthritis. eLife 2013; 2:e01202. [PMID: 24192039 PMCID: PMC3816614 DOI: 10.7554/elife.01202] [Citation(s) in RCA: 1418] [Impact Index Per Article: 118.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Rheumatoid arthritis (RA) is a prevalent systemic autoimmune disease, caused by a combination of genetic and environmental factors. Animal models suggest a role for intestinal bacteria in supporting the systemic immune response required for joint inflammation. Here we performed 16S sequencing on 114 stool samples from rheumatoid arthritis patients and controls, and shotgun sequencing on a subset of 44 such samples. We identified the presence of Prevotella copri as strongly correlated with disease in new-onset untreated rheumatoid arthritis (NORA) patients. Increases in Prevotella abundance correlated with a reduction in Bacteroides and a loss of reportedly beneficial microbes in NORA subjects. We also identified unique Prevotella genes that correlated with disease. Further, colonization of mice revealed the ability of P. copri to dominate the intestinal microbiota and resulted in an increased sensitivity to chemically induced colitis. This work identifies a potential role for P. copri in the pathogenesis of RA. DOI:http://dx.doi.org/10.7554/eLife.01202.001 We share our bodies with a diverse set of microorganisms, known collectively as the human microbiome. Indeed, estimates suggest that our bodies contain 10 times as many microbial cells as human cells. Our stomach and intestines alone are home to many hundreds and possibly thousands of microbial species that break down indigestible compounds and help to prevent the growth of harmful bacteria. The immune system must therefore learn to tolerate these microorganisms, while retaining the ability to launch attacks against microorganisms that cause harm. Failure of this process may increase the risk of autoimmune diseases in which the body mistakenly attacks its own cells and tissues. Rheumatoid arthritis is a chronic autoimmune disease marked by inflammation of the joints. Although the causes of rheumatoid arthritis are unknown, mice with mutations that increase the risk of the disease remain healthy if they are kept under sterile conditions. However, if these mice are exposed to certain species of bacteria sometimes found in the gut, they begin to show signs of joint inflammation. Here, Scher et al. used genome sequencing to compare gut bacteria from patients with rheumatoid arthritis and healthy controls. A bacterial species called Prevotella copri was more abundant in patients suffering from untreated rheumatoid arthritis than in healthy individuals. Moreover, the presence of P. copri corresponded to a reduction in the abundance of other bacterial groups—including a number of beneficial microbes. In a mouse model of gut inflammation, animals colonized with P. copri had more severe disease than controls, consistent with a pro-inflammatory function of this organism. Current treatments for rheumatoid arthritis target symptoms. However, by highlighting the role played by gut bacteria, the work of Scher et al. suggests that novel treatment options focused on curbing the spread of P. copri in the gut could delay or prevent the onset of this disease. DOI:http://dx.doi.org/10.7554/eLife.01202.002
Collapse
Affiliation(s)
- Jose U Scher
- Department of Medicine, New York University School of Medicine and Hospital for Joint Diseases, New York, United States
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
242
|
Scher JU, Sczesnak A, Longman RS, Segata N, Ubeda C, Bielski C, Rostron T, Cerundolo V, Pamer EG, Abramson SB, Huttenhower C, Littman DR. Expansion of intestinal Prevotella copri correlates with enhanced susceptibility to arthritis. eLife 2013; 2:e01202. [PMID: 24192039 DOI: 10.7554/elife.01202.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2023] Open
Abstract
Rheumatoid arthritis (RA) is a prevalent systemic autoimmune disease, caused by a combination of genetic and environmental factors. Animal models suggest a role for intestinal bacteria in supporting the systemic immune response required for joint inflammation. Here we performed 16S sequencing on 114 stool samples from rheumatoid arthritis patients and controls, and shotgun sequencing on a subset of 44 such samples. We identified the presence of Prevotella copri as strongly correlated with disease in new-onset untreated rheumatoid arthritis (NORA) patients. Increases in Prevotella abundance correlated with a reduction in Bacteroides and a loss of reportedly beneficial microbes in NORA subjects. We also identified unique Prevotella genes that correlated with disease. Further, colonization of mice revealed the ability of P. copri to dominate the intestinal microbiota and resulted in an increased sensitivity to chemically induced colitis. This work identifies a potential role for P. copri in the pathogenesis of RA. DOI: http://dx.doi.org/10.7554/eLife.01202.001.
