151
|
Gopal A, Chidambaram IS, Devaraj N, Devaraj H. Shigella dysenteriae infection activates proinflammatory response through β-catenin/NF-κB signaling pathway. PLoS One 2017; 12:e0174943. [PMID: 28430783 PMCID: PMC5400225 DOI: 10.1371/journal.pone.0174943] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 03/17/2017] [Indexed: 11/18/2022] Open
Abstract
Shigella dysenteriae (S.dysenteriae) the causative agent of bacillary dysentery invades the human colonic epithelium resulting in severe intestinal inflammatory response and epithelial destruction. However, the mechanism by which S.dysenteriae infection regulates proinflammatory cytokines during intestinal inflammation is still obscure. In this study, we evaluated whether the interaction of β-catenin and NF-κB regulates proinflammatory cytokines TNF-α and IL-8 by modulating GSK-3β activity during S.dysenteriae infection in rat ileal loop model. Here we demonstrated that S.dysenteriae infection stimulate β-catenin degradation which in turn decreased the association between NF-κB and β-catenin. Also, we showed that S.dysenteriae infection increased GSK-3β kinase activity which in turn phosphorylates β-catenin for its degradation by ubiquitination and upregulates IL-8 through NF-κB activation thereby leading to inflammation. Thus these findings revealed the role of β-catenin/ NF-κB and GSK-3β in modulating the inflammatory response during bacterial infection and also showed that β-catenin acts as a critical regulator of inflammation.
Collapse
Affiliation(s)
- Ashidha Gopal
- Unit of Biochemistry, Department of Zoology, University of Madras, Chennai, Tamilnadu, India
| | - Iyer Soumya Chidambaram
- Unit of Biochemistry, Department of Zoology, University of Madras, Chennai, Tamilnadu, India
| | - Niranjali Devaraj
- Department of Biochemistry, University of Madras, Chennai, Tamilnadu, India
| | - Halagowder Devaraj
- Unit of Biochemistry, Department of Zoology, University of Madras, Chennai, Tamilnadu, India
- * E-mail:
| |
Collapse
|
152
|
Morvay PL, Baes M, Van Veldhoven PP. Differential activities of peroxisomes along the mouse intestinal epithelium. Cell Biochem Funct 2017; 35:144-155. [PMID: 28370438 DOI: 10.1002/cbf.3255] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 01/10/2017] [Accepted: 01/26/2017] [Indexed: 02/01/2023]
Abstract
The presence of peroxisomes in mammalian intestine has been revealed formerly by catalase staining combined with electron microscopy. Despite the central role of intestine in lipid uptake and the established importance of peroxisomes in different lipid-related pathways, few data are available on the physiological role of peroxisomes in intestinal metabolism, more specifically, α-, β-oxidation, and etherlipid synthesis. Hence, the peroxisomal compartment was analyzed in more detail in mouse intestine. On the basis of immunohistochemistry, the organelles are mainly confined to the epithelial cells. The expression of the classical peroxisome marker catalase was highest in the proximal part of jejunum and decreased along the tract. PEX14 showed a similar profile, but was still substantial expressed in large intestinal epithelium. Immunoblotting of epithelial cells, isolated from the different segments, showed also such gradient for some enzymes, ie, catalase, ACOX1, and D-specific multifunctional protein 2, and for the ABCD1 transporter, being high in small and low or absent in large intestine. Other peroxisomal enzymes (PHYH, HACL1, and ACAA1), the ABCD2 and ABCD3 transporters, and peroxins PEX13 and PEX14, however, did not follow this pattern, displaying rather constant signals throughout the intestinal epithelium. The small intestine displayed the highest peroxisomal β-oxidation activity and is particularly active on dicarboxylic acids. Etherlipid synthesis was high in the large intestine, and colonic cells had the highest content of plasmalogens. Overall, these data suggest that peroxisomes exert different functions according to the intestinal segment.
Collapse
Affiliation(s)
- Petruta L Morvay
- Lipid Biochemistry and Protein Interactions (LIPIT), KU Leuven, Leuven, Belgium
| | | | | |
Collapse
|
153
|
Loss of the EPH receptor B6 contributes to colorectal cancer metastasis. Sci Rep 2017; 7:43702. [PMID: 28262839 PMCID: PMC5337985 DOI: 10.1038/srep43702] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 01/26/2017] [Indexed: 01/03/2023] Open
Abstract
Although deregulation of EPHB signaling has been shown to be an important step in colorectal tumorigenesis, the role of EPHB6 in this process has not been investigated. We found here that manipulation of EPHB6 levels in colon cancer cell lines has no effect on their motility and growth on a solid substrate, soft agar or in a xenograft mouse model. We then used an EphB6 knockout mouse model to show that EphB6 inactivation does not efficiently initiate tumorigenesis in the intestinal tract. In addition, when intestinal tumors are initiated genetically or pharmacologically in EphB6+/+ and EphB6−/− mice, no differences were observed in animal survival, tumor multiplicity, size or histology, and proliferation of intestinal epithelial cells or tumor cells. However, reintroduction of EPHB6 into colon cancer cells significantly reduced the number of lung metastasis after tail-vein injection in immunodeficient mice, while EPHB6 knockdown in EPHB6-expressing cells increased their metastatic spread. Consistently, although EPHB6 protein expression in a series of 130 primary colorectal tumors was not associated with patient survival, EPHB6 expression was significantly lower in lymph node metastases compared to primary tumors. Our results indicate that the loss of EPHB6 contributes to the metastatic process of colorectal cancer.
Collapse
|
154
|
Structural environment built by AKAP12+ colon mesenchymal cells drives M2 macrophages during inflammation recovery. Sci Rep 2017; 7:42723. [PMID: 28205544 PMCID: PMC5311874 DOI: 10.1038/srep42723] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 01/13/2017] [Indexed: 01/22/2023] Open
Abstract
Macrophages exhibit phenotypic plasticity, as they have the ability to switch their functional phenotypes during inflammation and recovery. Simultaneously, the mechanical environment actively changes. However, how these dynamic alterations affect the macrophage phenotype is unknown. Here, we observed that the extracellular matrix (ECM) constructed by AKAP12+ colon mesenchymal cells (CMCs) generated M2 macrophages by regulating their shape during recovery. Notably, rounded macrophages were present in the linear and loose ECM of inflamed colons and polarized to the M1 phenotype. In contrast, ramified macrophages emerged in the contracted ECM of recovering colons and mainly expressed M2 macrophage markers. These contracted structures were not observed in the inflamed colons of AKAP12 knockout (KO) mice. Consequently, the proportion of M2 macrophages in inflamed colons was lower in AKAP12 KO mice than in WT mice. In addition, clinical symptoms and histological damage were more severe in AKAP12 KO mice than in WT mice. In experimentally remodeled collagen gels, WT CMCs drove the formation of a more compacted structure than AKAP12 KO CMCs, which promoted the polarization of macrophages toward an M2 phenotype. These results demonstrated that tissue contraction during recovery provides macrophages with the physical cues that drive M2 polarization.
Collapse
|
155
|
Hua G, Wang C, Pan Y, Zeng Z, Lee SG, Martin ML, Haimovitz-Friedman A, Fuks Z, Paty PB, Kolesnick R. Distinct Levels of Radioresistance in Lgr5 + Colonic Epithelial Stem Cells versus Lgr5 + Small Intestinal Stem Cells. Cancer Res 2017; 77:2124-2133. [PMID: 28202528 DOI: 10.1158/0008-5472.can-15-2870] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 01/19/2017] [Accepted: 01/19/2017] [Indexed: 12/31/2022]
Abstract
Although small and large intestines possess seemingly similar Wnt-driven leucine-rich repeat-containing G protein-coupled receptor 5 (Lgr5)+ adult epithelial stem cells, we report here that the two organs exhibit distinct mechanisms of tissue response to ionizing radiation. Employing Lgr5-lacZ transgenic mice and Lgr5 in situ hybridization, we found colonic epithelial stem cells (CESC) markedly more radioresistant in vivo than small intestinal crypt base columnar stem cells (CBC; D0 = 6.0 ± 0.3 Gy vs. 1.3 ± 0.1, respectively; P < 0.01). Accordingly, CESCs survived 30 Gy exposure, while CBCs were completely depleted after 15 Gy. EdU incorporation studies indicated that after 19 Gy, CBCs exited growth arrest at 12 hours, resuming normal mitotic activity despite 60% of this population displaying residual γH2AX foci, indicative of persistent unrepaired DNA damage. Checkpoint recovery before complete double-strand break (DSB) repair represents the sine qua non of a newly defined potentially lethal pathophysiology termed checkpoint adaptation. In the small intestinal mucosa, checkpoint adaptation resulted in CBCs succumbing to an 8-fold increase in the incidence of highly lethal chromosomal aberrations and mitotic catastrophe by 48 hours postradiation. In contrast, Lgr5+ CESCs displayed delayed checkpoint recovery at 48 hours post-19 Gy, coordinated with complete DSB repair and regeneration of colonic mucosa originating, at least in part, from surviving CESCs. The discovery that small intestinal CBCs succumb to checkpoint adaptation is the first demonstration that this aberrant cell-cycle response may drive mammalian tissue radiosensitivity. Cancer Res; 77(8); 2124-33. ©2017 AACR.
Collapse
Affiliation(s)
- Guoqiang Hua
- Institute of Radiation Medicine, Fudan University, Shanghai, China. .,Laboratory of Signal Transduction, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Chu Wang
- Institute of Radiation Medicine, Fudan University, Shanghai, China
| | - Yan Pan
- Institute of Radiation Medicine, Fudan University, Shanghai, China.,Laboratory of Signal Transduction, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Zhaoshi Zeng
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sang Gyu Lee
- Laboratory of Signal Transduction, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Maria Laura Martin
- Laboratory of Signal Transduction, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Zvi Fuks
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Philip B Paty
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Richard Kolesnick
- Laboratory of Signal Transduction, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
156
|
Andretta E, Cartón-García F, Martínez-Barriocanal Á, de Marcondes PG, Jimenez-Flores LM, Macaya I, Bazzocco S, Bilic J, Rodrigues P, Nieto R, Landolfi S, Ramon y Cajal S, Schwartz S, Brown A, Dopeso H, Arango D. Investigation of the role of tyrosine kinase receptor EPHA3 in colorectal cancer. Sci Rep 2017; 7:41576. [PMID: 28169277 PMCID: PMC5294649 DOI: 10.1038/srep41576] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 12/22/2016] [Indexed: 12/23/2022] Open
Abstract
EPH signaling deregulation has been shown to be important for colorectal carcinogenesis and genome-wide sequencing efforts have identified EPHA3 as one of the most frequently mutated genes in these tumors. However, the role of EPHA3 in colorectal cancer has not been thoroughly investigated. We show here that ectopic expression of wild type EPHA3 in colon cancer cells did not affect their growth, motility/invasion or metastatic potential in vivo. Moreover, overexpression of mutant EPHA3 or deletion of the endogenous mutant EPHA3 in colon cancer cells did not affect their growth or motility. EPHA3 inactivation in mice did not initiate the tumorigenic process in their intestine, and had no effects on tumor size/multiplicity after tumor initiation either genetically or pharmacologically. In addition, immunohistochemical analysis of EPHA3 tumor levels did not reveal associations with survival or clinicopathological features of colorectal cancer patients. In conclusion, we show that EPHA3 does not play a major role in colorectal tumorigenesis. These results significantly contribute to our understanding of the role of EPH signaling during colorectal carcinogenesis, and highlighting the need for detailed functional studies to confirm the relevance of putative cancer driver genes identified in sequencing efforts of the cancer genome.
Collapse
Affiliation(s)
- Elena Andretta
- Group of Biomedical Research in Digestive Tract Tumors, CIBBIM-Nanomedicine, Vall d’Hebron University Hospital, Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d’Hebron, 119-129, 08035 Barcelona, Spain
| | - Fernando Cartón-García
- Group of Biomedical Research in Digestive Tract Tumors, CIBBIM-Nanomedicine, Vall d’Hebron University Hospital, Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d’Hebron, 119-129, 08035 Barcelona, Spain
| | - Águeda Martínez-Barriocanal
- Group of Biomedical Research in Digestive Tract Tumors, CIBBIM-Nanomedicine, Vall d’Hebron University Hospital, Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d’Hebron, 119-129, 08035 Barcelona, Spain
| | - Priscila Guimarães de Marcondes
- Group of Biomedical Research in Digestive Tract Tumors, CIBBIM-Nanomedicine, Vall d’Hebron University Hospital, Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d’Hebron, 119-129, 08035 Barcelona, Spain
| | - Lizbeth M. Jimenez-Flores
- Group of Biomedical Research in Digestive Tract Tumors, CIBBIM-Nanomedicine, Vall d’Hebron University Hospital, Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d’Hebron, 119-129, 08035 Barcelona, Spain
| | - Irati Macaya
- Group of Biomedical Research in Digestive Tract Tumors, CIBBIM-Nanomedicine, Vall d’Hebron University Hospital, Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d’Hebron, 119-129, 08035 Barcelona, Spain
| | - Sarah Bazzocco
- Group of Biomedical Research in Digestive Tract Tumors, CIBBIM-Nanomedicine, Vall d’Hebron University Hospital, Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d’Hebron, 119-129, 08035 Barcelona, Spain
| | - Josipa Bilic
- Group of Biomedical Research in Digestive Tract Tumors, CIBBIM-Nanomedicine, Vall d’Hebron University Hospital, Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d’Hebron, 119-129, 08035 Barcelona, Spain
| | - Paulo Rodrigues
- Group of Biomedical Research in Digestive Tract Tumors, CIBBIM-Nanomedicine, Vall d’Hebron University Hospital, Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d’Hebron, 119-129, 08035 Barcelona, Spain
| | - Rocio Nieto
- Group of Biomedical Research in Digestive Tract Tumors, CIBBIM-Nanomedicine, Vall d’Hebron University Hospital, Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d’Hebron, 119-129, 08035 Barcelona, Spain
| | | | | | - Simo Schwartz
- Group of Drug Delivery and Targeting, CIBBIM-Nanomedicine, Vall d’Hebron University Hospital, Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - Arthur Brown
- Robarts Research Institute, London, Ontario, Canada
| | - Higinio Dopeso
- Group of Biomedical Research in Digestive Tract Tumors, CIBBIM-Nanomedicine, Vall d’Hebron University Hospital, Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d’Hebron, 119-129, 08035 Barcelona, Spain
| | - Diego Arango
- Group of Biomedical Research in Digestive Tract Tumors, CIBBIM-Nanomedicine, Vall d’Hebron University Hospital, Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d’Hebron, 119-129, 08035 Barcelona, Spain
| |
Collapse
|
157
|
Du X, Chen W, Wang Y, Chen C, Guo L, Ju R, Li J, Zhang D, Zhu L, Ye C. Therapeutic efficacy of carboxyamidotriazole on 2,4,6-trinitrobenzene sulfonic acid-induced colitis model is associated with the inhibition of NLRP3 inflammasome and NF-κB activation. Int Immunopharmacol 2017; 45:16-25. [PMID: 28152446 DOI: 10.1016/j.intimp.2017.01.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 12/07/2016] [Accepted: 01/10/2017] [Indexed: 01/08/2023]
Abstract
Excess proinflammatory cytokines owing to the activation of NF-κB and NLRP3 inflammasome play the key role in inflammatory bowel disease (IBD). Previously, we reported the anti-inflammatory activity of carboxyamidotriazole (CAI) resulting from decreasing cytokines. Therefore, we investigated the therapeutic effects of CAI in 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced rat colitis and the involvement of CAI action with NLRP3 inflammasome and NF-κB pathway. CAI was orally administered to TNBS-induced colitis rat. The severity of colitis was assessed, and NLRP3 inflammasome, NF-κB pathway and cytokines were determined. Our results showed that CAI significantly reduced weight loss and disease activity index (DAI) scores in colitis rats and alleviated the colonic macroscopic signs and pathological damage. In addition, the intestinal inflammatory markers and permeability index were markedly ameliorated by CAI treatment. The decreased levels of tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, IL-6, IL-18 were also detected in the colon tissues of CAI-treated colitis rats. Moreover, the activation of NLRP3 inflammasome in inflamed colon was significantly suppressed by showing an obvious reduction in the NLRP3 and activated caspase-1 levels. Furthermore, CAI reduced NF-κB p65 expression and IκBα phosphorylation and degradation in colitis rats. Therefore, CAI attenuates TNBS-induced colitis, which may be attributed to its inhibition of NLRP3 inflammasome and NF-κB activation, and down-regulation of proinflammatory cytokines. These results provide further understanding of the intestinal anti-inflammatory effect of CAI and highlight it as a potential drug for the treatment of IBD.
