1
|
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]
|
2
|
Tahiri M, Johnsrud C, Steffensen IL. Evidence and hypotheses on adverse effects of the food additives carrageenan (E 407)/processed Eucheuma seaweed (E 407a) and carboxymethylcellulose (E 466) on the intestines: a scoping review. Crit Rev Toxicol 2023; 53:521-571. [PMID: 38032203 DOI: 10.1080/10408444.2023.2270574] [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] [Received: 06/13/2023] [Revised: 10/02/2023] [Accepted: 10/02/2023] [Indexed: 12/01/2023]
Abstract
This scoping review provides an overview of publications reporting adverse effects on the intestines of the food additives carrageenan (CGN) (E 407)/processed Eucheuma seaweed (PES) (E 407a) and carboxymethylcellulose (CMC) (E 466). It includes evidence from human, experimental mammal and in vitro research publications, and other evidence. The databases Medline, Embase, Scopus, Web of Science Core Collection, Cochrane Database of Systematic Reviews and Epistemonikos were searched without time limits, in addition to grey literature. The publications retrieved were screened against predefined criteria. From two literature searches, 2572 records were screened, of which 224 records were included, as well as 38 records from grey literature, making a total of 262 included publications, 196 on CGN and 101 on CMC. These publications were coded and analyzed in Eppi-Reviewer and data gaps presented in interactive maps. For CGN, five, 69 and 33 research publications on humans, experimental mammals and in vitro experiments were found, further separated as degraded or native (non-degraded) CGN. For CMC, three human, 20 animal and 14 in vitro research publications were obtained. The most studied adverse effects on the intestines were for both additives inflammation, the gut microbiome, including fermentation, intestinal permeability, and cancer and metabolic effects, and immune effects for CGN. Further studies should focus on native CGN, in the form and molecular weight used as food additive. For both additives, randomized controlled trials of sufficient power and with realistic dietary exposure levels of single additives, performed in persons of all ages, including potentially vulnerable groups, are needed.
Collapse
Affiliation(s)
- Mirlinda Tahiri
- Department of Food Safety, Division of Climate and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Celine Johnsrud
- Department of Food Safety, Division of Climate and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Inger-Lise Steffensen
- Department of Food Safety, Division of Climate and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| |
Collapse
|
3
|
Ephraim R, Fraser S, Devereaux J, Stavely R, Feehan J, Eri R, Nurgali K, Apostolopoulos V. Differential Gene Expression of Checkpoint Markers and Cancer Markers in Mouse Models of Spontaneous Chronic Colitis. Cancers (Basel) 2023; 15:4793. [PMID: 37835487 PMCID: PMC10571700 DOI: 10.3390/cancers15194793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 09/17/2023] [Accepted: 09/21/2023] [Indexed: 10/15/2023] Open
Abstract
The presence of checkpoint markers in cancer cells aids in immune escape. The identification of checkpoint markers and early cancer markers is of utmost importance to gain clarity regarding the relationship between colitis and progressive inflammation leading to cancer. Herein, the gene expression levels of checkpoint makers, cancer-related pathways, and cancer genes in colon tissues of mouse models of chronic colitis (Winnie and Winnie-Prolapse mice) using next-generation sequencing are determined. Winnie mice are a result of a Muc2 missense mutation. The identification of such genes and their subsequent expression and role at the protein level would enable novel markers for the early diagnosis of cancer in IBD patients. The differentially expressed genes in the colonic transcriptome were analysed based on the Kyoto Encyclopedia of Genes and Genomes pathway. The expression of several oncogenes is associated with the severity of IBD, with Winnie-Prolapse mice expressing a large number of key genes associated with development of cancer. This research presents a number of new targets to evaluate for the development of biomarkers and therapeutics.
Collapse
Affiliation(s)
- Ramya Ephraim
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia; (R.E.); (S.F.); (J.D.); (J.F.); (K.N.)
| | - Sarah Fraser
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia; (R.E.); (S.F.); (J.D.); (J.F.); (K.N.)
| | - Jeannie Devereaux
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia; (R.E.); (S.F.); (J.D.); (J.F.); (K.N.)
| | - Rhian Stavely
- Pediatric Surgery Research Laboratories, Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA;
| | - Jack Feehan
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia; (R.E.); (S.F.); (J.D.); (J.F.); (K.N.)
- Immunology Program, Australian Institute of Musculoskeletal Science (AIMSS), Melbourne, VIC 3021, Australia
| | - Rajaraman Eri
- STEM/School of Science, RMIT University, Melbourne, VIC 3001, Australia;
| | - Kulmira Nurgali
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia; (R.E.); (S.F.); (J.D.); (J.F.); (K.N.)
- Department of Medicine Western Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC 3010, Australia
- Regenerative Medicine and Stem Cells Program, Australian Institute of Musculoskeletal Science (AIMSS), Melbourne, VIC 3021, Australia
| | - Vasso Apostolopoulos
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia; (R.E.); (S.F.); (J.D.); (J.F.); (K.N.)
- Immunology Program, Australian Institute of Musculoskeletal Science (AIMSS), Melbourne, VIC 3021, Australia
| |
Collapse
|
4
|
Chemically Induced Colitis-Associated Cancer Models in Rodents for Pharmacological Modulation: A Systematic Review. J Clin Med 2022; 11:jcm11102739. [PMID: 35628865 PMCID: PMC9146029 DOI: 10.3390/jcm11102739] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 04/29/2022] [Accepted: 05/10/2022] [Indexed: 02/01/2023] Open
Abstract
Animal models for colitis-associated colorectal cancer (CACC) represent an important tool to explore the mechanistic basis of cancer-related inflammation, providing important evidence that several inflammatory mediators play specific roles in the initiation and perpetuation of colitis and CACC. Although several original articles have been published describing the CACC model in rodents, there is no consensus about the induction method. This review aims to identify, summarize, compare, and discuss the chemical methods for the induction of CACC through the PRISMA methodology. METHODS We searched MEDLINE via the Pubmed platform for studies published through March 2021, using a highly sensitive search expression. The inclusion criteria were only original articles, articles where a chemically-induced animal model of CACC is described, preclinical studies in vivo with rodents, and articles published in English. RESULTS Chemically inducible models typically begin with the administration of a carcinogenic compound (as azoxymethane (AOM) or 1,2-dimethylhydrazine (DMH)), and inflammation is caused by repeated cycles of colitis-inducing agents (such as 2,4,6-trinitrobenzenesulfonic acid (TNBS) or dextran sulfate sodium (DSS)). The strains mostly used are C57BL/6 and Balb/c with 5-6 weeks. To characterize the preclinical model, the parameters more used include body weight, stool consistency and morbidity, inflammatory biomarkers such as tumor necrosis factor (TNF)-α, interleukin (IL)-6 and IL-1β, angiogenesis markers such as proliferating cell nuclear antigen (PCNA), marker of proliferation Ki-67, and caspase 3, the presence of ulcers, thickness or hyperemia in the colon, and histological evaluation of inflammation. CONCLUSION The AOM administration seems to be important to the CACC induction method, since the carcinogenic effect is achieved with just one administration. DSS has been the more used inflammatory agent; however, the TNBS contribution should be more studied, since it allows a reliable, robust, and a highly reproducible animal model of intestinal inflammation.
Collapse
|
5
|
Wang S, Bai M, Shu Q, Liu Z, Shao Y, Xu K, Xiong X, Liu H, Li Y. Modulating Effect of Paeonol on Piglets With Ulcerative Colitis. Front Nutr 2022; 9:846684. [PMID: 35495936 PMCID: PMC9045399 DOI: 10.3389/fnut.2022.846684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 02/15/2022] [Indexed: 11/13/2022] Open
Abstract
Piglet enteritis is a major problem that needs to be solved urgently in modern pig production. Paeonol (Pae) has been used as a novel treatment option due to its good medicinal value. This study purported to elucidate the regulatory mechanism of Pae on dextran sodium sulfate (DSS)-induced ulcerative colitis (UC) in weaned piglets. A total of 36 crossbred (Duroc × Landrace × Yorkshire) weaned piglets were stochastically split into six groups: the control group, DSS group, 0.2% Pae group, 0.4% Pae group, 0.8% Pae group, and mesalazine group. The control and DSS groups were fed with a basic diet, the three Pae and mesalazine groups were fed with 0.2, 0.4, 0.8%, and 2 g mesalazine per kilogram of basic diet throughout the study. On the 15th day of the test period, the control group was gavaged with 10 ml of normal saline, while the remaining five groups were gavaged with 10 ml 5% DSS solution for 13 days. The study lasted for 27 days. The results showed that the 0.8% Pae group significantly increased the average daily feed intake (ADFI) and Occludin mRNA expression in the colon of piglets (P < 0.05). The 0.2% Pae group markedly increased the average daily gain (ADG) and zonula occludens-1 (ZO-1) mRNA expression (P < 0.05). In the 0.2% and 0.4% Pae groups, the feed-to-gain ratio (F/G) was significantly reduced and the mRNA expression levels of Caspase-8, respectively, markedly enhanced the mRNA expression levels of transforming growth factor-β (TGF-β) and interleukins-4 (IL-4) (P < 0.05). In the 0.8% Pae group, the relative abundance of Campilobacterota was significantly reduced (P < 0.05). In the 0.4% Pae group, the relative abundance of Firmicutes was notably increased (P < 0.05). In the 0.2 and 0.8% Pae groups, the relative abundance of Prevotella was markedly increased (P < 0.05). In the 0.2% Pae group, the contents of propionic acid, butyric acid, and valerate acid were markedly higher (P < 0.05). Thus, it is speculated that Pae may regulate the balance of anti-inflammatory/pro-inflammatory factors, improve intestinal tight junction expression, reduce apoptosis, and improve intestinal microflora structure and growth performance of piglets, thereby restoring intestinal barrier function and alleviating DSS-induced UC in piglets.
Collapse
Affiliation(s)
- Shanshan Wang
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Miaomiao Bai
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production; Key Laboratory of Agro-ecological Processes in Subtropical Region; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Qingyan Shu
- Key Laboratory of Plant Resources, Institute of Botany, Chinese Academy of Sciences (CAS), Beijing, China
| | - Zhengan Liu
- Key Laboratory of Plant Resources, Institute of Botany, Chinese Academy of Sciences (CAS), Beijing, China
| | - Yirui Shao
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production; Key Laboratory of Agro-ecological Processes in Subtropical Region; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Kang Xu
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production; Key Laboratory of Agro-ecological Processes in Subtropical Region; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Xia Xiong
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production; Key Laboratory of Agro-ecological Processes in Subtropical Region; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Hongnan Liu
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production; Key Laboratory of Agro-ecological Processes in Subtropical Region; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Yao Li
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| |
Collapse
|
6
|
Tong G, Qian H, Li D, Li J, Chen J, Li X. Establishment and evaluation of a specific antibiotic-induced inflammatory bowel disease model in rats. PLoS One 2022; 17:e0264194. [PMID: 35192646 PMCID: PMC8863245 DOI: 10.1371/journal.pone.0264194] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 02/06/2022] [Indexed: 12/03/2022] Open
Abstract
Physical and chemical methods for generating rat models of enteritis have been established; however, antibiotic induction has rarely been used for this purpose. The present study aimed to establish and evaluate a rat model of inflammatory bowel disease (IBD) using antibiotics. A total of 84 Sprague-Dawley (SD) rats were divided into the following groups, according to the dosage and method of administration of the antibiotics: A, control; B, low-dose clindamycin; C, medium-dose clindamycin; D, high-dose clindamycin; E, low-dose clindamycin, ampicillin and streptomycin; F, medium-dose clindamycin, ampicillin and streptomycin; and G, high-dose clindamycin, ampicillin and streptomycin. Antibiotic administration was stopped on day 7; the modeling period covered days 1-7, and the recovery period covered days 8-15. Half of the animals were dissected on day 11, with the remaining animals dissected on day 15. Food and water intake, body weight and fecal weight were recorded. Intestinal flora was analyzed via microbial culture and quantitative PCR. The content of TNF-α, IL1-β, IL-6 and C-reactive protein (CRP) was assessed in abdominal aorta blood. Colonic and rectal tissues were examined pathologically via hematoxylin-eosin staining to assess leukocyte infiltration and intestinal mucosal changes as indicators of inflammation. Rat weight, food intake, water intake and 2-h fecal weight were significantly different across the experimental groups (P = 0.040, P = 0.016, P<0.001 and P = 0.009, respectively). Microbial cultures revealed no significant differences between group A and B,C (P = 0.546,0.872) but significant differences betwenn group A and the other experimental groups (all P<0.001). Furthermore, significant differences in the levels of Bacteroides, Faecalibacterium prausnitzii and Dialister invisus on day 4 between groups A, C and F (P = 0.033, P = 0.025 and P = 0.034, respectively). Significant differences were detected in the levels of TNF-α, IL1-β, IL-6 and CRP between the groups (all P<0.001). The colonic and rectal pathological inflammation scores of the experimental groups were significantly different compared with group A (B vs. A, P = 0.002; others, all P<0.001). These findings indicated that an antibiotic-induced IBD model was successfully established in SD rats; this animal model may serve as a useful model for clinical IBD research.
Collapse
Affiliation(s)
- Guojun Tong
- Departments of General Surgery, Huzhou Central Hospital, Huzhou, Zhejiang, China
- Central Laboratory, Huzhou Central Hospital, Huzhou, Zhejiang, China
| | - Hai Qian
- Departments of General Surgery, Huzhou Central Hospital, Huzhou, Zhejiang, China
| | - Dongli Li
- Central Laboratory, Huzhou Central Hospital, Huzhou, Zhejiang, China
| | - Jing Li
- Central Laboratory, Huzhou Central Hospital, Huzhou, Zhejiang, China
| | - Jing Chen
- Central Laboratory, Huzhou Central Hospital, Huzhou, Zhejiang, China
| | - Xiongfeng Li
- Orthopedic Surgery, Huzhou Central Hospital, Huzhou, Zhejiang, China
| |
Collapse
|
7
|
Ramírez-Rico G, Drago-Serrano ME, León-Sicairos N, de la Garza M. Lactoferrin: A Nutraceutical with Activity against Colorectal Cancer. Front Pharmacol 2022; 13:855852. [PMID: 35264972 PMCID: PMC8899398 DOI: 10.3389/fphar.2022.855852] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 01/31/2022] [Indexed: 12/29/2022] Open
Abstract
Homeostasis in the human body results from the tight regulation of several events, since too little inflammation disrupts the process of tissue repair and remodeling, whereas too much exerts a collateral effect by causing tissue damage with life-threatening consequences. In some clinical conditions, such as inflammatory bowel disease (IBD), inflammation functions as a double-edged sword by either enabling or inhibiting cancer development and progression. Generally, cancer develops through evasion mechanisms that regulate cell growth, causing a high rate of uncontrolled proliferation, and mechanisms for evading cell death, such as apoptosis. Moreover, chronic inflammation is a factor that contributes to colorectal cancer (CRC), as observed in individuals with IBD; all these conditions favor an increased rate of angiogenesis and eventual metastasis. Lactoferrin (Lf) is a mammalian iron-binding multifunctional glycoprotein regarded as a natural compound that up- and downregulates both humoral and cellular components of immunity involved in regulating the inflammatory response and maintaining gut homeostasis. Human and bovine Lf share high sequence homology and have very similar antimicrobial, anti-inflammatory, and immunomodulatory activities. Bovine Lf from milk is considered a safe molecule and is commercially available in large quantities. This review mainly focuses on the regulatory effects of orally administered bovine Lf on the inflammatory response associated with CRC; this approach indicates that CRC is one of the most frequently diagnosed cancers and affects the intestinal tract with high clinical and epidemiologic relevance. Thus, this review may provide foundations for the potential use of bovine Lf alone or as a natural adjunct agent to increase the effectiveness and reduce the side effects of anticancer chemotherapy.