Collapse
Affiliation(s)
- Jose U Scher
- Department of Medicine, New York University School of Medicine and Hospital for Joint Diseases, New York, United States
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
243
|
Bär F, Bochmann W, Widok A, von Medem K, Pagel R, Hirose M, Yu X, Kalies K, König P, Böhm R, Herdegen T, Reinicke AT, Büning J, Lehnert H, Fellermann K, Ibrahim S, Sina C. Mitochondrial gene polymorphisms that protect mice from colitis. Gastroenterology 2013; 145:1055-1063.e3. [PMID: 23872498 DOI: 10.1053/j.gastro.2013.07.015] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 06/11/2013] [Accepted: 07/20/2013] [Indexed: 12/30/2022]
Abstract
BACKGROUND & AIMS Dysregulated energy homeostasis in the intestinal mucosa frequently is observed in patients with ulcerative colitis (UC). Intestinal tissues from these patients have reduced activity of the mitochondrial oxidative phosphorylation (OXPHOS) complex, so mitochondrial dysfunction could contribute to the pathogenesis of UC. However, little is known about the mechanisms by which OXPHOS activity could be altered. We used conplastic mice, which have identical nuclear but different mitochondrial genomes, to investigate activities of the OXPHOS complex. METHODS Colitis was induced in C57BL/6J wild-type (B6.B6) and 3 strains of conplastic mice (B6.NZB, B6.NOD, and B6.AKR) by administration of dextran sodium sulfate or rectal application of trinitrobenzene sulfonate. Colon tissues were collected and analyzed by histopathology, immunohistochemical analysis, and immunoblot analysis; we also measured mucosal levels of adenosine triphosphate (ATP) and reactive oxygen species, OXPHOS complex activity, and epithelial cell proliferation and apoptosis. RESULTS We identified mice with increased mucosal OXPHOS complex activities and levels of ATP. These mice developed less-severe colitis after administration of dextran sodium sulfate or trinitrobenzene sulfonate than mice with lower mucosal levels of ATP. Colon tissues from these mice also had increased enterocyte proliferation and transcription factor nuclear factor-κB activity, which have been shown to protect the mucosal barrier-defects in these processes have been associated with inflammatory bowel disease. CONCLUSIONS Variants in mitochondrial DNA that increase mucosal levels of ATP protect mice from colitis. Increasing mitochondrial ATP synthesis in intestinal epithelial cells could be a therapeutic approach for UC.
Collapse
Affiliation(s)
- Florian Bär
- Medical Department I, University Hospital Schleswig-Holstein, Lübeck, Germany; Institute of Anatomy, University of Lübeck, Lübeck, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
244
|
Kodani T, Rodriguez-Palacios A, Corridoni D, Lopetuso L, Di Martino L, Marks B, Pizarro J, Pizarro T, Chak A, Cominelli F. Flexible colonoscopy in mice to evaluate the severity of colitis and colorectal tumors using a validated endoscopic scoring system. J Vis Exp 2013:e50843. [PMID: 24193215 DOI: 10.3791/50843] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
The use of modern endoscopy for research purposes has greatly facilitated our understanding of gastrointestinal pathologies. In particular, experimental endoscopy has been highly useful for studies that require repeated assessments in a single laboratory animal, such as those evaluating mechanisms of chronic inflammatory bowel disease and the progression of colorectal cancer. However, the methods used across studies are highly variable. At least three endoscopic scoring systems have been published for murine colitis and published protocols for the assessment of colorectal tumors fail to address the presence of concomitant colonic inflammation. This study develops and validates a reproducible endoscopic scoring system that integrates evaluation of both inflammation and tumors simultaneously. This novel scoring system has three major components: 1) assessment of the extent and severity of colorectal inflammation (based on perianal findings, transparency of the wall, mucosal bleeding, and focal lesions), 2) quantitative recording of tumor lesions (grid map and bar graph), and 3) numerical sorting of clinical cases by their pathological and research relevance based on decimal units with assigned categories of observed lesions and endoscopic complications (decimal identifiers). The video and manuscript presented herein were prepared, following IACUC-approved protocols, to allow investigators to score their own experimental mice using a well-validated and highly reproducible endoscopic methodology.