Collapse
Affiliation(s)
- Xiaowan Du
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Wei Chen
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Yufeng Wang
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Chen Chen
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Lei Guo
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Rui Ju
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Juan Li
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Dechang Zhang
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Lei Zhu
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China.
| | - Caiying Ye
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China.
| |
Collapse
|
158
|
Nguyen TT, Savory JGA, Brooke-Bisschop T, Ringuette R, Foley T, Hess BL, Mulatz KJ, Trinkle-Mulcahy L, Lohnes D. Cdx2 Regulates Gene Expression through Recruitment of Brg1-associated Switch-Sucrose Non-fermentable (SWI-SNF) Chromatin Remodeling Activity. J Biol Chem 2017; 292:3389-3399. [PMID: 28082674 DOI: 10.1074/jbc.m116.752774] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 01/10/2017] [Indexed: 12/22/2022] Open
Abstract
The packaging of genomic DNA into nucleosomes creates a barrier to transcription that can be relieved through ATP-dependent chromatin remodeling via complexes such as the switch-sucrose non-fermentable (SWI-SNF) chromatin remodeling complex. The SWI-SNF complex remodels chromatin via conformational or positional changes of nucleosomes, thereby altering the access of transcriptional machinery to target genes. The SWI-SNF complex has limited ability to bind to sequence-specific elements, and, therefore, its recruitment to target loci is believed to require interaction with DNA-associated transcription factors. The Cdx family of homeodomain transcript ion factors (Cdx1, Cdx2, and Cdx4) are essential for a number of developmental programs in the mouse. Cdx1 and Cdx2 also regulate intestinal homeostasis throughout life. Although a number of Cdx target genes have been identified, the basis by which Cdx members impact their transcription is poorly understood. We have found that Cdx members interact with the SWI-SNF complex and make direct contact with Brg1, a catalytic member of SWI-SNF. Both Cdx2 and Brg1 co-occupy a number of Cdx target genes, and both factors are necessary for transcriptional regulation of such targets. Finally, Cdx2 and Brg1 occupancy occurs coincident with chromatin remodeling at some of these loci. Taken together, our findings suggest that Cdx transcription factors regulate target gene expression, in part, through recruitment of Brg1-associated SWI-SNF chromatin remodeling activity.
Collapse
Affiliation(s)
- Thinh T Nguyen
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Joanne G A Savory
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Travis Brooke-Bisschop
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Randy Ringuette
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Tanya Foley
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Bradley L Hess
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Kirk J Mulatz
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Laura Trinkle-Mulcahy
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - David Lohnes
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.
| |
Collapse
|
159
|
Kuper CF, Wijnands MVW, Zander SAL. Mucosa-Associated Lymphoid Tissues. IMMUNOPATHOLOGY IN TOXICOLOGY AND DRUG DEVELOPMENT 2017. [DOI: 10.1007/978-3-319-47385-7_4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
160
|
Hasebe T, Matsukawa J, Ringus D, Miyoshi J, Hart J, Kaneko A, Yamamoto M, Kono T, Fujiya M, Kohgo Y, Wang CZ, Yuan CS, Bissonnette M, Musch MW, Chang EB. Daikenchuto (TU-100) Suppresses Tumor Development in the Azoxymethane and APC min/+ Mouse Models of Experimental Colon Cancer. Phytother Res 2017; 31:90-99. [PMID: 27730672 PMCID: PMC5590753 DOI: 10.1002/ptr.5735] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 08/26/2016] [Accepted: 09/18/2016] [Indexed: 02/06/2023]
Abstract
Chemopreventative properties of traditional medicines and underlying mechanisms of action are incompletely investigated. This study demonstrates that dietary daikenchuto (TU-100), comprised of ginger, ginseng, and Japanese pepper effectively suppresses intestinal tumor development and progression in the azoxymethane (AOM) and APCmin/+ mouse models. For the AOM model, TU-100 was provided after the first of six biweekly AOM injections. Mice were sacrificed at 30 weeks. APCmin/+ mice were fed diet without or with TU-100 starting at 6 weeks, and sacrificed at 24 weeks. In both models, dietary TU-100 decreased tumor size. In APC min/+ mice, the number of small intestinal tumors was significantly decreased. In the AOM model, both TU-100 and Japanese ginseng decreased colon tumor numbers. Decreased Ki-67 and β-catenin immunostaining and activation of numerous transduction pathways involved in tumor initiation and progression were observed. EGF receptor expression and stimulation/phosphorylation in vitro were investigated in C2BBe1 cells. TU-100, ginger, and 6-gingerol suppressed EGF receptor induced Akt activation. TU-100 and ginseng and to a lesser extent ginger or 6-gingerol inhibited EGF ERK1/2 activation. TU-100 and some of its components and metabolites of these components inhibit tumor progression in two mouse models of colon cancer by blocking downstream pathways of EGF receptor activation. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Takumu Hasebe
- Department of Medicine, Knapp Center for Biomedical Discovery, University of Chicago, Chicago, IL, USA
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Jun Matsukawa
- Department of Medicine, Knapp Center for Biomedical Discovery, University of Chicago, Chicago, IL, USA
| | - Daina Ringus
- Department of Medicine, Knapp Center for Biomedical Discovery, University of Chicago, Chicago, IL, USA
| | - Jun Miyoshi
- Department of Medicine, Knapp Center for Biomedical Discovery, University of Chicago, Chicago, IL, USA
| | - John Hart
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Atsushi Kaneko
- Tsumura Research Laboratories, Tsumura and Co., Ami, Ibaraki, Japan
| | | | - Toru Kono
- Center for Clinical and Biomedical Research, Sapporo Higashi Tokushukai Hospital, Sapporo, Hokkaido, Japan
- Division of Gastroenterologic and General Surgery, Department of Surgery, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Mikihiro Fujiya
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Yutaka Kohgo
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Chong-Zi Wang
- Tang Center for Herbal Medicine Research, Department of Anesthesia and Critical Care, University of Chicago, Chicago, IL, USA
| | - Chun-Su Yuan
- Tang Center for Herbal Medicine Research, Department of Anesthesia and Critical Care, University of Chicago, Chicago, IL, USA
| | - Marc Bissonnette
- Department of Medicine, Knapp Center for Biomedical Discovery, University of Chicago, Chicago, IL, USA
| | - Mark W. Musch
- Department of Medicine, Knapp Center for Biomedical Discovery, University of Chicago, Chicago, IL, USA
| | - Eugene B. Chang
- Department of Medicine, Knapp Center for Biomedical Discovery, University of Chicago, Chicago, IL, USA
| |
Collapse
|
161
|
Fiette L, Slaoui M, Bauchet AL. Procedures of Necropsy and Tissue Sampling. Methods Mol Biol 2017; 1641:71-100. [PMID: 28748458 DOI: 10.1007/978-1-4939-7172-5_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Necropsy is a major step of most studies using laboratory animals. During necropsy, tissue and organ noticeable grossly changes should be recorded and critical tissue samples may be stored for the subsequent evaluation. It is therefore important that the personnel in charge of this key experimental step to be adequately trained and aware of the study endpoints.
Collapse
Affiliation(s)
| | - Mohamed Slaoui
- Preclinical Safety, Sanofi R&D, 13, Quai Jules Guesde, 94403, Vitry-sur-Seine, France.
| | - Anne-Laure Bauchet
- Translational Medicine and Early Development, Biomarkers and Clinical Bioanalyses, Sanofi R&D, 13, Quai Jules Guesde, 94403, Vitry-sur-Seine, France
| |
Collapse
|
162
|
Slaoui M, Bauchet AL, Fiette L. Tissue Sampling and Processing for Histopathology Evaluation. Methods Mol Biol 2017; 1641:101-114. [PMID: 28748459 DOI: 10.1007/978-1-4939-7172-5_4] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Histological procedures aim at providing good-quality sections that can be used for a light microscopic evaluation of tissue. These are applicable to identify either spontaneous or diseases-induced changes. Routinely, tissues are fixed with neutral formalin 10%, embedded in paraffin, and manually sectioned with a microtome to obtain 4-5 μm thick paraffin sections. Dewaxed sections are then stained with HE&S (hematoxylin-eosin and saffron) or can be used for other purposes (special stains, immunohistochemistry, in situ hybridization, etc.). During this processing, many steps and procedures are critical to ensure standard and interpretable sections. This chapter provides key recommendations to efficiently achieve this objective.
Collapse
Affiliation(s)
- Mohamed Slaoui
- Preclinical Safety, Sanofi R&D, 13, Quai Jules Guesde, BP14, 94403, Vitrysur-Seine, France.
| | - Anne-Laure Bauchet
- Translational Medicine and Early Development, Biomarkers and Clinical Bioanalysis, Sanofi R&D, Vitry-sur-Seine, France
| | | |
Collapse
|
163
|
Haller S, Nunez V, Havnar C, Safra N, Foreman O. Using the HistoPress embedding tool to process murine colon samples for histopathology. J Histotechnol 2016. [DOI: 10.1080/01478885.2016.1211227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Susan Haller
- Department of Research Pathology, Genentech Inc, South San Francisco, CA, USA
| | - Victor Nunez
- Department of Research Pathology, Genentech Inc, South San Francisco, CA, USA
| | - Charles Havnar
- Department of Research Pathology, Genentech Inc, South San Francisco, CA, USA
| | - Noa Safra
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - Oded Foreman
- Department of Research Pathology, Genentech Inc, South San Francisco, CA, USA
| |
Collapse
|
164
|
van Esch E, de Rijk EP, Buse E, Zöller M, Cline JM. Recommendations for Routine Sampling, Trimming, and Paraffin-Embedding of Female Reproductive Organs, Mammary Gland, and Placenta in the Cynomolgus Monkey. Toxicol Pathol 2016. [DOI: 10.1177/0192623308326152] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In toxicity studies, the nonhuman primate is often the species of choice to evaluate the toxicologic potential of chemicals and drugs. Especially in the case of effects on female reproductive organs and mammary glands, other animal species are less predictive for man. To enable reliable histopathologic interpretation allowing a solid safety assessment, it is a prerequisite to obtain material of consistently high quality. Standardization of autopsy techniques, tissue sampling, and fixation and staining procedures will help significantly to obtain the quality that is needed. For this purpose, a detailed description of the procedures from necropsy to microscopic slide preparation of the female reproductive organs of the cynomolgus monkey is given. Procedures to sample and process the placenta are included. These recommendations can be used to achieve consistent, high-quality tissue preparations, allowing pathologists to conduct sensitive, accurate, and meaningful evaluations of the study material. Competing Interests: This article was sponsored by Covance Inc. and Schering- Plough. Martina Zölle and Eberhard Buse are employed by Covance Inc. Eveline P. C. T. de Rijk and Eric Van Esch are employed by Schering-Plough. No other competing interests were declared.
Collapse
|
165
|
Mitochondrial function controls intestinal epithelial stemness and proliferation. Nat Commun 2016; 7:13171. [PMID: 27786175 PMCID: PMC5080445 DOI: 10.1038/ncomms13171] [Citation(s) in RCA: 144] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 09/09/2016] [Indexed: 02/07/2023] Open
Abstract
Control of intestinal epithelial stemness is crucial for tissue homeostasis. Disturbances in epithelial function are implicated in inflammatory and neoplastic diseases of the gastrointestinal tract. Here we report that mitochondrial function plays a critical role in maintaining intestinal stemness and homeostasis. Using intestinal epithelial cell (IEC)-specific mouse models, we show that loss of HSP60, a mitochondrial chaperone, activates the mitochondrial unfolded protein response (MT-UPR) and results in mitochondrial dysfunction. HSP60-deficient crypts display loss of stemness and cell proliferation, accompanied by epithelial release of WNT10A and RSPO1. Sporadic failure of Cre-mediated Hsp60 deletion gives rise to hyperproliferative crypt foci originating from OLFM4+ stem cells. These effects are independent of the MT-UPR-associated transcription factor CHOP. In conclusion, compensatory hyperproliferation of HSP60+ escaper stem cells suggests paracrine release of WNT-related factors from HSP60-deficient, functionally impaired IEC to be pivotal in the control of the proliferative capacity of the stem cell niche. It is unclear what role mitochondrial function plays in maintaining intestinal epithelial cell (IEC) homeostasis. Here, the authors deplete a mitochondrial chaperone, heat shock protein 60 (HSP60) in IEC and observe a loss of stemness and cell proliferation, and suggest this is accompanied by a compensatory release of WNT-related factors.
