1
|
Ye C, Wu C, Li Y, Chen C, Li X, Zhang J, Xu Z, Chen H, Guo Y. Traditional medicine Xianglian pill suppresses high-fat diet-related colorectal cancer via inactivating TLR4/MyD88 by remodeling gut microbiota composition and bile acid metabolism. JOURNAL OF ETHNOPHARMACOLOGY 2024; 333:118411. [PMID: 38824980 DOI: 10.1016/j.jep.2024.118411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 05/23/2024] [Accepted: 05/29/2024] [Indexed: 06/04/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Previous studies have revealed that a high-fat diet (HFD) promotes the progression of colorectal cancer (CRC) in close association with disturbances in the intestinal flora and metabolic disorders. Xianglian pill (XLP) is a well-established traditional prescription with unique advantages in controlling intestinal flora imbalance and inflammation. However, its therapeutic effects on HFD-related CRC remain largely unknown. AIM OF THE STUDY The primary objective of this research was to investigate the anticancer mechanism of XLP in countering HFD-related CRC. MATERIALS AND METHODS The protective effect of XLP was evaluated using azoxymethane (AOM) and dextran sulfate sodium (DSS)-induced CRC model of mice exposed to a HFD. The degree of colorectal carcinogenesis, including body weight, colon length, and histopathology, was measured in mice treated with XLP and untreated mice. The effect of XLP on gut microbiota and its metabolites was detected using 16S rDNA and liquid chromatography/mass spectrometry analysis. Furthermore, a "pseudo-sterile" mouse model was constructed using antibiotics (Abx) to verify whether the gut microbiota and metabolites play a role in the pathogenesis of CRC. RESULTS XLP inhibited colorectal tumorigenesis in a dose-dependent fashion. Our findings also highlighted that XLP protected the integrity of the intestinal barrier by reducing the expression of pro-inflammatory cytokines, such as IL-6 and TNF-α, as well as the infiltration of pro-inflammatory macrophages. Mechanistically, XLP inhibited the TLR4/MyD88 pathway. Notably, the XLP treatment increased the proportion of probiotics (particularly Akkermansia) and significantly reduced fecal deoxycholic acid (DCA), a microbiota-derived metabolite of bile acids (BA) closely related to Muribaculaceae. Furthermore, after Abx treatment, XLP showed no clear antitumor effects on CRC. Simultaneously, DCA-supplemented feedings promoted colorectal tumorigenesis and provoked obvious colonic inflammation, M1 macrophage infiltration, and colonic injury. In vitro, the results of RAW-264.7 macrophages and normal intestinal epithelial cells treated with DCA corroborated our in vivo findings, demonstrating consistent patterns in inflammatory responses and intestinal barrier protein expression. CONCLUSION Our findings suggest that XLP inhibits colorectal cancer associated with HFD via inactivating TLR4/MyD88 by remodeling gut microbiota composition and BA metabolism.
Collapse
Affiliation(s)
- Chenxiao Ye
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Changhong Wu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Yan Li
- Department of Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310006, Zhejiang, China
| | - Chao Chen
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China; Department of Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310006, Zhejiang, China
| | - Xinrong Li
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China; Department of Integrative Medicine & Medical Oncology, Shengzhou People's Hospital (the First Affiliated Hospital of Zhejiang University Shengzhou Branch), Shengzhou, 312400, Zhejiang, China
| | - Jin Zhang
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China; Department of Traditional Chinese Medicine, The Second Hospital Affiliated to Air Force Medical University, Xi'an, 710038, Shaanxi, China
| | - Zhili Xu
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China; Department of Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310006, Zhejiang, China
| | - Haitao Chen
- Department of Integrated Chinese and Western Medicine, Zhejiang Cancer Hospital, Hangzhou, 310022, Zhejiang, China.
| | - Yong Guo
- Department of Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310006, Zhejiang, China.
| |
Collapse
|
2
|
Wu H, Ma W, Wang Y, Wang Y, Sun X, Zheng Q. Gut microbiome-metabolites axis: A friend or foe to colorectal cancer progression. Biomed Pharmacother 2024; 173:116410. [PMID: 38460373 DOI: 10.1016/j.biopha.2024.116410] [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: 01/09/2024] [Accepted: 03/06/2024] [Indexed: 03/11/2024] Open
Abstract
An expanding corpus of research robustly substantiates the complex interrelation between gut microbiota and the onset, progression, and metastasis of colorectal cancer. Investigations in both animal models and human subjects have consistently underscored the role of gut bacteria in a variety of metabolic activities, driven by dietary intake. These activities include amino acid metabolism, carbohydrate fermentation, and the generation and regulation of bile acids. These metabolic derivatives, in turn, have been identified as significant contributors to the progression of colorectal cancer. This thorough review meticulously explores the dynamic interaction between gut bacteria and metabolites derived from the breakdown of amino acids, fatty acid metabolism, and bile acid synthesis. Notably, bile acids have been recognized for their potential carcinogenic properties, which may expedite tumor development. Extensive research has revealed a reciprocal influence of gut microbiota on the intricate spectrum of colorectal cancer pathologies. Furthermore, strategies to modulate gut microbiota, such as dietary modifications or probiotic supplementation, may offer promising avenues for both the prevention and adjunctive treatment of colorectal cancer. Nevertheless, additional research is imperative to corroborate these findings and enhance our comprehension of the underlying mechanisms in colorectal cancer development.
Collapse
Affiliation(s)
- Hao Wu
- Department of Immunology, Basic Medicine College, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China
| | - Wenmeng Ma
- Department of Immunology, Basic Medicine College, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China
| | - Yiyao Wang
- Department of Immunology, Basic Medicine College, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China
| | - Yuanyuan Wang
- Department of anesthesiology, The Fourth Affiliated Hospital, China Medical University, Shenyang, Liaoning Province, PR China
| | - Xun Sun
- Department of Immunology, Basic Medicine College, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China.
| | - Qianqian Zheng
- Department of Pathophysiology, Basic Medicine College, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China.
| |
Collapse
|
3
|
Fiecke C, Simsek S, Sharma AK, Gallaher DD. Effect of red wheat, aleurone, and testa layers on colon cancer biomarkers, nitrosative stress, and gut microbiome composition in rats. Food Funct 2023; 14:9617-9634. [PMID: 37814914 DOI: 10.1039/d3fo03438k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/11/2023]
Abstract
We previously found greater reduction of colon cancer (CC) biomarkers for red wheat compared to white wheat regardless of refinement state. In the present study we examined whether the phenolic-rich aleurone and testa layers are drivers of chemoprevention by red wheat and their influence on gut microbiota composition using a 1,2-dimethylhydrazine-induced CC rat model. Rats were fed a low-fat diet (16% of energy as fat), high-fat diet (50% of energy as fat), or high-fat diet containing whole red wheat, refined red wheat, refined white wheat, or aleurone- or testa-enriched fractions for 12 weeks. Morphological markers (aberrant crypt foci, ACF) were assessed after methylene blue staining and biochemical markers (3-nitrotyrosine [3-NT], Dclk1) by immunohistochemical determination of staining positivity within aberrant crypts. Gut microbiota composition was evaluated from 16S rRNA gene sequencing of DNA extracted from cecal contents. Relative to the high-fat diet, the whole and refined red wheat, refined white wheat, and testa-enriched fraction decreased ACF, while only the refined red wheat and aleurone-enriched fraction decreased 3-NT. No significant differences were observed for Dclk1. An increase in microbial diversity was observed for the aleurone-enriched fraction (ACE index) and whole red wheat (Inverse Simpson Index). The diet groups significantly modified overall microbiome composition, including altered abundances of Lactobacillus, Mucispirillum, Phascolarctobacterium, and Blautia coccoides. These results suggest that red wheat may reduce CC risk through modifications to the gut microbiota and nitrosative stress, which may be due, in part, to the influence of dietary fiber and the phenolic-rich aleurone layer.
Collapse
Affiliation(s)
- Chelsey Fiecke
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, MN, 55108, USA.
| | - Senay Simsek
- North Dakota State University, Department of Plant Sciences, Cereal Science Graduate Program, Fargo, ND, 58105, USA
| | - Ashok Kumar Sharma
- Department of Animal Science, University of Minnesota, St. Paul, MN, 55108, USA
| | - Daniel D Gallaher
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, MN, 55108, USA.
| |
Collapse
|
4
|
Andini KD, Nielsen M, Suerink M, Helderman NC, Koornstra JJ, Ahadova A, Kloor M, Mourits MJ, Kok K, Sijmons RH, Bajwa–ten Broeke SW. PMS2-associated Lynch syndrome: Past, present and future. Front Oncol 2023; 13:1127329. [PMID: 36895471 PMCID: PMC9989154 DOI: 10.3389/fonc.2023.1127329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/01/2023] [Indexed: 02/25/2023] Open
Abstract
Carriers of any pathogenic variant in one of the MMR genes (path_MMR carriers) were traditionally thought to be at comparable risk of developing a range of different malignancies, foremost colorectal cancer (CRC) and endometrial cancer. However, it is now widely accepted that their cancer risk and cancer spectrum range notably depending on which MMR gene is affected. Moreover, there is increasing evidence that the MMR gene affected also influences the molecular pathogenesis of Lynch syndrome CRC. Although substantial progress has been made over the past decade in understanding these differences, many questions remain unanswered, especially pertaining to path_PMS2 carriers. Recent findings show that, while the cancer risk is relatively low, PMS2-deficient CRCs tend to show more aggressive behaviour and have a worse prognosis than other MMR-deficient CRCs. This, together with lower intratumoral immune infiltration, suggests that PMS2-deficient CRCs might have more in common biologically with sporadic MMR-proficient CRCs than with other MMR-deficient CRCs. These findings could have important consequences for surveillance, chemoprevention and therapeutic strategies (e.g. vaccines). In this review we discuss the current knowledge, current (clinical) challenges and knowledge gaps that should be targeted by future studies.
Collapse
Affiliation(s)
- Katarina D. Andini
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Maartje Nielsen
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Manon Suerink
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Noah C. Helderman
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Jan Jacob Koornstra
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Aysel Ahadova
- Department of Applied Tumour Biology, Institute of Pathology, Heidelberg University Hospital, and Clinical Cooperation Unit Applied Tumor Biology, German Cancer Research Center, Heidelberg, Germany
| | - Matthias Kloor
- Department of Applied Tumour Biology, Institute of Pathology, Heidelberg University Hospital, and Clinical Cooperation Unit Applied Tumor Biology, German Cancer Research Center, Heidelberg, Germany
| | - Marian J.E. Mourits
- Department of Gynaecology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Klaas Kok
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Rolf H. Sijmons
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Sanne W. Bajwa–ten Broeke
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
5
|
Dietary-Induced Bacterial Metabolites Reduce Inflammation and Inflammation-Associated Cancer via Vitamin D Pathway. Int J Mol Sci 2023; 24:ijms24031864. [PMID: 36768196 PMCID: PMC9914969 DOI: 10.3390/ijms24031864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 12/23/2022] [Accepted: 01/09/2023] [Indexed: 01/19/2023] Open
Abstract
Environmental factors, including westernised diets and alterations to the gut microbiota, are considered risk factors for inflammatory bowel diseases (IBD). The mechanisms underpinning diet-microbiota-host interactions are poorly understood in IBD. We present evidence that feeding a lard-based high-fat (HF) diet can protect mice from developing DSS-induced acute and chronic colitis and colitis-associated cancer (CAC) by significantly reducing tumour burden/incidence, immune cell infiltration, cytokine profile, and cell proliferation. We show that HF protection was associated with increased gut microbial diversity and a significant reduction in Proteobacteria and an increase in Firmicutes and Clostridium cluster XIVa abundance. Microbial functionality was modulated in terms of signalling fatty acids and bile acids (BA). Faecal secondary BAs were significantly induced to include moieties that can activate the vitamin D receptor (VDR), a nuclear receptor richly represented in the intestine and colon. Indeed, colonic VDR downstream target genes were upregulated in HF-fed mice and in combinatorial lipid-BAs-treated intestinal HT29 epithelial cells. Collectively, our data indicate that HF diet protects against colitis and CAC risk through gut microbiota and BA metabolites modulating vitamin D targeting pathways. Our data highlights the complex relationship between dietary fat-induced alterations of microbiota-host interactions in IBD/CAC pathophysiology.
