1
|
Tozzi M, Fiore A, Travaglione S, Marcon F, Rainaldi G, Germinario EAP, Laterza I, Donati S, Macchia D, Spada M, Leoni O, Quattrini MC, Pietraforte D, Tomasoni S, Torrigiani F, Verin R, Matarrese P, Gambardella L, Spadaro F, Carollo M, Pietrantoni A, Carlini F, Panebianco C, Pazienza V, Colella F, Lucchetti D, Sgambato A, Sistigu A, Moschella F, Guidotti M, Vincentini O, Maroccia Z, Biffoni M, De Angelis R, Bracci L, Fabbri A. E. Coli cytotoxic necrotizing factor-1 promotes colorectal carcinogenesis by causing oxidative stress, DNA damage and intestinal permeability alteration. J Exp Clin Cancer Res 2025; 44:29. [PMID: 39876002 PMCID: PMC11776187 DOI: 10.1186/s13046-024-03271-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 12/31/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Bacterial toxins are emerging as promising hallmarks of colorectal cancer (CRC) pathogenesis. In particular, Cytotoxic Necrotizing Factor 1 (CNF1) from E. coli deserves special consideration due to the significantly higher prevalence of this toxin gene in CRC patients with respect to healthy subjects, and to the numerous tumor-promoting effects that have been ascribed to the toxin in vitro. Despite this evidence, a definitive causal link between CNF1 and CRC was missing. Here we investigated whether CNF1 plays an active role in CRC onset by analyzing pro-carcinogenic key effects specifically induced by the toxin in vitro and in vivo. METHODS Viability assays, confocal microscopy of γH2AX and 53BP1 molecules and cytogenetic analysis were carried out to assess CNF1-induced genotoxicity on non-neoplastic intestinal epithelial cells. Caco-2 monolayers and 3D Caco-2 spheroids were used to evaluate permeability alterations specifically induced by CNF1, either in the presence or in the absence of inflammation. In vivo, an inflammatory bowel disease (IBD) model was exploited to evaluate the carcinogenic potential of CNF1. Immunohistochemistry and immunofluorescence stainings of formalin-fixed paraffin-embedded (FFPE) colon tissue were carried out as well as fecal microbiota composition analysis by 16 S rRNA gene sequencing. RESULTS CNF1 induces the release of reactive oxidizing species and chromosomal instability in non-neoplastic intestinal epithelial cells. In addition, CNF1 modifies intestinal permeability by directly altering tight junctions' distribution in 2D Caco-2 monolayers, and by hindering the differentiation of 3D Caco-2 spheroids with an irregular arrangement of these junctions. In vivo, repeated intrarectal administration of CNF1 induces the formation of dysplastic aberrant crypt foci (ACF), and produces the formation of colorectal adenomas in an IBD model. These effects are accompanied by the increased neutrophilic infiltration in colonic tissue, by a mixed pro-inflammatory and anti-inflammatory cytokine milieu, and by the pro-tumoral modulation of the fecal microbiota. CONCLUSIONS Taken together, our results support the hypothesis that the CNF1 toxin from E. coli plays an active role in colorectal carcinogenesis. Altogether, these findings not only add new knowledge to the contribution of bacterial toxins to CRC, but also pave the way to the implementation of current screening programs and preventive strategies.
Collapse
Affiliation(s)
- Michela Tozzi
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Alessia Fiore
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, Rome, Italy
| | - Sara Travaglione
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, Rome, Italy
| | - Francesca Marcon
- Department of Environment and Health, Istituto Superiore di Sanità, Rome, Italy
| | - Gabriella Rainaldi
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Elena Angela Pia Germinario
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, Rome, Italy
| | - Ilenia Laterza
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, Rome, Italy
| | - Simona Donati
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Daniele Macchia
- Center of Animal Research and Welfare, Istituto Superiore di Sanità, Rome, Italy
| | - Massimo Spada
- Center of Animal Research and Welfare, Istituto Superiore di Sanità, Rome, Italy
| | - Omar Leoni
- Center of Animal Research and Welfare, Istituto Superiore di Sanità, Rome, Italy
| | | | | | - Sofia Tomasoni
- Department of Comparative Biomedicine and Food Science, BCA-University of Padua, Legnaro, PD, Italy
| | - Filippo Torrigiani
- Department of Comparative Biomedicine and Food Science, BCA-University of Padua, Legnaro, PD, Italy
| | - Ranieri Verin
- Department of Comparative Biomedicine and Food Science, BCA-University of Padua, Legnaro, PD, Italy
| | - Paola Matarrese
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | | | | | - Maria Carollo
- Core Facilities, Istituto Superiore di Sanità, Rome, Italy
| | | | - Francesca Carlini
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, Rome, Italy
| | - Concetta Panebianco
- Division of Gastroenterology, Fondazione IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, FG, Italy
| | - Valerio Pazienza
- Division of Gastroenterology, Fondazione IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, FG, Italy
| | - Filomena Colella
- Multiplex Spatial Profiling Center, Fondazione Policlinico Universitario "A. Gemelli" - IRCCS, Rome, Italy
| | - Donatella Lucchetti
- Multiplex Spatial Profiling Center, Fondazione Policlinico Universitario "A. Gemelli" - IRCCS, Rome, Italy
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Alessandro Sgambato
- Multiplex Spatial Profiling Center, Fondazione Policlinico Universitario "A. Gemelli" - IRCCS, Rome, Italy
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Antonella Sistigu
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario "A. Gemelli" - IRCCS, Rome, Italy
| | - Federica Moschella
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Marco Guidotti
- Department of Food Safety, Nutrition and Veterinary Public Health, Istituto Superiore di Sanità, Rome, Italy
| | - Olimpia Vincentini
- Department of Food Safety, Nutrition and Veterinary Public Health, Istituto Superiore di Sanità, Rome, Italy
| | - Zaira Maroccia
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, Rome, Italy
| | - Mauro Biffoni
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Roberta De Angelis
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Laura Bracci
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy.