Collapse
|
166
|
Dusaulcy R, Handgraaf S, Skarupelova S, Visentin F, Vesin C, Heddad-Masson M, Reimann F, Gribble F, Philippe J, Gosmain Y. Functional and Molecular Adaptations of Enteroendocrine L-Cells in Male Obese Mice Are Associated With Preservation of Pancreatic α-Cell Function and Prevention of Hyperglycemia. Endocrinology 2016; 157:3832-3843. [PMID: 27547850 PMCID: PMC7228810 DOI: 10.1210/en.2016-1433] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Glucose homeostasis depends on the coordinated secretion of glucagon, insulin, and Glucagon-like peptide (GLP)-1 by pancreas and intestine. Obesity, which is associated with an increased risk of developing insulin resistance and type 2 diabetes, affects the function of these organs. Here, we investigate the functional and molecular adaptations of proglucagon-producing cells in obese mice to better define their involvement in type 2 diabetes development. We used GLU-Venus transgenic male mice specifically expressing Venus fluorochrome in proglucagon-producing cells. Mice were subjected to 16 weeks of low-fat diet or high-fat diet (HFD) and then subdivided by measuring glycated hemoglobin (HbA1c) in 3 groups: low-fat diet mice and I-HFD (glucose-intolerant) mice with similar HbA1c and H-HFD (hyperglycemic) mice, which exhibited higher HbA1c. At 16 weeks, both HFD groups exhibited similar weight gain, hyperinsulinemia, and insulin resistance. However, I-HFD mice exhibited better glucose tolerance compared with H-HFD mice. I-HFD mice displayed functional and molecular adaptations of enteroendocrine L-cells resulting in increased intestinal GLP-1 biosynthesis and release as well as maintained pancreatic α- and β-cell functions. By contrast, H-HFD mice exhibited dysfunctional L, α- and β-cells with increased β- and L-cell numbers. Administration of the GLP-1R antagonist Exendin9-39 in I-HFD mice led to hyperglycemia and alterations of glucagon secretion without changes in insulin secretion. Our results highlight the cross-talk between islet and intestine endocrine cells and indicate that a compensatory adaptation of L-cell function in obesity plays an important role in preserving glucose homeostasis through the control of pancreatic α-cell functions.
Collapse
Affiliation(s)
- Rodolphe Dusaulcy
- Molecular Diabetes Laboratory, Division of Endocrinology, Diabetes, Hypertension and Nutrition; University Hospital/Diabetes Center/University of Geneva Medical School, 1211 Geneva, Switzerland
| | - Sandra Handgraaf
- Molecular Diabetes Laboratory, Division of Endocrinology, Diabetes, Hypertension and Nutrition; University Hospital/Diabetes Center/University of Geneva Medical School, 1211 Geneva, Switzerland
| | - Svetlana Skarupelova
- Molecular Diabetes Laboratory, Division of Endocrinology, Diabetes, Hypertension and Nutrition; University Hospital/Diabetes Center/University of Geneva Medical School, 1211 Geneva, Switzerland
| | - Florian Visentin
- Molecular Diabetes Laboratory, Division of Endocrinology, Diabetes, Hypertension and Nutrition; University Hospital/Diabetes Center/University of Geneva Medical School, 1211 Geneva, Switzerland
| | - Christian Vesin
- Department of Cell Physiology and Metabolism, University of Geneva School of Medicine, 1211 Geneva, Switzerland
| | - Mounia Heddad-Masson
- Molecular Diabetes Laboratory, Division of Endocrinology, Diabetes, Hypertension and Nutrition; University Hospital/Diabetes Center/University of Geneva Medical School, 1211 Geneva, Switzerland
| | - Frank Reimann
- Wellcome Trust/MRC Institute of Metabolic Science and MRC Metabolic Diseases Unit, University of Cambridge, Addenbrooke's Hospital, Cambridge, U.K
| | - Fiona Gribble
- Wellcome Trust/MRC Institute of Metabolic Science and MRC Metabolic Diseases Unit, University of Cambridge, Addenbrooke's Hospital, Cambridge, U.K
| | - Jacques Philippe
- Molecular Diabetes Laboratory, Division of Endocrinology, Diabetes, Hypertension and Nutrition; University Hospital/Diabetes Center/University of Geneva Medical School, 1211 Geneva, Switzerland
| | - Yvan Gosmain
- Molecular Diabetes Laboratory, Division of Endocrinology, Diabetes, Hypertension and Nutrition; University Hospital/Diabetes Center/University of Geneva Medical School, 1211 Geneva, Switzerland
- Address correspondence to: Yvan Gosmain, Molecular Diabetes Laboratory, University Hospital, 1211 Geneva 14, Switzerland, Tel. +41 22 372 42 37 ; Fax. +41 22 372 93 26,
| |
Collapse
|
167
|
Radnai B, Sturm EM, Stančić A, Jandl K, Labocha S, Ferreirós N, Grill M, Hasenoehrl C, Gorkiewicz G, Marsche G, Heinemann Á, Högenauer C, Schicho R. Eosinophils Contribute to Intestinal Inflammation via Chemoattractant Receptor-homologous Molecule Expressed on Th2 Cells, CRTH2, in Experimental Crohn's Disease. J Crohns Colitis 2016; 10:1087-95. [PMID: 26928963 PMCID: PMC4892354 DOI: 10.1093/ecco-jcc/jjw061] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 02/09/2016] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND AIMS Prostaglandin [PG] D2 activates two receptors, DP and CRTH2. Antagonism of CRTH2 has been shown to promote anti-allergic and anti-inflammatory effects. We investigated whether CRTH2 may play a role in Crohn's disease [CD], focusing on eosinophils which are widely present in the inflamed mucosa of CD patients and express both receptors. METHODS Using the 2,4,6-trinitrobenzenesulfonic acid [TNBS]-induced colitis model, involvement of CRTH2 in colitis was investigated by pharmacological antagonism, immunohistochemistry, Western blotting, immunoassay, and leukocyte recruitment. Chemotactic assays were performed with isolated human eosinophils. Biopsies and serum samples of CD patients were examined for presence of CRTH2 and ligands, respectively. RESULTS High amounts of CRTH2-positive cells, including eosinophils, are present in the colonic mucosa of mice with TNBS colitis and in human CD. The CRTH2 antagonist OC-459, but not the DP antagonist MK0524, reduced inflammation scores and decreased TNF-α, IL-1β, and IL-6 as compared with control mice. OC-459 inhibited recruitment of eosinophils into the colon and also inhibited CRTH2-induced chemotaxis of human eosinophils in vitro. Eosinophil-depleted ΔdblGATA knockout mice were less sensitive to TNBS-induced colitis, whereas IL-5 transgenic mice with lifelong eosinophilia were more severely affected than wild types. In addition, we show that serum levels of PGD2 and Δ(12)-PGJ2 were increased in CD patients as compared with control individuals. CONCLUSIONS CRTH2 plays a pro-inflammatory role in TNBS-induced colitis. Eosinophils contribute to the severity of the inflammation, which is improved by a selective CRTH2 antagonist. CRTH2 may, therefore, represent an important target in the pharmacotherapy of CD.
Collapse
Affiliation(s)
- Balázs Radnai
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Austria
| | - Eva M Sturm
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Austria
| | - Angela Stančić
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Austria
| | - Katharina Jandl
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Austria
| | - Sandra Labocha
- Institute of Clinical Pharmacology, Goethe University, Frankfurt/Main, Germany
| | - Nerea Ferreirós
- Institute of Clinical Pharmacology, Goethe University, Frankfurt/Main, Germany
| | - Magdalena Grill
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Austria
| | - Carina Hasenoehrl
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Austria
| | | | - Gunther Marsche
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Austria
| | - Ákos Heinemann
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Austria
| | | | - Rudolf Schicho
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Austria
| |
Collapse
|
168
|
Chiacchiera F, Rossi A, Jammula S, Zanotti M, Pasini D. PRC2 preserves intestinal progenitors and restricts secretory lineage commitment. EMBO J 2016; 35:2301-2314. [PMID: 27585866 DOI: 10.15252/embj.201694550] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 08/10/2016] [Indexed: 11/09/2022] Open
Abstract
Chromatin modifications shape cell heterogeneity by activating and repressing defined sets of genes involved in cell proliferation, differentiation and development. Polycomb-repressive complexes (PRCs) act synergistically during development and differentiation by maintaining transcriptional repression of common genes. PRC2 exerts this activity by catalysing H3K27 trimethylation. Here, we show that in the intestinal epithelium PRC2 is required to sustain progenitor cell proliferation and the correct balance between secretory and absorptive lineage differentiation programs. Using genetic models, we show that PRC2 activity is largely dispensable for intestinal stem cell maintenance but is strictly required for radiation-induced regeneration by preventing Cdkn2a transcription. Combining these models with genomewide molecular analysis, we further demonstrate that preferential accumulation of secretory cells does not result from impaired proliferation of progenitor cells induced by Cdkn2a activation but rather from direct regulation of transcription factors responsible for secretory lineage commitment. Overall, our data uncover a dual role of PRC2 in intestinal homeostasis highlighting the importance of this repressive layer in controlling cell plasticity and lineage choices in adult tissues.
Collapse
Affiliation(s)
- Fulvio Chiacchiera
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Alessandra Rossi
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - SriGanesh Jammula
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Marika Zanotti
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Diego Pasini
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| |
Collapse
|
169
|
Bernier-Latmani J, Petrova TV. High-resolution 3D analysis of mouse small-intestinal stroma. Nat Protoc 2016; 11:1617-29. [PMID: 27560169 DOI: 10.1038/nprot.2016.092] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Here we detail a protocol for whole-mount immunostaining of mouse small-intestinal villi that can be used to generate high-resolution 3D images of all gut cell types, including blood and lymphatic vessel cells, neurons, smooth muscle cells, fibroblasts and immune cells. The procedure describes perfusion, fixation, dissection, immunostaining, mounting, clearing, confocal imaging and quantification, using intestinal vasculature as an example. As intestinal epithelial cells prevent visualization with some antibodies, we also provide an optional protocol to remove these cells before fixation. In contrast to alternative current techniques, our protocol enables the entire villus to be visualized with increased spatial resolution of cell location, morphology and cell-cell interactions, thus allowing for easy quantification of phenotypes. The technique, which takes 7 d from mouse dissection to microscopic examination, will be useful for researchers who are interested in most aspects of intestinal biology, including mucosal immunology, infection, nutrition, cancer biology and intestinal microbiota.
Collapse
Affiliation(s)
- Jeremiah Bernier-Latmani
- Department of Fundamental Oncology, Ludwig Institute for Cancer Research and Institute of Pathology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Tatiana V Petrova
- Department of Fundamental Oncology, Ludwig Institute for Cancer Research and Institute of Pathology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.,Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences. Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
170
|
Molendijk I, Barnhoorn MC, de Jonge-Muller ESM, Mieremet-Ooms MAC, van der Reijden JJ, van der Helm D, Hommes DW, van der Meulen-de Jong AE, Verspaget HW. Intraluminal Injection of Mesenchymal Stromal Cells in Spheroids Attenuates Experimental Colitis. J Crohns Colitis 2016; 10:953-64. [PMID: 26896084 DOI: 10.1093/ecco-jcc/jjw047] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 02/11/2016] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS In recent years, mesenchymal stromal cells [MSCs] emerged as a promising therapeutic option for various diseases, due to their immunomodulatory properties. We previously observed that intraperitoneally injected MSCs in experimental colitis form spherical shaped aggregates. Therefore, we aggregated MSCs in vitro into spheroids and injected them intraluminally in mice with established colitis, to investigate whether these MSC spheroids could alleviate the colitis. METHODS We injected 0.5 x 10(6) MSCs in spheroids, 2.0 x 10(6) MSCs in spheroids, or phosphate-buffered saline [PBS] as a treatment control, via an enema in mice with established dextran sulphate sodium [DSS]-induced colitis. Body weight was measured daily and disease activity score was determined at sacrifice. Endoscopy was performed to evaluate mucosal healing. After sacrifice, both systemic and local inflammatory responses were evaluated. RESULTS Intraluminally injected MSC spheroids alleviated DSS-induced colitis, resulting in significantly less body weight loss and lower disease activity score at sacrifice when a high dose of MSC spheroids was administered. However, the percentage of mucosal lesions in the distal colon and endoscopy scores were not significantly lower after treatment with 2.0 x 10(6) MSCs in spheroids compared with PBS-treated mice. Systemic inflammation marker serum amyloid A [SAA] was significantly reduced after treatment with 2.0 x 10(6) MSCs in spheroids. In addition, local cytokine levels of IFN-ɣ, TNF-α, IL-6, and IL-17a, as well as numbers of macrophages and neutrophils, showed a clear decrease-though not always significant-after intraluminal injection of the MSC spheroids. CONCLUSION Intraluminally injected MSC spheroids at least partially attenuate experimental colitis, with fewer phagocytes and proinflammmatory cytokines, when a high dose of MSCs in spheroids was administered.
Collapse
Affiliation(s)
- Ilse Molendijk
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marieke C Barnhoorn
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Marij A C Mieremet-Ooms
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Johan J van der Reijden
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Danny van der Helm
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Daniel W Hommes
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands Division of Digestive Diseases, University of California Los Angeles, Los Angeles, CA, USA
| | | | - Hein W Verspaget
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
171
|
Kalanxhi E, Risberg K, Barua IS, Dueland S, Waagene S, Andersen SN, Pettersen SJ, Lindvall JM, Redalen KR, Flatmark K, Ree AH. Induction of Apoptosis in Intestinal Toxicity to a Histone Deacetylase Inhibitor in a Phase I Study with Pelvic Radiotherapy. Cancer Res Treat 2016; 49:374-386. [PMID: 27488871 PMCID: PMC5398387 DOI: 10.4143/crt.2016.080] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Accepted: 06/28/2016] [Indexed: 12/24/2022] Open
Abstract
Purpose When integrating molecularly targeted compounds in radiotherapy, synergistic effects of the systemic agent and radiation may extend the limits of patient tolerance, increasing the demand for understanding the pathophysiological mechanisms of treatment toxicity. In this Pelvic Radiation and Vorinostat (PRAVO) study, we investigated mechanisms of adverse effects in response to the histone deacetylase (HDAC) inhibitor vorinostat (suberoylanilide hydroxamic acid, SAHA) when administered as a potential radiosensitiser. Materials and Methods This phase I study for advanced gastrointestinal carcinoma was conducted in sequential patient cohorts exposed to escalating doses of vorinostat combined with standard-fractionated palliative radiotherapy to pelvic target volumes. Gene expression microarray analysis of the study patient peripheral blood mononuclear cells (PBMC) was followed by functional validation in cultured cell lines and mice treated with SAHA. Results PBMC transcriptional responses to vorinostat, including induction of apoptosis, were confined to the patient cohort reporting dose-limiting intestinal toxicities. At relevant SAHA concentrations, apoptotic features (annexin V staining and caspase 3/7 activation, but not poly-(ADP-ribose)-polymerase cleavage) were observed in cultured intestinal epithelial cells. Moreover, SAHA-treated mice displayed significant weight loss. Conclusion The PRAVO study design implemented a strategy to explore treatment toxicity caused by an HDAC inhibitor when combined with radiotherapy and enabled the identification of apoptosis as a potential mechanism responsible for the dose-limiting effects of vorinostat. To the best of our knowledge, this is the first report deciphering mechanisms of normal tissue adverse effects in response to an HDAC inhibitor within a combined-modality treatment regimen.