Collapse
Affiliation(s)
- Gerardo Ramírez-Rico
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados Del Instituto Politécnico Nacional (CINVESTAV-IPN), México City, Mexico
- Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México (UNAM), México City, Mexico
| | - Maria Elisa Drago-Serrano
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana Unidad Xochimilco, Mexico City, Mexico
| | - Nidia León-Sicairos
- Centro de Investigación Aplicada a La Salud Pública (CIASaP), Facultad de Medicina, Universidad Autónoma de Sinaloa, Culiacán, Mexico
- Hospital Pediátrico de Sinaloa, Culiacán, Mexico
| | - Mireya de la Garza
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados Del Instituto Politécnico Nacional (CINVESTAV-IPN), México City, Mexico
- *Correspondence: Mireya de la Garza,
| |
Collapse
|
8
|
Ma B, Kamle S, Akosman B, Khan H, Lee CM, Lee CG, Elias JA. CHI3L1 enhances melanoma lung metastasis via regulation of T cell co-stimulators and CTLA-4/B7 axis. Front Immunol 2022; 13:1056397. [PMID: 36618349 PMCID: PMC9812560 DOI: 10.3389/fimmu.2022.1056397] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022] Open
Abstract
ICOS/ICOSL and CD28/B7-1/B7-2 are T cell co-stimulators and CTLA-4 is an immune checkpoint inhibitor that play critical roles in the pathogenesis of neoplasia. Chitinase 3-like-1 (CHI3L1) is induced in many cancers where it portends a poor prognosis and contributes to tumor metastasis. Here we demonstrate that CHI3L1 inhibits the expression of ICOS, ICOSL and CD28 while stimulating CTLA-4 and the B7 moieties in melanoma lung metastasis. We also demonstrate that RIG-like helicase innate immune activation augments T cell co-stimulation, inhibits CTLA-4 and suppresses pulmonary metastasis. At least additive antitumor responses were seen in melanoma lung metastasis treated with anti-CTLA-4 and anti-CHI3L1 antibodies in combination. Synergistic cytotoxic T cell-induced tumor cell death and the heightened induction of the tumor suppressor PTEN were seen in co-cultures of T and tumor cells treated with bispecific antibodies that target both CHI3L1 and CTLA-4. Thus, CHI3L1 contributes to pulmonary metastasis by inhibiting T cell co-stimulation and stimulating CTLA-4. The simultaneous targeting of CHI3L1 and the CTLA-4 axis with individual and, more powerfully with bispecific antibodies, represent promising therapeutic strategies for pulmonary metastasis.
Collapse
Affiliation(s)
- Bing Ma
- Molecular Microbiology and Immunology, Brown University, Providence, RI, United States
| | - Suchitra Kamle
- Molecular Microbiology and Immunology, Brown University, Providence, RI, United States
| | - Bedia Akosman
- Molecular Microbiology and Immunology, Brown University, Providence, RI, United States
| | - Hina Khan
- Division of Hematology-Oncology, Warren Alpert Medical School, Brown University, Providence, RI, United States
| | - Chang-Min Lee
- Molecular Microbiology and Immunology, Brown University, Providence, RI, United States
| | - Chun Geun Lee
- Molecular Microbiology and Immunology, Brown University, Providence, RI, United States
| | - Jack A. Elias
- Molecular Microbiology and Immunology, Brown University, Providence, RI, United States
- Department of Medicine, Brown University, Providence, RI, United States
- *Correspondence: Jack A. Elias,
| |
Collapse
|
9
|
Decreased IL-10 accelerates B-cell leukemia/lymphoma in a mouse model of pediatric lymphoid leukemia. Blood Adv 2021; 6:854-865. [PMID: 34727170 PMCID: PMC8945291 DOI: 10.1182/bloodadvances.2021005522] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 10/04/2021] [Indexed: 11/25/2022] Open
Abstract
B-cell deficiency and DNA damage in the bone marrow of Il10−/− mice are associated with inflammation and mitigated by antibiotics. In ETV6-RUNX1+Cdkn2a−/− mice, low IL-10 accelerates the development of B-cell leukemia/lymphoma in a dose-dependent manner.
Exposures to a wide repertoire of common childhood infections and strong inflammatory responses to those infections are associated with the risk of pediatric B-cell acute lymphoblastic leukemia (B-ALL) in opposing directions. Neonatal inflammatory markers are also related to risk by unknown mechanism(s). Here, we demonstrate that interleukin-10 (IL-10) deficiency, which is associated with childhood B-ALL, indirectly impairs B lymphopoiesis and increases B-cell DNA damage in association with a module of 6 proinflammatory/myeloid-associated cytokines (IL-1α, IL-6, IL-12p40, IL-13, macrophage inflammatory protein-1β/CCL4, and granulocyte colony-stimulating factor). Importantly, antibiotics attenuated inflammation and B-cell defects in preleukemic Cdkn2a−/−Il10−/− mice. In an ETV6-RUNX1+ (E6R1+) Cdkn2a−/− mouse model of B-ALL, decreased levels of IL-10 accelerated B-cell neoplasms in a dose-dependent manner and altered the mutational profile of these neoplasms. Our results illuminate a mechanism through which a low level of IL-10 can create a risk for leukemic transformation and support developing evidence that microbial dysbiosis contributes to pediatric B-ALL.
Collapse
|
10
|
Ma B, Akosman B, Kamle S, Lee CM, He CH, Koo JS, Lee CG, Elias JA. CHI3L1 regulates PD-L1 and anti-CHI3L1-PD-1 antibody elicits synergistic antitumor responses. J Clin Invest 2021; 131:137750. [PMID: 34720089 DOI: 10.1172/jci137750] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 09/02/2021] [Indexed: 12/24/2022] Open
Abstract
Evasion of the immune response is a hallmark of cancer, and programmed cell death 1 (PD-1) and PD-1 ligand 1 (PD-L1) are major mediators of this immunosuppression. Chitinase 3-like 1 (CHI3L1) is induced in many cancers, where it portends a poor prognosis and contributes to tumor metastasis and spread. However, the mechanism(s) that CHI3L1 uses in metastasis have not been defined. Here we demonstrate that CHI3L1 regulates the expression of PD-L1, PD-L2, PD-1, LAG3, and TIM3 and plays a critical role in melanoma progression and lymphatic spread. CHI3L1 also contributed to IFN-γ-stimulated macrophage PD-L1 expression, and RIG-like helicase innate immunity suppressed CHI3L1, PD-L1, and melanoma progression. Individual antibodies against CHI3L1 or PD-1 had discrete antitumor effects and additive antitumor responses in metastasis models and T cell-tumor cell cocultures when administered simultaneously. Synergistic cytotoxic tumor cell death was seen in T cell-tumor cell cocultures, and significantly enhanced antitumor responses were seen in in vivo tumor models treated with bispecific antibodies that simultaneously target CHI3L1 and PD-1. CHI3L1 contributes to tumor progression by stimulating the PD-1/PD-L1 axis and other checkpoint molecules. The simultaneous targeting of CHI3L1 and the PD-1/PD-L1 axis with individual and, more powerfully, with bispecific antibodies represents a promising therapy for pulmonary metastasis and progression.
Collapse
Affiliation(s)
- Bing Ma
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Bedia Akosman
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Suchitra Kamle
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Chang-Min Lee
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Chuan Hua He
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Ja Seok Koo
- Section of Medical Oncology, Department of Internal Medicine, Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut, USA
| | - Chun Geun Lee
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Jack A Elias
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA.,Department of Medicine, Brown University, Providence, Rhode Island, USA
| |
Collapse
|
11
|
Mahalhal A, Burkitt MD, Duckworth CA, Hold GL, Campbell BJ, Pritchard DM, Probert CS. Long-Term Iron Deficiency and Dietary Iron Excess Exacerbate Acute Dextran Sodium Sulphate-Induced Colitis and Are Associated with Significant Dysbiosis. Int J Mol Sci 2021; 22:3646. [PMID: 33807459 PMCID: PMC8037348 DOI: 10.3390/ijms22073646] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 03/25/2021] [Accepted: 03/26/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Oral iron supplementation causes gastrointestinal side effects. Short-term alterations in dietary iron exacerbate inflammation and alter the gut microbiota, in murine models of colitis. Patients typically take supplements for months. We investigated the impact of long-term changes in dietary iron on colitis and the microbiome in mice. METHODS We fed mice chow containing differing levels of iron, reflecting deficient (100 ppm), normal (200 ppm), and supplemented (400 ppm) intake for up to 9 weeks, both in absence and presence of dextran sodium sulphate (DSS)-induced chronic colitis. We also induced acute colitis in mice taking these diets for 8 weeks. Impact was assessed (i) clinically and histologically, and (ii) by sequencing the V4 region of 16S rRNA. RESULTS In mice with long-term changes, the iron-deficient diet was associated with greater weight loss and histological inflammation in the acute colitis model. Chronic colitis was not influenced by altering dietary iron however there was a change in the microbiome in DSS-treated mice consuming 100 ppm and 400 ppm iron diets, and control mice consuming the 400 ppm iron diet. Proteobacteria levels increased significantly, and Bacteroidetes levels decreased, in the 400 ppm iron DSS group at day-63 compared to baseline. CONCLUSIONS Long-term dietary iron alterations affect gut microbiota signatures but do not exacerbate chronic colitis, however acute colitis is exacerbated by such dietary changes. More work is needed to understand the impact of iron supplementation on IBD. The change in the microbiome, in patients with colitis, may arise from the increased luminal iron and not simply from colitis.
Collapse
Affiliation(s)
- Awad Mahalhal
- Department of Molecular and Cellular Cancer Medicine, Institute of Systems, Molecular and Integrated Biology, University of Liverpool, Liverpool L69 3GE, UK; (A.M.); (D.M.P.)
- Department of Anatomy and Histology, Faculty of Medicine, Benghazi University, Benghazi, Libya
| | - Michael D. Burkitt
- Division of Diabetes Endocrinology and Gastroenterology, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PL, UK;
| | - Carrie A. Duckworth
- Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GE, UK;
| | - Georgina L. Hold
- Microbiome Research Centre, St George & Sutherland Clinical School, Clinical Sciences (Pitney) Building, University of New South Wales Sydney, Kogarah, NSW 2217, Australia;
| | - Barry J. Campbell
- Department of Infection Biology & Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 3GE, UK;
| | - David Mark Pritchard
- Department of Molecular and Cellular Cancer Medicine, Institute of Systems, Molecular and Integrated Biology, University of Liverpool, Liverpool L69 3GE, UK; (A.M.); (D.M.P.)
| | - Chris S. Probert
- Department of Molecular and Cellular Cancer Medicine, Institute of Systems, Molecular and Integrated Biology, University of Liverpool, Liverpool L69 3GE, UK; (A.M.); (D.M.P.)
| |
Collapse
|
12
|
MicroRNA Biomarkers in IBD-Differential Diagnosis and Prediction of Colitis-Associated Cancer. Int J Mol Sci 2020; 21:ijms21217893. [PMID: 33114313 PMCID: PMC7660644 DOI: 10.3390/ijms21217893] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 12/14/2022] Open
Abstract
Inflammatory bowel disease (IBD) includes Crohn's disease (CD) and ulcerative colitis (UC). These are chronic autoimmune diseases of unknown etiology affecting the gastrointestinal tract. The IBD population includes a heterogeneous group of patients with varying disease courses requiring personalized treatment protocols. The complexity of the disease often delays the diagnosis and the initiation of appropriate treatments. In a subset of patients, IBD leads to colitis-associated cancer (CAC). MicroRNAs are single-stranded regulatory noncoding RNAs of 18 to 22 nucleotides with putative roles in the pathogenesis of IBD and colorectal cancer. They have been explored as biomarkers and therapeutic targets. Both tissue-derived and circulating microRNAs have emerged as promising biomarkers in the differential diagnosis and in the prognosis of disease severity of IBD as well as predictive biomarkers in drug resistance. In addition, knowledge of the cellular localization of differentially expressed microRNAs is a prerequisite for deciphering the biological role of these important epigenetic regulators and the cellular localization may even contribute to an alternative repertoire of biomarkers. In this review, we discuss findings based on RT-qPCR, microarray profiling, next generation sequencing and in situ hybridization of microRNA biomarkers identified in the circulation and in tissue biopsies.
Collapse
|
13
|
He CH, Lee CG, Ma B, Kamle S, Choi AMK, Elias JA. N-Glycosylation Regulates Chitinase 3-like-1 and IL-13 Ligand Binding to IL-13 Receptor α2. Am J Respir Cell Mol Biol 2020; 63:386-395. [PMID: 32402213 DOI: 10.1165/rcmb.2019-0446oc] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Chitinase 3-like-1 (Chi3l1) and IL-13 are both ligands of IL-13 receptor α2 (IL-13Rα2). The binding of the former activates mitogen-activated protein kinase, AKT, and Wnt/β-catenin signaling, and plays important roles in innate and adaptive immunity, cellular apoptosis, oxidative injury, allergic inflammation, tumor metastasis and wound healing, fibrosis, and repair in the lung. In contrast, the latter binding is largely a decoy event that diminishes the effects of IL-13. Here, we demonstrate that IL-13Rα2 N-glycosylation is a critical determinant of which ligand binds. Structure-function evaluations demonstrated that Chi3l1-IL-13Rα2 binding was increased when sites of N-glycosylation are mutated, and studies with tunicamycin and Peptide:N-glycosidase F (PNGase F) demonstrated that Chi3l1-IL-13Rα2 binding and signaling were increased when N-glycosylation was diminished. In contrast, structure-function experiments demonstrated that IL-13 binding to IL-13Rα2 was dependent on each of the four sites of N-glycosylation in IL-13Rα2, and experiments with tunicamycin and PNGase F demonstrated that IL-13-IL-13Rα2 binding was decreased when IL-13Rα2 N-glycosylation was diminished. Studies with primary lung epithelial cells also demonstrated that Chi3l1 inhibited, whereas IL-13 stimulated, N-glycosylation as evidenced by the ability of Chi3l1 to inhibit and IL-13 to stimulate the subunits of the oligosaccharide complex A and B (STT3A and STT3B). These studies demonstrate that N-glycosylation is a critical determinant of Chi3l1 and IL-13 binding to IL-13Rα2, and highlight the ability of Chi3l1 and IL-13 to alter key elements of the N-glycosylation apparatus in a manner that would augment their respective binding.