Collapse
|
6
|
Chatelaine HA, Ramazani CA, Spencer K, Olivo‐Marston S, Bailey MT, McElroy J, Hatzakis E, Mathé EA, Kopec RE. Dietary Energy Intake and Presence of Aberrant Crypt Foci Are Associated with Phospholipid, Purine, and Taurine Metabolite Abundances in C57BL/6N Mouse Colon. Mol Nutr Food Res 2022; 66:e2200180. [PMID: 35969485 PMCID: PMC9787839 DOI: 10.1002/mnfr.202200180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 07/12/2022] [Indexed: 12/31/2022]
Abstract
SCOPE Colon metabolomes associated with high-fat (H) versus energy-restricted (E) diets in early colorectal cancer (CRC) models have never been directly compared. The objectives of this study are to elucidate metabolites associated with diet, aberrant crypt foci (ACF), and diet:ACF interaction, using a lifetime murine model. METHODS AND RESULTS Three-week-old mice consumed control (C), E, or H initiation diets for 18 weeks. ACF formation is initiated weeks 16-21 with azoxymethane injections, followed by progression diet crossover (to C, E, or H) through week 60. Colon extracts are analyzed using ultra-high-performance liquid chromatography-high resolution mass spectrometry (UHPLC-HRMS). Metabolites associated with diet, ACF, or diet:ACF are determined using regression models (FDR-adjusted p-value <0.05). No metabolites are significantly associated with initiation diets, but concentrations of acylcarnitines and phospholipids are associated with C, E, and H progression diets. Purines, taurine, and phospholipids are associated with ACF presence. No significant associations between metabolites and diet:ACF interaction are observed. CONCLUSIONS These results suggest that recent, rather than early-life, diet is more closely associated with the colon metabolome, particularly lipid metabolism. Results from this study also provide candidate biomarkers of early CRC development and provide support for the importance of early diet on influencing pre-CRC risk.
Collapse
Affiliation(s)
- Haley A. Chatelaine
- OSU Interdisciplinary Nutrition PhD Program (OSUN)Department of Human SciencesThe Ohio State University1787 Neil AveColumbusOH43210United States
- Division of Preclinical Innovation Informatics CoreNational Center for Advancing Translational Sciences9800 Medical Center DriveRockvilleMD20850USA
| | - Cynthia A. Ramazani
- Department of Biomedical InformaticsThe Ohio State UniversityColumbusOH43210USA
- Big Data for Indiana State UniversityIndiana State UniversityTerre HauteIN47807USA
| | - Kyle Spencer
- Division of Preclinical Innovation Informatics CoreNational Center for Advancing Translational Sciences9800 Medical Center DriveRockvilleMD20850USA
- Department of Biomedical InformaticsThe Ohio State UniversityColumbusOH43210USA
- Nationwide Children's HospitalColumbusOH43205USA
| | - Susan Olivo‐Marston
- Division of EpidemiologyCollege of Public HealthThe Ohio State UniversityColumbusOH43210USA
- Southern Illinois University School of MedicineSpringfieldIL62794USA
| | - Michael T. Bailey
- Nationwide Children's HospitalColumbusOH43205USA
- Department of PediatricsThe Ohio State University College of MedicineColumbusOH43210USA
- Center for Microbial PathogenesisNationwide Children's HospitalColumbusOH43205USA
- Oral and GI Research Affinity GroupNationwide Children's HospitalColumbusOH43205USA
| | - Joseph McElroy
- Department of Biomedical InformaticsThe Ohio State UniversityColumbusOH43210USA
| | - Emmanuel Hatzakis
- Department of Food Science and TechnologyThe Ohio State UniversityColumbusOH43210USA
- Foods for Health Discovery ThemeThe Ohio State UniversityColumbusOH43210USA
| | - Ewy A. Mathé
- Division of Preclinical Innovation Informatics CoreNational Center for Advancing Translational Sciences9800 Medical Center DriveRockvilleMD20850USA
- Department of Biomedical InformaticsThe Ohio State UniversityColumbusOH43210USA
- Comprehensive Cancer CenterThe Ohio State UniversityColumbusOH43210USA
- Translational Data Analytics InstituteThe Ohio State UniversityColumbusOH43210USA
| | - Rachel E. Kopec
- OSU Interdisciplinary Nutrition PhD Program (OSUN)Department of Human SciencesThe Ohio State University1787 Neil AveColumbusOH43210United States
- Foods for Health Discovery ThemeThe Ohio State UniversityColumbusOH43210USA
| |
Collapse
|
7
|
Zou Q, Lei X, Xu A, Li Z, He Q, Huang X, Xu G, Tian F, Ding Y, Zhu W. Chemokines in progression, chemoresistance, diagnosis, and prognosis of colorectal cancer. Front Immunol 2022; 13:724139. [PMID: 35935996 PMCID: PMC9353076 DOI: 10.3389/fimmu.2022.724139] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 06/27/2022] [Indexed: 12/24/2022] Open
Abstract
Plenty of factors affect the oncogenesis and progression of colorectal cancer in the tumor microenvironment, including various immune cells, stromal cells, cytokines, and other factors. Chemokine is a member of the cytokine superfamily. It is an indispensable component in the tumor microenvironment. Chemokines play an antitumor or pro-tumor role by recruitment or polarization of recruiting immune cells. Meanwhile, chemokines, as signal molecules, participate in the formation of a cross talk among signaling pathways and non-coding RNAs, which may be involved in promoting tumor progression. In addition, they also function in immune escape. Chemokines are related to drug resistance of tumor cells and may even provide reference for the diagnosis, therapy, and prognosis of patients with colorectal cancer.
Collapse
Affiliation(s)
- Qian Zou
- Department of Pathology, Guangdong Medical University, Dongguan, China
| | - Xue Lei
- Department of Pathology, Guangdong Medical University, Dongguan, China
| | - Aijing Xu
- Department of Genetics and Endocrinology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Ziqi Li
- Department of Pathology, Guangdong Medical University, Dongguan, China
| | - Qinglian He
- Department of Pathology, Guangdong Medical University, Dongguan, China
| | - Xiujuan Huang
- Department of Pathology, Guangdong Medical University, Dongguan, China
- Department of Hematology, Longgang District People’s Hospital of Shenzhen, Shenzhen, China
| | - Guangxian Xu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Institute of Clinical Laboratory, Guangdong Medical University, Dongguan, China
| | - Faqing Tian
- Department of Pathology, Guangdong Medical University, Dongguan, China
- Department of Genetics and Endocrinology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
- *Correspondence: Faqing Tian, ; Yuanlin Ding, ; Wei Zhu,
| | - Yuanlin Ding
- School of Public Health, Guangdong Medical University, Dongguan, China
- *Correspondence: Faqing Tian, ; Yuanlin Ding, ; Wei Zhu,
| | - Wei Zhu
- Department of Pathology, Guangdong Medical University, Dongguan, China
- *Correspondence: Faqing Tian, ; Yuanlin Ding, ; Wei Zhu,
| |
Collapse
|
8
|
Cholesterol Promotes Colorectal Cancer Growth by Activating the PI3K/AKT Pathway. JOURNAL OF ONCOLOGY 2022; 2022:1515416. [PMID: 35528239 PMCID: PMC9076305 DOI: 10.1155/2022/1515416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/05/2022] [Accepted: 04/09/2022] [Indexed: 11/18/2022]
Abstract
Globally, the incidence of colorectal cancer (CRC) increases each year, with an unhealthy diet representing one of the major pathogenic risk factors for CRC. Cholesterol is a vital dietary ingredient required to maintain the normal function of the body; however, disturbances in cholesterol levels have been discovered to exert a significant role in tumorigenesis. The present study is aimed at investigating the role of cholesterol in the occurrence of CRC. Briefly, CRC model mice were established through an intraperitoneal injection of azoxyemethane (AOM) and were subsequently either fed a normal diet (ND), high-fat diet (HFD), or high-fat high-cholesterol diet (HFHC). Furthermore, in vitro experiments were performed following the treatment of SW480 and HCT116 cells with cholesterol, and the cell viability and colony formation rate of CRC cells were analyzed. The findings identified that cholesterol levels were increased in CRC tissues compared with adjacent normal tissues. In contrast, the serum levels of cholesterol were decreased in patients with CRC compared with the healthy controls; however, no significant differences were observed in the cholesterol levels between stage I + II and stage III + IV patients with CRC. Notably, CRC model mice fed with an HFD or HFHC recorded a larger body weight compared with those mice fed a ND; however, no significant differences were reported in the number of tumors formed in each group. Furthermore, the tumor size in the HFHC group was discovered to be increased compared with the ND and HFD groups, and HGD and the pathological morphology were the most pronounced in the HFHC group. Moreover, mice in the HFHC group presented the highest ratio of Ki-67-positive staining and the lowest ratio of TUNEL-positive staining compared with those in the two other groups. Cholesterol treatment also increased the cell viability and clonality of SW480 and HCT116 cells. In addition, the protein expression levels of phosphorylated-AKT were upregulated in cholesterol-induced CRC cells and tissues, whereas the treatment with BAY80-6946 attenuated the cholesterol-induced increases in the cell viability, colony formation ability, and tumor size. In conclusion, the findings of the present study suggested that cholesterol may stimulate the progression of CRC by activating the PI3K/AKT signaling pathway; however, cholesterol may not affect the number of tumors formed in CRC. In addition, cholesterol was discovered to mainly affect the advanced stages of CRC rather than the early stages.
Collapse
|
9
|
Lee H. Obesity-Associated Cancers: Evidence from Studies in Mouse Models. Cells 2022; 11:cells11091472. [PMID: 35563777 PMCID: PMC9102145 DOI: 10.3390/cells11091472] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/21/2022] [Accepted: 04/25/2022] [Indexed: 02/04/2023] Open
Abstract
Obesity, one of the major problems in modern human society, is correlated with various diseases, including type 2 diabetes mellitus (T2DM). In particular, epidemiological and experimental evidence indicates that obesity is closely linked to at least 13 different types of cancer. The mechanisms that potentially explain the link between obesity and cancer include hyperactivation of the IGF pathway, metabolic dysregulation, dysfunctional angiogenesis, chronic inflammation, and interaction between pro-inflammatory cytokines, endocrine hormones, and adipokines. However, how the largely uniform morbidity of obesity leads to different types of cancer still needs to be investigated. To study the link between obesity and cancer, researchers have commonly used preclinical animal models, particularly mouse models. These models include monogenic models of obesity (e.g., ob/ob and db/db mice) and genetically modified mouse models of human cancers (e.g., Kras-driven pancreatic cancer, Apc-mutated colorectal cancer, and Her2/neu-overexpressing breast cancer). The experimental results obtained using these mouse models revealed strong evidence of a link between obesity and cancer and suggested their underlying mechanisms.
Collapse
Affiliation(s)
- Ho Lee
- Division of Cancer Biology, Research Institute, National Cancer Center, Goyang 10408, Korea; ; Tel.: +82-31-920-2274; Fax: +82-31-920-2279
- Graduate School of Cancer Science and Policy, National Cancer Center, Goyang 10408, Korea
| |
Collapse
|
10
|
3,4-Dihydroxyphenylethanol (DPE or Hydroxytyrosol) Counteracts ERK1/2 and mTOR Activation, Pro-Inflammatory Cytokine Release, Autophagy and Mitophagy Reduction Mediated by Benzo[a]pyrene in Primary Human Colonic Epithelial Cells. Pharmaceutics 2022; 14:pharmaceutics14030663. [PMID: 35336037 PMCID: PMC8948646 DOI: 10.3390/pharmaceutics14030663] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/10/2022] [Accepted: 03/16/2022] [Indexed: 02/04/2023] Open
Abstract
Understanding the effects induced by carcinogens on primary colonic epithelial cells and how to counteract them might help to prevent colon cancer, which is one of the most frequent and aggressive cancers. In this study, we exposed primary human colonic epithelial cells (HCoEpC) to Benzo[a]pyrene (B[a]P) and found that it led to an increased production of pro-inflammatory cytokines and activated ERK1/2 and mTOR. These pathways are known to be involved in inflammatory bowel disease (IBD), which represents a colon cancer risk factor. Moreover, B[a]P reduced autophagy and mitophagy, processes whose dysregulation has been clearly demonstrated to predispose to cancer either by in vitro or in vivo studies. Interestingly, all the effects induced by B[a]P could be counteracted by 3,4-Dihydroxyphenylethanol (DPE or Hydroxytyrosol, H), the most powerful anti-inflammatory and antioxidant compound contained in olive oil. This study sheds light on the mechanisms that could be involved in colon carcinogenesis induced by a chemical carcinogen and identifies a safe natural product that may help to prevent them.