Collapse
Affiliation(s)
- Erta Kalanxhi
- Department of Oncology, Akershus University Hospital, Lørenskog, Norway.,Institute of Clinical Molecular Biology, Akershus University Hospital, Lørenskog, Norway
| | - Karianne Risberg
- Department of Oncology, Akershus University Hospital, Lørenskog, Norway.,Institute of Clinical Molecular Biology, Akershus University Hospital, Lørenskog, Norway
| | - Imon S Barua
- Institute of Clinical Molecular Biology, Akershus University Hospital, Lørenskog, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Svein Dueland
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Stein Waagene
- Department of Tumour Biology, Oslo University Hospital, Oslo, Norway
| | - Solveig Norheim Andersen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Pathology, Akershus University Hospital, Lørenskog, Norway
| | | | - Jessica M Lindvall
- Institute of Clinical Molecular Biology, Akershus University Hospital, Lørenskog, Norway
| | | | - Kjersti Flatmark
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Tumour Biology, Oslo University Hospital, Oslo, Norway.,Department of Gastroenterological Surgery, Oslo University Hospital, Oslo, Norway
| | - Anne Hansen Ree
- Department of Oncology, Akershus University Hospital, Lørenskog, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
172
|
Ibiza S, García-Cassani B, Ribeiro H, Carvalho T, Almeida L, Marques R, Misic AM, Bartow-McKenney C, Larson DM, Pavan WJ, Eberl G, Grice EA, Veiga-Fernandes H. Glial-cell-derived neuroregulators control type 3 innate lymphoid cells and gut defence. Nature 2016; 535:440-443. [PMID: 27409807 PMCID: PMC4962913 DOI: 10.1038/nature18644] [Citation(s) in RCA: 288] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 06/13/2016] [Indexed: 02/08/2023]
Abstract
Group 3 innate lymphoid cells (ILC3) are major regulators of inflammation and infection at mucosal barriers. ILC3 development is thought to be programmed, but how ILC3 perceive, integrate and respond to local environmental signals remains unclear. Here we show that ILC3 in mice sense their environment and control gut defence as part of a glial–ILC3–epithelial cell unit orchestrated by neurotrophic factors. We found that enteric ILC3 express the neuroregulatory receptor RET. ILC3-autonomous Ret ablation led to decreased innate interleukin-22 (IL-22), impaired epithelial reactivity, dysbiosis and increased susceptibility to bowel inflammation and infection. Neurotrophic factors directly controlled innate Il22 downstream of the p38 MAPK/ERK-AKT cascade and STAT3 activation. Notably, ILC3 were adjacent to neurotrophic-factor-expressing glial cells that exhibited stellate-shaped projections into ILC3 aggregates. Glial cells sensed microenvironmental cues in a MYD88-dependent manner to control neurotrophic factors and innate IL-22. Accordingly, glial-intrinsic Myd88 deletion led to impaired production of ILC3-derived IL-22 and a pronounced propensity towards gut inflammation and infection. Our work sheds light on a novel multi-tissue defence unit, revealing that glial cells are central hubs of neuron and innate immune regulation by neurotrophic factor signals.
Collapse
Affiliation(s)
- Sales Ibiza
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Av. Prof. Egas Moniz, Edifício Egas Moniz, 1649-028 Lisboa, Portugal
| | - Bethania García-Cassani
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Av. Prof. Egas Moniz, Edifício Egas Moniz, 1649-028 Lisboa, Portugal
| | - Hélder Ribeiro
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Av. Prof. Egas Moniz, Edifício Egas Moniz, 1649-028 Lisboa, Portugal
| | - Tânia Carvalho
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Av. Prof. Egas Moniz, Edifício Egas Moniz, 1649-028 Lisboa, Portugal
| | - Luís Almeida
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Av. Prof. Egas Moniz, Edifício Egas Moniz, 1649-028 Lisboa, Portugal
| | - Rute Marques
- Microenvironment and Immunity Unit, Institut Pasteur, 25 Rue du Docteur Roux, 75724 Paris, France
| | - Ana M Misic
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, 421 Curie Blvd, 1007 Biomedical Research Building, Philadelphia, PA 19104, US
| | - Casey Bartow-McKenney
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, 421 Curie Blvd, 1007 Biomedical Research Building, Philadelphia, PA 19104, US
| | - Denise M Larson
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, US
| | - William J Pavan
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, US
| | - Gérard Eberl
- Microenvironment and Immunity Unit, Institut Pasteur, 25 Rue du Docteur Roux, 75724 Paris, France
| | - Elizabeth A Grice
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, 421 Curie Blvd, 1007 Biomedical Research Building, Philadelphia, PA 19104, US
| | - Henrique Veiga-Fernandes
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Av. Prof. Egas Moniz, Edifício Egas Moniz, 1649-028 Lisboa, Portugal
- Champalimaud Research. Champalimaud Centre for the Unknown. 1400-038 Lisbon, Portugal
| |
Collapse
|
173
|
CdtR Regulates TcdA and TcdB Production in Clostridium difficile. PLoS Pathog 2016; 12:e1005758. [PMID: 27414650 PMCID: PMC4944984 DOI: 10.1371/journal.ppat.1005758] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 06/18/2016] [Indexed: 12/18/2022] Open
Abstract
Clostridium difficile is a global health burden and the leading cause of antibiotic-associated diarrhoea worldwide, causing severe gastrointestinal disease and death. Three well characterised toxins are encoded by this bacterium in two genetic loci, specifically, TcdB (toxin B) and TcdA (toxin A) in the Pathogenicity Locus (PaLoc) and binary toxin (CDT) in the genomically distinct CDT locus (CdtLoc). Toxin production is controlled by regulators specific to each locus. The orphan response regulator, CdtR, encoded within the CdtLoc, up-regulates CDT production. Until now there has been no suggestion that CdtR influences TcdA and TcdB production since it is not carried by all PaLoc-containing strains and CdtLoc is not linked genetically to PaLoc. Here we show that, in addition to CDT, CdtR regulates TcdA and TcdB production but that this effect is strain dependent. Of clinical relevance, CdtR increased the production of TcdA, TcdB and CDT in two epidemic ribotype 027 human strains, modulating their virulence in a mouse infection model. Strains traditionally from animal lineages, notably ribotype 078 strains, are increasingly being isolated from humans and their genetic and phenotypic analysis is critical for future studies on this important pathogen. Here we show that CdtR-mediated toxin regulation did not occur in other strain backgrounds, including a ribotype 078 animal strain. The finding that toxin gene regulation is strain dependent highlights the regulatory diversity between C. difficile isolates and the importance of studying virulence regulation in diverse lineages and clinically relevant strains. Our work provides the first evidence that TcdA, TcdB and CDT production is linked by a common regulatory mechanism and that CdtR may act as a global regulator of virulence in epidemic 027 strains. Clostridium difficile is the leading cause of antibiotic-associated diarrhoea. The TcdB, TcdA and binary toxins produced by C. difficile are encoded in two genomically distinct loci: TcdB and TcdA in the Pathogenicity Locus (PaLoc) and binary toxin (CDT) in the CDT locus (CdtLoc). Toxin production is primarily controlled by regulators specific to each locus. Because the presence of these loci varies amongst different strains of C. difficile, no rational link for their co-regulation has ever been proposed. Here we have shown that the regulator of CDT production, CdtR, also regulates production of TcdA and TcdB in a strain dependent manner. These results represent the first evidence that TcdA and TcdB production is linked to the production of CDT by a common regulatory mechanism. Collectively, our results establish CdtR as an important virulence regulator in two clinically important, epidemic strains of C. difficile, and further highlights the need to investigate regulatory mechanisms of important virulence factors in diverse strain backgrounds.
Collapse
|
174
|
Bialkowska AB, Ghaleb AM, Nandan MO, Yang VW. Improved Swiss-rolling Technique for Intestinal Tissue Preparation for Immunohistochemical and Immunofluorescent Analyses. J Vis Exp 2016. [PMID: 27501188 DOI: 10.3791/54161] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Understanding the role of factors that regulate intestinal epithelial homeostasis and response to injury and regeneration is important. The current literature describes several different methodological approaches to obtain images of intestinal tissues for data validation. In this paper, we delineate a common protocol relating to the derivation and processing of mouse intestinal tissues. Proper fixation of intestinal tissues and Swiss-roll techniques that enhance intestinal epithelial morphology are discussed. Postresection processing and reorientation of embedded intestinal tissues are critical in obtaining paraffin-embedded blocks that display intact intestinal structural features after sectioning. The Swiss-rolling technique helps in histological assessment of the complete intestinal or colonic sections examined. An ability to differentiate intestinal structural features can be vital in quantitative measurements of intestinal inflammation and tumorigenesis along the entire length. Finally, paraffin-embedded sections are ideal for robust processing using both immunohistochemical and immunofluorescent detection methods. Nonfluorescent immunohistochemical sections provide a vibrant image of the tissue detailing different cellular structural features but do not provide flexibility for intracellular co-localization experiments. Multiple fluorescent channels can be appropriately utilized with immunofluorescent detection for co-localization experiments, lending support to mechanistic studies.
Collapse
Affiliation(s)
| | - Amr M Ghaleb
- Department of Medicine, Stony Brook University School of Medicine
| | | | - Vincent W Yang
- Department of Medicine, Stony Brook University School of Medicine; Department of Physiology & Biophysics, Stony Brook University School of Medicine;
| |
Collapse
|
175
|
Kuper CF, Van Zijverden M, Klaassen C, Tegelenbosch-Schouten M, Wolterbeek APM. Effects of Cyclosporin A and Cyclophosphamide on Peyer’s Patches in Rat, Exposed in utero and Neonatally or During Adult Age. Toxicol Pathol 2016; 35:226-32. [PMID: 17366316 DOI: 10.1080/01926230601156245] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The effects of cyclosporin A (CY) and cyclophosphamide (CPS) on Peyer’s patches (PP) were studied in Wistar rats, exposed in utero and neonatally or during adult age. In one study, pregnant dams received 5 or 15 mg/kg bw/day CY from gestation day 6 to day 21 of lactation. In two other studies, animals were exposed at young adult age: female rats received orally 5 or 20 mg/kg/day CY or 5 or 10 mg/kg bw CPS for 4 weeks; males received orally 5 mg/kg bw CPS for 4 weeks, or a single iv injection of 50 mg/kg bw CPS. Upon in utero and neonatal exposure, the numbers of grossly observed PP were increased in male pups from the high-dose CY dams at 70 days of age. Exposure to high-dose CY at adult age only tended to decrease the numbers of PP; germinal center development was reduced in the PP from the middle segment of the small intestines, as examined microscopically. Exposure to both doses CPS at adult age reduced the numbers of PP and reduced germinal centre development and the number of lymphocytes in all compartments of PP. It was concluded that the effects of CPS and CY could be established by counting the number of grossly visible PP and by microscopic observation of PP, provided that regional differences of PP were taken into account. Moreover, the type of effects of an immunotoxic agent may vary with age of exposure.
Collapse
Affiliation(s)
- C Frieke Kuper
- TNO Quality of Life, Toxicology and Applied Pharmacology, AJ Zeist, 3700, The Netherlands.
| | | | | | | | | |
Collapse
|
176
|
Phosphorylations of Serines 21/9 in Glycogen Synthase Kinase 3α/β Are Not Required for Cell Lineage Commitment or WNT Signaling in the Normal Mouse Intestine. PLoS One 2016; 11:e0156877. [PMID: 27284979 PMCID: PMC4902192 DOI: 10.1371/journal.pone.0156877] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 05/22/2016] [Indexed: 11/19/2022] Open
Abstract
The WNT signalling pathway controls many developmental processes and plays a key role in maintenance of intestine renewal and homeostasis. Glycogen Synthase Kinase 3 (GSK3) is an important component of the WNT pathway and is involved in regulating β-catenin stability and expression of WNT target genes. The mechanisms underpinning GSK3 regulation in this context are not completely understood, with some evidence suggesting this occurs through inhibitory N-terminal serine phosphorylation in a similar way to GSK3 inactivation in insulin signaling. To investigate this in a physiologically relevant context, we have analysed the intestinal phenotype of GSK3 knockin mice in which N-terminal serines 21/9 of GSK3α/β have been mutated to non-phosphorylatable alanine residues. We show that these knockin mutations have very little effect on overall intestinal integrity, cell lineage commitment, β-catenin localization or WNT target gene expression although a small increase in apoptosis at villi tips is observed. Our results provide in vivo evidence that GSK3 is regulated through mechanisms independent of N-terminal serine phosphorylation in order for β-catenin to be stabilised.
Collapse
|
177
|
Gross S, Garofalo DC, Balderes DA, Mastracci TL, Dias JM, Perlmann T, Ericson J, Sussel L. The novel enterochromaffin marker Lmx1a regulates serotonin biosynthesis in enteroendocrine cell lineages downstream of Nkx2.2. Development 2016; 143:2616-28. [PMID: 27287799 DOI: 10.1242/dev.130682] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 05/26/2016] [Indexed: 12/17/2022]
Abstract
Intestinal hormone-producing cells represent the largest endocrine system in the body, but remarkably little is known about enteroendocrine cell type specification in the embryo and adult. We analyzed stage- and cell type-specific deletions of Nkx2.2 and its functional domains in order to characterize its role in the development and maintenance of enteroendocrine cell lineages in the mouse duodenum and colon. Although Nkx2.2 regulates enteroendocrine cell specification in the duodenum at all stages examined, it controls the differentiation of progressively fewer enteroendocrine cell populations when deleted from Ngn3(+) progenitor cells or in the adult duodenum. During embryonic development Nkx2.2 regulates all enteroendocrine cell types, except gastrin and preproglucagon. In developing Ngn3(+) enteroendocrine progenitor cells, Nkx2.2 is not required for the specification of neuropeptide Y and vasoactive intestinal polypeptide, indicating that a subset of these cell populations derive from an Nkx2.2-independent lineage. In adult duodenum, Nkx2.2 becomes dispensable for cholecystokinin and secretin production. In all stages and Nkx2.2 mutant conditions, serotonin-producing enterochromaffin cells were the most severely reduced enteroendocrine lineage in the duodenum and colon. We determined that the transcription factor Lmx1a is expressed in enterochromaffin cells and functions downstream of Nkx2.2. Lmx1a-deficient mice have reduced expression of Tph1, the rate-limiting enzyme for serotonin biosynthesis. These data clarify the function of Nkx2.2 in the specification and homeostatic maintenance of enteroendocrine populations, and identify Lmx1a as a novel enterochromaffin cell marker that is also essential for the production of the serotonin biosynthetic enzyme Tph1.