Collapse
Affiliation(s)
- Tomohiro Kodani
- Division of Gastroenterology, Case Western Reserve University School of Medicine, Cleveland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
245
|
Morón B, Spalinger M, Kasper S, Atrott K, Frey-Wagner I, Fried M, McCole DF, Rogler G, Scharl M. Activation of protein tyrosine phosphatase non-receptor type 2 by spermidine exerts anti-inflammatory effects in human THP-1 monocytes and in a mouse model of acute colitis. PLoS One 2013; 8:e73703. [PMID: 24040033 PMCID: PMC3767590 DOI: 10.1371/journal.pone.0073703] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 07/18/2013] [Indexed: 12/19/2022] Open
Abstract
Background Spermidine is a dietary polyamine that is able to activate protein tyrosine phosphatase non-receptor type 2 (PTPN2). As PTPN2 is known to be a negative regulator of interferon-gamma (IFN-γ)-induced responses, and IFN-γ stimulation of immune cells is a critical process in the immunopathology of inflammatory bowel disease (IBD), we wished to explore the potential of spermidine for reducing pro-inflammatory effects in vitro and in vivo. Methods Human THP-1 monocytes were treated with IFN-γ and/or spermidine. Protein expression and phosphorylation were analyzed by Western blot, cytokine expression by quantitative-PCR, and cytokine secretion by ELISA. Colitis was induced in mice by dextran sodium sulfate (DSS) administration. Disease severity was assessed by recording body weight, colonoscopy and histology. Results Spermidine increased expression and activity of PTPN2 in THP-1 monocytes and reduced IFN-γ-induced phosphorylation of signal transducer and activator of transcription (STAT) 1 and 3, as well as p38 mitogen-activated protein kinase (MAPK) in a PTPN2 dependent manner. Subsequently, IFN-γ-induced expression/secretion of intracellular cell adhesion molecule (ICAM)-1 mRNA, monocyte chemoattractant protein (MCP)-1, and interleukin (IL)-6 was reduced in spermidine-treated cells. The latter effects were absent in PTPN2-knockdown cells. In mice with DSS-induced colitis, spermidine treatment resulted in ameliorated weight loss and decreased mucosal damage indicating reduced disease severity. Conclusions Activation of PTPN2 by spermidine ameliorates IFN-γ-induced inflammatory responses in THP-1 cells. Furthermore, spermidine treatment significantly reduces disease severity in mice with DSS-induced colitis; hence, spermidine supplementation and subsequent PTPN2 activation may be helpful in the treatment of chronic intestinal inflammation such as IBD.
Collapse
Affiliation(s)
- Belén Morón
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Marianne Spalinger
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Stephanie Kasper
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Kirstin Atrott
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Isabelle Frey-Wagner
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Michael Fried
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Declan F. McCole
- Division of Biomedical Sciences, University of California Riverside, Riverside, California, United States of America
| | - Gerhard Rogler
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
- Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Michael Scharl
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
- Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
- * E-mail:
| |
Collapse
|
246
|
Putoczki TL, Thiem S, Loving A, Busuttil RA, Wilson NJ, Ziegler PK, Nguyen PM, Preaudet A, Farid R, Edwards KM, Boglev Y, Luwor RB, Jarnicki A, Horst D, Boussioutas A, Heath JK, Sieber OM, Pleines I, Kile BT, Nash A, Greten FR, McKenzie BS, Ernst M. Interleukin-11 is the dominant IL-6 family cytokine during gastrointestinal tumorigenesis and can be targeted therapeutically. Cancer Cell 2013; 24:257-71. [PMID: 23948300 DOI: 10.1016/j.ccr.2013.06.017] [Citation(s) in RCA: 319] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Revised: 05/17/2013] [Accepted: 06/27/2013] [Indexed: 02/08/2023]
Abstract
Among the cytokines linked to inflammation-associated cancer, interleukin (IL)-6 drives many of the cancer "hallmarks" through downstream activation of the gp130/STAT3 signaling pathway. However, we show that the related cytokine IL-11 has a stronger correlation with elevated STAT3 activation in human gastrointestinal cancers. Using genetic mouse models, we reveal that IL-11 has a more prominent role compared to IL-6 during the progression of sporadic and inflammation-associated colon and gastric cancers. Accordingly, in these models and in human tumor cell line xenograft models, pharmacologic inhibition of IL-11 signaling alleviated STAT3 activation, suppressed tumor cell proliferation, and reduced the invasive capacity and growth of tumors. Our results identify IL-11 signaling as a potential therapeutic target for the treatment of gastrointestinal cancers.