Collapse
|
11
|
Sun Y, Liu B, Chen Y, Xing Y, Zhang Y. Multi-Omics Prognostic Signatures Based on Lipid Metabolism for Colorectal Cancer. Front Cell Dev Biol 2022; 9:811957. [PMID: 35223868 PMCID: PMC8874334 DOI: 10.3389/fcell.2021.811957] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 12/08/2021] [Indexed: 12/12/2022] Open
Abstract
Background: The potential biological processes and laws of the biological components in malignant tumors can be understood more systematically and comprehensively through multi-omics analysis. This study elaborately explored the role of lipid metabolism in the prognosis of colorectal cancer (CRC) from the metabonomics and transcriptomics. Methods: We performed K-means unsupervised clustering algorithm and t test to identify the differential lipid metabolites determined by liquid chromatography tandem mass spectrometry (LC-MS/MS) in the serum of 236 CRC patients of the First Hospital of Jilin University (JLUFH). Cox regression analysis was used to identify prognosis-associated lipid metabolites and to construct multi-lipid-metabolite prognostic signature. The composite nomogram composed of independent prognostic factors was utilized to individually predict the outcome of CRC patients. Glycerophospholipid metabolism was the most significant enrichment pathway for lipid metabolites in CRC, whose related hub genes (GMRHGs) were distinguished by gene set variation analysis (GSVA) and weighted gene co-expression network analysis (WGCNA). Cox regression and least absolute shrinkage and selection operator (LASSO) regression analysis were utilized to develop the prognostic signature. Results: Six-lipid-metabolite and five-GMRHG prognostic signatures were developed, indicating favorable survival stratification effects on CRC patients. Using the independent prognostic factors as variables, we established a composite nomogram to individually evaluate the prognosis of CRC patients. The AUCs of one-, three-, and five-year ROC curves were 0.815, 0.815, and 0.805, respectively, showing auspicious prognostic accuracy. Furthermore, we explored the potential relationship between tumor microenvironment (TME) and immune infiltration. Moreover, the mutational frequency of TP53 in the high-risk group was significantly higher than that in the low-risk group (p < 0.001), while in the coordinate mutational status of TP53, the overall survival of CRC patients in the high-risk group was significantly lower than that in low-risk group with statistical differences. Conclusion: We identified the significance of lipid metabolism for the prognosis of CRC from the aspects of metabonomics and transcriptomics, which can provide a novel perspective for promoting individualized treatment and revealing the potential molecular biological characteristics of CRC. The composite nomogram including a six-lipid-metabolite prognostic signature is a promising predictor of the prognosis of CRC patients.
Collapse
|
12
|
Aku AM, Patil A. Understanding cancer etiology: A review of the evidence-based Ayurvedic framework of cancer etiologies. Ayu 2022; 43:1-7. [PMID: 37554416 PMCID: PMC10405889 DOI: 10.4103/ayu.ayu_318_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 04/18/2022] [Accepted: 05/01/2023] [Indexed: 08/10/2023] Open
Abstract
Background of the Study As many as 10 million people have died from cancer globally in 2020, making it the top cause of mortality in the globe. Cancer develops as a result of the multi-stage process by which normal cells convert into tumor cells, progressing from a precancerous lesion to a malignant tumor. By avoiding risk factors and applying existing evidence-based preventative techniques, 30%-50% of malignancies may be averted. In order to avoid cancer, it is essential to know the specific causes of the disease. Nidanas, or etiologies, are well-described in Ayurvedic science. Here, the causes of cancer have been outlined so that the knowledge may be used effectively to avoid the disease. Aims Identification of cancer etiologies that have been described in classics. Evidence-based understanding of these etiologies and to comprehend the significance of etiologies in cancer prevention. Materials and methods A thorough evaluation of literature, including ancient Ayurvedic classics, modern medical texts, and articles published in reputable journals, was conducted to meet the study's goals and objectives. Results According to Ayurveda, there are three distinct phases of tumorigenesis: chronic inflammation, precancerous development, Granthi (a benign glandular swelli ng), and Arbuda (a precancerous tumor) (definite malignancy). A growing body of evidence suggests that the tumor microenvironment, which is predominantly controlled by inflammatory cells, is an essential player in the neoplastic process. Conclusion Although inflammation is coming into the picture just now in the contemporary world, Ayurveda has described this as a leading cause 5000 years back. It is evident that diet and lifestyle play a crucial role in the etiology of Shoth (inflammation).
Collapse
Affiliation(s)
- Amulya Murthy Aku
- Department of PG Studies in Swasthavritta, KAHER's Shri B.M.K. Ayurveda Mahavidyalaya, Post-Graduation Studies and Research Centre, Belagavi, Karnataka, India
| | - Ashok Patil
- Department of PG Studies in Swasthavritta, KAHER's Shri B.M.K. Ayurveda Mahavidyalaya, Post-Graduation Studies and Research Centre, Belagavi, Karnataka, India
| |
Collapse
|
13
|
Inflammatory Mediators and Gut Microbial Toxins Drive Colon Tumorigenesis by IL-23 Dependent Mechanism. Cancers (Basel) 2021; 13:cancers13205159. [PMID: 34680308 PMCID: PMC8533859 DOI: 10.3390/cancers13205159] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/03/2021] [Accepted: 10/08/2021] [Indexed: 01/01/2023] Open
Abstract
Simple Summary Western-style diet, rich in high fat, is the major cause of obesity and enhanced risk of colon cancer in the USA and worldwide. The inflammatory molecules are a well-established link between obesity and the modulation of colon tumorigenesis. In particular, IL-23 plays an important role in the impact of a western-style diet on obesity, the gut microbiome, and colon tumorigenesis. However, the underlying mechanism of IL-23 production for colon tumor progression and whether IL-23 can be a potential target is not clear. Our findings signify the role of pro-tumorigenic innate immune cells, including dendritic cells and macrophages in IL-23 production by bacterial toxins and eicosanoids. IL-23 knockdown in the tumorigenic dendritic cells and macrophages inhibited the colon tumor cell and organoids growth. Taken together, targeting IL-23 may be a promising option for the prevention and treatment of high-fat/obesity-associated colon cancer in clinical trials. Abstract Obesity-associated chronic inflammation predisposes colon cancer risk development. Interleukin-23 (IL-23) is a potential inflammatory mediator linking obesity to chronic colonic inflammation, altered gut microbiome, and colon carcinogenesis. We aimed to elucidate the role of pro-inflammatory eicosanoids and gut bacterial toxins in priming dendritic cells and macrophages for IL-23 secretion to promote colon tumor progression. To investigate the association of IL-23 with obesity and colon tumorigenesis, we utilized TCGA data set and colonic tumors from humans and preclinical models. To understand IL-23 production by inflammatory mediators and gut microbial toxins, we performed several in vitro mechanistic studies to mimic the tumor microenvironment. Colonic tumors were utilized to perform the ex vivo experiments. Our findings showed that IL-23 is elevated in obese individuals, colonic tumors and correlated with reduced disease-free survival. In vitro studies showed that IL-23 treatment increased the colon tumor cell self-renewal, migration, and invasion while disrupting epithelial barrier permeability. Co-culture experiments of educated dendritic cells/macrophages with colon cancer cells significantly increased the tumor aggression by increasing the secretory levels of IL-23, and these observations are further supported by ex vivo rat colonic tumor organotypic experiments. Our results demonstrate gut microbe toxins and eicosanoids facilitate IL-23 production, which plays an important role in obesity-associated colonic tumor progression. This newly identified nexus represents a potential target for the prevention and treatment of obesity-associated colon cancer.
Collapse
|
14
|
Jin BR, Kim HJ, Sim SA, Lee M, An HJ. Anti-Obesity Drug Orlistat Alleviates Western-Diet-Driven Colitis-Associated Colon Cancer via Inhibition of STAT3 and NF-κB-Mediated Signaling. Cells 2021; 10:cells10082060. [PMID: 34440829 PMCID: PMC8394553 DOI: 10.3390/cells10082060] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/05/2021] [Accepted: 08/08/2021] [Indexed: 12/31/2022] Open
Abstract
Many researchers have argued that Western diet (WD)-induced obesity accelerates inflammation and that inflammation is a link between obesity and colorectal cancer (CRC). This study investigated the effect of WDs on the development and progression of colitis-associated colon cancer (CAC) and the efficacy of the anti-obesity agent orlistat on WD-driven CAC in mice. The results revealed that the WD exacerbated CAC in azoxymethane (AOM)/dextran sulfate sodium (DSS)-induced mice, which showed increased mortality, tumor formation, and aggravation of tumor progression. Furthermore, WD feeding also upregulated inflammation, hyperplasia, and tumorigenicity levels through the activation of STAT3 and NF-κB signaling in an AOM/DSS-induced mouse model. In contrast, treatment with orlistat increased the survival rate and alleviated the symptoms of CAC, including a recovery in colon length and tumor production decreases in WD-driven AOM/DSS-induced mice. Additionally, orlistat inhibited the extent of inflammation, hyperplasia, and tumor progression via the inhibition of STAT3 and NF-κB activation. Treatment with orlistat also suppressed the β-catenin, slug, XIAP, Cdk4, cyclin D, and Bcl-2 protein levels in WD-driven AOM/DSS-induced mice. The results of this study indicate that orlistat alleviates colon cancer promotion in WD-driven CAC mice by suppressing inflammation, especially by inhibiting STAT3 and NF-κB activation.
Collapse
Affiliation(s)
- Bo-Ram Jin
- Department of Pharmacology, College of Korean Medicine, Sangji University, 83 Sangjidae-gil, Wonju-si 26339, Gangwon-do, Korea; (B.-R.J.); (H.-J.K.); (S.-A.S.)
| | - Hyo-Jung Kim
- Department of Pharmacology, College of Korean Medicine, Sangji University, 83 Sangjidae-gil, Wonju-si 26339, Gangwon-do, Korea; (B.-R.J.); (H.-J.K.); (S.-A.S.)
| | - Seo-Ah Sim
- Department of Pharmacology, College of Korean Medicine, Sangji University, 83 Sangjidae-gil, Wonju-si 26339, Gangwon-do, Korea; (B.-R.J.); (H.-J.K.); (S.-A.S.)
| | - Minho Lee
- Department of Life Science, Dongguk University-Seoul, 32 Dongguk-ro, Ilsandong-gu, Goyang-si 10326, Gyeonggi-do, Korea
- Correspondence: (M.L.); (H.-J.A.); Tel.: +82-33-738-7503 (H.-J.A.); Fax: +82-33-730-0679 (H.-J.A.)
| | - Hyo-Jin An
- Department of Pharmacology, College of Korean Medicine, Sangji University, 83 Sangjidae-gil, Wonju-si 26339, Gangwon-do, Korea; (B.-R.J.); (H.-J.K.); (S.-A.S.)
- Correspondence: (M.L.); (H.-J.A.); Tel.: +82-33-738-7503 (H.-J.A.); Fax: +82-33-730-0679 (H.-J.A.)
| |
Collapse
|
15
|
Dłubek J, Rysz J, Jabłonowski Z, Gluba-Brzózka A, Franczyk B. The Correlation between Lipid Metabolism Disorders and Prostate Cancer. Curr Med Chem 2021; 28:2048-2061. [PMID: 32767911 DOI: 10.2174/0929867327666200806103744] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 07/12/2020] [Accepted: 07/19/2020] [Indexed: 11/22/2022]
Abstract
Prostate cancer is the second most common cancer affecting the male population all over the world. The existence of a correlation between lipid metabolism disorders and cancer of the prostate gland has been widely known for a long time. According to hypotheses, cholesterol may contribute to prostate cancer progression as a result of its participation as a signaling molecule in prostate growth and differentiation via numerous biologic mechanisms including Akt signaling and de novo steroidogenesis. The results of some studies suggest that increased cholesterol levels may be associated with a higher risk of a more aggressive course of the disease. The aforementioned alterations in the synthesis of fatty acids are a unique feature of cancer and, therefore, constitute an attractive target for therapeutic intervention in the treatment of prostate cancer. Pharmacological or gene therapy aims to reduce the activity of enzymes involved in de novo synthesis of fatty acids, FASN, ACLY (ATP citrate lyase) or SCD-1 (Stearoyl-CoA Desaturase) in particular, that may result in cells growth arrest. Nevertheless, not all cancers are unequivocally associated with hypocholesterolaemia. It cannot be ruled out that the relationship between prostate cancer and lipid disorders is not a direct quantitative correlation between carcinogenesis and the amount of circulating cholesterol. Perhaps the correspondence is more sophisticated and connected to the distribution of cholesterol fractions or even sub-fractions of e.g. HDL cholesterol.