| | - Alessia Fabbri
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
2
|
McKnight CA, Diehl LJ, Bergin IL. Digestive Tract and Salivary Glands. HASCHEK AND ROUSSEAUX' S HANDBOOK OF TOXICOLOGIC PATHOLOGY 2024:1-148. [DOI: 10.1016/b978-0-12-821046-8.00001-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
3
|
Le Naour J, Montégut L, Pan Y, Scuderi SA, Cordier P, Joseph A, Sauvat A, Iebba V, Paillet J, Ferrere G, Brechard L, Mulot C, Dubourg G, Zitvogel L, Pol JG, Vacchelli E, Puig PL, Kroemer G. Formyl peptide receptor-1 (FPR1) represses intestinal oncogenesis. Oncoimmunology 2023; 12:2237354. [PMID: 37492227 PMCID: PMC10364666 DOI: 10.1080/2162402x.2023.2237354] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 07/27/2023] Open
Abstract
Formyl peptide receptor-1 (FPR1) is a pattern recognition receptor that is mostly expressed by myeloid cells. In patients with colorectal cancer (CRC), a loss-of-function polymorphism (rs867228) in the gene coding for FPR1 has been associated with reduced responses to chemotherapy or chemoradiotherapy. Moreover, rs867228 is associated with accelerated esophageal and colorectal carcinogenesis. Here, we show that dendritic cells from Fpr1-/- mice exhibit reduced migration in response to chemotherapy-treated CRC cells. Moreover, Fpr1-/- mice are particularly susceptible to chronic ulcerative colitis and colorectal oncogenesis induced by the mutagen azoxymethane followed by oral dextran sodium sulfate, a detergent that induces colitis. These experiments were performed after initial co-housing of Fpr1-/- mice and wild-type controls, precluding major Fpr1-driven differences in the microbiota. Pharmacological inhibition of Fpr1 by cyclosporin H also tended to increase intestinal oncogenesis in mice bearing the ApcMin mutation, and this effect was reversed by the anti-inflammatory drug sulindac. We conclude that defective FPR1 signaling favors intestinal tumorigenesis through the modulation of the innate inflammatory/immune response.
Collapse
Affiliation(s)
- Julie Le Naour
- Centre de Recherche des Cordeliers, Equipe Labellisée Par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Villejuif, France
- Faculty of Medicine Kremlin Bicêtre, Université Paris Saclay, Le Kremlin Bicêtre, France
| | - Léa Montégut
- Centre de Recherche des Cordeliers, Equipe Labellisée Par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Villejuif, France
- Faculty of Medicine Kremlin Bicêtre, Université Paris Saclay, Le Kremlin Bicêtre, France
| | - Yuhong Pan
- Centre de Recherche des Cordeliers, Equipe Labellisée Par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Villejuif, France
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Sarah Adriana Scuderi
- Centre de Recherche des Cordeliers, Equipe Labellisée Par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Villejuif, France
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Pierre Cordier
- Laboratory of Proliferation, Stress and Liver Physiopathology, Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, Paris, France
| | - Adrien Joseph
- Centre de Recherche des Cordeliers, Equipe Labellisée Par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Villejuif, France
- Faculty of Medicine Kremlin Bicêtre, Université Paris Saclay, Le Kremlin Bicêtre, France
| | - Allan Sauvat
- Centre de Recherche des Cordeliers, Equipe Labellisée Par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Villejuif, France
| | - Valerio Iebba
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Juliette Paillet
- Centre de Recherche des Cordeliers, Equipe Labellisée Par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Villejuif, France
- Faculty of Medicine Kremlin Bicêtre, Université Paris Saclay, Le Kremlin Bicêtre, France
| | - Gladys Ferrere
- Institut National de la Santé Et de la Recherche Medicale (INSERM) U1015 and Equipe Labellisée–Ligue Nationale Contre le Cancer, Villejuif, France
| | - Ludivine Brechard
- Aix Marseille Univ, IRD, AP-HM, MEPHI, IHU Méditerranée Infection, Marseille, France
| | - Claire Mulot
- Centre de Recherche des Cordeliers, Equipe Labélisée Ligue Contre le Cancer, Sorbonne Université, Université Paris Cité, INSERM, Paris, France
| | - Grégory Dubourg
- Aix Marseille Univ, IRD, AP-HM, MEPHI, IHU Méditerranée Infection, Marseille, France
| | - Laurence Zitvogel
- Faculty of Medicine Kremlin Bicêtre, Université Paris Saclay, Le Kremlin Bicêtre, France
- Center of Clinical Investigations BIOTHERIS, INSERM CIC1428, Gustave Roussy, Villejuif, France
- Institut National de la Santé Et de la Recherche Médicale, UMR1015, Gustave Roussy, Villejuif, France
- Gustave Roussy Cancer Center, Villejuif, France
| | - Jonathan G. Pol
- Centre de Recherche des Cordeliers, Equipe Labellisée Par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Villejuif, France