Collapse
Affiliation(s)
- Tracy L Putoczki
- Ludwig Institute for Cancer Research, Melbourne, VIC 3050, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
247
|
Chakilam S, Gandesiri M, Rau TT, Agaimy A, Vijayalakshmi M, Ivanovska J, Wirtz RM, Schulze-Luehrmann J, Benderska N, Wittkopf N, Chellappan A, Ruemmele P, Vieth M, Rave-Fränk M, Christiansen H, Hartmann A, Neufert C, Atreya R, Becker C, Steinberg P, Schneider-Stock R. Death-associated protein kinase controls STAT3 activity in intestinal epithelial cells. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 182:1005-20. [PMID: 23438478 DOI: 10.1016/j.ajpath.2012.11.026] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Revised: 10/30/2012] [Accepted: 11/15/2012] [Indexed: 12/20/2022]
Abstract
The TNF-IL-6-STAT3 pathway plays a crucial role in promoting ulcerative colitis-associated carcinoma (UCC). To date, the negative regulation of STAT3 is poorly understood. Interestingly, intestinal epithelial cells of UCC in comparison to ulcerative colitis show high expression levels of anti-inflammatory death-associated protein kinase (DAPK) and low levels of pSTAT3. Accordingly, epithelial DAPK expression was enhanced in STAT3(IEC-KO) mice. To unravel a possible regulatory mechanism, we used an in vitro TNF-treated intestinal epithelial cell model. We identified a new function of DAPK in suppressing TNF-induced STAT3 activation as DAPK siRNA knockdown and treatment with a DAPK inhibitor potentiated STAT3 activation, IL-6 mRNA expression, and secretion. DAPK attenuated STAT3 activity directly by physical interaction shown in three-dimensional structural modeling. This model suggests that DAPK-induced conformational changes in the STAT3 dimer masked its nuclear localization signal. Alternatively, pharmacological inactivation of STAT3 led to an increase in DAPK mRNA and protein levels. Chromatin immunoprecipitation showed that STAT3 restricted DAPK expression by promoter binding, thereby reinforcing its own activation by inducing IL-6. This novel negative regulation principle might balance TNF-induced inflammation and seems to play an important role in the inflammation-associated transformation process as confirmed in an AOM+DSS colon carcinogenesis mouse model. DAPK as a negative regulator of STAT3 emerges as therapeutic option in the treatment of ulcerative colitis and UCC.
Collapse
Affiliation(s)
- Saritha Chakilam
- Experimental Tumor Pathology, University of Erlangen-Nuremberg, Erlangen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
248
|
Jirkof P, Leucht K, Cesarovic N, Caj M, Nicholls F, Rogler G, Arras M, Hausmann M. Burrowing is a sensitive behavioural assay for monitoring general wellbeing during dextran sulfate sodium colitis in laboratory mice. Lab Anim 2013; 47:274-83. [PMID: 23828853 DOI: 10.1177/0023677213493409] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
An impaired intestinal epithelial barrier is thought to be a major factor in the pathogenesis of human inflammatory bowel disease (IBD). IBD is frequently investigated by inducing a damaged barrier in murine models of colitis. This can be done by feeding mice with dextran sulfate sodium (DSS) polymers in their drinking water. Refinement measures should focus on alleviating unnecessary suffering during this probably painful condition. Appropriate parameters are needed to decide when to terminate the experiments. Our aim was to investigate whether a change in burrowing behaviour is a sensitive measure of animal welfare in murine models of colitis. Acute colitis was induced in C57BL/6 mice with 2.0% DSS over nine days. The burrowing test is based on the species-typical behaviour of mice to spontaneously displace items from tubes within their home cage. As a burrowing apparatus, a water bottle (250 mL, 150 mm length, 55 mm diameter) filled with 138-142 g of pellets of the animal's diet was used. The presence of intestinal inflammation as a result of acute DSS-induced colitis was confirmed by a decrease in body weight, colon length and an increase of murine endoscopic index of colitis severity, histological score and spleen weight in the group receiving DSS as compared with the control group. An onset of intestinal inflammation correlated with a significant decrease in burrowing behaviour (P < 0.05). Altered adrenal gland histology indicated stress as a result of acute colitis. Our findings provide evidence that changes of spontaneous burrowing behaviour correlate with the onset of inflammation in acute DSS-induced colitis.