Collapse
Affiliation(s)
- Justyna Dłubek
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, Zeromskiego 113, 90-549 Lodz, Poland
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, Zeromskiego 113, 90-549 Lodz, Poland
| | - Zbigniew Jabłonowski
- Department of Urology, Medical University of Lodz, Zeromskiego 113, 90-549 Lodz, Poland
| | - Anna Gluba-Brzózka
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, Zeromskiego 113, 90-549 Lodz, Poland
| | - Beata Franczyk
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, Zeromskiego 113, 90-549 Lodz, Poland
| |
Collapse
|
16
|
Iranpour S, Al-Mosawi AKM, Bahrami AR, Sadeghian H, Matin MM. Investigating the effects of two novel 4-MMPB analogs as potent lipoxygenase inhibitors for prostate cancer treatment. ACTA ACUST UNITED AC 2021; 28:10. [PMID: 33947474 PMCID: PMC8097893 DOI: 10.1186/s40709-021-00141-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 04/16/2021] [Indexed: 01/06/2023]
Abstract
Background Lipoxygenases are one of the critical signaling mediators which can be targeted for human prostate cancer (PC) therapy. In this study, 4-methyl-2-(4-methylpiperazinyl)pyrimido[4,5-b]benzothiazine (4-MMPB) and its two analogs, 4-propyl-2-(4-methylpiperazinyl)pyrimido[4,5-b]benzothiazine (4-PMPB) and 4-ethyl-2-(4-methylpiperazinyl)pyrimido[4,5-b]benzothiazine (4-EMPB), were proposed to have anti-tumor properties in prostate cancer. Methods After synthesizing the compounds, cytotoxic effects of 4-MMPB and its two analogs against PC-3 cancerous and HDF normal cells were investigated by 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) assay and then mechanism of cell death was assessed by flow cytometry. Finally, the anti-tumor effects of the mentioned compounds were investigated in an immunocompromised C57BL/6 mouse model. Results 4-PMPB and 4-EMPB had similar anti-cancer effects on PC-3 cells as compared with 4-MMPB, while they were not effective on normal cells. Moreover, apoptosis and ferroptosis were the main mechanisms of induced cell death in these cancerous cells. Furthermore, in vivo results indicated that both analogs had similar anti-cancer effects as 4-MMPB, leading to delayed tumor growth without any noticeable side effects in weight loss and histological investigations. Conclusion Thus, our results suggest that specific targeting of lipoxygenases via 4-MMPB analogs can be considered as a treatment of choice for PC therapy, although it requires further investigations.
Collapse
Affiliation(s)
- Sonia Iranpour
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Aseel Kamil Mohammed Al-Mosawi
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran.,Department of Biology, Faculty of Science, University of Thi-Qar, Nasiriyah, Iraq
| | - Ahmad Reza Bahrami
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran.,Industrial Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Hamid Sadeghian
- Applied Biomedical Research Center, Department of Laboratory Sciences, School of Paramedical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam M Matin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran. .,Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| |
Collapse
|
17
|
Messex JK, Liou GY. Targeting BTK Signaling in the Microenvironment of Solid Tumors as a Feasible Cancer Therapy Option. Cancers (Basel) 2021; 13:cancers13092198. [PMID: 34063667 PMCID: PMC8124209 DOI: 10.3390/cancers13092198] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/23/2021] [Accepted: 04/30/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Protein tyrosine kinase BTK is essential for B cell maturation and proliferation. Dysregulation of BTK signaling in B cells leads to B cell lymphoma. In addition to B cells, BTK is also expressed in other types of immune cells including MDSC, dendritic cells, mast cells and macrophages, all of which comprise the tumor microenvironment in solid cancers. Although BTK inhibitors have been FDA-approved as the front-line treatment for B cell malignancy CLL/SLL, studies have been reluctant to report on BTKs role within the tumor microenvironment during solid cancer development limiting the possibility of using these BTK inhibitors as an adjuvant treatment option for solid cancers. Here, we review BTK signaling within the cells found in the tumor microenvironment as well as summarizing clinical trials using BTK inhibitors which target the tumor microenvironment in an attempt to combat solid tumors. Abstract The cell environment plays a pivotal role in determining cellular outcome, as well as cancer initiation, progression, and dissemination. Within this environment, in addition to the structural components, such as the extracellular matrix, there are various types of cells surrounding the tumor cells. Communication among these cells and the tumor cells via signaling pathways is important for tumor growth. Originally discovered in patients with immunodeficiency X-linked gammaglobulinemia, the Bruton’s tyrosine kinase (BTK) signaling pathway, known for its role in B cell maturation, is critical to cancer cell proliferation, metastasis and evasion of cancer eliminating cells. Given that BTK inhibitors have been FDA approved for chronic lymphocytic leukemia/small lymphocytic lymphoma and that the majority of BTK studies have been focused on B cells, the use of BTK inhibitors as a future treatment strategy of solid tumors has yet to be evaluated. In this review, we summarize studies analyzing BTK signaling within the cells found in the tumor microenvironment, as well as clinical trial where BTK inhibitors are currently being used to target the tumor microenvironment as a way to combat solid tumors.
Collapse
Affiliation(s)
- Justin K. Messex
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA;
| | - Geou-Yarh Liou
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA;
- Department of Biological Sciences, Clark Atlanta University, Atlanta, GA 30314, USA
- Correspondence: ; Tel.: +1-(404)-880-6981; Fax: +1-(404)-880-6756
| |
Collapse
|
18
|
Abstract
Obesity is epidemiologically linked to 13 forms of cancer. The local and systemic obese environment is complex and likely affect tumors through multiple avenues. This includes modulation of cancer cell phenotypes and the composition of the tumor microenvironment. A molecular understanding of how obesity links to cancer holds promise for identifying candidate genes for targeted therapy for obese cancer patient. Herein, we review both the cell-autonomous and non-cell-autonomous mechanisms linking obesity and cancer as well as provide an overview of the mouse model systems applied to study this.
Collapse
Affiliation(s)
- Xiao-Zheng Liu
- Department of Biomedicine, University of Bergen, N-5020 Bergen, Norway
| | - Line Pedersen
- Department of Biomedicine, University of Bergen, N-5020 Bergen, Norway
| | - Nils Halberg
- Department of Biomedicine, University of Bergen, N-5020 Bergen, Norway
| |
Collapse
|
19
|
Tong Y, Gao H, Qi Q, Liu X, Li J, Gao J, Li P, Wang Y, Du L, Wang C. High fat diet, gut microbiome and gastrointestinal cancer. Theranostics 2021; 11:5889-5910. [PMID: 33897888 PMCID: PMC8058730 DOI: 10.7150/thno.56157] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 03/09/2021] [Indexed: 12/12/2022] Open
Abstract
Gastrointestinal cancer is currently one of the main causes of cancer death, with a large number of cases and a wide range of lesioned sites. A high fat diet, as a public health problem, has been shown to be correlated with various digestive system diseases and tumors, and can accelerate the occurrence of cancer due to inflammation and altered metabolism. The gut microbiome has been the focus of research in recent years, and associated with cell damage or tumor immune microenvironment changes via direct or extra-intestinal effects; this may facilitate the occurrence and development of gastrointestinal tumors. Based on research showing that both a high fat diet and gut microbes can promote the occurrence of gastrointestinal tumors, and that a high fat diet imbalances intestinal microbes, we propose that a high fat diet drives gastrointestinal tumors by changing the composition of intestinal microbes.
Collapse
Affiliation(s)
- Yao Tong
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Huiru Gao
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Qiuchen Qi
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Xiaoyan Liu
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Juan Li
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Jie Gao
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Peilong Li
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yunshan Wang
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Lutao Du
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Chuanxin Wang
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Shandong Engineering & Technology Research Center for Tumor Marker Detection, Jinan, Shandong, China
- Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Jinan, Shandong, China
| |
Collapse
|
20
|
The Dark Side of the Force: When the Immune System Is the Fuel of Tumor Onset. Int J Mol Sci 2021; 22:ijms22031224. [PMID: 33513730 PMCID: PMC7865698 DOI: 10.3390/ijms22031224] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/19/2021] [Accepted: 01/22/2021] [Indexed: 12/26/2022] Open
Abstract
Nowadays, it is well accepted that inflammation is a critical player in cancer, being, in most cases, the main character of the process. Different types of tumor arise from sites of infection or chronic inflammation. This non-resolving inflammation is responsible for tumor development at different levels: it promotes tumor initiation, as well as tumor progression, stimulating both tumor growth and metastasis. Environmental factors, lifestyle and infections are the three main triggers of chronic immune activation that promote or increase the risk of many different cancers. In this review, we focus our attention on tumor onset; in particular, we summarize the knowledge about the cause and the mechanisms behind the inflammation-driven cancer development.
Collapse
|
21
|
Bader J, Carson M, Enos R, Velazquez K, Sougiannis A, Singh U, Becker W, Nagarkatti M, Fan D, Murphy A. High-fat diet-fed ovariectomized mice are susceptible to accelerated subcutaneous tumor growth potentially through adipose tissue inflammation, local insulin-like growth factor release, and tumor associated macrophages. Oncotarget 2020; 11:4554-4569. [PMID: 33346251 PMCID: PMC7733624 DOI: 10.18632/oncotarget.27832] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 11/03/2020] [Indexed: 12/20/2022] Open
Abstract
Background: The association between obesity and colorectal cancer (CRC) risk has been well established. This relationship appears to be more significant in men than in women, which may be attributable to sex hormones. However, controlled animal studies to substantiate these claims and the mechanisms involved are lacking. Materials and Methods: MC38 murine colon adenocarcinoma cells were injected subcutaneously into high-fat diet (HFD) fed male, female and ovariectomized (OVX) female C57BL/6 mice. Results: HFD increased tumor growth (main effect) that was consistent with metabolic perturbations (P < 0.01). HFD OVX mice exhibited the most significant tumor growth compared to HFD male and female mice (p < 0.05) and this was associated with increased subcutaneous adipose tissue (p < 0.05). Further, the subcutaneous adipose tissue depots within HFD OVX mice exhibited more severe macrophage associated inflammation compared to female (P < 0.01), but not male mice. Conditioned media from subcutaneous adipose tissue of HFD OVX contained higher IGF-1 levels compared to male (P < 0.01), but not female mice. Finally, HFD OVX mice had increased M2-like gene expression in their tumor-associated macrophages (TAMs) compared to female mice (P < 0.01). Conclusions: This work provides evidences suggesting adiposity, adipose specific IGF-1, macrophage associated adipose inflammation, and TAMs as potential mechanisms driving obesity-enhanced CRC in females lacking ovarian hormones.
Collapse
Affiliation(s)
- Jackie Bader
- Department of Pathology, Microbiology, & Immunology, School of Medicine, University of South Carolina, Columbia, SC 29209, USA
| | - Meredith Carson
- Department of Pathology, Microbiology, & Immunology, School of Medicine, University of South Carolina, Columbia, SC 29209, USA
| | - Reilly Enos
- Department of Pathology, Microbiology, & Immunology, School of Medicine, University of South Carolina, Columbia, SC 29209, USA
| | - Kandy Velazquez
- Department of Pathology, Microbiology, & Immunology, School of Medicine, University of South Carolina, Columbia, SC 29209, USA
| | - Alexander Sougiannis
- Department of Pathology, Microbiology, & Immunology, School of Medicine, University of South Carolina, Columbia, SC 29209, USA
| | - Udai Singh
- Department of Medicine, University of Virginia Health Systems, Charlottesville, VA 22908, USA
| | - William Becker
- Department of Pathology, Microbiology, & Immunology, School of Medicine, University of South Carolina, Columbia, SC 29209, USA
| | - Mitzi Nagarkatti
- Department of Pathology, Microbiology, & Immunology, School of Medicine, University of South Carolina, Columbia, SC 29209, USA
| | - Daping Fan
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC 29209, USA
| | - Angela Murphy
- Department of Pathology, Microbiology, & Immunology, School of Medicine, University of South Carolina, Columbia, SC 29209, USA
| |
Collapse
|
22
|
Loke YL, Chew MT, Ngeow YF, Lim WWD, Peh SC. Colon Carcinogenesis: The Interplay Between Diet and Gut Microbiota. Front Cell Infect Microbiol 2020; 10:603086. [PMID: 33364203 PMCID: PMC7753026 DOI: 10.3389/fcimb.2020.603086] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 10/28/2020] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) incidence increases yearly, and is three to four times higher in developed countries compared to developing countries. The well-known risk factors have been attributed to low physical activity, overweight, obesity, dietary consumption including excessive consumption of red processed meats, alcohol, and low dietary fiber content. There is growing evidence of the interplay between diet and gut microbiota in CRC carcinogenesis. Although there appears to be a direct causal role for gut microbes in the development of CRC in some animal models, the link between diet, gut microbes, and colonic carcinogenesis has been established largely as an association rather than as a cause-and-effect relationship. This is especially true for human studies. As essential dietary factors influence CRC risk, the role of proteins, carbohydrates, fat, and their end products are considered as part of the interplay between diet and gut microbiota. The underlying molecular mechanisms of colon carcinogenesis mediated by gut microbiota are also discussed. Human biological responses such as inflammation, oxidative stress, deoxyribonucleic acid (DNA) damage can all influence dysbiosis and consequently CRC carcinogenesis. Dysbiosis could add to CRC risk by shifting the effect of dietary components toward promoting a colonic neoplasm together with interacting with gut microbiota. It follows that dietary intervention and gut microbiota modulation may play a vital role in reducing CRC risk.