| | - Erika Vacchelli
- Centre de Recherche des Cordeliers, Equipe Labellisée Par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Villejuif, France
| | - Pierre-Laurent Puig
- Centre de Recherche des Cordeliers, Equipe Labélisée Ligue Contre le Cancer, Sorbonne Université, Université Paris Cité, INSERM, Paris, France
- Institut du Cancer Paris CARPEM, APHP. Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe Labellisée Par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Villejuif, France
- Institut du Cancer Paris CARPEM, APHP. Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| |
Collapse
|
4
|
Arthrospira (Spirulina) platensis feeding reduces the early stage of chemically induced rat colon carcinogenesis. Br J Nutr 2023; 129:395-405. [PMID: 35506448 DOI: 10.1017/s0007114522001350] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Colorectal cancer is the third most diagnosed cancer worldwide and linked to dietary/lifestyle factors. Arthrospira (Spirulina) platensis (AP) contains bioactive compounds with beneficial effects in vivo/in vitro. We evaluated the effects of AP feeding against 1,2-dimethylhydrazine (DMH)-induced colon carcinogenesis. Male Sprague Dawley rats were given subcutaneous injections of DMH (4 × 40 mg/kg body weight) (G1-G3) or vehicle (G4-G5) twice a week (weeks 3-4). During weeks 1-4, animals were fed a diet containing 1 % (G2) or 2 % (G3-G4) AP powder (w/w). After this period, all groups received a balanced diet until week 12. Some animals were euthanised after the last DMH injection (week 4) for histological, immunohistochemical (Ki-67, γ-H2AX and caspase-3) and molecular analyses (real time-PCR for 91 genes), while other animals were euthanised at week 12 for preneoplastic aberrant crypt foci (ACF) analysis. Both AP treatments (G2-G3) significantly decreased the DMH-induced increase in γ-H2AX (DNA damage) and caspase 3 (DNA damage-induced cell death) in colonic crypts at week 4. In addition, Cyp2e1 (Drug metabolism), Notch1, Notch2 and Jag1 genes (Notch pathway) and Atm, Wee1, Chek2, Mgmt, Ogg1 and Xrcc6 genes (DNA repair) were also down-regulated by 2 % AP feeding (G3) at week 4. A significant reduction in ACF development was observed in both AP-treated groups (G2-G3) at week 12. In conclusion, findings indicate that AP feeding reduced acute colonic damage after DMH, resulting in fewer preneoplastic lesions. Our study provided mechanistic insights on dietary AP-preventive effects against early colon carcinogenesis.
Collapse
|
5
|
Madka V, Kumar G, Pathuri G, Panneerselvam J, Zhang Y, Ganta V, Lightfoot S, Lubet RA, Suen CS, Steele VE, Janakiram NB, Mohammed A, Rao CV. Proton pump inhibitor omeprazole suppresses carcinogen induced colonic adenoma progression to adenocarcinoma in F344 rat. Cancer Prev Res (Phila) 2021; 14:1009-1020. [PMID: 34341012 DOI: 10.1158/1940-6207.capr-21-0057] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 05/25/2021] [Accepted: 07/09/2021] [Indexed: 11/16/2022]
Abstract
Colorectal cancer (CRC) causes over 53,000 deaths annually in the United States. Its rising incidences worldwide and particularly in young adults is a major concern. Here, we evaluated the efficacy of omeprazole (OME) that is clinically approved for treating acid-reflux, to enable its repurposing for CRC prevention. In the azoxymethane (AOM)-induced rat CRC model, dietary OME (250 and 500 ppm) was administered at early adenoma stage (8 weeks after AOM) to assess the progression of early lesions to adenocarcinoma. Administration of OME at 250 ppm or 500 ppm doses led to suppression of total colon adenocarcinoma incidence by 15.7% and 32% (p<0.01), respectively. Importantly, invasive carcinoma incidence was reduced by 59% (p<0.0005) and 90% (p<0.0001) in OME administered rats in a dose-dependent manner. There was also a strong and dose-dependent inhibition in the adenocarcinoma multiplicity in rats exposed to OME. Administration of 250 and 500 ppm OME inhibited total colon adenocarcinoma multiplicity by ~49% and ~65% (p<0.0001), respectively. While non-invasive adenocarcinomas multiplicity was suppressed by ~34% to ~48% (p<0.02), the invasive carcinomas multiplicity was reduced by ~74% to ~94% (p<0.0001) in OME exposed rats in comparison to the untreated rats. Biomarker analysis results showed a decrease in cell proliferation and anti-apoptotic/pro-survival proteins with an increase in apoptosis. Transcriptome analysis of treated tumors revealed a significant increase in adenocarcinoma inhibitory genes (Olmf4; Spink4) expression and down regulation of progression promoting genes (SerpinA1, MMP21, IL6). In summary, OME showed significant protection against the progression of adenoma to adenocarcinoma.