Collapse
Affiliation(s)
- Chuan Hua He
- Department of Molecular Microbiology and Immunology and
| | - Chun Geun Lee
- Department of Molecular Microbiology and Immunology and
| | - Bing Ma
- Department of Molecular Microbiology and Immunology and
| | | | - Augustine M K Choi
- Department of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, New York
| | - Jack A Elias
- Department of Molecular Microbiology and Immunology and.,Department of Medicine, Brown University, Providence, Rhode Island; and
| |
Collapse
|
14
|
Song H, Tang X, Li X, Wang Y, Deng A, Wang W, Zhang H, Qin H, Wu L. HLJ2 Effectively Ameliorates Colitis-Associated Cancer via Inhibition of NF-κB and Epithelial-Mesenchymal Transition. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:4291-4302. [PMID: 33116416 PMCID: PMC7573331 DOI: 10.2147/dddt.s262806] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 09/02/2020] [Indexed: 01/24/2023]
Abstract
Introduction Colitis-associated cancer (CAC) accounts for approximately 15% of IBD patient mortalities. However, currently available anti-CAC drugs possess many disadvantages including safety, specificity and side effects. Therefore, the development of novel anti-CAC compounds is imperative. HLJ2 was a monomeric compound synthesized by our institute and reported to have an effect on ulcer colitis. Methods In vivo the AOM/DSS-induced CAC model was used to evaluate the effects of HLJ2 on ameliorating CAC symptoms, immunohistochemical analysis was used to analyze the pathological damage to colons and epithelial–mesenchymal transition was for changes of cytokines. In vitro, flow cytometric analysis, immunofluorescence and Western blot were used to detect the inhibition effect of HLJ2 on nuclear factor-κB and epithelial–mesenchymal transition in TGF-β1-stimulated SW480 cells. Results In the AOM/DSS animal model, HLJ2 was demonstrated to inhibit the secretion of inflammatory cytokines and nuclear factor-κB, levels of tumorigenesis-related proteins including snail, and finally inhibited a key step in metastasis, epithelial–mesenchymal transition. In vitro, HLJ2 was also shown to inhibit nuclear factor-κB and epithelial–mesenchymal transition in TGF-β1-stimulated SW480 cells in accordance with in vivo results. Meanwhile, the nuclear factor-κB inhibitor could interrupt the effect of HLJ2 on epithelial–mesenchymal transition. Discussion HLJ2 may ameliorate CAC through inhibiting nuclear factor-κB and then downstream epithelial–mesenchymal transition. The combination of the obvious improvement in effects on CAC without obvious side effects suggests that HLJ2 could be developed as a potential CAC therapeutic candidate.
Collapse
Affiliation(s)
- Huachen Song
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Xiaonan Tang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Xiang Li
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Yufei Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Anjun Deng
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Wenjie Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Haijing Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Hailin Qin
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - LianQiu Wu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| |
Collapse
|
15
|
Zhao Y, Jiang Q. Roles of the Polyphenol-Gut Microbiota Interaction in Alleviating Colitis and Preventing Colitis-Associated Colorectal Cancer. Adv Nutr 2020; 12:546-565. [PMID: 32905583 PMCID: PMC8009754 DOI: 10.1093/advances/nmaa104] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 07/07/2020] [Accepted: 07/29/2020] [Indexed: 12/17/2022] Open
Abstract
Accumulating evidence indicates that the gut microbiota can promote or inhibit colonic inflammation and carcinogenesis. Promotion of beneficial gut bacteria is considered a promising strategy to alleviate colonic diseases including colitis and colorectal cancer. Interestingly, dietary polyphenols, which have been shown to attenuate colitis and inhibit colorectal cancer in animal models and some human studies, appear to reach relatively high concentrations in the large intestine and to interact with the gut microbial community. This review summarizes the modulatory effects of polyphenols on the gut microbiota in humans and animals under healthy and diseased conditions including colitis and colitis-associated colorectal cancer (CAC). Existing human and animal studies indicate that polyphenols and polyphenol-rich whole foods are capable of elevating butyrate producers and probiotics that alleviate colitis and inhibit CAC, such as Lactobacillus and Bifidobacterium. Studies in colitis and CAC models indicate that polyphenols decrease opportunistic pathogenic or proinflammatory microbes and counteract disease-induced dysbiosis. Consistently, polyphenols also change microbial functions, including increasing butyrate formation. Moreover, polyphenol metabolites produced by the gut microbiota appear to have anticancer and anti-inflammatory activities, protect gut barrier integrity, and mitigate inflammatory conditions in cells and animal models. Based on these results, we conclude that polyphenol-mediated alteration of microbial composition and functions, together with polyphenol metabolites produced by the gut microbiota, likely contribute to the protective effects of polyphenols on colitis and CAC. Future research is needed to validate the causal role of the polyphenol-gut microbiota interaction in polyphenols' anti-colitis and anti-CAC effects, and to further elucidate mechanisms underlying such interaction.
Collapse
Affiliation(s)
- Yiying Zhao
- Department of Nutrition Science, Purdue University, West Lafayette, IN, USA
| | | |
Collapse
|
16
|
Song M, Lan Y, Wu X, Han Y, Wang M, Zheng J, Li Z, Li F, Zhou J, Xiao J, Cao Y, Xiao H. The chemopreventive effect of 5-demethylnobiletin, a unique citrus flavonoid, on colitis-driven colorectal carcinogenesis in mice is associated with its colonic metabolites. Food Funct 2020; 11:4940-4952. [PMID: 32459257 PMCID: PMC10726105 DOI: 10.1039/d0fo00616e] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2023]
Abstract
5-Demethylnobiletin (5DN) is a unique flavonoid mainly found in citrus fruits. In this study, we determined the chemopreventive effects of 5DN and its major colonic metabolites on both a colitis-driven colon carcinogenesis mouse model and a human colon cancer cell model. In azoxymethane/dextran sulfate sodium-treated mice, dietary 5DN (0.05% w/w in the diet) significantly decreased the tumor incidence, multiplicity and burden, and showed potent anti-proliferative, proapoptotic, and anti-inflammatory activities in mouse colon tissue. Three major metabolites of 5DN, named 5,3'-didemethylnobiletin (M1), 5,4'-didemethylnobiletin (M2) and 5,3',4'-tridemethylnobiletin (M3), were found in the colonic mucosa of 5DN-treated mice, and the combined level of these metabolites in mouse colonic mucosa was 1.56-fold higher than that of 5DN. Cell culture studies revealed that 5DN and its colonic metabolites profoundly inhibited the growth of human colon cancer cells by inducing cell cycle arrest, triggering apoptosis and modulating key signaling proteins related to cell proliferation and apoptosis. Importantly, the colonic metabolites, especially M1, showed much stronger effects than those produced by 5DN itself. Overall, our results demonstrated that dietary 5DN significantly inhibited colitis-driven colon carcinogenesis in mice, and this chemopreventive effect was associated with its metabolites in the colon.
Collapse
Affiliation(s)
- Mingyue Song
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, Guangdong, P.R. China and Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, Guangdong, P.R. China and Department of Food Science, University of Massachusetts, Amherst, MA, USA.
| | - Yaqi Lan
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, Guangdong, P.R. China and Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, Guangdong, P.R. China
| | - Xian Wu
- Department of Food Science, University of Massachusetts, Amherst, MA, USA. and Department of Kinesiology and Health, Miami University, Oxford, OH, USA
| | - Yanhui Han
- Department of Food Science, University of Massachusetts, Amherst, MA, USA.
| | - Minqi Wang
- Department of Food Science, University of Massachusetts, Amherst, MA, USA.
| | - Jinkai Zheng
- Department of Food Science, University of Massachusetts, Amherst, MA, USA. and Institute of Agro-Products Processing Science and Technology, Chinese Academy of Agricultural Sciences, Beijing, P.R. China
| | - Zhengze Li
- Department of Food Science, University of Massachusetts, Amherst, MA, USA.
| | - Fang Li
- Department of Food Science, University of Massachusetts, Amherst, MA, USA.
| | - Jiazhi Zhou
- Department of Food Science, University of Massachusetts, Amherst, MA, USA.
| | - Jie Xiao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, Guangdong, P.R. China and Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, Guangdong, P.R. China
| | - Yong Cao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, Guangdong, P.R. China and Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, Guangdong, P.R. China
| | - Hang Xiao
- Department of Food Science, University of Massachusetts, Amherst, MA, USA.
| |
Collapse
|
17
|
Meng S, Li Y, Zang X, Jiang Z, Ning H, Li J. Effect of TLR2 on the proliferation of inflammation-related colorectal cancer and sporadic colorectal cancer. Cancer Cell Int 2020; 20:95. [PMID: 32256204 PMCID: PMC7104506 DOI: 10.1186/s12935-020-01184-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 03/23/2020] [Indexed: 02/07/2023] Open
Abstract
Background Colitis-associated cancer (CAC) is a complication of inflammatory bowel disease (IBD) with a poor prognosis because it is often diagnosed in advanced stages with local progression or metastasis. Compared with the more common polyp-induced sporadic colorectal cancer (sCRC), CAC has different molecular mechanisms. Toll-like receptor 2 (TLR2) expression is not limited to cells related to inflammation and immune function. High levels of TLR2 expression in tumor tissues of colorectal cancer (CRC) patients have been reported. This report is to investigate the effects of knockout and knockdown of the TLR2 gene on the proliferation of CAC and sCRC. Methods Twelve C57BL/6 J wild-type mice (WT) and 12 TLR2 knockout mice (TLR2-/-) were used to rapidly establish a colitis-associated cancer (CAC) model via the 1,2-dimethylhydrazine-dextran sodium sulfate (DMH-DSS) method and were divided into the normal WT control group (NC), TLR2 knockout control group (KC), normal wild-type tumor modeling group (NT), and TLR2 knockout tumor modeling group (KT), with 6 mice in each group. The general performance of the mice during modeling, the gross changes of the colon and the rectum, and the pathological score of HE staining were used to observe tumor growth. The expression of TLR2 was detected by immunohistochemistry, and tumor proliferation was detected by Ki67 labeling. Lentivirus carrying TLR2-RNAi was used to stably infect colorectal cancer cells (HCT116 and HT29) to knock down TLR2 gene expression. The experimental groups included the uninfected control group, negative control group, and gene knockdown group. After infection, the expression of TLR2 protein was detected by Western blot, and cell proliferation and the cell cycle were detected by the CCK-8 method and fluorescence-activated cell sorting. Western blot was used to detect the expression levels of p- NF-κβ, cyclin D1 and cyclin D3 protein in each group of cells. Results TLR2 knockout in the CAC model resulted in greater changes in body weight and more severe diarrhea and colorectal hemorrhage. However, knocking out the TLR2 gene reduced the shortening of colorectal length, the number of tumors, and the total tumor volume and inhibited the growth of CAC. Knocking out the TLR2 gene also reduced the pathological score and tumor severity. TLR2 was localized in the cell membrane of the colorectal epithelium of the NC group and of the colorectal tumors of the NT group and was highly expressed in the NT group, while antigen Ki67 was localized in the nucleus of the colorectal tumor cells of the NT group and the KT group, and its expression was reduced in the KT group. In an in vitro sporadic colorectal cancer cell experiment, TLR2 protein in the TLR2 knockdown group was significantly downregulated, and TLR2 knockdown significantly inhibited the proliferation of HCT116 and HT29 colorectal cancer cells, resulting in G1 phase arrest. The expression levels of p-NF-κβ, cyclin D1 and cyclin D3 proteins in TLR2 gene knockdown group cells were significantly reduced. Conclusion Knockout and knockdown of TLR2 can inhibit the proliferation of inflammation-related colorectal cancer and sporadic colorectal cancer.
Collapse
Affiliation(s)
- Shuang Meng
- 1Department of Gastroenterology, The First Affiliated Hospital of Jinzhou Medical University, No. 2, Section 5 Renmin Street, Guta District, Jinzhou City, 121001 Liaoning Province China.,2Jinzhou Medical University, No. 40, Section 3, Songpo Road, Linghe District, Jinzhou City, 121001 Liaoning Province China
| | - Yingjie Li
- 1Department of Gastroenterology, The First Affiliated Hospital of Jinzhou Medical University, No. 2, Section 5 Renmin Street, Guta District, Jinzhou City, 121001 Liaoning Province China
| | - Xiaozhen Zang
- 1Department of Gastroenterology, The First Affiliated Hospital of Jinzhou Medical University, No. 2, Section 5 Renmin Street, Guta District, Jinzhou City, 121001 Liaoning Province China
| | - Zheng Jiang
- Jinzhou No. 2 Hospital, No. 2, Section 6, Nanjing Road, Linghe District, Jinzhou City, 121001 Liaoning Province China
| | - Huahan Ning
- 1Department of Gastroenterology, The First Affiliated Hospital of Jinzhou Medical University, No. 2, Section 5 Renmin Street, Guta District, Jinzhou City, 121001 Liaoning Province China
| | - Jing Li
- 1Department of Gastroenterology, The First Affiliated Hospital of Jinzhou Medical University, No. 2, Section 5 Renmin Street, Guta District, Jinzhou City, 121001 Liaoning Province China
| |
Collapse
|
18
|
Kuznietsova H, Dziubenko N, Byelinska I, Hurmach V, Bychko A, Lynchak O, Milokhov D, Khilya O, Rybalchenko V. Pyrrole derivatives as potential anti-cancer therapeutics: synthesis, mechanisms of action, safety. J Drug Target 2019; 28:547-563. [PMID: 31814456 DOI: 10.1080/1061186x.2019.1703189] [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] [Indexed: 01/01/2023]
Abstract
Pyrrole derivatives (PDs) chloro-1-(4-chlorobenzyl)-4-((3-(trifluoromethyl)phenyl)amino)-1H-pyrrole-2,5-dione (MI-1) and 5-amino-4-(1,3-benzothyazol-2-yn)-1-(3-methoxyphenyl)-1,2-dihydro-3H-pyrrole-3-one (D1) were synthesised as inhibitors of several protein kinases including EGFR and VEGFR. The aim of the study was to reveal the exact mechanisms of PDs' action EGFR and VEGFR are involved in. We observed, that both PDs could bind with EGFR and VEGFR and form stable complexes. PDs entered into electrostatic interactions with polar groups of phospholipid heads in cell membrane, and the power of interaction depended on the nature of PD radical substituents (greater for MI-1 and smaller for D1). Partial intercalation of MI-1 into the membrane hydrophobic zone also occurred. PDs concentrations induced apoptosis in malignant cells but normal ones had different sensitivity to those. MI-1 and D1 acted like antioxidants in inflamed colonic tissue, as evidenced by reduce of lipid and protein peroxidation products (by 43-67%) and increase of superoxide dismutase activity (by 40 and 58%) with restoring these values to control ones. MI-1 restored reduced haemoglobin and normalised elevated platelets and monocytes in settings of colorectal cancer, whereas D1 normalised only platelets. Thus, MI-1 and D1 could be used as competitive inhibitors of EGFR and VEGFR and antioxidants, which might contribute to realisation of their anti-inflammatory, proapoptotic and antitumor activity.