Collapse
Affiliation(s)
- Yean Leng Loke
- Centre for Biomedical Physics, School of Healthcare and Medical Sciences, Sunway University, Petaling Jaya, Malaysia
| | - Ming Tsuey Chew
- Centre for Biomedical Physics, School of Healthcare and Medical Sciences, Sunway University, Petaling Jaya, Malaysia
| | - Yun Fong Ngeow
- Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Kajang, Malaysia.,Centre for Research on Communicable Diseases, Universiti Tunku Abdul Rahman, Kajang, Malaysia
| | - Wendy Wan Dee Lim
- Department of Gastroenterology, Sunway Medical Centre, Petaling Jaya, Malaysia
| | - Suat Cheng Peh
- Ageing Health and Well-Being Research Centre, Sunway University, Petaling Jaya, Malaysia.,Department of Medical Sciences, School of Healthcare and Medical Sciences, Sunway University, Petaling Jaya, Malaysia
| |
Collapse
|
23
|
Medina-Reyes EI, Delgado-Buenrostro NL, Díaz-Urbina D, Rodríguez-Ibarra C, Déciga-Alcaraz A, González MI, Reyes JL, Villamar-Duque TE, Flores-Sánchez ML, Hernández-Pando R, Mancilla-Díaz JM, Chirino YI, Pedraza-Chaverri J. Food-grade titanium dioxide (E171) induces anxiety, adenomas in colon and goblet cells hyperplasia in a regular diet model and microvesicular steatosis in a high fat diet model. Food Chem Toxicol 2020; 146:111786. [PMID: 33038453 DOI: 10.1016/j.fct.2020.111786] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/09/2020] [Accepted: 09/25/2020] [Indexed: 12/19/2022]
Abstract
Food-grade titanium dioxide (E171) is a white additive widely used in solid and liquid food products. There is still debate about E171 toxic effects after oral consumption since this additive is deposited in colon, liver, spleen, testis and brain. The consumption of E171 commonly occurs with Western diets that are characterized by a high fat content. Thus, E171 could worsen adverse effects associated with a high fat diet (HFD) such as anxiety, colon diseases and testicular damage. We aimed to evaluate the effects of E171 on anxiety-like behavior, colon, liver and testis and to analyze if the administration of a HFD could exacerbate adverse effects. E171 was administered at ~5 mg/kgbw by drinking water for 16 weeks and mice were fed with a Regular Diet or a HFD. E171 promoted anxiety, induced adenomas in colon, goblet cells hypertrophy and hyperplasia and mucins overexpression, but had no toxic effects on testicular tissue or spermatozoa in regular diet fed-mice. Additionally, E171 promoted microvesicular steatosis in liver in HFD fed-mice and the only HFD administration decreased the spermatozoa concentration and motility. In conclusion, E171 administration increases the number of adenomas in colon, induces hypertrophy and hyperplasia in goblet cells and microvesicular steatosis.
Collapse
Affiliation(s)
- Estefany I Medina-Reyes
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México. Ciudad Universitaria, Coyoacán, CP 04510, Ciudad de México, Mexico.
| | - Norma L Delgado-Buenrostro
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Av. de los Barrios No. 1, Tlalnepantla de Baz, CP 54090, Estado de México, Mexico
| | - Daniel Díaz-Urbina
- Laboratorio de Neurobiología de la Alimentación. Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Av. de los Barrios No. 1, Tlalnepantla de Baz, CP 54090, Estado de México, Mexico
| | - Carolina Rodríguez-Ibarra
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Av. de los Barrios No. 1, Tlalnepantla de Baz, CP 54090, Estado de México, Mexico
| | - Alejandro Déciga-Alcaraz
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Av. de los Barrios No. 1, Tlalnepantla de Baz, CP 54090, Estado de México, Mexico
| | - Marisol I González
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Av. de los Barrios No. 1, Tlalnepantla de Baz, CP 54090, Estado de México, Mexico
| | - José L Reyes
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Av. de los Barrios No. 1, Tlalnepantla de Baz, CP 54090, Estado de México, Mexico
| | - Tomás E Villamar-Duque
- Bioterio de la Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Av. de los Barrios No. 1, Tlalnepantla de Baz, CP 54090, Estado de México, Mexico
| | - María Lo Flores-Sánchez
- Bioterio de la Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Av. de los Barrios No. 1, Tlalnepantla de Baz, CP 54090, Estado de México, Mexico
| | - Rogelio Hernández-Pando
- Sección de Patología Experimental, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga 15, Tlalpan, CP 14000, Ciudad de México, Mexico
| | - Juan M Mancilla-Díaz
- Laboratorio de Neurobiología de la Alimentación. Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Av. de los Barrios No. 1, Tlalnepantla de Baz, CP 54090, Estado de México, Mexico
| | - Yolanda I Chirino
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Av. de los Barrios No. 1, Tlalnepantla de Baz, CP 54090, Estado de México, Mexico
| | - José Pedraza-Chaverri
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México. Ciudad Universitaria, Coyoacán, CP 04510, Ciudad de México, Mexico
| |
Collapse
|
24
|
Yang Y, Gao G, Shi J, Zhang J. Increased Blood Lipid Level is Associated with Cancer-Specific Mortality and All-Cause Mortality in Patients with Colorectal Cancer (≥65 Years): A Population-Based Prospective Cohort Study. Risk Manag Healthc Policy 2020; 13:855-863. [PMID: 32801961 PMCID: PMC7399450 DOI: 10.2147/rmhp.s260113] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 07/08/2020] [Indexed: 12/12/2022] Open
Abstract
Background Hyperlipidaemia is related to the development of many cancers. The aim of this study was to explore whether blood lipid levels were associated with increased rates of cancer-specific mortality and all-cause mortality in patients with colorectal cancer (CRC). Methods Data on 8504 participants from The Irish Longitudinal Study on Ageing (TILDA) were analysed. A total of 304 participants with CRC who had experienced curative surgery were included. Logistic regression analysis was performed to analyse the relationship between blood lipid levels and CRC severity. Cox regression analysis was performed to assess the association between blood lipid levels and cancer-specific mortality and all-cause mortality in patients with CRC. Results In 304 patients with CRC, the average age was 67.8±5.4 years. The logistic regression analysis indicated that elevated levels of total cholesterol (2.104 [1.358–3.650]; P-trend<0.001), triglycerides (1.665 [1.337–2.076]; P-trend=0.005) and LDL (2.127 [1.446–4.099]; P-trend<0.001) but not HDL (0.688 [0.409–1.252]; P-trend=0.124) were associated with an increased risk of higher CRC stage after adjustments were made for age, sex, marital status, BMI, drinking status, smoking status, education, physical activity, antilipidaemic medications and self-reported CVDs (≥2). Cox proportional hazard analysis showed that higher blood lipid levels of total cholesterol, triglycerides and LDL were independently associated with higher rates of cancer-specific mortality and all-cause mortality. Similar results persisted in the sensitivity analysis using antilipidaemic medications as an additional covariate and the stratification analysis using antilipidaemic medications as a stratified variable. Conclusion Increased blood lipid levels were associated with an increased risk of cancer-specific mortality and all-cause mortality in patients with CRC after adjusting for potential confounding factors. Clinicians should pay more attention to the prognostic value of increased blood lipids in patients with CRC for the risk of death.
Collapse
Affiliation(s)
- Yong Yang
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, People's Republic of China
| | - Ge Gao
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, People's Republic of China
| | - Jun Shi
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, People's Republic of China
| | - Jiangnan Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, People's Republic of China
| |
Collapse
|
25
|
Intestinal Macrophages at the Crossroad between Diet, Inflammation, and Cancer. Int J Mol Sci 2020; 21:ijms21144825. [PMID: 32650452 PMCID: PMC7404402 DOI: 10.3390/ijms21144825] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/03/2020] [Accepted: 07/04/2020] [Indexed: 12/11/2022] Open
Abstract
Intestinal macrophages are key players in the regulation of the oral tolerance, controlling gut homeostasis by discriminating innocuous antigens from harmful pathogens. Diet exerts a significant impact on human health, influencing the composition of gut microbiota and the developing of several non-communicable diseases, including cancer. Nutrients and microbiota are able to modify the profile of intestinal macrophages, shaping their key function in the maintenance of the gut homeostasis. Intestinal disease often occurs as a breakdown of this balance: defects in monocyte-macrophage differentiation, wrong dietary habits, alteration of microbiota composition, and impairment in the resolution of inflammation may contribute to the development of intestinal chronic inflammation and colorectal cancer. Accordingly, dietary interventions and macrophage-targeted therapies are emerging as innovative tools for the treatment of several intestinal pathologies. In this review, we will describe the delicate balance between diet, microbiota and intestinal macrophages in homeostasis and how the perturbation of this equilibrium may lead to the occurrence of inflammatory conditions in the gut. The understanding of the molecular pathways and dietary factors regulating the activity of intestinal macrophages might result in the identification of innovative targets for the treatments of intestinal pathologies.
Collapse
|
26
|
Shen CJ, Chang KY, Lin BW, Lin WT, Su CM, Tsai JP, Liao YH, Hung LY, Chang WC, Chen BK. Oleic acid-induced NOX4 is dependent on ANGPTL4 expression to promote human colorectal cancer metastasis. Am J Cancer Res 2020; 10:7083-7099. [PMID: 32641980 PMCID: PMC7330862 DOI: 10.7150/thno.44744] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 05/20/2020] [Indexed: 12/13/2022] Open
Abstract
Background: Colorectal cancer (CRC) progression and related mortality are highly associated with metabolic disorders. However, the molecular mechanism involved in the regulation of hyperlipidemia-associated CRC metastasis remains unclear. This study aimed to investigate the effects of angiopoietin-like 4 (ANGPTL4) on NADPH oxidase 4 (NOX4) expression and reactive oxygen species (ROS) production, which might provide new targets for improving outcomes in patients with hyperlipidemia-associated CRC metastasis. Methods: The clinical relevance of relationship between NOX4 expression and ANGPTL4 was examined in CRC patients by the Oncomine and TCGA data set. Expressions of NOX4, epithelial-mesenchymal transition (EMT) markers, and gene regulation of NOX4 in free fatty acids (FFAs)-treated CRC cells were determined. The FFAs-triggered metastatic ability of CRC cells under treatments of antioxidants or knockdown of NOX4, ANGPTL4, and MMPs was evaluated in vitro and in vivo. In addition, effects of antioxidants and depletion of metastasis-associated molecules on the correlation between ROS production and FFAs-promoted CRC metastasis were also clarified. Results: In this study, we found that the induction of NOX4, followed by the increased ROS was essential for oleic acid (OA)-promoted CRC cell metastasis. The depletion of ANGPTL4 significantly inhibited c-Jun-mediated transactivation of NOX4 expression, accompanied with reduced levels of ROS, MMP-1, and MMP-9, resulting in the disruption of OA-promoted CRC cell metastasis. Moreover, knockdown of ANGPTL4, NOX4, MMP-1, and MMP-9 or the treatment of antioxidants dramatically inhibited circulating OA-enhanced tumor cell extravasation and metastatic seeding of tumor cells in lungs, indicating that the ANGPTL4/NOX4 axis was critical for dyslipidemia-associated tumor metastasis. Conclusion: The coincident expression of NOX4 and ANGPTL4 in CRC tumor specimens provides the insight into the potential therapeutic targets for the treatment of dyslipidemia-associated CRC metastasis.