Collapse
Affiliation(s)
- Venkateshwar Madka
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hematology/Oncology Section, University of Oklahoma Health Sciences Center
| | - Gaurav Kumar
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hematology/Oncology Section, University of Oklahoma Health Sciences Center
| | - Gopal Pathuri
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hematology/Oncology Section, University of Oklahoma Health Sciences Center
| | - Janani Panneerselvam
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hematology/Oncology Section, University of Oklahoma Health Sciences Center
| | - Yuting Zhang
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hematology/Oncology Section, University of Oklahoma Health Sciences Center
| | - Vishal Ganta
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hematology/Oncology Section, University of Oklahoma Health Sciences Center
| | - Stanley Lightfoot
- Pathology-Retired, Center for Cancer Prevention and Drug Development
| | - Ronald A Lubet
- Division of Cancer Prevention, National Cancer Institute
| | - Chen S Suen
- Cancer Prevention, National Cancer Institute
| | | | | | - Altaf Mohammed
- Division of Cancer Prevention, National Cancer Institute
| | - Chinthalapally V Rao
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hematology/Oncology Section, University of Oklahoma Health Sciences Center
| |
Collapse
|
6
|
Zhao Q, Bi Y, Zhong J, Ren Z, Liu Y, Jia J, Yu M, Tan Y, Zhang Q, Yu X. Pristimerin suppresses colorectal cancer through inhibiting inflammatory responses and Wnt/β-catenin signaling. Toxicol Appl Pharmacol 2019; 386:114813. [PMID: 31715269 DOI: 10.1016/j.taap.2019.114813] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 11/01/2019] [Accepted: 11/08/2019] [Indexed: 02/07/2023]
Abstract
Pristimerin, a triterpenoid, has exhibited potential anti-inflammatory and anti-tumor activities. Nevertheless, the role and mechanism of pristimerin in intestinal inflammation and colon cancer require further investigation. Here, we found that pristimerin protected mice from dextran sulfate sodium (DSS)-induced colitis, restoring epithelial damage and reducing tissue inflammation and inflammatory cell infiltration. In addition, pristimerin dramatically reduced the number and size of the tumors in a azoxymethane (AOM)/DSS-induced colitis-associated colorectal cancer (CAC) model. Furthermore, we found that pristimerin suppressed Wnt/β-catenin signaling by RNA-Seq. Pristimerin inhibited Wnt/β-catenin signaling via activation of GSK3β, thereby suppressing Wnt target gene expression in colon cancer HCT116 and HT-29 cells. In HCT116 colon cancer xenografts and APCmin/+ mice, which undergo spontaneous intestinal tumorigenesis, administration of pristimerin reduced the tumor progression and decreased the expression of phosphorylated GSK3β Ser 9, β-catenin, cyclin D1 and c-Myc. These results suggest that pristimerin is a potent agent for preventing colon inflammation and carcinogenesis.
Collapse
Affiliation(s)
- Qun Zhao
- Laboratory of Inflammation and Molecular Pharmacology, School of Basic Medical Sciences & Biomedical Research Institute, Hubei University of Medicine, Shiyan 442000, China; Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan 442000, China
| | - Yun Bi
- Laboratory of Inflammation and Molecular Pharmacology, School of Basic Medical Sciences & Biomedical Research Institute, Hubei University of Medicine, Shiyan 442000, China
| | - Jing Zhong
- Laboratory of Inflammation and Molecular Pharmacology, School of Basic Medical Sciences & Biomedical Research Institute, Hubei University of Medicine, Shiyan 442000, China; Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China
| | - Ziting Ren
- Laboratory of Inflammation and Molecular Pharmacology, School of Basic Medical Sciences & Biomedical Research Institute, Hubei University of Medicine, Shiyan 442000, China
| | - Yingxiang Liu
- Laboratory of Inflammation and Molecular Pharmacology, School of Basic Medical Sciences & Biomedical Research Institute, Hubei University of Medicine, Shiyan 442000, China
| | - Junjun Jia
- Laboratory of Inflammation and Molecular Pharmacology, School of Basic Medical Sciences & Biomedical Research Institute, Hubei University of Medicine, Shiyan 442000, China
| | - Mengting Yu
- Laboratory of Inflammation and Molecular Pharmacology, School of Basic Medical Sciences & Biomedical Research Institute, Hubei University of Medicine, Shiyan 442000, China
| | - Yan Tan
- Laboratory of Inflammation and Molecular Pharmacology, School of Basic Medical Sciences & Biomedical Research Institute, Hubei University of Medicine, Shiyan 442000, China
| | - Qiufang Zhang
- Laboratory of Inflammation and Molecular Pharmacology, School of Basic Medical Sciences & Biomedical Research Institute, Hubei University of Medicine, Shiyan 442000, China
| | - Xianjun Yu
- Laboratory of Inflammation and Molecular Pharmacology, School of Basic Medical Sciences & Biomedical Research Institute, Hubei University of Medicine, Shiyan 442000, China; Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan 442000, China.