Collapse
Affiliation(s)
- Stefanie Gross
- Department of Genetics and Development, Columbia University Medical Center, New York, NY, USA
| | - Diana C Garofalo
- Department of Genetics and Development, Columbia University Medical Center, New York, NY, USA
| | - Dina A Balderes
- Department of Genetics and Development, Columbia University Medical Center, New York, NY, USA
| | - Teresa L Mastracci
- Department of Genetics and Development, Columbia University Medical Center, New York, NY, USA
| | - José M Dias
- Department of Cell and Molecular Biology, Karolinska Institute, von Eulers v. 3, 171 77, Stockholm, Sweden
| | - Thomas Perlmann
- Department of Cell and Molecular Biology, Karolinska Institute, von Eulers v. 3, 171 77, Stockholm, Sweden Ludwig Institute for Cancer Research, Stockholm Branch, Nobels v. 3, 171 77, Stockholm, Sweden
| | - Johan Ericson
- Department of Cell and Molecular Biology, Karolinska Institute, von Eulers v. 3, 171 77, Stockholm, Sweden
| | - Lori Sussel
- Department of Genetics and Development, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
178
|
Cho Y, Gutierrez L, Bordonaro M, Russo D, Anzelmi F, Hooven JT, Cerra C, Lazarova DL. Effects of propolis and gamma-cyclodextrin on intestinal neoplasia in normal weight and obese mice. Cancer Med 2016; 5:2448-58. [PMID: 27265242 PMCID: PMC4898980 DOI: 10.1002/cam4.787] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 04/28/2016] [Accepted: 04/29/2016] [Indexed: 12/20/2022] Open
Abstract
Obesity is associated with colorectal cancer (CRC). This effect might be attributed to adipokine‐supported signaling. We have established that propolis suppresses survival signaling in CRC cells in vitro; therefore, we ascertained the ability of a propolis supplement to modulate intestinal neoplastic development in C57BL/6J‐ApcMin/+/J mice in the lean and obese state. To induce obesity, mice were fed with a Western diet containing 40% fat. Since the propolis supplement includes gamma‐cyclodextrin, the interventions included diets supplemented with or without gamma‐cyclodextrin. The animals were administered the following diets: (1) control diet, (2) control diet/gamma‐cyclodextrin, (3) control diet/propolis, (4) Western diet, (5) Western diet/gamma‐cyclodextrin, and (6) Western diet/propolis. Western diet, resulting in obesity, accelerated neoplastic progression, as evidenced by the larger size and higher grade dysplasia of the neoplasms. In the context of normal weight, gamma‐cyclodextrin and propolis affected neoplastic progression, as determined by the size of the lesions and their grade of dysplasia. A statistically significant decrease in the number of adenomas was detected in mice fed a control diet with the propolis supplement (61.8 ± 10.6 vs. 35.3 ± 7.6, P = 0.008). Although there was no significant difference in the polyp numbers between the six groups, the mice with the lowest number and size of adenomas were those fed a Western diet with gamma‐cyclodextrin. This unexpected outcome might be explained by the increased levels of apoptosis detected in the intestinal tissues of these obese mice. We posit that butyrate derived from the metabolism of gamma‐cyclodextrin may contribute to the decreased neoplastic burden in the context of obesity; however, future studies are required to address this possibility.
Collapse
Affiliation(s)
- Youngjin Cho
- The Commonwealth Medical College, Scranton, Pennsylvania, 18509
| | | | | | - Daniel Russo
- The Commonwealth Medical College, Scranton, Pennsylvania, 18509
| | - Frank Anzelmi
- The Commonwealth Medical College, Scranton, Pennsylvania, 18509
| | - Jayde T Hooven
- Penn State Hershey College of Medicine, Hershey, Pennsylvania, 17033
| | - Carmine Cerra
- The Commonwealth Medical College, Scranton, Pennsylvania, 18509
| | | |
Collapse
|
179
|
Intestinal Preparation Techniques for Histological Analysis in the Mouse. ACTA ACUST UNITED AC 2016; 6:148-168. [DOI: 10.1002/cpmo.2] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
180
|
Bortolotti M, Ventura G, Jegatheesan P, Choisy C, Cynober L, De Bandt JP. Impact of qualitative and quantitative variations in nitrogen supply on catch-up growth in food-deprived-refed young rats. Clin Nutr 2016; 35:669-78. [DOI: 10.1016/j.clnu.2015.04.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 03/09/2015] [Accepted: 04/22/2015] [Indexed: 02/07/2023]
|
181
|
Rocha-Pereira J, Kolawole AO, Verbeken E, Wobus CE, Neyts J. Post-exposure antiviral treatment of norovirus infections effectively protects against diarrhea and reduces virus shedding in the stool in a mortality mouse model. Antiviral Res 2016; 132:76-84. [PMID: 27252124 DOI: 10.1016/j.antiviral.2016.05.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 05/27/2016] [Accepted: 05/27/2016] [Indexed: 11/30/2022]
Abstract
Noroviruses are a leading cause of gastroenteritis across the world in all age groups and are linked to increased hospitalization and mortality in children, the elderly and immunocompromised. The development of specific antiviral treatment for norovirus gastroenteritis is urgently needed. We explored in a mouse model whether an inhibitor of norovirus replication could be used therapeutically post murine norovirus (MNV)-infection of mice. Using the MNV, we previously discovered that the viral polymerase inhibitor 2'-C-methylcytidine (2CMC) is able to protect against diarrhea and mortality in mice when used prophylactically and to block the transmission of MNV between mice. Here, we investigated whether 2CMC could be used therapeutically, starting treatment between 12 h and 3 days post-infection with 2CMC. Post-exposure treatment of MNV-infected mice with 2CMC was efficient up to 2 days after infection, preventing norovirus-induced diarrhea, delaying and reducing MNV shedding in stool of treated mice. Rehydration of 2CMC-treated animals did not result in a further improvement of the disease evolution compared to antiviral treatment only. The presence of MNV antigens and inflammation in the small intestine of infected mice inversely correlated with the effectiveness of delayed antiviral treatment. Anti-MNV IgGs were detected in re-challenged mice 10 weeks after the first contact, these protected the mice from re-infection. We here demonstrate the benefit of antiviral treatment in ongoing norovirus infections.
Collapse
Affiliation(s)
- Joana Rocha-Pereira
- KU Leuven, University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Abimbola O Kolawole
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Eric Verbeken
- KU Leuven - University of Leuven, Department of Imaging & Pathology, Translational Cell & Tissue Research, Leuven, Belgium
| | - Christiane E Wobus
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Johan Neyts
- KU Leuven, University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Leuven, Belgium.
| |
Collapse
|
182
|
Li J, Song J, Zaytseva YY, Liu Y, Rychahou P, Jiang K, Starr ME, Kim JT, Harris JW, Yiannikouris FB, Katz WS, Nilsson PM, Orho-Melander M, Chen J, Zhu H, Fahrenholz T, Higashi RM, Gao T, Morris AJ, Cassis LA, Fan TWM, Weiss HL, Dobner PR, Melander O, Jia J, Evers BM. An obligatory role for neurotensin in high-fat-diet-induced obesity. Nature 2016; 533:411-5. [PMID: 27193687 PMCID: PMC5484414 DOI: 10.1038/nature17662] [Citation(s) in RCA: 200] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 03/10/2016] [Indexed: 12/15/2022]
Abstract
Obesity and its associated comorbidities (for example, diabetes mellitus and hepatic steatosis) contribute to approximately 2.5 million deaths annually and are among the most prevalent and challenging conditions confronting the medical profession. Neurotensin (NT; also known as NTS), a 13-amino-acid peptide predominantly localized in specialized enteroendocrine cells of the small intestine and released by fat ingestion, facilitates fatty acid translocation in rat intestine, and stimulates the growth of various cancers. The effects of NT are mediated through three known NT receptors (NTR1, 2 and 3; also known as NTSR1, 2, and NTSR3, respectively). Increased fasting plasma levels of pro-NT (a stable NT precursor fragment produced in equimolar amounts relative to NT) are associated with increased risk of diabetes, cardiovascular disease and mortality; however, a role for NT as a causative factor in these diseases is unknown. Here we show that NT-deficient mice demonstrate significantly reduced intestinal fat absorption and are protected from obesity, hepatic steatosis and insulin resistance associated with high fat consumption. We further demonstrate that NT attenuates the activation of AMP-activated protein kinase (AMPK) and stimulates fatty acid absorption in mice and in cultured intestinal cells, and that this occurs through a mechanism involving NTR1 and NTR3 (also known as sortilin). Consistent with the findings in mice, expression of NT in Drosophila midgut enteroendocrine cells results in increased lipid accumulation in the midgut, fat body, and oenocytes (specialized hepatocyte-like cells) and decreased AMPK activation. Remarkably, in humans, we show that both obese and insulin-resistant subjects have elevated plasma concentrations of pro-NT, and in longitudinal studies among non-obese subjects, high levels of pro-NT denote a doubling of the risk of developing obesity later in life. Our findings directly link NT with increased fat absorption and obesity and suggest that NT may provide a prognostic marker of future obesity and a potential target for prevention and treatment.
Collapse
Affiliation(s)
- Jing Li
- Department of Surgery, University of Kentucky, Lexington, Kentucky 40536, USA
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Jun Song
- Department of Surgery, University of Kentucky, Lexington, Kentucky 40536, USA
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Yekaterina Y Zaytseva
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky 40536, USA
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Yajuan Liu
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Piotr Rychahou
- Department of Surgery, University of Kentucky, Lexington, Kentucky 40536, USA
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Kai Jiang
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Marlene E Starr
- Department of Surgery, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Ji Tae Kim
- Department of Surgery, University of Kentucky, Lexington, Kentucky 40536, USA
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Jennifer W Harris
- Department of Surgery, University of Kentucky, Lexington, Kentucky 40536, USA
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Frederique B Yiannikouris
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Wendy S Katz
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Peter M Nilsson
- Department of Clinical Sciences, Lund University, Malmö, 221 00 Lund, Sweden
- Department of Internal Medicine, Skåne University Hospital, Malmö, 205 02 Malmö, Sweden
| | - Marju Orho-Melander
- Department of Clinical Sciences, Lund University, Malmö, 221 00 Lund, Sweden
| | - Jing Chen
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky 40536, USA
- Center for Structural Biology, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Haining Zhu
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky 40536, USA
- Center for Structural Biology, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Timothy Fahrenholz
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky 40536, USA
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky 40536, USA
- Center for Environmental and Systems Biochemistry, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Richard M Higashi
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky 40536, USA
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky 40536, USA
- Center for Environmental and Systems Biochemistry, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Tianyan Gao
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky 40536, USA
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Andrew J Morris
- Division of Cardiovascular Medicine, Gill Heart Institute, University of Kentucky and Lexington Veterans Affairs Medical Center, Lexington, Kentucky 40536, USA
| | - Lisa A Cassis
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Teresa W-M Fan
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky 40536, USA
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky 40536, USA
- Center for Environmental and Systems Biochemistry, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Heidi L Weiss
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky 40536, USA
- Department of Biostatistics, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Paul R Dobner
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA
| | - Olle Melander
- Department of Clinical Sciences, Lund University, Malmö, 221 00 Lund, Sweden
- Department of Internal Medicine, Skåne University Hospital, Malmö, 205 02 Malmö, Sweden
| | - Jianhang Jia
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky 40536, USA
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky 40536, USA
| | - B Mark Evers
- Department of Surgery, University of Kentucky, Lexington, Kentucky 40536, USA
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky 40536, USA
| |
Collapse
|
183
|
Acute blockade of IL-25 in a colitis associated colon cancer model leads to increased tumor burden. Sci Rep 2016; 6:25643. [PMID: 27165713 PMCID: PMC4863374 DOI: 10.1038/srep25643] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 04/19/2016] [Indexed: 12/19/2022] Open
Abstract
Chronic inflammation within the gastrointestinal tract results in an increased risk for developing colorectal cancer. Epithelial cytokines, including interleukin-25 (IL-25), are produced in the colon and are critical for protection from parasites, but can also be pathogenic in the context of inflammatory bowel diseases and allergy. Whether IL-25 is involved in the progression from inflammation to cancer is still largely unexplored. Using a well-established murine model for colitis-induced colon cancer; we aimed to determine the role of IL-25 in this process. We found that acute IL-25 blockade resulted in greater tumor burdens compared to isotype control treated mice. Histologically, α-IL-25 treated mice had increased colitis scores compared to mice receiving isotype control antibody, as well as decreased eosinophilia. This is the first study to explore the therapeutic potential of using an IL-25 blocking antibody during a chronic inflammatory setting. Taken together these data suggest that IL-25 plays an inhibitory role in the growth and development of colonic tumors.
Collapse
|
184
|
Intestinal knockout of Nedd4 enhances growth of Apc min tumors. Oncogene 2016; 35:5839-5849. [PMID: 27086928 DOI: 10.1038/onc.2016.125] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 03/07/2016] [Accepted: 03/11/2016] [Indexed: 12/24/2022]
Abstract
Nedd4 (Nedd4-1) is an E3 ubiquitin ligase that belongs to the HECT family and comprises a C2-WW(n)-HECT domain architecture. Although it has been reported to regulate growth factor receptors and cellular signaling, its role in cancer development has been controversial, with some studies proposing that it promotes cancer while others suggest it inhibits tumor growth. Here, we tested the effect of Nedd4 on intestinal tumor formation and growth using Nedd4-knockout mice (Nedd4 floxed (fl) mice crossed to villin-Cre mice). Although we find that knockout of Nedd4 on its own does not cause tumor growth, its knockout in the context of Apc+/min-derived colorectal tumors leads to augmentation of tumor growth, suggesting that Nedd4 normally suppresses intestinal WNT signaling and growth of colonic tumors. WNT signaling microarray, immunoblotting and immunohistochemistry analyses of tumors derived from the Villin-Cre;Nedd4fl/fl;Apc+/min colons demonstrated elevated expression of the WNT upstream effectors LEF1 (full length) and YY1 in these tumors relative to control (Apc+/min alone) tumors. Together, these results suggest that Nedd4 suppresses colonic WNT signaling and tumor growth, at least in part, by suppressing the transcription factors LEF1 and YY1.
Collapse
|
185
|
Winek K, Engel O, Koduah P, Heimesaat MM, Fischer A, Bereswill S, Dames C, Kershaw O, Gruber AD, Curato C, Oyama N, Meisel C, Meisel A, Dirnagl U. Depletion of Cultivatable Gut Microbiota by Broad-Spectrum Antibiotic Pretreatment Worsens Outcome After Murine Stroke. Stroke 2016; 47:1354-63. [PMID: 27056982 PMCID: PMC4839545 DOI: 10.1161/strokeaha.115.011800] [Citation(s) in RCA: 159] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 03/07/2016] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND PURPOSE Antibiotics disturbing microbiota are often used in treatment of poststroke infections. A bidirectional brain-gut microbiota axis was recently suggested as a modulator of nervous system diseases. We hypothesized that gut microbiota may be an important player in the course of stroke. METHODS We investigated the outcome of focal cerebral ischemia in C57BL/6J mice after an 8-week decontamination with quintuple broad-spectrum antibiotic cocktail. These microbiota-depleted animals were subjected to 60 minutes middle cerebral artery occlusion or sham operation. Infarct volume was measured using magnetic resonance imaging, and mice were monitored clinically throughout the whole experiment. At the end point, tissues were preserved for further analysis, comprising histology and immunologic investigations using flow cytometry. RESULTS We found significantly decreased survival in the middle cerebral artery occlusion microbiota-depleted mice when the antibiotic cocktail was stopped 3 days before surgery (compared with middle cerebral artery occlusion specific pathogen-free and sham-operated microbiota-depleted mice). Moreover, all microbiota-depleted animals in which antibiotic treatment was terminated developed severe acute colitis. This phenotype was rescued by continuous antibiotic treatment or colonization with specific pathogen-free microbiota before surgery. Further, infarct volumes on day one did not differ between any of the experimental groups. CONCLUSIONS Conventional microbiota ensures intestinal protection in the mouse model of experimental stroke and prevents development of acute and severe colitis in microbiota-depleted mice not given antibiotic protection after cerebral ischemia. Our experiments raise the clinically important question as to whether microbial colonization or specific microbiota are crucial for stroke outcome.