Collapse
Affiliation(s)
- P Jirkof
- Institute of Laboratory Animal Science, University of Zurich, Zurich, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
249
|
Dothel G, Vasina V, Barbara G, De Ponti F. Animal models of chemically induced intestinal inflammation: predictivity and ethical issues. Pharmacol Ther 2013; 139:71-86. [PMID: 23563278 DOI: 10.1016/j.pharmthera.2013.04.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Accepted: 03/19/2013] [Indexed: 02/08/2023]
Abstract
The debate about the ethical and scientific issues regarding the use of animals in research is mainly focused on these questions: a) whether preclinical studies in animals are still ethically acceptable; b) whether it is possible to establish more soundly their predictivity; c) what measures should be taken to reduce the clinical attrition often due to biased preclinical assessment of potential efficacy of new drugs. This review aims at a critical revision of animal models of chemically induced intestinal inflammation in drug development. These models, notwithstanding differences among species, still represent a major source of information about biological systems and can have undisputable translational value, provided that appropriate measures are taken to ensure that experiments are both scientifically and ethically justified. These measures include: a) more stringent application to preclinical experiments of standards used in clinical studies (such as sample size, randomization, inclusion/exclusion criteria, blinding); b) selection of the animal model after careful pathophysiological scrutiny bearing in mind inherent limitations of each model (e.g. acute self-limiting vs chronic disease, animal species, role of the intestinal immune system and microbiome); and c) experimental design duly considering the specific pharmacological profile of each agent to be screened (such as bioavailability, route of administration, full consideration of the pharmacological spectrum). In this perspective, the new European legislation is an opportunity to fully apply these standards so that in vivo animal models can provide an invaluable mean to study complex physiological and biochemical interactions, which cannot be completely simulated in silico and/or in vitro.
Collapse
Affiliation(s)
- Giovanni Dothel
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | | | | | | |
Collapse
|
250
|
Miyamoto S, Nakanishi M, Rosenberg DW. Suppression of colon carcinogenesis by targeting Notch signaling. Carcinogenesis 2013; 34:2415-23. [PMID: 23729655 DOI: 10.1093/carcin/bgt191] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Recent studies have shown that aberrant Notch signaling contributes to the pathogenesis of colorectal cancer (CRC). However, the potential therapeutic benefits of Notch pathway inhibitors, including gamma-secretase inhibitors (GSIs) on colon carcinogenesis are still unclear. In this study, the effects of the GSI, N-[N-3,5-difluorophenacetyl]-L-alanyl-S-phenylglycine methyl ester (DAPM) on colon carcinogenesis were investigated. In vitro, DAPM suppressed cell proliferation and induced the expression of Krüppel-like factor 4 (KLF4) and p21 in human colon cancer cells. Interestingly, p21-null HCT 116 cells were largely resistant to the suppressive effects of DAPM on cell proliferation compared with the parental cells. To investigate the effects of DAPM in vivo, colonoscopy was performed to establish the presence of colon tumors 9 weeks after azoxymethane treatment. After tumors were identified, mice were injected intraperitoneally every other day with either DAPM or vehicle for 4 weeks. The frequency of both large (>4mm) and small (<1mm) colon tumors was significantly reduced by DAPM treatment. Colon tumors in the DAPM-treated mice displayed increased levels of KLF4 and p21, accompanied by reduced Ki-67 staining compared with controls. Notably, in human colon tumor biopsies, KLF4 and p21 expressions were present within hyperplastic polyps, but the levels of both proteins were markedly reduced in tubular adenomas. Our results suggest that inhibition of Notch signaling by DAPM provides a potential chemopreventive strategy for patients with tubular adenomas, in part via activation of the KLF4-p21 axis.
Collapse
Affiliation(s)
- Shingo Miyamoto
- Center for Molecular Medicine, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT 06030-3103, USA
| | | | | |
Collapse
|