| |
Collapse
|
7
|
Knoblaugh SE, Hohl TM, La Perle KMD. Pathology Principles and Practices for Analysis of Animal Models. ILAR J 2019; 59:40-50. [PMID: 31053847 DOI: 10.1093/ilar/ilz001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 01/03/2019] [Indexed: 12/18/2022] Open
Abstract
Over 60% of NIH extramural funding involves animal models, and approximately 80% to 90% of these are mouse models of human disease. It is critical to translational research that animal models are accurately characterized and validated as models of human disease. Pathology analysis, including histopathology, is essential to animal model studies by providing morphologic context to in vivo, molecular, and biochemical data; however, there are many considerations when incorporating pathology endpoints into an animal study. Mice, and in particular genetically modified models, present unique considerations because these modifications are affected by background strain genetics, husbandry, and experimental conditions. Comparative pathologists recognize normal pathobiology and unique phenotypes that animals, including genetically modified models, may present. Beyond pathology, comparative pathologists with research experience offer expertise in animal model development, experimental design, optimal specimen collection and handling, data interpretation, and reporting. Critical pathology considerations in the design and use of translational studies involving animals are discussed, with an emphasis on mouse models.
Collapse
Affiliation(s)
- Sue E Knoblaugh
- Department of Veterinary Biosciences, and Comparative Pathology & Mouse Phenotyping Shared Resource, The Ohio State University, Columbus, Ohio
| | - Tobias M Hohl
- Infectious Diseases Service, Memorial Sloan Kettering Cancer Center, New York City, New York
| | - Krista M D La Perle
- Department of Veterinary Biosciences, and Comparative Pathology & Mouse Phenotyping Shared Resource, The Ohio State University, Columbus, Ohio
| |
Collapse
|
8
|
Croes L, Fransen E, Hylebos M, Buys K, Hermans C, Broeckx G, Peeters M, Pauwels P, Op de Beeck K, Van Camp G. Determination of the Potential Tumor-Suppressive Effects of Gsdme in a Chemically Induced and in a Genetically Modified Intestinal Cancer Mouse Model. Cancers (Basel) 2019; 11:cancers11081214. [PMID: 31434357 PMCID: PMC6721630 DOI: 10.3390/cancers11081214] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 08/06/2019] [Accepted: 08/13/2019] [Indexed: 02/06/2023] Open
Abstract
Gasdermin E (GSDME), also known as deafness autosomal dominant 5 (DFNA5) and previously identified to be an inducer of regulated cell death, is frequently epigenetically inactivated in different cancer types, suggesting that GSDME is a tumor suppressor gene. In this study, we aimed to evaluate the tumor-suppressive effects of GSDME in two intestinal cancer mouse models. To mimic the silencing of GSDME by methylation as observed in human cancers, a Gsdme knockout (KO) mouse was developed. The effect of GSDME on tumorigenesis was studied both in a chemically induced and in a genetic intestinal cancer mouse model, as strong evidence shows that GSDME plays a role in human colorectal cancer and representative mouse models for intestinal cancer are available. Azoxymethane (AOM) was used to induce colorectal tumors in the chemically induced intestinal cancer model (n = 100). For the genetic intestinal cancer model, Apc1638N/+ mice were used (n = 37). In both experiments, the number of mice bearing microscopic proliferative lesions, the number and type of lesions per mouse and the histopathological features of the adenocarcinomas were compared between Gsdme KO and wild type (WT) mice. Unfortunately, we found no major differences between Gsdme KO and WT mice, neither for the number of affected mice nor for the multiplicity of proliferative lesions in the mice. However, recent breakthroughs on gasdermin function indicate that GSDME is an executioner of necrotic cell death. Therefore, it is possible that GSDME may be important for creating an inflammatory microenvironment around the tumor. This is in line with the trend towards more severe inflammation in WT compared to Gsdme KO mice, that we observed in our study. We conclude that the effect of GSDME in tumor biology is probably more subtle than previously thought.