Collapse
Affiliation(s)
- Katarzyna Winek
- From the Department of Experimental Neurology (K.W., O.E., P.K., N.O., A.M., U.D.), NeuroCure Clinical Research (K.W., C.C., A.M., U.D.), Center for Stroke Research Berlin (K.W., O.E., P.K., A.M., U.D.), Department of Microbiology and Hygiene (M.M.H., A.F., S.B.), Institute for Medical Immunology (C.D., C.M.), and Department of Neurology (A.M., U.D.), Charité - Universitätsmedizin Berlin, Germany; Institute of Veterinary Pathology, Faculty of Veterinary Medicine, Freie Universität Berlin, Germany (O.K., A.D.G.); German Rheumatism Research Center (DRFZ), Berlin, Germany (C.C.); and German Center for Neurodegeneration Research (DZNE), partner site Berlin, Germany (U.D.)
| | - Odilo Engel
- From the Department of Experimental Neurology (K.W., O.E., P.K., N.O., A.M., U.D.), NeuroCure Clinical Research (K.W., C.C., A.M., U.D.), Center for Stroke Research Berlin (K.W., O.E., P.K., A.M., U.D.), Department of Microbiology and Hygiene (M.M.H., A.F., S.B.), Institute for Medical Immunology (C.D., C.M.), and Department of Neurology (A.M., U.D.), Charité - Universitätsmedizin Berlin, Germany; Institute of Veterinary Pathology, Faculty of Veterinary Medicine, Freie Universität Berlin, Germany (O.K., A.D.G.); German Rheumatism Research Center (DRFZ), Berlin, Germany (C.C.); and German Center for Neurodegeneration Research (DZNE), partner site Berlin, Germany (U.D.)
| | - Priscilla Koduah
- From the Department of Experimental Neurology (K.W., O.E., P.K., N.O., A.M., U.D.), NeuroCure Clinical Research (K.W., C.C., A.M., U.D.), Center for Stroke Research Berlin (K.W., O.E., P.K., A.M., U.D.), Department of Microbiology and Hygiene (M.M.H., A.F., S.B.), Institute for Medical Immunology (C.D., C.M.), and Department of Neurology (A.M., U.D.), Charité - Universitätsmedizin Berlin, Germany; Institute of Veterinary Pathology, Faculty of Veterinary Medicine, Freie Universität Berlin, Germany (O.K., A.D.G.); German Rheumatism Research Center (DRFZ), Berlin, Germany (C.C.); and German Center for Neurodegeneration Research (DZNE), partner site Berlin, Germany (U.D.)
| | - Markus M Heimesaat
- From the Department of Experimental Neurology (K.W., O.E., P.K., N.O., A.M., U.D.), NeuroCure Clinical Research (K.W., C.C., A.M., U.D.), Center for Stroke Research Berlin (K.W., O.E., P.K., A.M., U.D.), Department of Microbiology and Hygiene (M.M.H., A.F., S.B.), Institute for Medical Immunology (C.D., C.M.), and Department of Neurology (A.M., U.D.), Charité - Universitätsmedizin Berlin, Germany; Institute of Veterinary Pathology, Faculty of Veterinary Medicine, Freie Universität Berlin, Germany (O.K., A.D.G.); German Rheumatism Research Center (DRFZ), Berlin, Germany (C.C.); and German Center for Neurodegeneration Research (DZNE), partner site Berlin, Germany (U.D.)
| | - André Fischer
- From the Department of Experimental Neurology (K.W., O.E., P.K., N.O., A.M., U.D.), NeuroCure Clinical Research (K.W., C.C., A.M., U.D.), Center for Stroke Research Berlin (K.W., O.E., P.K., A.M., U.D.), Department of Microbiology and Hygiene (M.M.H., A.F., S.B.), Institute for Medical Immunology (C.D., C.M.), and Department of Neurology (A.M., U.D.), Charité - Universitätsmedizin Berlin, Germany; Institute of Veterinary Pathology, Faculty of Veterinary Medicine, Freie Universität Berlin, Germany (O.K., A.D.G.); German Rheumatism Research Center (DRFZ), Berlin, Germany (C.C.); and German Center for Neurodegeneration Research (DZNE), partner site Berlin, Germany (U.D.)
| | - Stefan Bereswill
- From the Department of Experimental Neurology (K.W., O.E., P.K., N.O., A.M., U.D.), NeuroCure Clinical Research (K.W., C.C., A.M., U.D.), Center for Stroke Research Berlin (K.W., O.E., P.K., A.M., U.D.), Department of Microbiology and Hygiene (M.M.H., A.F., S.B.), Institute for Medical Immunology (C.D., C.M.), and Department of Neurology (A.M., U.D.), Charité - Universitätsmedizin Berlin, Germany; Institute of Veterinary Pathology, Faculty of Veterinary Medicine, Freie Universität Berlin, Germany (O.K., A.D.G.); German Rheumatism Research Center (DRFZ), Berlin, Germany (C.C.); and German Center for Neurodegeneration Research (DZNE), partner site Berlin, Germany (U.D.)
| | - Claudia Dames
- From the Department of Experimental Neurology (K.W., O.E., P.K., N.O., A.M., U.D.), NeuroCure Clinical Research (K.W., C.C., A.M., U.D.), Center for Stroke Research Berlin (K.W., O.E., P.K., A.M., U.D.), Department of Microbiology and Hygiene (M.M.H., A.F., S.B.), Institute for Medical Immunology (C.D., C.M.), and Department of Neurology (A.M., U.D.), Charité - Universitätsmedizin Berlin, Germany; Institute of Veterinary Pathology, Faculty of Veterinary Medicine, Freie Universität Berlin, Germany (O.K., A.D.G.); German Rheumatism Research Center (DRFZ), Berlin, Germany (C.C.); and German Center for Neurodegeneration Research (DZNE), partner site Berlin, Germany (U.D.)
| | - Olivia Kershaw
- From the Department of Experimental Neurology (K.W., O.E., P.K., N.O., A.M., U.D.), NeuroCure Clinical Research (K.W., C.C., A.M., U.D.), Center for Stroke Research Berlin (K.W., O.E., P.K., A.M., U.D.), Department of Microbiology and Hygiene (M.M.H., A.F., S.B.), Institute for Medical Immunology (C.D., C.M.), and Department of Neurology (A.M., U.D.), Charité - Universitätsmedizin Berlin, Germany; Institute of Veterinary Pathology, Faculty of Veterinary Medicine, Freie Universität Berlin, Germany (O.K., A.D.G.); German Rheumatism Research Center (DRFZ), Berlin, Germany (C.C.); and German Center for Neurodegeneration Research (DZNE), partner site Berlin, Germany (U.D.)
| | - Achim D Gruber
- From the Department of Experimental Neurology (K.W., O.E., P.K., N.O., A.M., U.D.), NeuroCure Clinical Research (K.W., C.C., A.M., U.D.), Center for Stroke Research Berlin (K.W., O.E., P.K., A.M., U.D.), Department of Microbiology and Hygiene (M.M.H., A.F., S.B.), Institute for Medical Immunology (C.D., C.M.), and Department of Neurology (A.M., U.D.), Charité - Universitätsmedizin Berlin, Germany; Institute of Veterinary Pathology, Faculty of Veterinary Medicine, Freie Universität Berlin, Germany (O.K., A.D.G.); German Rheumatism Research Center (DRFZ), Berlin, Germany (C.C.); and German Center for Neurodegeneration Research (DZNE), partner site Berlin, Germany (U.D.)
| | - Caterina Curato
- From the Department of Experimental Neurology (K.W., O.E., P.K., N.O., A.M., U.D.), NeuroCure Clinical Research (K.W., C.C., A.M., U.D.), Center for Stroke Research Berlin (K.W., O.E., P.K., A.M., U.D.), Department of Microbiology and Hygiene (M.M.H., A.F., S.B.), Institute for Medical Immunology (C.D., C.M.), and Department of Neurology (A.M., U.D.), Charité - Universitätsmedizin Berlin, Germany; Institute of Veterinary Pathology, Faculty of Veterinary Medicine, Freie Universität Berlin, Germany (O.K., A.D.G.); German Rheumatism Research Center (DRFZ), Berlin, Germany (C.C.); and German Center for Neurodegeneration Research (DZNE), partner site Berlin, Germany (U.D.)
| | - Naoki Oyama
- From the Department of Experimental Neurology (K.W., O.E., P.K., N.O., A.M., U.D.), NeuroCure Clinical Research (K.W., C.C., A.M., U.D.), Center for Stroke Research Berlin (K.W., O.E., P.K., A.M., U.D.), Department of Microbiology and Hygiene (M.M.H., A.F., S.B.), Institute for Medical Immunology (C.D., C.M.), and Department of Neurology (A.M., U.D.), Charité - Universitätsmedizin Berlin, Germany; Institute of Veterinary Pathology, Faculty of Veterinary Medicine, Freie Universität Berlin, Germany (O.K., A.D.G.); German Rheumatism Research Center (DRFZ), Berlin, Germany (C.C.); and German Center for Neurodegeneration Research (DZNE), partner site Berlin, Germany (U.D.)
| | - Christian Meisel
- From the Department of Experimental Neurology (K.W., O.E., P.K., N.O., A.M., U.D.), NeuroCure Clinical Research (K.W., C.C., A.M., U.D.), Center for Stroke Research Berlin (K.W., O.E., P.K., A.M., U.D.), Department of Microbiology and Hygiene (M.M.H., A.F., S.B.), Institute for Medical Immunology (C.D., C.M.), and Department of Neurology (A.M., U.D.), Charité - Universitätsmedizin Berlin, Germany; Institute of Veterinary Pathology, Faculty of Veterinary Medicine, Freie Universität Berlin, Germany (O.K., A.D.G.); German Rheumatism Research Center (DRFZ), Berlin, Germany (C.C.); and German Center for Neurodegeneration Research (DZNE), partner site Berlin, Germany (U.D.)
| | - Andreas Meisel
- From the Department of Experimental Neurology (K.W., O.E., P.K., N.O., A.M., U.D.), NeuroCure Clinical Research (K.W., C.C., A.M., U.D.), Center for Stroke Research Berlin (K.W., O.E., P.K., A.M., U.D.), Department of Microbiology and Hygiene (M.M.H., A.F., S.B.), Institute for Medical Immunology (C.D., C.M.), and Department of Neurology (A.M., U.D.), Charité - Universitätsmedizin Berlin, Germany; Institute of Veterinary Pathology, Faculty of Veterinary Medicine, Freie Universität Berlin, Germany (O.K., A.D.G.); German Rheumatism Research Center (DRFZ), Berlin, Germany (C.C.); and German Center for Neurodegeneration Research (DZNE), partner site Berlin, Germany (U.D.)
| | - Ulrich Dirnagl
- From the Department of Experimental Neurology (K.W., O.E., P.K., N.O., A.M., U.D.), NeuroCure Clinical Research (K.W., C.C., A.M., U.D.), Center for Stroke Research Berlin (K.W., O.E., P.K., A.M., U.D.), Department of Microbiology and Hygiene (M.M.H., A.F., S.B.), Institute for Medical Immunology (C.D., C.M.), and Department of Neurology (A.M., U.D.), Charité - Universitätsmedizin Berlin, Germany; Institute of Veterinary Pathology, Faculty of Veterinary Medicine, Freie Universität Berlin, Germany (O.K., A.D.G.); German Rheumatism Research Center (DRFZ), Berlin, Germany (C.C.); and German Center for Neurodegeneration Research (DZNE), partner site Berlin, Germany (U.D.).
| |
Collapse
|
186
|
An Inducible, Large-Intestine-Specific Transgenic Mouse Model for Colitis and Colitis-Induced Colon Cancer Research. Dig Dis Sci 2016; 61:1069-79. [PMID: 26631394 PMCID: PMC5476293 DOI: 10.1007/s10620-015-3971-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 11/08/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND Animal models are an important tool to understand intestinal biology. Our laboratory previously generated C57BL/6-Tg(Car1-cre)5Flt transgenic mice (CAC) with large-intestine-specific Cre recombinase (Cre) expression as a model to study colon health. AIM To expand the utility of the CAC mouse model by determining the impact of chemically induced colitis on CAC transgene expression. METHODS CAC mice were crossed to Rosa reporter mice (Rosa26R (flox/flox) ) with a lox-STOP-lox signal controlling β-galactosidase (βgal) expression and then further crossed with Apc(CKO/CKO) mice in some experiments to delete Apc alleles (Apc (Δ580) ). Initially, 8-week-old CAC(Tg/WT);Rosa26R (flox/WT) ;Apc (Δ580/WT) mice were treated with dextran sulfate sodium (DSS) in drinking water (5 days, 0, 0.65, 1.35, or 2.0 %). Colon tissue damage and βgal labeling were analyzed 10 day after stopping DSS. Next, 8-week-old CAC(Tg/WT);Rosa26R(flox/flox) mice were treated with 0 or 1.35 % DSS, and colonic βgal labeling was assessed at 30 day post-DSS treatment. Finally, 10-week-old CAC(Tg/WT);Apc (Δ580/WT) mice were treated with DSS (0 or 2 %) for 5 days and colonic tumors were analyzed at 20 weeks. RESULTS CAC(Tg/WT);Rosa26R (flox/WT) ;Apc (Δ580/WT) mice had a DSS dose-dependent increase in colon epithelial damage that correlated with increased epithelial βgal labeling at 10 days (r (2) = 0.9, β = 0.75). The βgal labeling in CAC(Tg/WT);Rosa26R(flox/flox) mice colon remained high at 30 days, especially in the crypts of the healed ulcer. DSS also increased colon tumor incidence and multiplicity in CAC(Tg/WT);Apc (Δ580/WT) mice. CONCLUSIONS DSS-mediated epithelial damage induces a persistent, Cre-mediated recombination of floxed alleles in CAC mice. This enables the examination of gene function in colon epithelium during experimental colitis and colitis-induced colon cancer.