Collapse
|
27
|
Aceto GM, Catalano T, Curia MC. Molecular Aspects of Colorectal Adenomas: The Interplay among Microenvironment, Oxidative Stress, and Predisposition. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1726309. [PMID: 32258104 PMCID: PMC7102468 DOI: 10.1155/2020/1726309] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 12/23/2019] [Accepted: 12/30/2019] [Indexed: 12/11/2022]
Abstract
The development of colorectal cancer (CRC) is a multistep process initiated by a benign polyp that has the potential to evolve into in situ carcinoma through the interactions between environmental and genetic factors. CRC incidence rates are constantly increased for young adult patients presenting an advanced tumor stage. The majority of CRCs arise from colonic adenomas originating from aberrant cell proliferation of colon epithelium. Endoscopic polypectomy represents a tool for early detection and removal of polyps, although the occurrence of cancers after negative colonoscopy shows a significant incidence. It has long been recognized that the aberrant regulation of Wingless/It (Wnt)/β-Catenin signaling in the pathogenesis of colorectal cancer is supported by its critical role in the differentiation of stem cells in intestinal crypts and in the maintenance of intestinal homeostasis. For this review, we will focus on the development of adenomatous polyps through the interplay between renewal signaling in the colon epithelium and reactive oxygen species (ROS) production. The current knowledge of molecular pathology allows us to deepen the relationships between oxidative stress and other risk factors as lifestyle, microbiota, and predisposition. We underline that the chronic inflammation and ROS production in the colon epithelium can impair the Wnt/β-catenin and/or base excision repair (BER) pathways and predispose to polyp development. In fact, the coexistence of oxidative DNA damage and errors in DNA polymerase can foster C>T transitions in various types of cancer and adenomas, leading to a hypermutated phenotype of tumor cells. Moreover, the function of Adenomatous Polyposis Coli (APC) protein in regulating DNA repair is very important as therapeutic implication making DNA damaging chemotherapeutic agents more effective in CRC cells that tend to accumulate mutations. Additional studies will determine whether approaches based on Wnt inhibition would provide long-term therapeutic value in CRC, but it is clear that APC disruption plays a central role in driving and maintaining tumorigenesis.
Collapse
Affiliation(s)
- Gitana Maria Aceto
- Department of Medical, Oral and Biotechnological Sciences, G. d'Annunzio University of Chieti-Pescara, 66100 Chieti, Italy
| | - Teresa Catalano
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy
| | - Maria Cristina Curia
- Department of Medical, Oral and Biotechnological Sciences, G. d'Annunzio University of Chieti-Pescara, 66100 Chieti, Italy
| |
Collapse
|
28
|
Garcia-Villatoro EL, DeLuca JAA, Callaway ES, Allred KF, Davidson LA, Hensel ME, Menon R, Ivanov I, Safe SH, Jayaraman A, Chapkin RS, Allred CD. Effects of high-fat diet and intestinal aryl hydrocarbon receptor deletion on colon carcinogenesis. Am J Physiol Gastrointest Liver Physiol 2020; 318:G451-G463. [PMID: 31905023 PMCID: PMC7137094 DOI: 10.1152/ajpgi.00268.2019] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Consumption of a high-fat diet has been associated with an increased risk of developing colorectal cancer (CRC). However, the effects of the interaction between dietary fat content and the aryl hydrocarbon receptor (AhR) on colorectal carcinogenesis remain unclear. Mainly known for its role in xenobiotic metabolism, AhR has been identified as an important regulator for maintaining intestinal epithelial homeostasis. Although previous research using whole body AhR knockout mice has revealed an increased incidence of colon and cecal tumors, the unique role of AhR activity in intestinal epithelial cells (IECs) and modifying effects of fat content in the diet at different stages of sporadic CRC development are yet to be elucidated. In the present study, we have examined the effects of a high-fat diet on IEC-specific AhR knockout mice in a model of sporadic CRC. Although loss of AhR activity in IECs significantly induced the development of premalignant lesions, in a separate experiment, no significant changes in colon mass incidence were observed. Moreover, consumption of a high-fat diet promoted cell proliferation in crypts at the premalignant colon cancer lesion stage and colon mass multiplicity as well as β-catenin expression and nuclear localization in actively proliferating cells in colon masses. Our data demonstrate the modifying effects of high-fat diet and AhR deletion in IECs on tumor initiation and progression.NEW & NOTEWORTHY Through the use of an intestinal-specific aryl hydrocarbon receptor (AhR) knockout mouse model, this study demonstrates that the expression of AhR in intestinal epithelial cells is required to reduce the formation of premalignant colon cancer lesions. Furthermore, consumption of a high-fat diet and the loss of AhR in intestinal epithelial cells influences the development of colorectal cancer at various stages.
Collapse
Affiliation(s)
| | - Jennifer A. A. DeLuca
- 1Department of Nutrition and Food Science, Texas A&M University, College Station, Texas
| | - Evelyn S. Callaway
- 2Department of Chemical Engineering, Texas A&M University, College Station, Texas
| | - Kimberly F. Allred
- 1Department of Nutrition and Food Science, Texas A&M University, College Station, Texas
| | - Laurie A. Davidson
- 1Department of Nutrition and Food Science, Texas A&M University, College Station, Texas,3Program in Integrative Nutrition & Complex Diseases, Texas A&M University, College Station, Texas
| | - Martha E. Hensel
- 4Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas
| | - Rani Menon
- 2Department of Chemical Engineering, Texas A&M University, College Station, Texas
| | - Ivan Ivanov
- 5Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas
| | - Stephen H. Safe
- 5Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas
| | - Arul Jayaraman
- 2Department of Chemical Engineering, Texas A&M University, College Station, Texas
| | - Robert S. Chapkin
- 1Department of Nutrition and Food Science, Texas A&M University, College Station, Texas,3Program in Integrative Nutrition & Complex Diseases, Texas A&M University, College Station, Texas
| | - Clinton D. Allred
- 1Department of Nutrition and Food Science, Texas A&M University, College Station, Texas
| |
Collapse
|
29
|
Slobodnick A, Krasnokutsky S, Lehmann RA, Keenan RT, Quach J, Francois F, Pillinger MH. Colorectal Cancer Among Gout Patients Undergoing Colonoscopy. J Clin Rheumatol 2019; 25:335-340. [PMID: 31764494 DOI: 10.1097/rhu.0000000000000893] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND/OBJECTIVE The connection between gout and various cancers remains unclear. We assessed the relationship between gout and colorectal cancer in a population of veterans. METHODS We reviewed the Computerized Patient Record System of the VA New York Harbor Health Care System to assess the 10-year occurrence of colorectal cancer in patients with gout undergoing colonoscopy, versus patients with osteoarthritis but no gout. RESULTS Gout and osteoarthritis subjects were similar in age, ethnicity, body mass index, and smoking history. Among 581 gout and 598 osteoarthritis subjects with documented colonoscopies, the 10-year prevalence of colorectal cancer was significantly lower in gout (0.8%) versus osteoarthritis (3.7%) (p = 0.0008) patients. Differences in colorectal cancer rates remained significant after stratifying for nonsteroidal anti-inflammatory drug use. Among gout subjects, use of colchicine and/or allopurinol, as well as the presence/absence of concomitant osteoarthritis, did not influence colorectal cancer occurrence. On subanalysis, differences in colorectal cancer occurrence between gout and osteoarthritis subjects persisted among those who underwent diagnostic (0.5% in gout vs 4.6% in osteoarthritis subjects, p < 0.001) but not screening (0.9% in gout subjects vs 1% in osteoarthritis subjects, p = 1.0) colonoscopy. There was no significant difference in nonmalignant colorectal polyp occurrence between gout and osteoarthritis subjects. CONCLUSIONS Subjects with gout had decreased colonoscopy-documented occurrence of colorectal cancer compared with osteoarthritis subjects, suggesting a possible protective effect.
Collapse
Affiliation(s)
- Anastasia Slobodnick
- From the Section of Rheumatology, VA New York Harbor Health Care System, New York Campus
- Crystal Diseases Study Group, Division of Rheumatology, New York University School of Medicine, New York, NY
| | - Svetlana Krasnokutsky
- From the Section of Rheumatology, VA New York Harbor Health Care System, New York Campus
- Crystal Diseases Study Group, Division of Rheumatology, New York University School of Medicine, New York, NY
| | - Robert A Lehmann
- From the Section of Rheumatology, VA New York Harbor Health Care System, New York Campus
- Crystal Diseases Study Group, Division of Rheumatology, New York University School of Medicine, New York, NY
| | - Robert T Keenan
- Division of Rheumatology, Duke University School of Medicine, Durham, NC
| | - Jonathan Quach
- Section of Gastroenterology, VA New York Harbor Health Care System, New York Campus
- Division of Gastroenterology, New York University School of Medicine, New York, NY
| | - Fritz Francois
- Section of Gastroenterology, VA New York Harbor Health Care System, New York Campus
- Division of Gastroenterology, New York University School of Medicine, New York, NY
| | - Michael H Pillinger
- From the Section of Rheumatology, VA New York Harbor Health Care System, New York Campus
- Crystal Diseases Study Group, Division of Rheumatology, New York University School of Medicine, New York, NY
| |
Collapse
|
30
|
Wang CZ, Zhang CF, Luo Y, Yao H, Yu C, Chen L, Yuan J, Huang WH, Wan JY, Zeng J, Sawadogo WR, Yuan CS. Baicalein, an enteric microbial metabolite, suppresses gut inflammation and cancer progression in Apc Min/+ mice. Clin Transl Oncol 2019; 22:1013-1022. [PMID: 31650468 DOI: 10.1007/s12094-019-02225-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 10/03/2019] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Chronic inflammation is recognized as a risk factor for colorectal cancer (CRC) development. Baicalin (BI), a major constituent in an anti-inflammatory herb Scutellaria baicalensis, can be biotransformed into baicalein (BE) by the intestinal microbiota. We evaluated the anti-inflammation and anti-CRC effects of the metabolite BE. METHODS The in vitro biotransformation by human intestinal microbiota from BI into BE has been determined with HPLC. Using a gut-specific ApcMin/+ mouse model, the effects of oral BE on the life span, organ index, and tumor multiplicity were evaluated. The expressions of inflammatory cytokines were determined using ELISA. To verify the in vivo data, the anti-inflammatory and antiproliferative effects of BE were determined with an in vitro cell model. RESULTS HPLC analysis showed that BI was quickly transformed into BE by the intestinal microbiota. Oral BE (30 mg/kg/day) significantly increased the life span, from 125.2 to 218.4 days (P < 0.01%). BE treatment also decreased intestine index and increased spleen index. Compared with the model group, following BE treatment, tumor numbers were significantly reduced in the small intestine and colon (P < 0.01, P < 0.05, respectively). In the gut tissues, BE treatment significantly reduced inflammatory cytokine levels such as IL-1β, IL-2, IL-6, IL-10, G-CSF, and GM-CSF. In vitro data supported our in vivo results that the anti-CRC effects of BE were via the inhibition of gut inflammation and induction of cancer cell death. CONCLUSION Our results suggest that the parent compound BI can be quickly converted into its microbial metabolite BE, which has stronger bioactive effects than BI. Baicalein is an active chemopreventive metabolite for inflammatory associated CRC.
Collapse
Affiliation(s)
- C-Z Wang
- Tang Center for Herbal Medicine Research, Pritzker School of Medicine, University of Chicago, 5841 S. Maryland Ave., MC 4028, Chicago, IL, 60637, USA.