Collapse
Affiliation(s)
| | | | | | - Vasyl Hurmach
- Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Andriy Bychko
- Bogomolets National Medical University, Kyiv, Ukraine
| | - Oksana Lynchak
- Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Demyd Milokhov
- Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Olga Khilya
- Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | | |
Collapse
|
19
|
Souris JS, Zhang HJ, Dougherty U, Chen NT, Waller JV, Lo LW, Hart J, Chen CT, Bissonnette M. A novel mouse model of sporadic colon cancer induced by combination of conditional Apc genes and chemical carcinogen in the absence of Cre recombinase. Carcinogenesis 2019; 40:1376-1386. [PMID: 30859181 PMCID: PMC6875902 DOI: 10.1093/carcin/bgz050] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 01/27/2019] [Accepted: 03/08/2019] [Indexed: 02/07/2023] Open
Abstract
Although valuable insights into colon cancer biology have been garnered from human colon cancer cell lines and primary colonic tissues, and animal studies using human colon cancer xenografts, immunocompetent mouse models of spontaneous or chemically induced colon cancer better phenocopy human disease. As most sporadic human colon tumors present adenomatous polyposis coli (APC) gene mutations, considerable effort has gone into developing mice that express mutant Apc alleles that mimic human colon cancer pathogenesis. A serious limitation of many of these Apc-mutant murine models, however, is that these mice develop numerous tumors in the small intestine but few, if any, in the colon. In this work, we examined three spontaneous mouse models of colon tumorigenesis based upon the widely used multiple intestinal neoplasia (Min) mouse: mice with either constitutive or conditional Apc mutations alone or in combination with caudal-related homeobox transcription factor CDX2P-Cre transgene - either with or without exposure to the potent colon carcinogen azoxymethane. Using the CDX2 promoter to drive Cre recombinase transgene expression effectively inactivated Apc in colonocytes, creating a model with earlier tumor onset and increased tumor incidence/burden, but without the Min mouse model's small intestine tumorigenesis and susceptibility to intestinal perforation/ulceration/hemorrhage. Most significantly, azoxymethane-treated mice with conditional Apc expression, but absent the Cre recombinase gene, demonstrated nearly 50% tumor incidence with two or more large colon tumors per mouse of human-like histology, but no small intestine tumors - unlike the azoxymethane-resistant C57BL/6J-background Min mouse model. As such this model provides a robust platform for chemoprevention studies.
Collapse
Affiliation(s)
- Jeffrey S Souris
- Department of Radiology, The University of Chicago, Chicago, IL, USA
| | - Hannah J Zhang
- Department of Radiology, The University of Chicago, Chicago, IL, USA
| | | | - Nai-Tzu Chen
- Institute of New Drug Development, China Medical University, Taichung, Taiwan
| | - Joseph V Waller
- Department of Radiology, The University of Chicago, Chicago, IL, USA
| | - Leu-Wei Lo
- Department of Radiology, The University of Chicago, Chicago, IL, USA
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan, Miaoli, Taiwan
| | - John Hart
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Chin-Tu Chen
- Department of Radiology, The University of Chicago, Chicago, IL, USA
| | - Marc Bissonnette
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
20
|
Vázquez-Arreguín K, Bensard C, Schell JC, Swanson E, Chen X, Rutter J, Tantin D. Oct1/Pou2f1 is selectively required for colon regeneration and regulates colon malignancy. PLoS Genet 2019; 15:e1007687. [PMID: 31059499 PMCID: PMC6522070 DOI: 10.1371/journal.pgen.1007687] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 05/16/2019] [Accepted: 04/16/2019] [Indexed: 12/22/2022] Open
Abstract
The transcription factor Oct1/Pou2f1 promotes poised gene expression states, mitotic stability, glycolytic metabolism and other characteristics of stem cell potency. To determine the effect of Oct1 loss on stem cell maintenance and malignancy, we deleted Oct1 in two different mouse gut stem cell compartments. Oct1 deletion preserved homeostasis in vivo and the ability to establish organoids in vitro, but blocked the ability to recover from treatment with dextran sodium sulfate, and the ability to maintain organoids after passage. In a chemical model of colon cancer, loss of Oct1 in the colon severely restricted tumorigenicity. In contrast, loss of one or both Oct1 alleles progressively increased tumor burden in a colon cancer model driven by loss-of-heterozygosity of the tumor suppressor gene Apc. The different outcomes are consistent with prior findings that Oct1 promotes mitotic stability, and consistent with differentially expressed genes between the two models. Oct1 ChIPseq using HCT116 colon carcinoma cells identifies target genes associated with mitotic stability, metabolism, stress response and malignancy. This set of gene targets overlaps significantly with genes differentially expressed in the two tumor models. These results reveal that Oct1 is selectively required for recovery after colon damage, and that Oct1 has potent effects in colon malignancy, with outcome (pro-oncogenic or tumor suppressive) dictated by tumor etiology.
Collapse
Affiliation(s)
- Karina Vázquez-Arreguín
- Department of Pathology and Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT, United States of America
| | - Claire Bensard
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, United States of America
| | - John C. Schell
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, United States of America
| | - Eric Swanson
- Department of Pathology and Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT, United States of America
| | - Xinjian Chen
- Department of Pathology and Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT, United States of America
| | - Jared Rutter
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, United States of America
- Howard Hughes Medical Institute, Salt Lake City, Utah, United States of America
| | - Dean Tantin
- Department of Pathology and Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT, United States of America
| |
Collapse
|
21
|
Dai F, Dong S, Rong Z, Xuan Q, Chen P, Chen M, Fan Y, Gao Q. Expression of inositol-requiring enzyme 1β is downregulated in azoxymethane/dextran sulfate sodium-induced mouse colonic tumors. Exp Ther Med 2019; 17:3181-3188. [PMID: 30936991 DOI: 10.3892/etm.2019.7317] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 11/22/2018] [Indexed: 12/28/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a risk factor in colon cancer. Endoplasmic reticulum (ER) stress is associated with IBD and cancer. In the current study an azoxymethane (AOM) and dextran sulfate sodium (DSS)-induced mouse colonic tumor model was established to analyze the expression of ER stress chaperone molecules. Female C57BL/6 mice were intraperitoneally injected with 12 mg/kg AOM. On the 7th day following AOM injection, mice were treated with 1% DSS supplemented to the drinking water for 7 days, then followed by 14 days of normal drinking water. The cycle of 7 days DSS plus 14 days normal water was repeated twice and colonic tumors were evaluated for their number and size. Mice in the control group were injected with saline and received normal drinking water for the course of the experiment. mRNA levels of cytokines, inositol-requiring enzyme (IRE)1α and 1β, their downstream targets X-box binding protein (XBP)1u, XBP1s and mucin (MUC) 2 and interleukin (IL)-6, IL-8 and tumor necrosis factor (TNF)-α were detected by reverse transcription-quantitative polymerase chain reaction. IRE1α, IRE1β and MUC2 protein expression was evaluated by immunohistochemistry, and IRE1α and IRE1β levels were further assessed by western blot analysis. It was observed that tumors developed in the distal colon of mice treated with AOM/DSS. IL-6, IL-8 and TNF-α mRNA levels were significantly increased in mice of the tumor group compared with mice of the control group. There were no significant differences in IRE1α mRNA and protein expression between the two groups and XBP1s mRNA levels were increased in the tumor compared with the control group. IRE1β and MUC2 mRNA levels were significantly decreased in the tumor compared with the control group (decreased by 42 and 30%, respectively). IRE1β and MUC2 proteins were predominately expressed in colonic epithelial cells and expression was decreased in the tumor compared with the control group. In conclusion, the downregulation of IRE1β and MUC2 may reduce the ability of colon tissues to resist inflammation, thus promoting the occurrence and development of colonic tumors.
Collapse
Affiliation(s)
- Faliang Dai
- Department of Gastroenterology and Hepatology, Beijing Rehabilitation Hospital, Capital Medical University, Beijing 100144, P.R. China.,Department of Gastroenterology and Hepatology, The First Affiliated Hospital and Clinical Medical College, Henan University of Science and Technology, Luoyang, Henan 471023, P.R. China.,Department of Surgery, The First Affiliated Hospital and Clinical Medical College, Henan University of Science and Technology, Luoyang, Henan 471023, P.R. China.,Department of Minimally Invasive Interventional Oncology, Caoxian People's Hospital, Heze, Shandong 274400, P.R. China
| | - Shizhen Dong
- Department of Clinical Laboratory Diagnosis, The First Affiliated Hospital and Clinical Medical College, Henan University of Science and Technology, Luoyang, Henan 471023, P.R. China
| | - Zhenhua Rong
- Department of Minimally Invasive Interventional Oncology, Caoxian People's Hospital, Heze, Shandong 274400, P.R. China
| | - Qingxia Xuan
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital and Clinical Medical College, Henan University of Science and Technology, Luoyang, Henan 471023, P.R. China.,Department of Ultrasound, Women and Children Health Care Center of Luoyang, Luoyang, Henan 471000, P.R. China
| | - Pan Chen
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital and Clinical Medical College, Henan University of Science and Technology, Luoyang, Henan 471023, P.R. China
| | - Menglu Chen
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital and Clinical Medical College, Henan University of Science and Technology, Luoyang, Henan 471023, P.R. China
| | - Yonggang Fan
- Department of Surgery, The First Affiliated Hospital and Clinical Medical College, Henan University of Science and Technology, Luoyang, Henan 471023, P.R. China
| | - Qiang Gao
- Department of Gastroenterology and Hepatology, Beijing Rehabilitation Hospital, Capital Medical University, Beijing 100144, P.R. China.,Department of Gastroenterology and Hepatology, The First Affiliated Hospital and Clinical Medical College, Henan University of Science and Technology, Luoyang, Henan 471023, P.R. China
| |
Collapse
|
22
|
Driscoll K, Deshpande A, Chapp A, Li K, Datta R, Ramakrishna W. Anti-inflammatory and immune-modulating effects of rice callus suspension culture (RCSC) and bioactive fractions in an in vitro inflammatory bowel disease model. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 57:364-376. [PMID: 30831485 DOI: 10.1016/j.phymed.2018.12.034] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 12/24/2018] [Accepted: 12/29/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Rice callus suspension culture (RCSC) has been shown to exhibit potent antiproliferative activity in multiple cancer cell lines. RCSC and its bioactive compounds can fill the need for drugs with no side effects. HYPOTHESIS/PURPOSE The anti-inflammatory potential of RCSC and its bioactive fractions on normal colon epithelial cell lines, was investigated. STUDY DESIGN Three cell lines, InEpC, NCM356 and CCD841-CoN were treated with proinflammatory cytokines followed by RCSC. Cytoplasmic and nuclear ROS were assayed with fluorescent microscopy and flow cytometer. Expression analysis of immune-related genes was performed in RCSC-treated cell lines. RCSC was fractionated using column chromatography and HPLC. Pooled fractions 10-18 was used to test for antiproliferative activity using colon adenocarcinoma cell line, SW620 and anti-inflammatory activity using CCD841-CoN. Mass spectrometric analysis was performed to identify candidate compounds in four fractions. RESULTS RCSC treatment showed differential effects with higher cytoplasmic ROS levels in NCM356 and CCD841-CoN and lower ROS levels in InEpC. Nuclear generated ROS levels increased in all three treated cell lines. Flow cytometry analysis of propidium iodide stained cells indicated mitigation of cell death caused by inflammation in RCSC treated groups in both NCM356 and CCD841-CoN. Genes encoding transcription factors and cytokines were differentially regulated in NCM356 and CCD841-CoN cell lines treated with RCSC which provided insights into possible pathways. Analysis of pooled fractions 10-18 by HPLC identified 8 peaks. Cell viability assay with fractions 10-18 using SW620 showed that the number of viable cells were greatly reduced which was similar to 6X and 33X RCSC with very little effect on normal cells which similar to 1X RCSC. RCSC fractions increased nuclear and cytoplasmic ROS vs. both untreated and inflammatory control. Analysis of four fractions by mass spectrometry identified 4-deoxyphloridzin, 5'-methoxycurcumin, piceid and lupeol as candidate compounds which are likely to be responsible for the antiproliferative, anti-inflammatory and immune-regulating properties of RCSC. CONCLUSION RCSC and its fractions showed anti-inflammatory activity on inflamed colon epithelial cells. Downstream target candidate genes which are likely to mediate RCSC effects were identified. Candidate compounds responsible for the antiproliferative and anti-inflammatory activity of RCSC and its fractions provide possible drug targets.
Collapse
Affiliation(s)
- Kyle Driscoll
- Department of Biological Sciences, Michigan Technological University, Houghton, MI, USA
| | - Aparna Deshpande
- Department of Biological Sciences, Michigan Technological University, Houghton, MI, USA
| | - Andrew Chapp
- Department of Biological Sciences, Michigan Technological University, Houghton, MI, USA
| | - Kefeng Li
- School of Medicine, University of California, San Diego, CA, USA
| | - Rupali Datta
- Department of Biological Sciences, Michigan Technological University, Houghton, MI, USA
| | - Wusirika Ramakrishna
- Department of Biological Sciences, Michigan Technological University, Houghton, MI, USA; Department of Biochemistry and Microbial Sciences, Central University of Punjab, Bathinda, Punjab, India.
| |
Collapse
|
23
|
Eaton K, Pirani A, Snitkin ES. Replication Study: Intestinal inflammation targets cancer-inducing activity of the microbiota. eLife 2018; 7:e34364. [PMID: 30295289 PMCID: PMC6175580 DOI: 10.7554/elife.34364] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 09/19/2018] [Indexed: 01/16/2023] Open
Abstract
As part of the Reproducibility Project: Cancer Biology we published a Registered Report (Eaton et al., 2015) that described how we intended to replicate selected experiments from the paper "Intestinal Inflammation Targets Cancer-Inducing Activity of the Microbiota" (Arthur et al., 2012). Here we report the results. We observed no impact on bacterial growth or colonization capacity when the polyketide synthase (pks) genotoxic island was deleted from E. coli NC101, similar to the original study (Supplementary Figure 7; Arthur et al., 2012). However, for the experiment that compared inflammation, invasion, and neoplasia in azoxymethane (AOM)-treated interleukin-10-deficient mice mono-associated with NC101 or NC101[Formula: see text] pks the experimental timing of the replication attempt was longer than that of the original study. This difference was because in the original study the methodology was not clearly stated and likely led to the increased mortality and severity of inflammation observed in this replication attempt. Additionally, early death occurred during AOM treatment with higher mortality observed in NC101[Formula: see text] pks mono-associated mice compared to NC101, which was in the same direction, but more severe than the original study (Suppleme1ntal Figure 10; Arthur et al., 2012). A meta-analysis suggests that mice mono-associated with NC101[Formula: see text] pks have higher mortality compared to NC101. While these data were unable to address whether, under the conditions of the original study, NC101 and NC101[Formula: see text] pks differ in inflammation, invasion, and neoplasia this replication attempt demonstrates that clear description of experimental methods is essential to ensure accurate reproduction of experimental studies.
Collapse
Affiliation(s)
- Kathryn Eaton
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, United States
| | - Ali Pirani
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, United States
| | - Evan S Snitkin
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, United States
| |
Collapse
|
24
|
Carvalho L, Gomes JRM, Tavares LC, Xavier AR, Klika KD, Holmdahl R, Carvalho RA, Souto-Carneiro MM. Reactive Oxygen Species Deficiency Due to Ncf1-Mutation Leads to Development of Adenocarcinoma and Metabolomic and Lipidomic Remodeling in a New Mouse Model of Dextran Sulfate Sodium-Induced Colitis. Front Immunol 2018; 9:701. [PMID: 29867918 PMCID: PMC5960697 DOI: 10.3389/fimmu.2018.00701] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 03/21/2018] [Indexed: 12/12/2022] Open
Abstract
Inflammatory bowel disease is characterized by chronic relapsing idiopathic inflammation of the gastrointestinal tract and persistent inflammation. Studies focusing on the immune-regulatory function of reactive oxygen species (ROS) are still largely missing. In this study, we analyzed an ROS-deficient mouse model leading to colon adenocarcinoma. Colitis was induced with dextran sulfate sodium (DSS) supplied via the drinking water in wild-type (WT) and Ncf1-mutant (Ncf1) B10.Q mice using two different protocols, one mimicking recovery after acute colitis and another simulating chronic colitis. Disease progression was monitored by evaluation of clinical parameters, histopathological analysis, and the blood serum metabolome using 1H nuclear magnetic resonance spectroscopy. At each experimental time point, colons and spleens from some mice were removed for histopathological analysis and internal clinical parameters. Clinical scores for weight variation, stool consistency, colorectal bleeding, colon length, and spleen weight were significantly worse for Ncf1 than for WT mice. Ncf1 mice with only a 7-day exposure to DSS followed by a 14-day resting period developed colonic distal high-grade dysplasia in contrast to the low-grade dysplasia found in the colon of WT mice. After a 21-day resting period, there was still β-catenin-rich inflammatory infiltration in the Ncf1 mice together with high-grade dysplasia and invasive well-differentiated adenocarcinoma, while in the WT mice, high-grade dysplasia was prominent without malignant invasion and only low inflammation. Although exposure to DSS generated less severe histopathological changes in the WT group, the blood serum metabolome revealed an increased fatty acid content with moderate-to-strong correlations to inflammation score, weight variation, colon length, and spleen weight. Ncf1 mice also displayed a similar pattern but with lower coefficients and showed consistently lower glucose and/or higher lactate levels which correlated with inflammation score, weight variation, and spleen weight. In our novel, DSS-induced colitis animal model, the lack of an oxidative burst ROS was sufficient to develop adenocarcinoma, and display altered blood plasma metabolic and lipid profiles. Thus, oxidative burst seems to be necessary to prevent evolution toward cancer and may confer a protective role in a ROS-mediated self-control mechanism.