Collapse
|
187
|
Davis JS, Gupta V, Gagea M, Wu X. An Advanced Histologic Method for Evaluation of Intestinal Adenomas in Mice Using Digital Slides. PLoS One 2016; 11:e0151463. [PMID: 26974326 PMCID: PMC4790911 DOI: 10.1371/journal.pone.0151463] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 02/29/2016] [Indexed: 11/19/2022] Open
Abstract
Background and Methods Mice are used for modelling the biology of many human diseases, including colorectal cancer (CRC). Mouse models recapitulate many aspects of human disease and are invaluable tools for studying the biology, treatment and prevention of CRC. Unlike humans, many mouse models develop lesions primarily in the small intestine, which necessitates removal and examination of this organ in order to evaluate treatment efficacy. Commonly, the small intestine is visually examined for gross lesions and then selectively embedded in paraffin blocks for further microscopic analysis. Unfortunately, this method suffers from inherent bias toward counting large lesions and simultaneously missing smaller lesions. Even more, this method leaves no permanent record of diagnosed and measured lesions. We evaluated inter-observer variability in a mouse model of CRC using visual examination, and directly compared the visual, gross examination with a histologic analytic method using digital slides of hematoxylin and eosin stained tissue sections. Results Using visual examination, there was a high degree of inter-observer variability. As this method does not provide a permanent record of measurements, there is no capability to arbitrate between differing observations. In contrast, histologic analysis allowed for the creation of a permanent record of lesion measurements taken. When compared directly, histologic analysis of annotated digital images has significantly improved accuracy. Using this method we were able to distinguish mutant mice from wild type littermates even at a very young age. With gross visual examination, this distinction was not possible. Conclusion Histologic analysis of digital images of murine intestinal tissue provides a vital improvement over the commonly used visual, gross examination method. Unlike visual gross examination, histologic analysis is not biased by the size of intestinal adenoma, misdiagnosis of another lesion type, or presence of a Peyer’s patch. It also provides accountability in the form of a permanent record of lesions counted. Histologic analysis using digital slides represents a critical improvement over the current, widely used method of visual gross examination and should be considered for future studies using mouse models of CRC.
Collapse
Affiliation(s)
- Jennifer S. Davis
- Department of Clinical Cancer Prevention, Unit 1013, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Vineet Gupta
- Department of Clinical Cancer Prevention, Unit 1013, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Mihai Gagea
- Department of Veterinary Medicine and Surgery, Unit 0063, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Xiangwei Wu
- Department of Clinical Cancer Prevention, Unit 1013, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
- * E-mail:
| |
Collapse
|
188
|
Sundberg JP, Berndt A, Sundberg BA, Silva KA, Kennedy V, Smith RS, Cooper TK, Schofield PN. Approaches to Investigating Complex Genetic Traits in a Large-Scale Inbred Mouse Aging Study. Vet Pathol 2016; 53:456-67. [PMID: 26936752 PMCID: PMC5297262 DOI: 10.1177/0300985815612556] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Inbred mice are a unique model system for studying aging because of the genetic homogeneity within inbred strains, the short life span of mice relative to humans, and the rich array of analytic tools that are available. A large-scale aging study was conducted on 28 inbred strains representing great genetic diversity to determine, via histopathology, the type and diversity of spontaneous diseases that aging mice develop. A total of 20 885 different diagnoses were made, with an average of 12 diagnoses per mouse in the study. Eighteen inbred strains have had their genomes sequenced, and many others have been partially sequenced to provide large repositories of data on genetic variation among the strains. This vast amount of genomic information can be utilized in genome-wide association studies to find candidate genes that are involved in the pathogenesis of spontaneous diseases. As an illustration, this article presents a genome-wide association study of the genetic associations of age-related intestinal amyloidosis, which implicated 3 candidate genes: translocating chain-associated membrane protein 1 (Tram1); splicing factor 3b, subunit 5 (Sf3b5); and syntaxin 11 (Stx11). Representative photomicrographs are available on the Mouse Tumor Biology Database and Pathbase to serve as a reference when evaluating inbred mice used in other genetic or experimental studies to rule out strain background lesions. Many of the age-related mouse diseases are similar, if not identical, to human diseases; therefore, the genetic discoveries have direct translational benefit.
Collapse
Affiliation(s)
| | - A Berndt
- University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | | | - K A Silva
- The Jackson Laboratory, Bar Harbor, ME, USA
| | - V Kennedy
- The Jackson Laboratory, Bar Harbor, ME, USA
| | - R S Smith
- The Jackson Laboratory, Bar Harbor, ME, USA
| | - T K Cooper
- Department of Comparative Medicine, Department of Pathology, Penn State Milton S. Hershey Medical Center, College of Medicine, Hershey, PA, USA
| | - P N Schofield
- The Jackson Laboratory, Bar Harbor, ME, USA Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, UK
| |
Collapse
|
189
|
Cheat S, Pinton P, Cossalter AM, Cognie J, Vilariño M, Callu P, Raymond-Letron I, Oswald IP, Kolf-Clauw M. The mycotoxins deoxynivalenol and nivalenol show in vivo synergism on jejunum enterocytes apoptosis. Food Chem Toxicol 2016; 87:45-54. [DOI: 10.1016/j.fct.2015.11.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 11/21/2015] [Accepted: 11/23/2015] [Indexed: 01/09/2023]
|
190
|
Gross S, Balderes D, Liu J, Asfaha S, Gu G, Wang TC, Sussel L. Nkx2.2 is expressed in a subset of enteroendocrine cells with expanded lineage potential. Am J Physiol Gastrointest Liver Physiol 2015; 309:G975-87. [PMID: 26492922 PMCID: PMC4683302 DOI: 10.1152/ajpgi.00244.2015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 10/19/2015] [Indexed: 01/31/2023]
Abstract
There are two major stem cell populations in the intestinal crypt region that express either Bmi1 or Lgr5; however, it has been shown that other populations in the crypt can regain stemness. In this study, we demonstrate that the transcription factor NK2 homeobox 2 (Nkx2.2) is expressed in enteroendocrine cells located in the villus and crypt of the intestinal epithelium and is coexpressed with the stem cell markers Bmi1 and Lgr5 in a subset of crypt cells. To determine whether Nkx2.2-expressing enteroendocrine cells display cellular plasticity and stem cell potential, we performed genetic lineage tracing of the Nkx2.2-expressing population using Nkx2.2(Cre/+);R26RTomato mice. These studies demonstrated that Nkx2.2+ cells are able to give rise to all intestinal epithelial cell types in basal conditions. The proliferative capacity of Nkx2.2-expressing cells was also demonstrated in vitro using crypt organoid cultures. Injuring the intestine with irradiation, systemic inflammation, and colitis did not enhance the lineage potential of Nkx2.2-expressing cells. These findings demonstrate that a rare mature enteroendocrine cell subpopulation that is demarcated by Nkx2.2 expression display stem cell properties during normal intestinal epithelial homeostasis, but is not easily activated upon injury.
Collapse
Affiliation(s)
- Stefanie Gross
- Department of Genetics and Development, Columbia University Medical Center, New York, New York
| | - Dina Balderes
- Department of Genetics and Development, Columbia University Medical Center, New York, New York
| | - Jing Liu
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Samuel Asfaha
- Department of Digestive and Liver Diseases, Columbia University Medical Center, New York, New York; and
| | - Guoqiang Gu
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Timothy C Wang
- Department of Digestive and Liver Diseases, Columbia University Medical Center, New York, New York; and Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York
| | - Lori Sussel
- Department of Genetics and Development, Columbia University Medical Center, New York, New York;
| |
Collapse
|
191
|
Häger C, Keubler LM, Biernot S, Dietrich J, Buchheister S, Buettner M, Bleich A. Time to Integrate to Nest Test Evaluation in a Mouse DSS-Colitis Model. PLoS One 2015; 10:e0143824. [PMID: 26637175 PMCID: PMC4670219 DOI: 10.1371/journal.pone.0143824] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 11/10/2015] [Indexed: 01/07/2023] Open
Abstract
Severity assessment in laboratory animals is an important issue regarding the implementation of the 3R concept into biomedical research and pivotal in current EU regulations. In mouse models of inflammatory bowel disease severity assessment is usually undertaken by clinical scoring, especially by monitoring reduction of body weight. This requires daily observance and handling of each mouse, which is time consuming, stressful for the animal and necessitates an experienced observer. The time to integrate to nest test (TINT) is an easily applicable test detecting disturbed welfare by measuring the time interval mice need to integrate nesting material to an existing nest. Here, TINT was utilized to assess severity in a mouse DSS-colitis model. TINT results depended on the group size of mice maintained per cage with most consistent time intervals measured when co-housing 4 to 5 mice. Colitis was induced with 1% or 1.5% DSS in group-housed WT and Cd14-deficient mice. Higher clinical scores and loss of body weight were detected in 1.5% compared to 1% DSS treated mice. TINT time intervals showed no dose dependent differences. However, increased clinical scores, body weight reductions, and increased TINT time intervals were detected in Cd14-/- compared to WT mice revealing mouse strain related differences. Therefore, TINT is an easily applicable method for severity assessment in a mouse colitis model detecting CD14 related differences, but not dose dependent differences. As TINT revealed most consistent results in group-housed mice, we recommend utilization as an additional method substituting clinical monitoring of the individual mouse.
Collapse
Affiliation(s)
- Christine Häger
- Institute of Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Hannover, Germany
| | - Lydia M. Keubler
- Institute of Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Hannover, Germany
| | - Svenja Biernot
- Institute of Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Hannover, Germany
| | - Jana Dietrich
- Institute of Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Hannover, Germany
| | - Stephanie Buchheister
- Institute of Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Hannover, Germany
| | - Manuela Buettner
- Institute of Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Hannover, Germany
| | - André Bleich
- Institute of Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Hannover, Germany
- * E-mail:
| |
Collapse
|
192
|
Sødring M, Gunnes G, Paulsen JE. Spontaneous initiation, promotion and progression of colorectal cancer in the novel A/J Min/+ mouse. Int J Cancer 2015; 138:1936-46. [PMID: 26566853 DOI: 10.1002/ijc.29928] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 11/02/2015] [Indexed: 12/17/2022]
Abstract
The C57BL/6J multiple intestinal neoplasia (Min/+) mouse is a widely used murine model for familial adenomatous polyposis, a hereditary form of human colorectal cancer. However, it is a questionable model partly because the vast majority of tumors arise in the small intestine, and partly because the fraction of tumors that progress to invasive carcinomas is minuscule. A/J mice are typically more susceptible to carcinogen-induced colorectal cancer than C57BL/6J mice. To investigate whether the novel Min/+ mouse on the A/J genetic background could be a better model for colorectal cancer, we examined the spontaneous intestinal tumorigenesis in 81 A/J Min/+ mice ranging in age from 4 to 60 weeks. The A/J Min/+ mouse exhibited a dramatic increase in number of colonic lesions when compared to what has been reported for the conventional Min/+ mouse; however, an increase in small intestinal lesions did not occur. In addition, this novel mouse model displayed a continual development of colonic lesions highlighted by the transition from early lesions (flat ACF) to tumors over time. In mice older than 40 weeks, 13 colonic (95% CI: 8.7-16.3) and 21 small intestinal (95% CI: 18.6-24.3) tumors were recorded. Notably, a considerable proportion of those lesions progressed to carcinomas in both the colon (21%) and small intestine (51%). These findings more closely reflect aspects of human colorectal carcinogenesis. In conclusion, the novel A/J Min/+ mouse may be a relevant model for initiation, promotion and progression of colorectal cancer.
Collapse
Affiliation(s)
- Marianne Sødring
- Department of Food Safety and Infection Biology, Norwegian University of Life Sciences, Oslo, Norway
| | - Gjermund Gunnes
- Department of Basic Sciences and Aquatic Medicine, Norwegian University of Life Sciences, Oslo, Norway
| | - Jan Erik Paulsen
- Department of Food Safety and Infection Biology, Norwegian University of Life Sciences, Oslo, Norway
| |
Collapse
|
193
|
Yu S, Hwang HE, Yun N, Goldenring JR, Nam KT. The mRNA and Protein Levels of Tubulin and β-Actin Are Greatly Reduced in the Proximal Duodenum of Mice Relative to the Rest of the Small Intestines. Dig Dis Sci 2015; 60:2670-6. [PMID: 25976623 DOI: 10.1007/s10620-015-3688-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 04/24/2015] [Indexed: 12/09/2022]
Abstract
To accurately quantify mRNA and protein levels, it is critical to choose appropriate internal standards. As the expression of housekeeping genes is assumed to remain constant, they are often employed to normalize signals to correct for sample-to-sample variations. However, recent studies have documented that β-actin and Glyceraldehyde 3-phosphate dehydrogenase (GAPDH) expression levels change in response to various stimuli during proliferation, activation, and differentiation. We investigated levels of α-, β-, γ-tubulin, β-actin, and GAPDH vary across the gastrointestinal tract of mice. We found that different regions of the small intestines had dramatically different expression profiles, as measured by western blot, quantitative Reverse transcription polymerase chain reaction (RT-PCR), and immunohistochemical staining. These results revealed that the expression levels of tubulins and β-actin were dramatically lower in the proximal duodenum, relative to the rest of the small intestines. These varying levels of housekeeping genes may reflect differences in the activities of specialized tissues and suggest unique requirements for tubulins in these tissue types. We conclude that the use of a single housekeeping gene to normalize gene expression in the gastrointestinal tracts of mice may introduce errors, as measured differences in gene expression may reflect regulation of the internal control rather than the mRNA or protein under investigation.
Collapse
Affiliation(s)
- Sungsook Yu
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 120-752, Korea
| | | | | | | | | |
Collapse
|
194
|
Abstract
Necropsy (also known as autopsy) is the post-mortem dissection of bodies after euthanasia or death and is a scientific examination conducted to observe and dissect the organs, collect tissues, and determine the extent of grossly evident disease. Research necropsies are conducted to obtain specific samples tailored according to study objectives. Diagnostic necropsy may be undertaken when unexpected illness or death occurs. The systematic collection of samples at necropsy is the critical first step in generating morphologic data from animal models. The morphologic (anatomic and histologic) data generates information on changes in cells, tissues, organs, and organ systems providing context for phenotypes (functional and morphological) to the level of the whole organism. Optimal insight into phenotype or pathophysiologic mechanisms is obtained when morphologic data is coupled with laboratory, medical, and molecular findings. This protocol provides a standard for an efficient routine mouse necropsy with brief comments on advanced or alternative techniques.