Collapse
Affiliation(s)
- Lieselot Croes
- Center of Medical Genetics, University of Antwerp, Prins Boudewijnlaan 43/6, Edegem, BE-2650 Antwerp, Belgium
- Center for Oncological Research, University of Antwerp, Universiteitsplein 1, Wilrijk, BE-2610 Antwerp, Belgium
| | - Erik Fransen
- Center of Medical Genetics, University of Antwerp, Prins Boudewijnlaan 43/6, Edegem, BE-2650 Antwerp, Belgium
- StatUa Center for Statistics, University of Antwerp, Prinsstraat 13, BE-2000 Antwerp, Belgium
| | - Marieke Hylebos
- Center of Medical Genetics, University of Antwerp, Prins Boudewijnlaan 43/6, Edegem, BE-2650 Antwerp, Belgium
- Center for Oncological Research, University of Antwerp, Universiteitsplein 1, Wilrijk, BE-2610 Antwerp, Belgium
| | - Kimberly Buys
- Center of Medical Genetics, University of Antwerp, Prins Boudewijnlaan 43/6, Edegem, BE-2650 Antwerp, Belgium
- Center for Oncological Research, University of Antwerp, Universiteitsplein 1, Wilrijk, BE-2610 Antwerp, Belgium
| | - Christophe Hermans
- Center for Oncological Research, University of Antwerp, Universiteitsplein 1, Wilrijk, BE-2610 Antwerp, Belgium
| | - Glenn Broeckx
- Department of Pathology, Antwerp University Hospital, Wilrijkstraat 10, Edegem, BE-2650 Antwerp, Belgium
| | - Marc Peeters
- Center for Oncological Research, University of Antwerp, Universiteitsplein 1, Wilrijk, BE-2610 Antwerp, Belgium
- Department of Medical Oncology, Antwerp University Hospital, Wilrijkstraat 10, Edegem, BE-2650 Antwerp, Belgium
| | - Patrick Pauwels
- Center for Oncological Research, University of Antwerp, Universiteitsplein 1, Wilrijk, BE-2610 Antwerp, Belgium
- Department of Pathology, Antwerp University Hospital, Wilrijkstraat 10, Edegem, BE-2650 Antwerp, Belgium
| | - Ken Op de Beeck
- Center of Medical Genetics, University of Antwerp, Prins Boudewijnlaan 43/6, Edegem, BE-2650 Antwerp, Belgium
- Center for Oncological Research, University of Antwerp, Universiteitsplein 1, Wilrijk, BE-2610 Antwerp, Belgium
| | - Guy Van Camp
- Center of Medical Genetics, University of Antwerp, Prins Boudewijnlaan 43/6, Edegem, BE-2650 Antwerp, Belgium.
- Center for Oncological Research, University of Antwerp, Universiteitsplein 1, Wilrijk, BE-2610 Antwerp, Belgium.
| |
Collapse
|
9
|
Romualdo GR, Rocha AB, Vinken M, Cogliati B, Moreno FS, Chaves MAG, Barbisan LF. Drinking for protection? Epidemiological and experimental evidence on the beneficial effects of coffee or major coffee compounds against gastrointestinal and liver carcinogenesis. Food Res Int 2019; 123:567-589. [PMID: 31285007 DOI: 10.1016/j.foodres.2019.05.029] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 05/15/2019] [Accepted: 05/20/2019] [Indexed: 02/07/2023]
Abstract
Recent meta-analyses indicate that coffee consumption reduces the risk for digestive tract (oral, esophageal, gastric and colorectal) and, especially, liver cancer. Coffee bean-derived beverages, as the widely-consumed espresso and "common" filtered brews, present remarkable historical, cultural and economic importance globally. These drinks have rich and variable chemical composition, depending on factors that vary from "seeding to serving". The alkaloids caffeine and trigonelline, as well as the polyphenol chlorogenic acid, are some of the most important bioactive organic compounds of these beverages, displaying high levels in both espresso and common brews and/or increased bioavailability after consumption. Thus, we performed a comprehensive literature overview of current knowledge on the effects of coffee beverages and their highly bioavailable compounds, describing: 1) recent epidemiological and experimental findings highlighting the beneficial effects against gastrointestinal/liver carcinogenesis, and 2) the main molecular mechanisms in these in vitro and in vivo bioassays. Findings predominantly address the protective effects of coffee beverages and their most common/bioavailable compounds individually on gastrointestinal and liver cancer development. Caffeine, trigonelline and chlorogenic acid modulate common molecular targets directly implicated in key cancer hallmarks, what could stimulate novel translational or population-based mechanistic investigations.