Collapse
Affiliation(s)
- Lina Carvalho
- Faculty of Medicine, Institute of Anatomic Pathology, University of Coimbra, Coimbra, Portugal
| | - Joana R M Gomes
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Ludgero C Tavares
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Ana R Xavier
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Karel D Klika
- Molecular Structure Analysis Department, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Rikard Holmdahl
- Department of Medical Biochemistry and Biophysics, Karolinska Instituite (KI), Stockholm, Sweden
| | - Rui A Carvalho
- Department of Life Sciences, Faculty of Science and Technology, Center for Functional Ecology, University of Coimbra, Coimbra, Portugal.,Department of Rhematology, Medical Clinic 5, Universitätsklinikum Heidelberg, Heidelberg, Germany
| | - M Margarida Souto-Carneiro
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Department of Rhematology, Medical Clinic 5, Universitätsklinikum Heidelberg, Heidelberg, Germany
| |
Collapse
|
25
|
Improvement of colitis by Lactobacillus plantarum LS/07 and inulin. ACTA VET BRNO 2018. [DOI: 10.2754/avb201786040399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The objective of this study was to investigate the effect of the probiotic strain Lactobacillus plantarum LS/07 and prebiotic inulin on the activity of β-glucuronidase, transcription nuclear factor kappa B (NFκB), myeloperoxidase (MPO), cytokine levels (IL-6, IL-8, IL-13), and counts of coliforms and lactobacilli in rats with dextran sulphate sodium (DSS) induced acute colitis. Sprague-Dawley rats were divided into groups: control (C), acute colitis (AC), prebiotic (Pre), and probiotic (Pro). Dextran sulphate sodium induced inflammatory process in the colonic tissue, increased the activity of β-glucuronidase (P < 0.05), increased counts of coliforms, decreased lactobacilli counts (P < 0.05), and activated production of the measured indicators NFκB, MPO, IL-6, IL-8, except of IL-13. Diet supplemented with L. plantarum and inulin alleviated DSS induced inflammatory process by inhibiting production of IL-6, IL-8, activities of NFκB and MPO, and by stimulation of IL-13. These results indicate that the dietary intake of Lactobacillus plantarum LS/07 and inulin suppressed the expression of markers playing an important role in the inflammatory process, which predisposes their use in prevention or treatment of acute colitis.
Collapse
|
26
|
Alteber Z, Sharbi-Yunger A, Pevsner-Fischer M, Blat D, Roitman L, Tzehoval E, Elinav E, Eisenbach L. The anti-inflammatory IFITM genes ameliorate colitis and partially protect from tumorigenesis by changing immunity and microbiota. Immunol Cell Biol 2018; 96:284-297. [PMID: 29356071 DOI: 10.1111/imcb.12000] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 12/04/2017] [Accepted: 12/06/2017] [Indexed: 12/19/2022]
Abstract
Inflammation plays pivotal roles in different stages of tumor development. Screening for predisposing genetic abnormalities and understanding the roles these genes play in the crosstalk between immune and cancer cells will provide new targets for cancer therapy and prevention. The interferon inducible transmembrane (IFITM) genes are involved in pathogenesis of the gastro-intestinal tract. We aimed at delineating the role of IFITM3 in colonic epithelial homeostasis, inflammation and colitis-associated tumorigenesis using IFITM3-deficient mice. Chemical induction of colitis in IFITM3-deficient mice results in significantly increased clinical signs of inflammation and induction of invasive tumorigenesis. Bone marrow transplantation showed that cells of the hematopoietic system are responsible for colitis deterioration. In these mice, impaired cytokine expression skewed inflammatory response toward pathogenic Th17 with reduced expression of the anti-inflammatory cytokine IL10 during the recovery phase. Intriguingly, mice lacking the entire IFITM locus developed spontaneous chronic colitis from the age of 14 weeks. Sequencing the 16S rRNA of naïve mice lacking IFITM3 gene, or the entire locus containing five IFITM genes, revealed these mice had significant bacterial differences from their wild-type littermates. Our novel results provide strong evidence for the essential role of IFITM genes in ameliorating colitis and colitis-associated tumorigenesis.
Collapse
Affiliation(s)
- Zoya Alteber
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Adi Sharbi-Yunger
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 76100, Israel
| | | | - Dan Blat
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Lior Roitman
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Esther Tzehoval
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Eran Elinav
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Lea Eisenbach
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 76100, Israel
| |
Collapse
|
27
|
Dobie R, MacRae VE, Pass C, Milne EM, Ahmed SF, Farquharson C. Suppressor of cytokine signaling 2 ( Socs2) deletion protects bone health of mice with DSS-induced inflammatory bowel disease. Dis Model Mech 2018; 11:dmm.028456. [PMID: 29343614 PMCID: PMC5818069 DOI: 10.1242/dmm.028456] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 11/06/2017] [Indexed: 12/15/2022] Open
Abstract
Individuals with inflammatory bowel disease (IBD) often present with poor bone health. The development of targeted therapies for this bone loss requires a fuller understanding of the underlying cellular mechanisms. Although bone loss in IBD is multifactorial, the altered sensitivity and secretion of growth hormone (GH) and insulin-like growth factor-1 (IGF-1) in IBD is understood to be a critical contributing mechanism. The expression of suppressor of cytokine signaling 2 (SOCS2), a well-established negative regulator of GH signaling, is stimulated by proinflammatory cytokines. Therefore, it is likely that SOCS2 expression represents a critical mediator through which proinflammatory cytokines inhibit GH/IGF-1 signaling and decrease bone quality in IBD. Using the dextran sodium sulfate (DSS) model of colitis, we reveal that endogenously elevated GH function in the Socs2−/− mouse protects the skeleton from osteopenia. Micro-computed tomography assessment of DSS-treated wild-type (WT) mice revealed a worsened trabecular architecture compared to control mice. Specifically, DSS-treated WT mice had significantly decreased bone volume, trabecular thickness and trabecular number, and a resulting increase in trabecular separation. In comparison, the trabecular bone of Socs2-deficient mice was partially protected from the adverse effects of DSS. The reduction in a number of parameters, including bone volume, was less, and no changes were observed in trabecular thickness or separation. This protected phenotype was unlikely to be a consequence of improved mucosal health in the DSS-treated Socs2−/− mice but rather a result of unregulated GH signaling directly on bone. These studies indicate that the absence of SOCS2 is protective against bone loss typical of IBD. This study also provides an improved understanding of the relative effects of GH/IGF-1 signaling on bone health in experimental colitis, information that is essential before these drugs are explored as bone protective agents in children and adults with IBD. Summary: Using a mouse model of inflammatory bowel disease, this article provides an improved understanding of the relative effects of GH/IGF-1 on bone health in experimental colitis.
Collapse
Affiliation(s)
- Ross Dobie
- Division of Developmental Biology, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Roslin, Midlothian, Edinburgh EH25 9RG, UK
| | - Vicky E MacRae
- Division of Developmental Biology, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Roslin, Midlothian, Edinburgh EH25 9RG, UK
| | - Chloe Pass
- Division of Developmental Biology, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Roslin, Midlothian, Edinburgh EH25 9RG, UK
| | - Elspeth M Milne
- Division of Developmental Biology, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Roslin, Midlothian, Edinburgh EH25 9RG, UK
| | - S Faisal Ahmed
- School of Medicine, University of Glasgow, Royal Hospital for Children, Govan Road, Glasgow G51 4TF, UK
| | - Colin Farquharson
- Division of Developmental Biology, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Roslin, Midlothian, Edinburgh EH25 9RG, UK
| |
Collapse
|
28
|
Abstract
Inflammatory bowel disease (IBD), including ulcerative colitis (UC) and Crohn's disease (CD), significantly increases the risk for development of colorectal cancer. Specifically, dysplasia and cancer associated with IBD (colitis-associated cancer or CAC) develop as a result of repeated cycles of injury and healing in the intestinal epithelium. Animal models are utilized to examine the mechanisms of CAC, the role of epithelial and immune cells in this process, as well as the development of novel therapeutic targets. These models typically begin with the administration of a carcinogenic compound, and inflammation is caused by repeated cycles of colitis-inducing agents. This review describes a common CAC model that utilizes the pro-carcinogenic compound azoxymethane (AOM) followed by dextran sulfate sodium (DSS) which induces the inflammatory insult.
Collapse
|
29
|
Lynchak OV, Prylutskyy YI, Rybalchenko VK, Kyzyma OA, Soloviov D, Kostjukov VV, Evstigneev MP, Ritter U, Scharff P. Comparative Analysis of the Antineoplastic Activity of C 60 Fullerene with 5-Fluorouracil and Pyrrole Derivative In Vivo. NANOSCALE RESEARCH LETTERS 2017; 12:8. [PMID: 28058641 PMCID: PMC5215998 DOI: 10.1186/s11671-016-1775-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 12/07/2016] [Indexed: 06/06/2023]
Abstract
The antitumor activity of pristine C60 fullerene aqueous solution (C60FAS) compared to 5-fluorouracil (5-FU) and pyrrole derivative 1-(4-Cl-benzyl)-3-Cl-4-(CF3-fenylamino)-1H-pyrrol-2.5-dione (MI-1) cytostatic drugs was investigated and analyzed in detail using the model of colorectal cancer induced by 1.2-dimethylhydrazine (DMH) in rats. The number, size, and location of the tumors were measured, and the pathology was examined. It was found that the number of tumors and total lesion area decreased significantly under the action of C60FAS and MI-1. Because these drugs have different mechanisms of action, their simultaneous administration can potentially increase the effectiveness and significantly reduce the side effects of antitumor therapy.
Collapse
Affiliation(s)
- O V Lynchak
- Taras Shevchenko National University of Kyiv, 64 Volodymyrska Str, 01601, Kyiv, Ukraine.
| | - Yu I Prylutskyy
- Taras Shevchenko National University of Kyiv, 64 Volodymyrska Str, 01601, Kyiv, Ukraine
| | - V K Rybalchenko
- Taras Shevchenko National University of Kyiv, 64 Volodymyrska Str, 01601, Kyiv, Ukraine
| | - O A Kyzyma
- Taras Shevchenko National University of Kyiv, 64 Volodymyrska Str, 01601, Kyiv, Ukraine
- Joint Institute for Nuclear Research, Dubna, Moscow Region, Russia
| | - D Soloviov
- Joint Institute for Nuclear Research, Dubna, Moscow Region, Russia
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, Russia
| | - V V Kostjukov
- Sevastopol State University, Sevastopol, Crimea, Ukraine
| | - M P Evstigneev
- Sevastopol State University, Sevastopol, Crimea, Ukraine
- Belgorod State University, 85 Pobedy Str, 308015, Belgorod, Russia
| | - U Ritter
- Institute of Chemistry and Biotechnology, Technical University of Ilmenau, 25 Weimarer Str, 98693, Ilmenau, Germany
| | - P Scharff
- Institute of Chemistry and Biotechnology, Technical University of Ilmenau, 25 Weimarer Str, 98693, Ilmenau, Germany
| |
Collapse
|
30
|
De Arcangelis A, Hamade H, Alpy F, Normand S, Bruyère E, Lefebvre O, Méchine-Neuville A, Siebert S, Pfister V, Lepage P, Laquerriere P, Dembele D, Delanoye-Crespin A, Rodius S, Robine S, Kedinger M, Van Seuningen I, Simon-Assmann P, Chamaillard M, Labouesse M, Georges-Labouesse E. Hemidesmosome integrity protects the colon against colitis and colorectal cancer. Gut 2017; 66:1748-1760. [PMID: 27371534 PMCID: PMC5595104 DOI: 10.1136/gutjnl-2015-310847] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Revised: 05/12/2016] [Accepted: 05/30/2016] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Epidemiological and clinical data indicate that patients suffering from IBD with long-standing colitis display a higher risk to develop colorectal high-grade dysplasia. Whereas carcinoma invasion and metastasis rely on basement membrane (BM) disruption, experimental evidence is lacking regarding the potential contribution of epithelial cell/BM anchorage on inflammation onset and subsequent neoplastic transformation of inflammatory lesions. Herein, we analyse the role of the α6β4 integrin receptor found in hemidesmosomes that attach intestinal epithelial cells (IECs) to the laminin-containing BM. DESIGN We developed new mouse models inducing IEC-specific ablation of α6 integrin either during development (α6ΔIEC) or in adults (α6ΔIEC-TAM). RESULTS Strikingly, all α6ΔIEC mutant mice spontaneously developed long-standing colitis, which degenerated overtime into infiltrating adenocarcinoma. The sequence of events leading to disease onset entails hemidesmosome disruption, BM detachment, IL-18 overproduction by IECs, hyperplasia and enhanced intestinal permeability. Likewise, IEC-specific ablation of α6 integrin induced in adult mice (α6ΔIEC-TAM) resulted in fully penetrant colitis and tumour progression. Whereas broad-spectrum antibiotic treatment lowered tissue pathology and IL-1β secretion from infiltrating myeloid cells, it failed to reduce Th1 and Th17 response. Interestingly, while the initial intestinal inflammation occurred independently of the adaptive immune system, tumourigenesis required B and T lymphocyte activation. CONCLUSIONS We provide for the first time evidence that loss of IECs/BM interactions triggered by hemidesmosome disruption initiates the development of inflammatory lesions that progress into high-grade dysplasia and carcinoma. Colorectal neoplasia in our mouse models resemble that seen in patients with IBD, making them highly attractive for discovering more efficient therapies.