- Department of Anesthesia and Critical Care, Pritzker School of Medicine, University of Chicago, Chicago, IL, 60637, USA.
| | - C-F Zhang
- Tang Center for Herbal Medicine Research, Pritzker School of Medicine, University of Chicago, 5841 S. Maryland Ave., MC 4028, Chicago, IL, 60637, USA
- Department of Anesthesia and Critical Care, Pritzker School of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | - Y Luo
- Tang Center for Herbal Medicine Research, Pritzker School of Medicine, University of Chicago, 5841 S. Maryland Ave., MC 4028, Chicago, IL, 60637, USA
- Department of Anesthesia and Critical Care, Pritzker School of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | - H Yao
- Tang Center for Herbal Medicine Research, Pritzker School of Medicine, University of Chicago, 5841 S. Maryland Ave., MC 4028, Chicago, IL, 60637, USA
- Department of Anesthesia and Critical Care, Pritzker School of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | - C Yu
- Tang Center for Herbal Medicine Research, Pritzker School of Medicine, University of Chicago, 5841 S. Maryland Ave., MC 4028, Chicago, IL, 60637, USA
- Department of Anesthesia and Critical Care, Pritzker School of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | - L Chen
- Tang Center for Herbal Medicine Research, Pritzker School of Medicine, University of Chicago, 5841 S. Maryland Ave., MC 4028, Chicago, IL, 60637, USA
- Department of Anesthesia and Critical Care, Pritzker School of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | - J Yuan
- Tang Center for Herbal Medicine Research, Pritzker School of Medicine, University of Chicago, 5841 S. Maryland Ave., MC 4028, Chicago, IL, 60637, USA
- Department of Anesthesia and Critical Care, Pritzker School of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | - W-H Huang
- Tang Center for Herbal Medicine Research, Pritzker School of Medicine, University of Chicago, 5841 S. Maryland Ave., MC 4028, Chicago, IL, 60637, USA
- Department of Anesthesia and Critical Care, Pritzker School of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | - J-Y Wan
- Tang Center for Herbal Medicine Research, Pritzker School of Medicine, University of Chicago, 5841 S. Maryland Ave., MC 4028, Chicago, IL, 60637, USA
- Department of Anesthesia and Critical Care, Pritzker School of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | - J Zeng
- Tang Center for Herbal Medicine Research, Pritzker School of Medicine, University of Chicago, 5841 S. Maryland Ave., MC 4028, Chicago, IL, 60637, USA
- Department of Anesthesia and Critical Care, Pritzker School of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | - W R Sawadogo
- Tang Center for Herbal Medicine Research, Pritzker School of Medicine, University of Chicago, 5841 S. Maryland Ave., MC 4028, Chicago, IL, 60637, USA
- Department of Anesthesia and Critical Care, Pritzker School of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | - C-S Yuan
- Tang Center for Herbal Medicine Research, Pritzker School of Medicine, University of Chicago, 5841 S. Maryland Ave., MC 4028, Chicago, IL, 60637, USA
- Department of Anesthesia and Critical Care, Pritzker School of Medicine, University of Chicago, Chicago, IL, 60637, USA
- Committee on Clinical Pharmacology and Pharmacogenomics, Pritzker School of Medicine, University of Chicago, Chicago, IL, 60637, USA
| |
Collapse
|
31
|
Rubinow KB, Houston B, Wang S, Goodspeed L, Ogimoto K, Morton GJ, McCarty C, Braun RE, Page ST. Androgen receptor deficiency in monocytes/macrophages does not alter adiposity or glucose homeostasis in male mice. Asian J Androl 2019; 20:276-283. [PMID: 29205180 PMCID: PMC5952483 DOI: 10.4103/aja.aja_54_17] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Androgen deprivation in men leads to increased adiposity, but the mechanisms underlying androgen regulation of fat mass have not been fully defined. Androgen receptor (AR) is expressed in monocytes/macrophages, which are resident in key metabolic tissues and influence energy metabolism in surrounding cells. Male mice bearing a cell-specific knockout of the AR in monocytes/macrophages (M-ARKO) were generated to determine whether selective loss of androgen signaling in these cells would lead to altered body composition. Wild-type (WT) and M-ARKO mice (12–22 weeks of age, n = 12 per group) were maintained on a regular chow diet for 8 weeks and then switched to a high-fat diet for 8 additional weeks. At baseline and on both the regular chow and high-fat diets, no differences in lean mass or fat mass were observed between groups. Consistent with the absence of differential body weight or adiposity, no differences in food intake (3.0 ± 0.5 g per day for WT mice vs 2.8 ± 0.4 g per day for M-ARKO mice) or total energy expenditure (0.6 ± 0.1 Kcal h−1 for WT mice vs 0.5 ± 0.1 Kcal h−1 for M-ARKO mice) were evident between groups during high-fat feeding. Liver weight was greater in M-ARKO than that in WT mice (1.5 ± 0.1 g vs 1.3 ± 0.0 g, respectively, P = 0.02). Finally, M-ARKO mice did not exhibit impairments in glucose tolerance or insulin sensitivity relative to WT mice at any study time point. In aggregate, these findings suggest that AR signaling specifically in monocytes/macrophages does not contribute to the regulation of systemic energy balance, adiposity, or insulin sensitivity in male mice.
Collapse
Affiliation(s)
- Katya B Rubinow
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Barbara Houston
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Shari Wang
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Leela Goodspeed
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Kayoko Ogimoto
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Gregory J Morton
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | | | | | - Stephanie T Page
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| |
Collapse
|
32
|
Goncalves MD, Hopkins BD, Cantley LC. Dietary Fat and Sugar in Promoting Cancer Development and Progression. ANNUAL REVIEW OF CANCER BIOLOGY-SERIES 2019. [DOI: 10.1146/annurev-cancerbio-030518-055855] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The uncontrolled cellular growth that characterizes tumor formation requires a constant delivery of nutrients. Since the 1970s, researchers have wondered if the supply of nutrients from the diet could impact tumor development. Numerous studies have assessed the impact of dietary components, specifically sugar and fat, to increased cancer risk. For the most part, data from these trials have been inconclusive; however, this does not indicate that dietary factors do not contribute to cancer progression. Rather, the dietary contribution may be dependent on tumor, patient, and context, making it difficult to detect in the setting of large trials. In this review, we combine data from prospective cohort trials with mechanistic studies in mice to argue that fat and sugar can play a role in tumorigenesis and disease progression. We find that certain tumors may respond directly to dietary sugar (colorectal and endometrial cancers) and fat (prostate cancer) or indirectly to the obese state (breast cancer).
Collapse
Affiliation(s)
- Marcus D. Goncalves
- Meyer Cancer Center, Department of Medicine, Weill Cornell Medical College, New York, NY 10021, USA;, ,
- Division of Endocrinology, Department of Medicine, Weill Cornell Medical College, New York, NY 10021, USA
| | - Benjamin D. Hopkins
- Meyer Cancer Center, Department of Medicine, Weill Cornell Medical College, New York, NY 10021, USA;, ,
| | - Lewis C. Cantley
- Meyer Cancer Center, Department of Medicine, Weill Cornell Medical College, New York, NY 10021, USA;, ,
| |
Collapse
|
33
|
Bader JE, Enos RT, Velázquez KT, Carson MS, Sougiannis AT, McGuinness OP, Robinson CM, Murphy EA. Repeated clodronate-liposome treatment results in neutrophilia and is not effective in limiting obesity-linked metabolic impairments. Am J Physiol Endocrinol Metab 2019; 316:E358-E372. [PMID: 30576244 PMCID: PMC6415716 DOI: 10.1152/ajpendo.00438.2018] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Depletion of macrophages is thought to be a therapeutic option for obesity-induced inflammation and metabolic dysfunction. However, whether the therapeutic effect is a direct result of reduced macrophage-derived inflammation or secondary to decreases in fat mass is controversial, as macrophage depletion has been shown to disrupt energy homeostasis. This study was designed to determine if macrophage depletion via clodronate-liposome (CLD) treatment could serve as an effective intervention to reduce obesity-driven inflammatory and metabolic impairments independent of changes in energy intake. After 16 wk on a high-fat diet (HFD) or the AIN-76A control (low-fat) diet (LFD) ( n = 30/diet treatment), male C57BL/6J mice were assigned to a CLD- or PBS-liposome treatment ( n = 15/group) for 4 wk. Liposomes were administered biweekly via intraperitoneal injections (8 administrations in total). PBS-liposome-treated groups were pair-fed to their CLD-treated dietary counterparts. Metabolic function was assessed before and after liposome treatment. Adipose tissue, as well as the liver, was investigated for macrophage infiltration and the presence of inflammatory mediators. Additionally, a complete blood count was performed. CLD treatment reduced energy intake. When controlling for energy intake, CLD treatment was unable to regress metabolic dysfunction or nonalcoholic fatty liver disease and impaired adipose tissue insulin action. Moreover, repeated CLD treatment induced neutrophilia and anemia, increased adipose tissue mRNA expression of the proinflammatory cytokines IL-6 and IL-1β, and augmented circulating IL-6 and monocyte chemoattractant protein-1 concentrations ( P < 0.05). This study suggests that repeated intraperitoneal administration of CLD to deplete macrophages attenuates obesity by limiting energy intake. Moreover, after controlling for the benefits of weight loss, the accompanying detrimental side effects limit regular CLD treatment as an effective therapeutic strategy.
Collapse
Affiliation(s)
- Jackie E Bader
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina , Columbia, South Carolina
| | - Reilly T Enos
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina , Columbia, South Carolina
| | - Kandy T Velázquez
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina , Columbia, South Carolina
| | - Meredith S Carson
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina , Columbia, South Carolina
| | - Alex T Sougiannis
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina , Columbia, South Carolina
| | - Owen P McGuinness
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine , Nashville, Tennessee
| | - Cory M Robinson
- Department of Microbiology, Immunology, and Cell Biology, School of Medicine, West Virginia University , Morgantown, West Virginia
| | - E Angela Murphy
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina , Columbia, South Carolina
| |
Collapse
|
34
|
Romagnolo DF, Donovan MG, Doetschman TC, Selmin OI. n-6 Linoleic Acid Induces Epigenetics Alterations Associated with Colonic Inflammation and Cancer. Nutrients 2019; 11:E171. [PMID: 30650553 PMCID: PMC6356359 DOI: 10.3390/nu11010171] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 01/03/2019] [Accepted: 01/10/2019] [Indexed: 02/06/2023] Open
Abstract
The farnesoid-X-receptor (FXR) protects against inflammation and cancer of the colon through maintenance of intestinal bile acid (BA) homeostasis. Conversely, higher levels of BA and cyclooxygenase-2 (COX-2) are risk factors for inflammation and cancer of the colon. In the United States, n-6 linoleic acid (LA) is the most commonly used dietary vegetable fat. Metabolism of n-6 fatty acids has been linked to a higher risk of intestinal cancer. The objectives of this study were to investigate in colonic mucosa the effects of a high-fat diet rich in LA (n-6HFD) on CpG methylation of Fxr and prostaglandin-endoperoxide synthase-2 (Ptsg-2) genes, and the impact on the expression of tumor suppressor adenomatous polyposis Coli (Apc) and proliferative cyclin D1 (Ccnd1) genes. Weaned C57BL/6J male mice were fed for 6 weeks either an n-6HFD containing 44% energy (44%E) from 22% safflower oil (SO, 76% LA by weight) or a 13% energy (13%E) control diet (Control) from SO (5% by weight). Mice fed the n-6HFD had reduced (60%) Fxr promoter CpG methylation and increased (~50%) Fxr mRNA. The expression of FXR-target ileal bile acid-binding protein (Ibabp), small heterodimer protein (Shp), and anti-inflammatory peroxisome proliferator-activated-γ1 genes was increased. The n-6HFD reduced Ptgs-2 CpG methylation, increased the expression of Cox-2, and increased Apc CpG methylation in colonic mucosa. Accordingly, reduced expression of Apc was coupled to accumulation of c-JUN and Ccnd1, respectively cofactor and gene targets for the β-catenin/Wnt signaling pathway. Finally, the n-6HFD reduced the expression of histone deacetylase-1 while favoring the accumulation of acetylated histone 3. We conclude that an n-6HFD epigenetically modifies Fxr, leading to the activation of downstream factors that participate in BA homeostasis. However, epigenetic activation of Ptsg-2 coupled with silencing of Apc and accumulation of C-JUN and Ccnd1 may increase the risk of inflammation and cancer of the colon.
Collapse
Affiliation(s)
- Donato F Romagnolo
- The University of Arizona Cancer Center, Tucson, AZ 85724, USA.
- Department of Nutritional Sciences, University of Arizona, Tucson, AZ 85721, USA.
| | - Micah G Donovan
- Interdisciplinary Cancer Biology Graduate Program, University of Arizona, Tucson, AZ 85724, USA.
| | - Tom C Doetschman
- Department of Cellular & Molecular Medicine, University of Arizona, Tucson, AZ 85724, USA.
| | - Ornella I Selmin
- The University of Arizona Cancer Center, Tucson, AZ 85724, USA.