Collapse
Affiliation(s)
| | | | - Mathieu Vinken
- Department of In Vitro Toxicology and Dermato-Cosmetology, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Bruno Cogliati
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo (USP), São Paulo, Brazil
| | - Fernando Salvador Moreno
- Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - María Angel García Chaves
- Department of Oncology, Biosanitary Research Institute of Granada (ibs.GRANADA), University Hospitals of Granada-University of Granada, Granada, Spain
| | - Luis Fernando Barbisan
- Department of Morphology, Biosciences Institute, São Paulo State University (UNESP), Botucatu, Brazil.
| |
Collapse
|
10
|
Chandrasekaran B, Pal D, Kolluru V, Tyagi A, Baby B, Dahiya NR, Youssef K, Alatassi H, Ankem MK, Sharma AK, Damodaran C. The chemopreventive effect of withaferin A on spontaneous and inflammation-associated colon carcinogenesis models. Carcinogenesis 2018; 39:1537-1547. [PMID: 30124785 PMCID: PMC6314334 DOI: 10.1093/carcin/bgy109] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 07/06/2018] [Accepted: 08/09/2018] [Indexed: 02/05/2023] Open
Abstract
Chemopreventive effects and associated mechanisms of withaferin A (WA) against intestinal and colon carcinogenesis remain unknown. We investigated the chemopreventive effect of WA on transgenic adenomatous polyposis coli (APCMin/+) mouse and chemically induced azoxymethane/dextran sodium sulfate (AOM/DSS) models of intestinal and colon carcinogenesis. Oral WA administration (4 and 3 mg/kg) inhibited tumor initiation and progression of intestinal polyps formation in APCMin/+ mice and colon carcinogenesis in the AOM/DSS mouse model. WA-administered mice showed a significant reduction in both number [duodenum, 33% (P > 0.05); jejunum, 32% (P < 0.025); ileum, 43% ( P < 0.001); and colon 59% (P < 0.01] and size of polyps in APCMin/+ mice compared with the respective controls. Similarly, tumor multiplicity was significantly reduced (P < 0.05) in the colon of WA-administered AOM/DSS mice. Pathological analysis showed reduced adenomas and tissue inflammation in WA-administered mouse models. Molecular studies suggested that WA inhibited the expression of inflammatory (interluekin-6, tumor necrosis factor-alpha and cyclooxygenase-2), pro-survival (pAKT, Notch1 and NF-κB) markers in APCMin/+ and AOM/DSS models. The results suggest that WA is a potent agent for preventing colon carcinogenesis and further investigation is required to show clinical utility of the agent.
Collapse
Affiliation(s)
| | - Deeksha Pal
- Department of Urology, University of Louisville, Louisville, KY, USA
| | - Venkatesh Kolluru
- Department of Urology, University of Louisville, Louisville, KY, USA
| | - Ashish Tyagi
- Department of Urology, University of Louisville, Louisville, KY, USA
| | - Becca Baby
- Department of Urology, University of Louisville, Louisville, KY, USA
| | - Nisha R Dahiya
- Department of Urology, University of Louisville, Louisville, KY, USA
| | - Khafateh Youssef
- Department of Pathology and Laboratory Medicine, University of Louisville, Louisville, KY, USA
| | - Houda Alatassi
- Department of Pathology and Laboratory Medicine, University of Louisville, Louisville, KY, USA
| | - Murali K Ankem
- Department of Urology, University of Louisville, Louisville, KY, USA
| | - Arun K Sharma
- Department of Pharmacology, Penn State Cancer Institute, Penn State College of Medicine, Hershey, PA, USA
| | - Chendil Damodaran
- Department of Urology, University of Louisville, Louisville, KY, USA
| |
Collapse
|
11
|
Cullen JM, Ward JM, Thompson CM. Reevaluation and Classification of Duodenal Lesions in B6C3F1 Mice and F344 Rats from 4 Studies of Hexavalent Chromium in Drinking Water. Toxicol Pathol 2016; 44:279-89. [PMID: 26538584 PMCID: PMC4785997 DOI: 10.1177/0192623315611501] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Thirteen-week and 2-year drinking water studies conducted by the National Toxicology Program (NTP) reported that hexavalent chromium (Cr(VI)) induced diffuse epithelial hyperplasia in the duodenum of B6C3F1 mice but not F344 rats. In the 2-year study, Cr(VI) exposure was additionally associated with duodenal adenomas and carcinomas in mice only. Subsequent 13-week Cr(VI) studies conducted by another group demonstrated non-neoplastic duodenal lesions in B6C3F1 mice similar to those of the NTP study as well as mild duodenal hyperplasia in F344 rats. Because intestinal lesions in mice are the basis for proposed safety standards for Cr(VI), and the histopathology data are relevant to the mode of action, consistency (an important Hill criterion for causality) was assessed across the aforementioned studies. Two veterinary pathologists applied uniform diagnostic criteria to the duodenal lesions in rats and mice from the 4 repeated-dose studies. Comparable non-neoplastic intestinal lesions were evident in mice and rats from all 4 studies; however, the incidence and severity of intestinal lesions were greater in mice than rats. These findings demonstrate consistency across studies and species and highlight the importance of standardized nomenclature for intestinal pathology. The differences in the severity of non-neoplastic lesions also likely contribute to the differential tumor response.