Collapse
Affiliation(s)
- Adèle De Arcangelis
- Department of Development and Stem Cells, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, Illkirch, France,Inserm, U964, Illkirch, France,CNRS, UMR 7104, Illkirch, France,Université de Strasbourg, Strasbourg, France
| | - Hussein Hamade
- Department of Development and Stem Cells, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, Illkirch, France,Inserm, U964, Illkirch, France,CNRS, UMR 7104, Illkirch, France,Université de Strasbourg, Strasbourg, France,Current address: F. Widjaja Foundation Inflammatory Bowel & Immunobiology Research Institute, Department of Medicine, Cedars Sinai Medical Center, Los Angeles, California, USA
| | - Fabien Alpy
- Inserm, U964, Illkirch, France,CNRS, UMR 7104, Illkirch, France,Université de Strasbourg, Strasbourg, France,Inserm, U1109, MNT3 Team, Strasbourg, France,Current address: Department of Functional Genomics and Cancer, IGBMC, Illkirch, France
| | - Sylvain Normand
- CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019—UMR 8204—CIIL—Centre d'Infection et d'Immunité de Lille, Université de Lille, Lille, France
| | - Emilie Bruyère
- Inserm, Université de Lille, CHRU Lille, UMR-S 1172—Jean-Pierre Aubert Research Center, Lille, France
| | - Olivier Lefebvre
- Université de Strasbourg, Strasbourg, France,Inserm, U1109, MNT3 Team, Strasbourg, France,LabEx Medalis, Université de Strasbourg, Strasbourg, France,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Agnès Méchine-Neuville
- Inserm, U1109, MNT3 Team, Strasbourg, France,CHRU Strasbourg, Hôpital de Hautepierre, Service d'anatomo-pathologie, Strasbourg, France,Current address: Département de Pathologie, Institut Bergonie, Bordeaux, France
| | - Stéphanie Siebert
- Department of Development and Stem Cells, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, Illkirch, France,Inserm, U964, Illkirch, France,CNRS, UMR 7104, Illkirch, France,Université de Strasbourg, Strasbourg, France
| | - Véronique Pfister
- Department of Development and Stem Cells, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, Illkirch, France,Inserm, U964, Illkirch, France,CNRS, UMR 7104, Illkirch, France,Université de Strasbourg, Strasbourg, France
| | - Patricia Lepage
- UMR1319—MICALIS Institute, INRA, AgroParisTech,Université Paris-Saclay, Jouy-en-Josas, France
| | - Patrice Laquerriere
- Université de Strasbourg, Strasbourg, France,CNRS, UMR 7178, Institut Pluridisciplinaire Hubert Curien,Strasbourg, France
| | - Doulaye Dembele
- Department of Development and Stem Cells, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, Illkirch, France,Inserm, U964, Illkirch, France,CNRS, UMR 7104, Illkirch, France,Université de Strasbourg, Strasbourg, France
| | - Anne Delanoye-Crespin
- CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019—UMR 8204—CIIL—Centre d'Infection et d'Immunité de Lille, Université de Lille, Lille, France
| | - Sophie Rodius
- Department of Development and Stem Cells, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, Illkirch, France,Inserm, U964, Illkirch, France,CNRS, UMR 7104, Illkirch, France,Université de Strasbourg, Strasbourg, France,Current address: NORLUX Neuro-Oncology Laboratory, CRP-Santé, Luxembourg
| | - Sylvie Robine
- Institut Curie, Paris, France,CNRS, UMR 144, Paris, France
| | - Michèle Kedinger
- Université de Strasbourg, Strasbourg, France,Inserm, U1109, MNT3 Team, Strasbourg, France
| | - Isabelle Van Seuningen
- Inserm, Université de Lille, CHRU Lille, UMR-S 1172—Jean-Pierre Aubert Research Center, Lille, France
| | - Patricia Simon-Assmann
- Université de Strasbourg, Strasbourg, France,Inserm, U1109, MNT3 Team, Strasbourg, France,LabEx Medalis, Université de Strasbourg, Strasbourg, France,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Mathias Chamaillard
- CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019—UMR 8204—CIIL—Centre d'Infection et d'Immunité de Lille, Université de Lille, Lille, France
| | - Michel Labouesse
- Department of Development and Stem Cells, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, Illkirch, France,Inserm, U964, Illkirch, France,CNRS, UMR 7104, Illkirch, France,Université de Strasbourg, Strasbourg, France,Current address: UMR7622, IBPS, Université Pierre et Marie Curie, Paris, France
| | - Elisabeth Georges-Labouesse
- Department of Development and Stem Cells, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, Illkirch, France,Inserm, U964, Illkirch, France,CNRS, UMR 7104, Illkirch, France,Université de Strasbourg, Strasbourg, France
| |
Collapse
|
31
|
Endharti AT, Permana S. Extract from mango mistletoes Dendrophthoe pentandra ameliorates TNBS-induced colitis by regulating CD4+ T cells in mesenteric lymph nodes. Altern Ther Health Med 2017; 17:468. [PMID: 28946886 PMCID: PMC5613399 DOI: 10.1186/s12906-017-1973-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Accepted: 09/12/2017] [Indexed: 12/19/2022]
Abstract
Background Mango mistletoes Dendrophthoe pentandra (MMDP) extract has attracted interest due to its pharmacological properties, including gastro protective effects. The aim of this study was to investigate whether MMDP extract could increase Foxp3 regulatory T cells and inhibits development of Th17 cells. Methods Colitis was induced in Balb/c mice by rectal administration of 2,4,6-trinitrobenzene sulfonic acid (TNBS). The mice were randomly divided into five groups comprising group1 receiving vehicle (the negative control), group 2–5 receiving TNBS, group 3–5 orally receiving either MMDP extract 150, 300 and 600 mg/kgBW for 7 days after TNBS administration. On day 8 of the experiment, the colon tissues were removed for histological examination, cytokine and myeloperoxidase (MPO) measurement. T-cells sub-population in mesenteric lymph nodes were analyzed by flow cytometer. Results MMDP extract potently suppressed colon shortening and MPO in mice with TNBS-induced colitis. Administration of the extract significantly decreased the severity of TNBS-induced colitis in a dose-dependent manner. The extract significantly attenuated the loss of body weight (p < 0.05). These effects were associated with a remarkable amelioration of the disruption of the colonic architecture, significant reduction of the colonic MPO (p < 0.05). The extract lowered the levels of Th17-associated cytokines but increased the production of Treg-associated cytokines in mesenteric lymph node cells. Conclusion Our results suggest that MMDP has the therapeutic potential to ameliorate TNBS-induced colitis symptoms revealed by histological change and inhibit IL-17 production.
Collapse
|
32
|
Yamauchi R, Kominato K, Mitsuyama K, Takedatsu H, Yoshioka S, Kuwaki K, Yamasaki H, Fukunaga S, Mori A, Akiba J, Tsuruta O, Torimura T. Stereomicroscopic features of colitis-associated tumors in mice: Evaluation of pit pattern. Oncol Lett 2017; 14:3675-3682. [PMID: 28927130 DOI: 10.3892/ol.2017.6645] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 03/03/2017] [Indexed: 12/27/2022] Open
Abstract
Patients with longstanding ulcerative colitis have an increased risk of colorectal cancer. Mouse models for colitis-associated tumors are indispensable for the development of novel strategies for prevention and intervention, as well as an improved understanding of the mechanisms underlying tumor formation. The present study examined whether stereomicroscopic observations with dye-application were able to detect and discriminate tumors in a colitis-associated tumor model in mice. Colonic tumors were induced in C57BL/6 mice by 15 cycles of treatment with dextran sulfate sodium (DSS) in drinking water. The mice were then divided into 4 groups: normal mice fed a control diet, normal mice fed an iron-supplemented diet, 0.7% DSS mice fed an iron diet and 1.5% DSS mice fed an iron diet. The entire colons were characterized with respect to both morphology and histology. The pit pattern architecture was analyzed using stereomicroscopy with dye agents (0.2% indigo carmine or 0.06% crystal violet). The tumor histology was graded as negative, indefinite or positive for dysplasia. The positive category was divided into two subcategories: low-grade dysplasia (LGD) and high-grade dysplasia (HGD). The tumor incidences and multiplicity were significantly higher in mice fed an iron diet and 1.5% DSS compared with in mice fed an iron diet and 0.7% DSS. Compared with LGD, HGD was predominantly located in the distal colon, was larger in size and had a higher incidence of elevated lesions (Is and IIa) and a lower incidence of flat lesions (IIb). In regards to the pit pattern, HGD had a high incidence of VI pits and a low incidence of IV, IIIL and II pits. In conclusion, evaluation of the pit pattern using stereomicroscopy with dye-application is useful for detecting and discriminating neoplastic changes in DSS mice and may further our understanding of the mechanisms that induce tumor formation in patients with ulcerative colitis and the characterization of pharmaceutical responses.
Collapse
Affiliation(s)
- Ryosuke Yamauchi
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Ken Kominato
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Keiichi Mitsuyama
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan.,Inflammatory Bowel Disease Center, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Hidetoshi Takedatsu
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Shinichiro Yoshioka
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Kotaro Kuwaki
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Hiroshi Yamasaki
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Shuhei Fukunaga
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Atsushi Mori
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Jun Akiba
- Department of Diagnostic Pathology, Kurume University Hospital, Kurume, Fukuoka 830-0011, Japan
| | - Osamu Tsuruta
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Takuji Torimura
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| |
Collapse
|
33
|
McCaffary D. STING signalling: an emerging common pathway in autoimmunity and cancer. Immunopharmacol Immunotoxicol 2017; 39:253-258. [PMID: 28724326 DOI: 10.1080/08923973.2017.1350704] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The equipoise between the disease states of cancer and autoinflammation has perhaps been underappreciated in clinical practice and biomedical research. However, since the discover of STING (stimulator of interferon genes) as an integral regulator of innate immunity, a wealth of information has implicated this signaling pathway in both of these diseases. Under cellular homeostasis, STING serves to detect - and promote immune defense against - DNA viruses and intracellular bacteria, as described in its initial discovery. The role of STING has since been expanded to include tumor surveillance and immune responses to cancer; indeed, defective STING responses are associated with certain cancers. Conversely, constitutive activation of this pathway can result in autoinflammatory disease, whereby STING is over-stimulated by self-DNA. This review explores the current state of STING research, concluding that further elucidation of the details of the STING pathway may offer novel therapeutics for these diseases, which are of considerable clinical gravity.
Collapse
Affiliation(s)
- David McCaffary
- a Medical Sciences Division , University of Oxford , Oxford , UK
| |
Collapse
|
34
|
Hardbower DM, Coburn LA, Asim M, Singh K, Sierra JC, Barry DP, Gobert AP, Piazuelo MB, Washington MK, Wilson KT. EGFR-mediated macrophage activation promotes colitis-associated tumorigenesis. Oncogene 2017; 36:3807-3819. [PMID: 28263971 PMCID: PMC5501754 DOI: 10.1038/onc.2017.23] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 01/12/2017] [Accepted: 01/25/2017] [Indexed: 02/08/2023]
Abstract
Epidermal growth factor receptor (EGFR) signaling is a known mediator of colorectal carcinogenesis. Studies have focused on the role of EGFR signaling in epithelial cells, although the exact nature of the role of EGFR in colorectal carcinogenesis remains a topic of debate. Here, we present evidence that EGFR signaling in myeloid cells, specifically macrophages, is critical for colon tumorigenesis in the azoxymethane-dextran sodium sulfate (AOM-DSS) model of colitis-associated carcinogenesis (CAC). In a human tissue microarray, colonic macrophages demonstrated robust EGFR activation in the pre-cancerous stages of colitis and dysplasia. Utilizing the AOM-DSS model, mice with a myeloid-specific deletion of Egfr had significantly decreased tumor multiplicity and burden, protection from high-grade dysplasia and significantly reduced colitis. Intriguingly, mice with gastrointestinal epithelial cell-specific Egfr deletion demonstrated no differences in tumorigenesis in the AOM-DSS model. The alterations in tumorigenesis in myeloid-specific Egfr knockout mice were accompanied by decreased macrophage, neutrophil and T-cell infiltration. Pro-tumorigenic M2 macrophage activation was diminished in myeloid-specific Egfr-deficient mice, as marked by decreased Arg1 and Il10 mRNA expression and decreased interleukin (IL)-4, IL10 and IL-13 protein levels. Surprisingly, diminished M1 macrophage activation was also detectable, as marked by significantly reduced Nos2 and Il1b mRNA levels and decreased interferon (IFN)-γ, tumor necrosis factor (TNF)-α and IL-1β protein levels. The alterations in M1 and M2 macrophage activation were confirmed in bone marrow-derived macrophages from mice with the myeloid-specific Egfr knockout. The combined effect of restrained M1 and M2 macrophage activation resulted in decreased production of pro-angiogenic factors, CXCL1 and vascular endothelial growth factor (VEGF), and reduced CD31+ blood vessels, which likely contributed to protection from tumorigenesis. These data reveal that EGFR signaling in macrophages, but not in colonic epithelial cells, has a significant role in CAC. EGFR signaling in macrophages may prove to be an effective biomarker of CAC or target for chemoprevention in patients with inflammatory bowel disease.
Collapse
Affiliation(s)
- Dana M. Hardbower
- Department of Pathology, Microbiology and Immunology; Vanderbilt University Medical Center; Nashville, TN, USA
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine; Vanderbilt University Medical Center; Nashville, TN, USA
| | - Lori A. Coburn
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine; Vanderbilt University Medical Center; Nashville, TN, USA
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center; Nashville, TN, USA
- Veterans Affairs Tennessee Valley Healthcare System; Nashville, TN, USA
| | - Mohammad Asim
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine; Vanderbilt University Medical Center; Nashville, TN, USA
| | - Kshipra Singh
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine; Vanderbilt University Medical Center; Nashville, TN, USA
| | - Johanna C. Sierra
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine; Vanderbilt University Medical Center; Nashville, TN, USA
| | - Daniel P. Barry
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine; Vanderbilt University Medical Center; Nashville, TN, USA
| | - Alain P. Gobert
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine; Vanderbilt University Medical Center; Nashville, TN, USA
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center; Nashville, TN, USA
| | - M. Blanca Piazuelo
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine; Vanderbilt University Medical Center; Nashville, TN, USA
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center; Nashville, TN, USA
| | - M. Kay Washington
- Department of Pathology, Microbiology and Immunology; Vanderbilt University Medical Center; Nashville, TN, USA
| | - Keith T. Wilson
- Department of Pathology, Microbiology and Immunology; Vanderbilt University Medical Center; Nashville, TN, USA
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine; Vanderbilt University Medical Center; Nashville, TN, USA
- Department of Cancer Biology; Vanderbilt University Medical Center; Nashville, TN, USA
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center; Nashville, TN, USA
- Vanderbilt Ingram Cancer Center; Vanderbilt University Medical Center; Nashville, TN, USA
- Veterans Affairs Tennessee Valley Healthcare System; Nashville, TN, USA
| |
Collapse
|
35
|
Menghini P, Di Martino L, Lopetuso LR, Corridoni D, Webster JC, Xin W, Arseneau KO, Lam M, Pizarro TT, Cominelli F. A novel model of colitis-associated cancer in SAMP1/YitFc mice with Crohn's disease-like ileitis. PLoS One 2017; 12:e0174121. [PMID: 28301579 PMCID: PMC5354461 DOI: 10.1371/journal.pone.0174121] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 03/04/2017] [Indexed: 12/19/2022] Open
Abstract
Patients with inflammatory bowel disease (IBD) are at increased risk for developing colorectal cancer. Evidence suggests that colonic dysplasia and colitis-associated cancer (CAC) are often linked to repeated cycles of epithelial cell injury and repair in the context of chronic production of inflammatory cytokines. Several mouse models of CAC have been proposed, including chemical induction through exposure to dextran sulfate sodium (DSS) with the genotoxic agents azoxymethane (AOM), 1,2-dymethylhydrazine (DHM) or targeted genetic mutations. However, such models are usually performed on healthy animals that usually lack the underlying genetic predisposition, immunological dysfunction and dysbiosis characteristic of IBD. We have previously shown that inbred SAMP1/YitFc (SAMP) mice develop a progressive Crohn’s disease (CD)-like ileitis in the absence of spontaneous colitis. We hypothesize that SAMP mice may be more susceptible to colonic tumorigenesis due to their predisposition to IBD. To test this hypothesis, we administered AOM/DSS to IBD-prone SAMP and their non-inflamed parental control strain, AKR mice. Our results showed that AOM/DSS treatment enhanced the susceptibility of colitis in SAMP compared to AKR mice, as assessed by endoscopic and histologic inflammatory scores, daily weight loss and disease activity index (DAI), during and after DSS administration. SAMP mice also showed increased colonic tumorigenesis, resulting in the occurrence of intramucosal carcinoma and a higher incidence of high-grade dysplasia and tumor burden. These phenomena occurred even in the absence of AOM and only upon repeated cycles of DSS. Taken together, our data demonstrate a heightened susceptibility to colonic inflammation and tumorigenesis in AOM/DSS-treated SAMP mice with CD-like ileitis. This novel model represents a useful tool to investigate relevant mechanisms of CAC, as well as for pre-clinical testing of potential IBD and colon cancer therapeutics.