Collapse
Affiliation(s)
- Piper M Treuting
- Department of Comparative Medicine & Comparative Pathology Program, School of Medicine, University of Washington, Seattle, Washington
| | - Jessica M Snyder
- Department of Comparative Medicine & Comparative Pathology Program, School of Medicine, University of Washington, Seattle, Washington
| |
Collapse
|
195
|
Oczypok EA, Milutinovic PS, Alcorn JF, Khare A, Crum LT, Manni ML, Epperly MW, Pawluk AM, Ray A, Oury TD. Pulmonary receptor for advanced glycation end-products promotes asthma pathogenesis through IL-33 and accumulation of group 2 innate lymphoid cells. J Allergy Clin Immunol 2015; 136:747-756.e4. [PMID: 25930197 PMCID: PMC4562894 DOI: 10.1016/j.jaci.2015.03.011] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 03/04/2015] [Accepted: 03/06/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND Single nucleotide polymorphisms in the human gene for the receptor for advanced glycation end-products (RAGE) are associated with an increased incidence of asthma. RAGE is highly expressed in the lung and has been reported to play a vital role in the pathogenesis of murine models of asthma/allergic airway inflammation (AAI) by promoting expression of the type 2 cytokines IL-5 and IL-13. IL-5 and IL-13 are prominently secreted by group 2 innate lymphoid cells (ILC2s), which are stimulated by the proallergic cytokine IL-33. OBJECTIVE We sought to test the hypothesis that pulmonary RAGE is necessary for allergen-induced ILC2 accumulation in the lung. METHODS AAI was induced in wild-type and RAGE knockout mice by using IL-33, house dust mite extract, or Alternaria alternata extract. RAGE's lung-specific role in type 2 responses was explored with bone marrow chimeras and induction of gastrointestinal type 2 immune responses. RESULTS RAGE was found to drive AAI by promoting IL-33 expression in response to allergen and by coordinating the inflammatory response downstream of IL-33. Absence of RAGE impedes pulmonary accumulation of ILC2s in models of AAI. Bone marrow chimera studies suggest that pulmonary parenchymal, but not hematopoietic, RAGE has a central role in promoting AAI. In contrast to the lung, the absence of RAGE does not affect IL-33-induced ILC2 influx in the spleen, type 2 cytokine production in the peritoneum, or mucus hypersecretion in the gastrointestinal tract. CONCLUSIONS For the first time, this study demonstrates that a parenchymal factor, RAGE, mediates lung-specific accumulation of ILC2s.
Collapse
Affiliation(s)
- Elizabeth A Oczypok
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - Pavle S Milutinovic
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - John F Alcorn
- Department of Pediatrics, Children's Hospital of Pittsburgh, Pittsburgh, Pa
| | - Anupriya Khare
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - Lauren T Crum
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - Michelle L Manni
- Department of Pediatrics, Children's Hospital of Pittsburgh, Pittsburgh, Pa
| | - Michael W Epperly
- Department of Radiation Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, Pa
| | - Adriane M Pawluk
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - Anuradha Ray
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - Tim D Oury
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pa.
| |
Collapse
|
196
|
Saijo H, Tatsumi N, Arihiro S, Kato T, Okabe M, Tajiri H, Hashimoto H. Microangiopathy triggers, and inducible nitric oxide synthase exacerbates dextran sulfate sodium-induced colitis. J Transl Med 2015; 95:728-48. [PMID: 25938626 DOI: 10.1038/labinvest.2015.60] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 03/07/2015] [Accepted: 03/31/2015] [Indexed: 02/08/2023] Open
Abstract
Ulcerative colitis (UC) is a representative clinical manifestation of inflammatory bowel disease that causes chronic gastrointestinal tract inflammation. Dextran sulfate sodium (DSS)-induced colitis mice have been used to investigate UC pathogenesis, and in this UC model, disturbance and impairment of the mucosal epithelium have been reported to cause colitis. However, how DSS sporadically breaks down the epithelium remains unclear. In this study, we focused on the colonic microcirculation and myenteric neurons of DSS-induced colitis. Moreover, we examined the potential of myenteric neurons as a target to prevent exacerbation of colitis. Fluorescent angiographic and histopathological studies revealed that DSS administration elicited blood vessel disruption before epithelial disorders appeared. Ischemic conditions in the lamina propria induced inducible nitric oxide synthase (iNOS) expression in myenteric neurons as colitis aggravated. When neuronal activity was inhibited with butylscopolamine, neuronal iNOS expression decreased, and the exacerbation of colitis was prevented. These results suggested that DSS-induced colitis was triggered by microcirculatory disturbance in the mucosa, and that excessive neuronal excitation aggravated colitis. During remission periods of human UC, endoscopic inspection of the colonic microcirculation may enable the early detection of disease recurrence, and inhibition of neuronal iNOS expression may prevent the disease from worsening.
Collapse
Affiliation(s)
- Hiroki Saijo
- 1] Department of Anatomy, The Jikei University School of Medicine, Tokyo, Japan [2] Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Norifumi Tatsumi
- Department of Anatomy, The Jikei University School of Medicine, Tokyo, Japan
| | - Seiji Arihiro
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Tomohiro Kato
- Department of Endoscopy, The Jikei University School of Medicine, Tokyo, Japan
| | - Masataka Okabe
- Department of Anatomy, The Jikei University School of Medicine, Tokyo, Japan
| | - Hisao Tajiri
- 1] Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan [2] Department of Endoscopy, The Jikei University School of Medicine, Tokyo, Japan
| | - Hisashi Hashimoto
- Department of Anatomy, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
197
|
Scialis RJ, Csanaky IL, Goedken MJ, Manautou JE. Multidrug Resistance-Associated Protein 3 Plays an Important Role in Protection against Acute Toxicity of Diclofenac. Drug Metab Dispos 2015; 43:944-950. [PMID: 25897176 PMCID: PMC4468432 DOI: 10.1124/dmd.114.061705] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 04/20/2015] [Indexed: 11/22/2022] Open
Abstract
Diclofenac (DCF) is a nonsteroidal anti-inflammatory drug commonly prescribed to reduce pain in acute and chronic inflammatory diseases. One of the main DCF metabolites is a reactive diclofenac acyl glucuronide (DCF-AG) that covalently binds to biologic targets and may contribute to adverse drug reactions arising from DCF use. Cellular efflux of DCF-AG is partially mediated by multidrug resistance-associated proteins (Mrp). The importance of Mrp2 during DCF-induced toxicity has been established, yet the role of Mrp3 remains largely unexplored. In the present work, Mrp3-null (KO) mice were used to study the toxicokinetics and toxicodynamics of DCF and its metabolites. DCF-AG plasma concentrations were 90% lower in KO mice than in wild-type (WT) mice, indicating that Mrp3 mediates DCF-AG basolateral efflux. In contrast, there were no differences in DCF-AG biliary excretion between WT and KO, suggesting that only DCF-AG basolateral efflux is compromised by Mrp3 deletion. Susceptibility to toxicity was also evaluated after a single high DCF dose. No signs of injury were detected in livers and kidneys; however, ulcers were found in the small intestines. Furthermore, the observed intestinal injuries were consistently more severe in KO compared with WT. DCF covalent adducts were observed in liver and small intestines; however, staining intensity did not correlate with the severity of injuries, implying that tissues respond differently to covalent modification. Overall, the data provide strong evidence that (1) in vivo Mrp3 plays an important role in DCF-AG disposition and (2) compromised Mrp3 function can enhance injury in the gastrointestinal tract after DCF treatment.
Collapse
Affiliation(s)
- Renato J Scialis
- University of Connecticut, School of Pharmacy, Department of Pharmaceutical Sciences, Storrs, Connecticut (R.J.S., J.E.M.); University of Kansas Medical Center, Department of Internal Medicine, Kansas City, Kansas (I.L.C.); and Office of Translational Science, Rutgers University, Piscataway, New Jersey (M.J.G.)
| | - Iván L Csanaky
- University of Connecticut, School of Pharmacy, Department of Pharmaceutical Sciences, Storrs, Connecticut (R.J.S., J.E.M.); University of Kansas Medical Center, Department of Internal Medicine, Kansas City, Kansas (I.L.C.); and Office of Translational Science, Rutgers University, Piscataway, New Jersey (M.J.G.)
| | - Michael J Goedken
- University of Connecticut, School of Pharmacy, Department of Pharmaceutical Sciences, Storrs, Connecticut (R.J.S., J.E.M.); University of Kansas Medical Center, Department of Internal Medicine, Kansas City, Kansas (I.L.C.); and Office of Translational Science, Rutgers University, Piscataway, New Jersey (M.J.G.)
| | - José E Manautou
- University of Connecticut, School of Pharmacy, Department of Pharmaceutical Sciences, Storrs, Connecticut (R.J.S., J.E.M.); University of Kansas Medical Center, Department of Internal Medicine, Kansas City, Kansas (I.L.C.); and Office of Translational Science, Rutgers University, Piscataway, New Jersey (M.J.G.)
| |
Collapse
|
198
|
Akbari P, Braber S, Alizadeh A, Verheijden KAT, Schoterman MHC, Kraneveld AD, Garssen J, Fink-Gremmels J. Galacto-oligosaccharides Protect the Intestinal Barrier by Maintaining the Tight Junction Network and Modulating the Inflammatory Responses after a Challenge with the Mycotoxin Deoxynivalenol in Human Caco-2 Cell Monolayers and B6C3F1 Mice. J Nutr 2015; 145:1604-13. [PMID: 26019243 DOI: 10.3945/jn.114.209486] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 05/08/2015] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND The integrity of the epithelial layer in the gastrointestinal tract protects organisms from exposure to luminal antigens, which are considered the primary cause of chronic intestinal inflammation and allergic responses. The common wheat-associated fungal toxin deoxynivalenol acts as a specific disruptor of the intestinal tight junction network and hence might contribute to the pathogenesis of inflammatory bowel diseases. OBJECTIVE The aim of the current study was to assess whether defined galacto-oligosaccharides (GOSs) can prevent deoxynivalenol-induced epithelial dysfunction. METHODS Human epithelial intestinal Caco-2 cells, pretreated with different concentrations of GOSs (0.5%, 1%, and 2%) for 24 h, were stimulated with 4.2-μM deoxynivalenol (24 h), and 6/7-wk-old male B6C3F1 mice were fed a diet supplemented with 1% GOSs for 2 wk before being orally exposed to deoxynivalenol (25 mg/kg body weight, 6 h). Barrier integrity was determined by measuring transepithelial electrical resistance (TEER) and intestinal permeability to marker molecules. A calcium switch assay was conducted to study the assembly of epithelial tight junction proteins. Alterations in tight junction and cytokine expression were assessed by quantitative reverse transcriptase-polymerase chain reaction, Western blot analysis, or ELISA, and their localization was visualized by immunofluorescence microscopy. Sections of the proximal and distal small intestine were stained with hematoxylin/eosin for histomorphometric analysis. RESULTS The in vitro data showed that medium supplemented with 2% GOSs improved tight junction assembly reaching an acceleration of 85% after 6 h (P < 0.05). In turn, GOSs prevented the deoxynivalenol-induced loss of epithelial barrier function as measured by TEER (114% of control), and paracellular flux of Lucifer yellow (82.7% of prechallenge values, P < 0.05). Moreover, GOSs stabilized the expression and cellular distribution of claudin3 and suppressed by >50% the deoxynivalenol-induced synthesis and release of interleukin-8 [IL8/chemokine CXC motif ligand (CXCL8)] (P < 0.05). In mice, GOSs prevented the deoxynivalenol-induced mRNA overexpression of claudin3 (P = 0.022) and CXCL8 homolog keratinocyte hemoattractant (Kc) (Cxcl1) (P = 0.06) as well as the deoxynivalenol-induced morphologic defects. CONCLUSIONS The results demonstrate that GOSs stimulate the tight junction assembly and in turn mitigate the deleterious effects of deoxynivalenol on the intestinal barrier of Caco-2 cells and on villus architecture of B6C3F1 mice.
Collapse
Affiliation(s)
- Peyman Akbari
- Divisions of Veterinary Pharmacy, Pharmacology, and Toxicology, and Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Saskia Braber
- Divisions of Veterinary Pharmacy, Pharmacology, and Toxicology, and
| | - Arash Alizadeh
- Divisions of Veterinary Pharmacy, Pharmacology, and Toxicology, and Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Kim A T Verheijden
- Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | | | - Aletta D Kraneveld
- Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Johan Garssen
- Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands; Nutricia Research, Utrecht, The Netherlands
| | | |
Collapse
|
199
|
Bioimage analysis of Shigella infection reveals targeting of colonic crypts. Proc Natl Acad Sci U S A 2015; 112:E3282-90. [PMID: 26056271 DOI: 10.1073/pnas.1509091112] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Few studies within the pathogenic field have used advanced imaging and analytical tools to quantitatively measure pathogenicity in vivo. In this work, we present a novel approach for the investigation of host-pathogen processes based on medium-throughput 3D fluorescence imaging. The guinea pig model for Shigella flexneri invasion of the colonic mucosa was used to monitor the infectious process over time with GFP-expressing S. flexneri. A precise quantitative imaging protocol was devised to follow individual S. flexneri in a large tissue volume. An extensive dataset of confocal images was obtained and processed to extract specific quantitative information regarding the progression of S. flexneri infection in an unbiased and exhaustive manner. Specific parameters included the analysis of S. flexneri positions relative to the epithelial surface, S. flexneri density within the tissue, and volume of tissue destruction. In particular, at early time points, there was a clear association of S. flexneri with crypts, key morphological features of the colonic mucosa. Numerical simulations based on random bacterial entry confirmed the bias of experimentally measured S. flexneri for early crypt targeting. The application of a correlative light and electron microscopy technique adapted for thick tissue samples further confirmed the location of S. flexneri within colonocytes at the mouth of crypts. This quantitative imaging approach is a novel means to examine host-pathogen systems in a tailored and robust manner, inclusive of the infectious agent.
Collapse
|
200
|
MET is required for the recruitment of anti-tumoural neutrophils. Nature 2015; 522:349-53. [PMID: 25985180 PMCID: PMC4594765 DOI: 10.1038/nature14407] [Citation(s) in RCA: 366] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Accepted: 03/13/2015] [Indexed: 01/15/2023]
Abstract
Mutations or amplification of the MET proto-oncogene are involved in the pathogenesis of several tumours1-4, which rely on the constitutive engagement of this pathway for their growth and survival1,5. However, MET is expressed not only by cancer cells but also by tumour-associated stromal cells although its precise role in this compartment is not well characterized6-11. Here, we show that MET is required for neutrophil chemoattraction and cytotoxicity in response to its ligand HGF. Met deletion in neutrophils enhances tumour growth and metastasis. This phenotype correlates with reduced neutrophil infiltration to both primary tumour and metastatic site. Similarly, Met is necessary for neutrophil transudation during colitis, skin rash or peritonitis. Mechanistically, Met is induced by tumour-derived TNF-α or other inflammatory stimuli in both mouse and human neutrophils. This induction is instrumental for neutrophil transmigration across an activated endothelium and iNOS production upon HGF stimulation. Consequently, HGF/MET-dependent nitric oxide release by neutrophils promotes cancer cell killing, which abates tumour growth and metastasis. Following systemic administration of a MET kinase inhibitor, we prove that the therapeutic benefit of MET targeting in cancer cells is partly countered by the pro-tumoural effect rising from MET blockade in neutrophils. Our work identifies an unprecedented role of MET in neutrophils, suggests a potential “Achilles’ heel” of MET-targeted therapies in cancer, and supports the rationale for evaluating anti-MET drugs in certain inflammatory diseases.
Collapse
|