Collapse
Affiliation(s)
- John M Cullen
- College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | | | | |
Collapse
|
12
|
Yoneda M, Molinolo AA, Ward JM, Kimura S, Goodlad RA. A Simple Device to Rapidly Prepare Whole Mounts of the Mouse Intestine. J Vis Exp 2015:e53042. [PMID: 26556642 DOI: 10.3791/53042] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Preparing whole mounts of the mouse small intestine and colon for subsequent analysis or quantification can be time consuming and difficult. We describe the use of a simple device to cut and 'roll' mouse intestines to rapidly prepare whole mount preparations of superior and uniform quality to that which can be achieved by hand. The device comprises a base that holds 4 stainless steel rods and a top, which acts a cutting guide. The rods are inserted into the lumen of the small intestine [divided into thirds] and the colon. The rods and samples are then placed over a piece of filter paper or card into the holding slots in the base of the device. The top of the device is then positioned and serves as a cutting guide. The two angled sections in the center of the top piece are used to guide a knife or scalpel and cut the intestines longitudinally on the top of the rods. Once the intestinal sections have been cut, the top is removed and the card, tissue and rods gently removed from the device and placed on the bench. The rods are then gently rolled sideways to flatten and stick the intestinal segments onto the underlying piece of filter paper or card. The final preparation can then be examined or fixed and stored for later analysis. The preparations are invaluable for the study of intestinal changes in normal or genetically modified mouse models. The preparations have been used for the study and quantification of the effects of inflammation (colitis), damage, pre-cancerous lesions (aberrant crypt foci (ACFs) and mucin depleted foci (MDFs)) and polyps or tumors.
Collapse
Affiliation(s)
- Mitsuhiro Yoneda
- National Cancer Institute, Laboratory of Metabolism, National Institutes of Health
| | - Alfredo A Molinolo
- National Institute of Dental and Craniofacial Research, Oral and Pharyngeal Cancer Branch, National Institutes of Health
| | | | - Shioko Kimura
- National Cancer Institute, Laboratory of Metabolism, National Institutes of Health;
| | | |
Collapse
|
13
|
Doulberis M, Angelopoulou K, Kaldrymidou E, Tsingotjidou A, Abas Z, Erdman SE, Poutahidis T. Cholera-toxin suppresses carcinogenesis in a mouse model of inflammation-driven sporadic colon cancer. Carcinogenesis 2014; 36:280-90. [PMID: 25550315 DOI: 10.1093/carcin/bgu325] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Human studies and clues from animal models have provided important links between gastrointestinal (GI) tract bacteria and colon cancer. Gut microbiota antigenic stimuli play an important role in shaping the intestinal immune responses. Therefore, especially in the case of inflammation-associated colon cancer, gut bacteria antigens may affect tumorigenesis. The present study aimed to investigate the effects of the oral administration of a bacterial product with known immunomodulatory properties on inflammation-driven colorectal neoplasmatogenesis. For that, we used cholera-toxin and a well-established mouse model of colon cancer in which neoplasia is initiated by a single dose of the genotoxic agent azoxymethane (AOM) and subsequently promoted by inflammation caused by the colitogenic substance dextran sodium sulfate (DSS). We found that a single, low, non-pathogenic dose of CT, given orally at the beginning of each DSS treatment cycle downregulated neutrophils and upregulated regulatory T-cells and IL-10 in the colonic mucosa. The CT-induced disruption of the tumor-promoting character of DSS-induced inflammation led to the reduction of the AOM-initiated colonic polypoidogenesis. This result adds value to the emerging notion that certain GI tract bacteria or their products affect the immune system and render the microenvironment of preneoplastic lesions less favorable for promoting their evolution to cancer.
Collapse
Affiliation(s)
- Michael Doulberis
- Laboratory of Pathology, Laboratory of Biochemistry and Toxicology and Laboratory of Anatomy, Histology and Embryology, Faculty of Health Sciences, School of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece, Department of Agricultural Development, Democritus University of Thrace, Orestiada 68200, Greece and Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | - Eleni Kaldrymidou
- Laboratory of Pathology, Laboratory of Biochemistry and Toxicology and Laboratory of Anatomy, Histology and Embryology, Faculty of Health Sciences, School of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece, Department of Agricultural Development, Democritus University of Thrace, Orestiada 68200, Greece and Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Anastasia Tsingotjidou
- Laboratory of Anatomy, Histology and Embryology, Faculty of Health Sciences, School of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | - Zaphiris Abas
- Department of Agricultural Development, Democritus University of Thrace, Orestiada 68200, Greece and
| | - Suzan E Erdman
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Theofilos Poutahidis
- Laboratory of Pathology, Laboratory of Biochemistry and Toxicology and Laboratory of Anatomy, Histology and Embryology, Faculty of Health Sciences, School of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece, Department of Agricultural Development, Democritus University of Thrace, Orestiada 68200, Greece and Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|