Collapse
Affiliation(s)
- Paola Menghini
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
- Digestive Health Research Institute, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Luca Di Martino
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
- Digestive Health Research Institute, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Loris R. Lopetuso
- Digestive Health Research Institute, Case Western Reserve University, Cleveland, Ohio, United States of America
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Daniele Corridoni
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
- Digestive Health Research Institute, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Joshua C. Webster
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
- Digestive Health Research Institute, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Wei Xin
- Digestive Health Research Institute, Case Western Reserve University, Cleveland, Ohio, United States of America
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Kristen O. Arseneau
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
- Digestive Health Research Institute, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Minh Lam
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
- Digestive Health Research Institute, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Theresa T. Pizarro
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
- Digestive Health Research Institute, Case Western Reserve University, Cleveland, Ohio, United States of America
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Fabio Cominelli
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
- Digestive Health Research Institute, Case Western Reserve University, Cleveland, Ohio, United States of America
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, United States of America
- * E-mail:
| |
Collapse
|
36
|
Yap SQ, Chin CF, Hong Thng AH, Pang YY, Ho HK, Ang WH. Finely Tuned Asymmetric Platinum(IV) Anticancer Complexes: Structure-Activity Relationship and Application as Orally Available Prodrugs. ChemMedChem 2017; 12:300-311. [PMID: 28028938 DOI: 10.1002/cmdc.201600577] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 12/22/2016] [Indexed: 01/12/2023]
Abstract
Platinum(IV) bis-carboxylates are highly versatile prodrug scaffolds with different axial ligands that can be functionalized while keeping the platinum(II) pharmacophore intact. Using a sequential acylation strategy, we developed a class of PtIV prodrugs of cisplatin with contrasting lipophilic and hydrophilic ligands. We investigated their stability, reduction rates, lipophilicity, aqueous solubility, and antiproliferative efficacies, and assessed for correlations among the parameters that could be useful in drug design. We showed that compounds with high lipophilicity result in better antiproliferative effects in vitro and in vivo, with one of the three compounds tested showing better efficacy than satraplatin against an animal model of colorectal cancer, owing to its higher solubility and lower reduction rates. Our asymmetric PtIV prodrugs may pave the way for a highly predictable, fine-tuned class of orally available PtIV prodrugs for the treatment of colorectal cancer.
Collapse
Affiliation(s)
- Siew Qi Yap
- Department of Chemistry, National University of Singapore, 3 Science Drive 3, Singapore, 117543, Singapore
| | - Chee Fei Chin
- Department of Chemistry, National University of Singapore, 3 Science Drive 3, Singapore, 117543, Singapore
| | - Agnes Hwee Hong Thng
- Department of Chemistry, National University of Singapore, 3 Science Drive 3, Singapore, 117543, Singapore
| | - Yi Yun Pang
- Department of Pharmacy, National University of Singapore, 18 Science Drive 4, Singapore, 117543, Singapore
| | - Han Kiat Ho
- Department of Pharmacy, National University of Singapore, 18 Science Drive 4, Singapore, 117543, Singapore.,NUS Graduate School of Integrative Sciences and Engineering, National University of Singapore, 28 Medical Drive, Singapore, 117456, Singapore
| | - Wee Han Ang
- Department of Chemistry, National University of Singapore, 3 Science Drive 3, Singapore, 117543, Singapore.,NUS Graduate School of Integrative Sciences and Engineering, National University of Singapore, 28 Medical Drive, Singapore, 117456, Singapore
| |
Collapse
|
37
|
Anti-Inflammatory Effects of Protein Kinase Inhibitor Pyrrol Derivate. ScientificWorldJournal 2016; 2016:2145753. [PMID: 28101521 PMCID: PMC5215602 DOI: 10.1155/2016/2145753] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 11/22/2016] [Accepted: 11/30/2016] [Indexed: 01/20/2023] Open
Abstract
In our previous studies we showed antitumor and anti-inflammatory activities of protein kinases inhibitor pyrrol derivate 1-(4-Cl-benzyl)-3-Cl-4-(CF3-fenylamino)-1H-pyrrol-2,5-dione (MI-1) on rat colon cancer model. Therefore anti-inflammatory effect of MI-1 on rat acetic acid induced ulcerative colitis (UC) model was aimed to be discovered. The anti-inflammatory effects of MI-1 (2.7 mg/kg daily) compared to reference drug Prednisolone (0.7 mg/kg daily) after 14-day usage were evaluated on macro- and light microscopy levels and expressed in 21-grade scale. Redox status of bowel mucosa was also estimated. It was shown that in UC group the grade of total injury (GTI) was equal to 9.6 (GTIcontrol = 0). Increase of malonic dialdehyde (MDA) by 89% and protein carbonyl groups (PCG) by 60% and decrease of superoxide dismutase (SOD) by 40% were also observed. Prednisolone decreased GTI to 3 and leveled SOD activity, but MDA and PCG remained higher than control ones by 52% and 42%, respectively. MI-1 restored colon mucosa integrity and decreased mucosa inflammation down to GTI = 0.5 and leveled PCG and SOD. Thus, MI-1 possessed anti-inflammatory properties, which were more expressed that Prednisolone ones, as well as normalized mucosa redox balance, and so has a prospect for correction of inflammatory processes.
Collapse
|
38
|
The dual anti-inflammatory and antioxidant activities of natural honey promote cell proliferation and neural regeneration in a rat model of colitis. Acta Histochem 2016; 118:588-595. [PMID: 27378376 DOI: 10.1016/j.acthis.2016.06.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 06/16/2016] [Accepted: 06/20/2016] [Indexed: 12/14/2022]
Abstract
A decreased antioxidant capacity and excessive inflammation are well-known features in the pathogenesis of ulcerative colitis (UC). Recent evidence has suggested a role of honey in reducing colitis-induced inflammatory and oxidative stress markers. In this study, we examined whether the anti-inflammatory and anti-oxidative properties of honey have a beneficial effect on the enteric innervation and cellular proliferation of UC in rat. The colitis was induced in rats by dextran sodium sulphate (DSS). The effect of natural honey on induced colitis was assessed by the following parameters in colonic samples: tissue injury, inflammatory infiltration, interleukin-1β and -6, superoxide dismutase and reduced glutathione. In addition, the expression of tumour necrosis factor-α, inducible NO synthase, caspase-3, CD34, Ki67, S100, c-kit, and neuron-specific enolase were examined by immunohistochemistry. Compared to the DSS-induced colitis group, the honey-treated group had significantly improved macroscopic and microscopic scores and exhibited the down-regulation of oxidative, inflammatory, and apoptotic markers. In addition, up-regulation of intrinsic muscular innervation and epithelial cellular proliferation markers was detected. These results provide new insight into the beneficial role of natural honey in the treatment of DSS-induced colitis via the inhibition of colonic motor dysfunction and the inflammatory-oxidative-apoptotic cascade. In addition, the role of honey in epithelial regeneration was clarified.
Collapse
|
39
|
Subramaniam R, Mizoguchi A, Mizoguchi E. Mechanistic roles of epithelial and immune cell signaling during the development of colitis-associated cancer. ACTA ACUST UNITED AC 2016; 2:1-21. [PMID: 27110580 DOI: 10.17980/2016.1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
To date, substantial evidence has shown a significant association between inflammatory bowel diseases (IBD) and development of colitis-associated cancer (CAC). The incidence/prevalence of IBD is higher in western countries including the US, Australia, and the UK. Although CAC development is generally characterized by stepwise accumulation of genetic as well as epigenetic changes, precise mechanisms of how chronic inflammation leads to the development of CAC are largely unknown. Preceding intestinal inflammation is one of the major influential factors for CAC tumorigenesis. Mucosal immune responses including activation of aberrant signaling pathways both in innate and adaptive immune cells play a pivotal role in CAC. Tumor progression and metastasis are shaped by a tightly controlled tumor microenvironment which is orchestrated by several immune cells and stromal cells including macrophages, neutrophils, dendritic cells, myeloid derived suppressor cells, T cells, and myofibroblasts. In this article, we will discuss the contributing factors of epithelial as well as immune cell signaling in initiation of CAC tumorigenesis and mucosal immune regulatory factors in the colonic tumor microenvironment. In depth understanding of these factors is necessary to develop novel anti-inflammatory and anti-cancer therapies for CAC in the near future.
Collapse
Affiliation(s)
- Renuka Subramaniam
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Atsushi Mizoguchi
- Department of Immunology, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Emiko Mizoguchi
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Department of Immunology, Kurume University School of Medicine, Kurume, Fukuoka, Japan; Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
40
|
Lee HA, Kim H, Lee KW, Park KY. Dead Nano-Sized Lactobacillus plantarum Inhibits Azoxymethane/Dextran Sulfate Sodium-Induced Colon Cancer in Balb/c Mice. J Med Food 2015; 18:1400-5. [PMID: 26595186 DOI: 10.1089/jmf.2015.3577] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The chemopreventive effects of dead nano-sized Lactobacillus plantarum (nLp) on colon carcinogenesis, induced by dextran sulfate sodium and azoxymethane, were evaluated using Balb/c mice and compared with the effects of pure live L. plantarum (pLp). nLp is a dead shrunken form of L. plantarum derived from kimchi and has a particle size of 0.5-1.0 μm. Animals fed nLp showed less weight loss, longer colons, lower colon weight/length ratios, and fewer colonic tumors compared with pLp. In addition, the administration of nLp significantly reduced the expression of inflammatory markers, mediated the expression of cell cycle and apoptotic markers in colon tissues, and elevated fecal IgA levels more than pLp. Accordingly, the present study shows that the anticolorectal cancer activities of nLp are greater than those of pLp and suggests this is due to the suppression of inflammation, the induction of cell cycle arrest and apoptosis, and enhanced IgA secretion.
Collapse
Affiliation(s)
- Hyun Ah Lee
- 1 Kimchi Research Institute, Pusan National University , Busan, Korea.,2 Department of Biotechnology, College of Life Science and Biotechnology, Korea University , Seoul, Korea
| | | | - Kwang-Won Lee
- 2 Department of Biotechnology, College of Life Science and Biotechnology, Korea University , Seoul, Korea
| | - Kun-Young Park
- 1 Kimchi Research Institute, Pusan National University , Busan, Korea
| |
Collapse
|
41
|
Van Der Kraak L, Gros P, Beauchemin N. Colitis-associated colon cancer: Is it in your genes? World J Gastroenterol 2015; 21:11688-11699. [PMID: 26556996 PMCID: PMC4631970 DOI: 10.3748/wjg.v21.i41.11688] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 07/02/2015] [Accepted: 09/15/2015] [Indexed: 02/06/2023] Open
Abstract
Colitis-associated colorectal cancer (CA-CRC) is the cause of death in 10%-15% of inflammatory bowel disease (IBD) patients. CA-CRC results from the accumulation of mutations in intestinal epithelial cells and progresses through a well-characterized inflammation to dysplasia to carcinoma sequence. Quantitative estimates of overall CA-CRC risks are highly variable ranging from 2% to 40% depending on IBD severity, duration and location, with IBD duration being the most significant risk factor associated with CA-CRC development. Recently, studies have identified IBD patients with similar patterns of colonic inflammation, but that differ with respect to CA-CRC development, suggesting a role for additional non-inflammatory risk factors in CA-CRC development. One suggestion is that select IBD patients carry polymorphisms in various low penetrance disease susceptibility genes, which pre-dispose them to CA-CRC development, although these loci have proven difficult to identify in human genome-wide association studies. Mouse models of CA-CRC have provided a viable alternative for the discovery, validation and study of individual genes in CA-CRC pathology. In this review, we summarize the current CA-CRC literature with a strong focus on genetic pre-disposition and highlight an emerging role for mouse models in the search for CA-CRC risk alleles.
Collapse
|
42
|
Abstract
BACKGROUND Intestinal microbiota influences the progression of colitis-associated colorectal cancer. With diet being a key determinant of the gut microbial ecology, dietary interventions are an attractive avenue for the prevention of colitis-associated colorectal cancer. Curcumin is the most active constituent of the ground rhizome of the Curcuma longa plant, which has been demonstrated to have anti-inflammatory, antioxidative, and antiproliferative properties. METHODS Il10 mice on 129/SvEv background were used as a model of colitis-associated colorectal cancer. Starting at 10 weeks of age, wild-type or Il10 mice received 6 weekly intraperitoneal injections of azoxymethane (AOM) or phosphate-buffered saline (PBS) and were started on either a control or a curcumin-supplemented diet. Stools were collected every 4 weeks for microbial community analysis. Mice were killed at 30 weeks of age. RESULTS Curcumin-supplemented diet increased survival, decreased colon weight/length ratio, and, at 0.5%, entirely eliminated tumor burden. Although colonic histology indicated improvement with curcumin, no effects of mucosal immune responses have been observed in PBS/Il10 mice and limited effects were seen in AOM/Il10 mice. In wild-type and in Il10 mice, curcumin increased bacterial richness, prevented age-related decrease in alpha diversity, increased the relative abundance of Lactobacillales, and decreased Coriobacterales order. Taxonomic profile of AOM/Il10 mice receiving curcumin was more similar to those of wild-type mice than those fed control diet. CONCLUSIONS In AOM/Il10 model, curcumin reduced or eliminated colonic tumor burden with limited effects on mucosal immune responses. The beneficial effect of curcumin on tumorigenesis was associated with the maintenance of a more diverse colonic microbial ecology.
Collapse
|
43
|
Effects of 5-Amyno-4-(1,3-benzothyazol-2-yn)-1-(3-methoxyphenyl)-1,2-dihydro-3H-pyrrol-3-one Intake on Digestive System in a Rat Model of Colon Cancer. ScientificWorldJournal 2015; 2015:376576. [PMID: 26504896 PMCID: PMC4609483 DOI: 10.1155/2015/376576] [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: 04/29/2015] [Revised: 09/02/2015] [Accepted: 09/09/2015] [Indexed: 12/30/2022] Open
Abstract
Introduction. Pyrrol derivate 5-amyno-4-(1,3-benzothyazol-2-yn)-1-(3-methoxyphenyl)-1,2-dihydro-3H-pyrrol-3-one (D1) has shown antiproliferative activities in vitro, so investigation of the impact of D1 intake on gut organs in rats that experienced colon cancer seems to be necessary. Materials and Methods. D1 at the dose of 2.3 mg/kg was administered per os daily for 27 (from the 1st day of experiment) or 7 (from the 21st week of experiment) weeks to rats that experienced 1,2-dimethylhydrazine (DMH)-induced colon cancer for 20 weeks. 5-Fluorouracil (5FU) was chosen as reference drug and was administered intraperitoneally weekly for 7 weeks (from the 21st week of experiment) at the dose of 45 mg/kg. Results. Antitumor activity of D1 comparable with the 5FU one against DMH-induced colon cancer in rats was observed (decrease of tumor number and tumor total area up to 46%). D1 attenuated the inflammation of colon, gastric and jejunal mucosa, and the liver, caused by DMH, unlike 5FU, aggravating the latter. In addition, D1 partially normalized mucosa morphometric parameters suggesting its functional restore. Conclusions. D1 possesses, comparable with 5-fluorouracil antitumor efficacy, less damaging effects on the tissues beyond cancerous areas and contributes to partial morphological and functional gut organs recovery.
Collapse
|
44
|
Sun Y, Lin LJ, Lin Y, Sang LX, Jiang M, Zheng CQ. Gingko biloba extract (Ginaton) ameliorates dextran sulfate sodium (DSS)-induced acute experimental colitis in mice via reducing IL-6/STAT3 and IL-23/IL-17. Int J Clin Exp Med 2015; 8:17235-17247. [PMID: 26770316 PMCID: PMC4694216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 10/09/2015] [Indexed: 06/05/2023]
Abstract
This study explored the underlying mechanism of Gingko biloba extract (Ginaton) on dextran sulfate sodium (DSS)-induced acute experimental colitis in mice. 40 male C57BL/6 mice were randomly divided into four groups: normal control group, Ginaton group, Ginaton treatment group, and DSS group. After 7 days administration, mice were sacrificed and colons were collected for H-E staining, immunohistochemistry, real-time PCR and Western blot. By observing clinical disease activity and histological damage, we assessed the effect of Ginaton on DSS-induced acute experimental colitis in mice and observed the effect of Ginaton on normal mice. We also explored the specific mechanism of Ginaton on DSS-induced acute experimental colitis in mice through examining the expression of inflammatory related mediators (gp130, STAT3, p-STAT3, ROR-γt) and cytokines (IL-6, IL-17, IL-23). Ginaton-treated DSS mice showed significant improvement over untreated DSS mice. Specifically, Ginaton improved clinical disease activity (DAI score, weight closs, colon shortening, and bloody stool) and histological damage, and reduced the expression of inflammatory-related mediators (p-STAT3, gp130, ROR-γt) and cytokines (IL-6, IL-17, IL-23). In addition, clinical disease activity, histological damage, the expression of inflammatory related mediators (STAT3, p-STAT3, gp130, ROR-t) and cytokines (IL-6, IL-17, IL-23) in mice of Ginaton group were similar to normal control group. In conclusion, Ginaton ameliorates DSS-induced acute experimental colitis in mice by reducing IL-17 production, which is at least partly involved in inhibiting IL-6/STAT3 signaling pathway and IL-23/IL-17 axis. Moreover, Ginaton itself does not cause inflammatory change in normal mice. These results support that Ginaton can be as a potential clinical treatment for ulcerative colitis (UC).
Collapse
Affiliation(s)
- Yan Sun
- Department of Gastroenterology, Shengjing Hospital of China Medical University39 Huaxiang Road, Tiexi District, Shenyang 110022, Liaoning Province, China
| | - Lian-Jie Lin
- Department of Gastroenterology, Shengjing Hospital of China Medical University39 Huaxiang Road, Tiexi District, Shenyang 110022, Liaoning Province, China
| | - Yan Lin
- Department of Gastroenterology, Shengjing Hospital of China Medical University39 Huaxiang Road, Tiexi District, Shenyang 110022, Liaoning Province, China
| | - Li-Xuan Sang
- Department of Gastroenterology, First Affiliated Hospital of China Medical University155 Nanjing North Street, Heping District, Shenyang 110001, Liaoning Province, China
| | - Min Jiang
- Department of Gastroenterology, First Affiliated Hospital of China Medical University155 Nanjing North Street, Heping District, Shenyang 110001, Liaoning Province, China
| | - Chang-Qing Zheng
- Department of Gastroenterology, Shengjing Hospital of China Medical University39 Huaxiang Road, Tiexi District, Shenyang 110022, Liaoning Province, China
| |
Collapse
|
45
|
Azzolini M, Mattarei A, La Spina M, Marotta E, Zoratti M, Paradisi C, Biasutto L. Synthesis and Evaluation as Prodrugs of Hydrophilic Carbamate Ester Analogues of Resveratrol. Mol Pharm 2015; 12:3441-54. [DOI: 10.1021/acs.molpharmaceut.5b00464] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Michele Azzolini
- Department of Biomedical Sciences, University of Padova, viale G. Colombo
3, 35131 Padova, Italy
- NÓOS Srl, via Campello sul Clitunno 34, 00181 Roma, Italy
| | - Andrea Mattarei
- Department
of Chemical Sciences, University of Padova, via Marzolo 1, 35121 Padova, Italy
| | - Martina La Spina
- Department of Biomedical Sciences, University of Padova, viale G. Colombo
3, 35131 Padova, Italy
| | - Ester Marotta
- Department
of Chemical Sciences, University of Padova, via Marzolo 1, 35121 Padova, Italy
| | - Mario Zoratti
- Department of Biomedical Sciences, University of Padova, viale G. Colombo
3, 35131 Padova, Italy
- CNR Neuroscience Institute, viale G. Colombo 3, 35131 Padova, Italy
| | - Cristina Paradisi
- Department
of Chemical Sciences, University of Padova, via Marzolo 1, 35121 Padova, Italy
| | - Lucia Biasutto
- Department of Biomedical Sciences, University of Padova, viale G. Colombo
3, 35131 Padova, Italy
- CNR Neuroscience Institute, viale G. Colombo 3, 35131 Padova, Italy
| |
Collapse
|
46
|
Aryl Hydrocarbon Receptor Activation Down-Regulates IL-7 and Reduces Inflammation in a Mouse Model of DSS-Induced Colitis. Dig Dis Sci 2015; 60:1958-66. [PMID: 25799939 DOI: 10.1007/s10620-015-3632-x] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 03/11/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS The pathogenesis of inflammatory bowel disease (IBD) is associated with dysregulation of intestinal immune system. Aryl hydrocarbon receptor (AHR) is believed to control the chronic inflammation in the gut. Besides, interleukin-7 (IL-7) is proved to be an important cytokine that activates mucosal inflammation in IBD. Moreover, intraepithelial lymphocytes (IELs) are one of the key immunological compartments involved in regulating intestinal inflammation. In this study, we investigated the function of 6-formylindolo (3,2-b) carbazole (Ficz), a ligand of AHR, on IL-7, colitis, and IEL phenotypes. METHODS Colitis was induced by administration of dextran sulfate sodium (DSS) to wild-type C57BL/6J mice for 7 days. Mice were weighted, colon tissues were collected and measured, and histology analyses were performed. IELs were isolated from colon, and the phenotype and activation of IELs were examined using flow cytometry detection. The expression of AHR and IL-7 was measured by immunofluorescence, Western blot, and RT-PCR. RESULTS Ficz down-regulated epithelial-derived IL-7 expression in mice with DSS-induced colitis and ameliorated DSS-induced colitis. Ficz also decreased CD8αβ(+) and CD8(+) IEL subpopulations, enhanced TCRγδ(+) IEL subpopulation, and reduced the percentage of activated CD4(+) and CD8(+) subpopulations. CONCLUSIONS Ficz could down-regulate epithelial-derived IL-7 expression in mice with DSS-induced colitis and inhibit inflammation in the gastrointestinal tract of mice. AHR-related compounds might be the new and promising therapeutic medicaments for the treatment of patients with IBD.
Collapse
|
47
|
Dheer R, Davies JM, Abreu MT. Inflammation and Colorectal Cancer. INTESTINAL TUMORIGENESIS 2015:211-256. [DOI: 10.1007/978-3-319-19986-3_8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
48
|
Charanya T, York T, Bloch S, Sudlow G, Liang K, Garcia M, Akers WJ, Rubin D, Gruev V, Achilefu S. Trimodal color-fluorescence-polarization endoscopy aided by a tumor selective molecular probe accurately detects flat lesions in colitis-associated cancer. JOURNAL OF BIOMEDICAL OPTICS 2014; 19:126002. [PMID: 25473883 PMCID: PMC4255434 DOI: 10.1117/1.jbo.19.12.126002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 10/24/2014] [Indexed: 05/10/2023]
Abstract
Colitis-associated cancer (CAC) arises from premalignant flat lesions of the colon, which are difficult to detect with current endoscopic screening approaches. We have developed a complementary fluorescence and polarization reporting strategy that combines the unique biochemical and physical properties of dysplasia and cancer for real-time detection of these lesions. Using azoxymethane-dextran sodium sulfate (AOM-DSS) treated mice, which recapitulates human CAC and dysplasia, we show that an octapeptide labeled with a near-infrared (NIR) fluorescent dye selectively identified all precancerous and cancerous lesions. A new thermoresponsive sol-gel formulation allowed topical application of the molecular probe during endoscopy. This method yielded high contrast-to-noise ratios (CNR) between adenomatous tumors (20.6 ± 1.65) and flat lesions (12.1 ± 1.03) and surrounding uninvolved colon tissue versus CNR of inflamed tissues (1.62±0.42) Incorporation of nanowire-filtered polarization imaging into NIR fluorescence endoscopy shows a high depolarization contrast in both adenomatous tumors and flat lesions in CAC, reflecting compromised structural integrity of these tissues. Together, the real-time polarization imaging provides real-time validation of suspicious colon tissue highlighted by molecular fluorescence endoscopy.
Collapse
Affiliation(s)
- Tauseef Charanya
- Washington University in St. Louis, Department of Radiology, 4525 Scott Avenue, East Building, St. Louis, Missouri 63110, United States
- Washington University in St. Louis, Department of Biomedical Engineering, 1 Brookings Drive, St. Louis, Missouri 63110, United States
| | - Timothy York
- Washington University in St. Louis, Department of Computer Science and Engineering, 1 Brookings Drive, St. Louis, Missouri 63110, United States
| | - Sharon Bloch
- Washington University in St. Louis, Department of Radiology, 4525 Scott Avenue, East Building, St. Louis, Missouri 63110, United States
| | - Gail Sudlow
- Washington University in St. Louis, Department of Radiology, 4525 Scott Avenue, East Building, St. Louis, Missouri 63110, United States
| | - Kexian Liang
- Washington University in St. Louis, Department of Radiology, 4525 Scott Avenue, East Building, St. Louis, Missouri 63110, United States
| | - Missael Garcia
- Washington University in St. Louis, Department of Computer Science and Engineering, 1 Brookings Drive, St. Louis, Missouri 63110, United States
| | - Walter J. Akers
- Washington University in St. Louis, Department of Radiology, 4525 Scott Avenue, East Building, St. Louis, Missouri 63110, United States
| | - Deborah Rubin
- Washington University in St. Louis, Department of Medicine, 660 South Euclid Avenue, St. Louis, Missouri 63110, United States
| | - Viktor Gruev
- Washington University in St. Louis, Department of Computer Science and Engineering, 1 Brookings Drive, St. Louis, Missouri 63110, United States
| | - Samuel Achilefu
- Washington University in St. Louis, Department of Radiology, 4525 Scott Avenue, East Building, St. Louis, Missouri 63110, United States
- Washington University in St. Louis, Department of Biomedical Engineering, 1 Brookings Drive, St. Louis, Missouri 63110, United States
- Washington University in St. Louis, Department of Biochemistry and Molecular Biophysics, 660 South Euclid Avenue, St. Louis, Missouri 63110, United States
- Address all correspondence to: Samuel Achilefu, E-mail:
| |
Collapse
|
49
|
Samuel T, Fadlalla K, Gales DN, Putcha BDK, Manne U. Variable NF-κB pathway responses in colon cancer cells treated with chemotherapeutic drugs. BMC Cancer 2014; 14:599. [PMID: 25134433 PMCID: PMC4152571 DOI: 10.1186/1471-2407-14-599] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 08/06/2014] [Indexed: 02/07/2023] Open
Abstract
Background The nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling pathway is activated in cells exposed to various stimuli, including those originating on the cell surface or in the nucleus. Activated NF-κB signaling is thought to enhance cell survival in response to these stimuli, which include chemotherapy and radiation. In the present effort, we determined which anticancer drugs preferentially activate NF-κB in colon cancer cells. Methods NF-κB reporter cells were established and treated with 5-fluorouracil (5-FU, DNA/RNA damaging), oxaliplatin (DNA damaging), camptothecin (CTP, topoisomerase inhibitor), phleomycin (radiomimetic), or erlotinib (EGFR inhibitor). The activation of NF-κB was assessed by immunofluorescence for p65 translocation, luciferase assays, and downstream targets of NF-κB activation (cIAP2, and Bcl-XL) were evaluated by immunoblotting, by ELISA (CXCL8 and IL-6 in culture supernatants), or by gene expression analysis. Results Colon cancer cells responded variably to different classes of therapeutic agents, and these agents initiated variable responses among different cell types. CPT activated NF-κB in SW480 colon cancer cells in a dose-dependent manner, but not in HCT116 cells that were either wild-type or deficient for p53. In SW480 colon cancer cells, NF-κB activation by CPT was accompanied by secretion of the cytokine CXCL8, but not by up-regulation of the anti-apoptotic genes, cIAP2 or Bcl-XL. On the contrary, treatment of HCT116 cells with CPT resulted in up-regulation of CXCR2, a receptor for CXCL8, without an increase in cytokine levels. In SW480 cells, NF-κB reporter activity, but not cytokine secretion, was inhibited by SM-7368, an NF-κB inhibitor. Conclusion The results show that, in response to cancer therapeutic agents, NF-κB activation varies with the cellular make up and that drug-induced NF-κB activation may be functionally uncoupled from anti-apoptotic outcomes found for other stimuli. Some cancer cells in a heterogeneous tumor tissue may, under therapeutic pressure, release soluble factors that have paracrine activity on neighboring cells that express the cognate receptors. Electronic supplementary material The online version of this article (doi:10.1186/1471-2407-14-599) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Temesgen Samuel
- School of Veterinary Medicine, Pathobiology Department and TU Center for Cancer Research, Tuskegee University, Tuskegee, AL 36088, USA.
| | | | | | | | | |
Collapse
|
50
|
Fleet JC. Animal models of gastrointestinal and liver diseases. New mouse models for studying dietary prevention of colorectal cancer. Am J Physiol Gastrointest Liver Physiol 2014; 307:G249-59. [PMID: 24875098 PMCID: PMC4121636 DOI: 10.1152/ajpgi.00019.2014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Colorectal cancer is a heterogeneous disease that is one of the major causes of cancer death in the U.S. There is evidence that lifestyle factors like diet can modulate the course of this disease. Demonstrating the benefit and mechanism of action of dietary interventions against colon cancer will require studies in preclinical models. Many mouse models have been developed to study colon cancer but no single model can reflect all types of colon cancer in terms of molecular etiology. In addition, many models develop only low-grade cancers and are confounded by development of the disease outside of the colon. This review will discuss how mice can be used to model human colon cancer and it will describe a variety of new mouse models that develop colon-restricted cancer as well as more advanced phenotypes for studies of late-state disease.
Collapse
Affiliation(s)
- James C. Fleet
- 1Department of Nutrition Science, Purdue University, West Lafayette, Indiana; and ,2Purdue University Center for Cancer Research, Purdue University, West Lafayette, Indiana
| |
Collapse
|