- Department of Nutritional Sciences, University of Arizona, Tucson, AZ 85721, USA.
| |
Collapse
|
35
|
Choi S, Snider AJ. Diet, lipids and colon cancer. CELLULAR NUTRIENT UTILIZATION AND CANCER 2019; 347:105-144. [DOI: 10.1016/bs.ircmb.2019.07.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
36
|
Using Mouse and Drosophila Models to Investigate the Mechanistic Links between Diet, Obesity, Type II Diabetes, and Cancer. Int J Mol Sci 2018; 19:ijms19124110. [PMID: 30567377 PMCID: PMC6320797 DOI: 10.3390/ijms19124110] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 12/12/2018] [Accepted: 12/14/2018] [Indexed: 02/06/2023] Open
Abstract
Many of the links between diet and cancer are controversial and over simplified. To date, human epidemiological studies consistently reveal that patients who suffer diet-related obesity and/or type II diabetes have an increased risk of cancer, suffer more aggressive cancers, and respond poorly to current therapies. However, the underlying molecular mechanisms that increase cancer risk and decrease the response to cancer therapies in these patients remain largely unknown. Here, we review studies in mouse cancer models in which either dietary or genetic manipulation has been used to model obesity and/or type II diabetes. These studies demonstrate an emerging role for the conserved insulin and insulin-like growth factor signaling pathways as links between diet and cancer progression. However, these models are time consuming to develop and expensive to maintain. As the world faces an epidemic of obesity and type II diabetes we argue that the development of novel animal models is urgently required. We make the case for Drosophila as providing an unparalleled opportunity to combine dietary manipulation with models of human metabolic disease and cancer. Thus, combining diet and cancer models in Drosophila can rapidly and significantly advance our understanding of the conserved molecular mechanisms that link diet and diet-related metabolic disorders to poor cancer patient prognosis.
Collapse
|
37
|
Shen Y, Wang C, Ren Y, Ye J. A comprehensive look at the role of hyperlipidemia in promoting colorectal cancer liver metastasis. J Cancer 2018; 9:2981-2986. [PMID: 30123367 PMCID: PMC6096362 DOI: 10.7150/jca.25640] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 06/09/2018] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most malignant cancers, and it tends to migrate to the liver and has a high mortality rate. Several mechanisms behind the metastasis of CRC have been identified, including hyperlipidemia. For example, hyperlipidemia can lead to enhanced stemness and neutrophil infiltration, which increases CRC metastasis. There are three primary aspects to the relationship between hyperlipidemia and CRC metastasis: hyperlipidemia (1) promotes the initial metastatic properties of CRC, (2) stimulates CRC cells to leave the vasculature, and (3) facilitates the development of CRC metastasis. In this study, we provide a comprehensive overview of the role that hyperlipidemia played in CRC metastasis to help reduce the mortality associated with CRC metastasis from the standpoint of metabolic. We also review cancer metastasis.
Collapse
Affiliation(s)
- Yimin Shen
- 1 Department of Endocrinology and Metabolism, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Caihua Wang
- 2 Department of Gastroenterology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Yuezhong Ren
- 1 Department of Endocrinology and Metabolism, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Jun Ye
- 2 Department of Gastroenterology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| |
Collapse
|
38
|
Guan F, Tabrizian T, Novaj A, Nakanishi M, Rosenberg DW, Huffman DM. Dietary Walnuts Protect Against Obesity-Driven Intestinal Stem Cell Decline and Tumorigenesis. Front Nutr 2018; 5:37. [PMID: 29904634 PMCID: PMC5990619 DOI: 10.3389/fnut.2018.00037] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 04/23/2018] [Indexed: 12/12/2022] Open
Abstract
Obesity can negatively impact intestinal homeostasis, and increase colon cancer risk and related mortality. Thus, given the alarmingly high rates of obesity in the US and globally, it is critical to identify practical strategies that can break the obesity-cancer link. Walnuts have been increasingly recognized to mitigate cancer risk, and contain many bioactive constituents with antioxidant and anti-inflammatory properties that could potentially counteract pathways thought to be initiators of obesity-related cancer. Therefore, the purpose of this study was to determine if walnuts could preserve intestinal homeostasis, and attenuate tumorigenesis and growth in the context of obesity and a high calorie diet. To this end, we studied effects of walnuts on these parameters under different dietary conditions in wildtype mice, two independent Apc models (Apc1638N/+ and ApcΔ14), and in MC38 colon cancer cells in vivo, respectively. Walnuts did not alter the metabolic phenotype or intestinal morphology in normal mice fed either a low-fat diet (LFD), LFD with 6% walnuts (LFD+W), high-fat diet (HFD), or HFD with 7.6% walnuts (HFD+W). However, walnuts did lead to a significant reduction in circulating CCL5 and preserved intestinal stem cell (ISC) function under HFD-fed conditions. Furthermore, walnuts reduced tumor multiplicity in Apc1638N/+ male HFD+W animals, as compared to HFD controls (3.7 ± 0.5 vs. 2.5 ± 0.3; P = 0.015), tended to reduce the number of adenocarcinomas (0.67 ± 0.16 vs. 0.29 ± 0.12; P = 0.07), and preferentially limited tumor growth in ApcΔ14 male mice (P = 0.019) fed a high-calorie western-style diet. In summary, these data demonstrate that walnuts confer significant protection against intestinal tumorigenesis and growth and preserve ISC function in the context of a high-calorie diet and obesity. Thus, these data add to the accumulating evidence connecting walnuts as a potentially effective dietary strategy to break the obesity-colon cancer link.
Collapse
Affiliation(s)
- Fangxia Guan
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, United States.,Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, United States.,School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Tahmineh Tabrizian
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, United States.,Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Ardijana Novaj
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, United States.,Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Masako Nakanishi
- School of Medicine, University of Connecticut Health, Farmington, CT, United States
| | - Daniel W Rosenberg
- School of Medicine, University of Connecticut Health, Farmington, CT, United States
| | - Derek M Huffman
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, United States.,Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, United States.,Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
39
|
Hintze KJ, Benninghoff AD, Cho CE, Ward RE. Modeling the Western Diet for Preclinical Investigations. Adv Nutr 2018; 9:263-271. [PMID: 29635305 PMCID: PMC5952921 DOI: 10.1093/advances/nmy002] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 01/08/2018] [Indexed: 02/07/2023] Open
Abstract
Rodent models have been invaluable for biomedical research. Preclinical investigations with rodents allow researchers to investigate diseases by using study designs that are not suitable for human subjects. The primary criticism of preclinical animal models is that results are not always translatable to humans. Some of this lack of translation is due to inherent differences between species. However, rodent models have been refined over time, and translatability to humans has improved. Transgenic animals have greatly aided our understanding of interactions between genes and disease and have narrowed the translation gap between humans and model animals. Despite the technological innovations of animal models through advances in genetics, relatively little attention has been given to animal diets. Namely, developing diets that replicate what humans eat will help make animal models more relevant to human populations. This review focuses on commonly used rodent diets that are used to emulate the Western dietary pattern in preclinical studies of obesity and type 2 diabetes, nonalcoholic liver disease, maternal nutrition, and colorectal cancer.
Collapse
Affiliation(s)
- Korry J Hintze
- Department of Nutrition, Dietetics, and Food Sciences, Utah State University, Logan, UT,USTAR Applied Nutrition Research, Utah State University, Logan, UT,Address correspondence to KJH (e-mail: )
| | - Abby D Benninghoff
- USTAR Applied Nutrition Research, Utah State University, Logan, UT,Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, UT
| | - Clara E Cho
- Department of Nutrition, Dietetics, and Food Sciences, Utah State University, Logan, UT,USTAR Applied Nutrition Research, Utah State University, Logan, UT
| | - Robert E Ward
- Department of Nutrition, Dietetics, and Food Sciences, Utah State University, Logan, UT,USTAR Applied Nutrition Research, Utah State University, Logan, UT
| |
Collapse
|
40
|
Long J, Zhang CJ, Zhu N, Du K, Yin YF, Tan X, Liao DF, Qin L. Lipid metabolism and carcinogenesis, cancer development. Am J Cancer Res 2018; 8:778-791. [PMID: 29888102 PMCID: PMC5992506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 04/08/2018] [Indexed: 06/08/2023] Open
Abstract
The disorder of lipid metabolism is pathologically linked to hyperlipidemia, lipid storage disease, obesity and other related diseases. Intriguingly, recent studies have revealed that lipid metabolism disorders play an important role in carcinogenesis and development as well, since they cause abnormal expression of various genes, proteins, and dysregulation of cytokines and signaling pathways. More importantly, lipid-lowering drugs and anti-lipid per-oxidation treatment have been showing their advantages in clinic, in comparison with other anti-cancer drugs with high toxicity. Thus, further elucidation of molecular mechanism between lipid metabolism and cancer is essential in developing novel diagnostic biomarkers and therapeutic targets of human cancers.
Collapse
Affiliation(s)
- Jia Long
- School of Pharmacy, Hunan University of Chinese MedicineChangsha, Hunan, China
| | - Chan-Juan Zhang
- School of Pharmacy, Hunan University of Chinese MedicineChangsha, Hunan, China
| | - Neng Zhu
- The First Affiliated Hospital, Hunan University of Chinese MedicineChangsha, Hunan, China
| | - Ke Du
- School of Pharmacy, Hunan University of Chinese MedicineChangsha, Hunan, China
| | - Yu-Fang Yin
- Department of Neuroscience and Pharmacology, School of Medicine, Southern Illinois UniversitySpringfield, Illinois, United States
| | - Xi Tan
- Department of Biochemistry & Molecular Biology, University of CalgaryCalgary, Alberta, Canada
| | - Duan-Fang Liao
- School of Pharmacy, Hunan University of Chinese MedicineChangsha, Hunan, China
- Division of Stem Cell Regulation and Application, Key Lab for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese MedicineChangsha, Hunan Province, China
| | - Li Qin
- School of Pharmacy, Hunan University of Chinese MedicineChangsha, Hunan, China
- Division of Stem Cell Regulation and Application, Key Lab for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese MedicineChangsha, Hunan Province, China
| |
Collapse
|
41
|
Li Y, Cui SX, Sun SY, Shi WN, Song ZY, Wang SQ, Yu XF, Gao ZH, Qu XJ. Chemoprevention of intestinal tumorigenesis by the natural dietary flavonoid myricetin in APCMin/+ mice. Oncotarget 2018; 7:60446-60460. [PMID: 27507058 PMCID: PMC5312395 DOI: 10.18632/oncotarget.11108] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 07/26/2016] [Indexed: 12/12/2022] Open
Abstract
Myricetin is a natural dietary flavonoid compound. We evaluated the efficacy of myricetin against intestinal tumorigenesis in adenomatous polyposis coli multiple intestinal neoplasia (APCMin/+) mice. Myricetin was given orally once a day for 12 consecutive weeks. APCMin/+ mice fed with myricetin developed fewer and smaller polyps without any adverse effects. Histopathological analysis showed a decreased number of dysplastic cells and degree of dysplasia in each polyp. Immunohistochemical and western blot analysis revealed that myricetin selectively inhibits cell proliferation and induces apoptosis in adenomatous polyps. The effects of myricetin were associated with a modulation the GSK-3β and Wnt/β-catenin pathways. ELISA analysis showed a reduced concentration of pro-inflammatory cytokines IL-6 and PGE2 in blood, which were elevated in APCMin/+ mice. The effect of myricetin treatment was more prominent in the adenomatous polyps originating in the colon. Further studies showed that myricetin downregulates the phosphorylated p38 MAPK/Akt/mTOR signaling pathways, which may be the mechanisms for the inhibition of adenomatous polyps by myricetin. Taken together, our data show that myricetin inhibits intestinal tumorigenesis through a collection of biological activities. Given these results, we suggest that myricetin could be used preventatively to reduce the risk of developing colon cancers.
Collapse
Affiliation(s)
- Ye Li
- Department of Pharmacology, School of Chemical Biology & Pharmaceutical Sciences, Capital Medical University, Beijing, China
| | - Shu-Xiang Cui
- Beijing Key Laboratory of Environmental Toxicology, Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, China
| | - Shi-Yue Sun
- Department of Pharmacology, Capital Medical University School of Basic Medical Sciences, Beijing, China
| | - Wen-Na Shi
- Department of Pharmacology, Capital Medical University School of Basic Medical Sciences, Beijing, China
| | - Zhi-Yu Song
- Department of Pharmacology, Capital Medical University School of Basic Medical Sciences, Beijing, China
| | - Shu-Qing Wang
- Department of Pharmacology, Capital Medical University School of Basic Medical Sciences, Beijing, China
| | - Xin-Feng Yu
- Department of Pharmacology, Capital Medical University School of Basic Medical Sciences, Beijing, China
| | - Zu-Hua Gao
- Department of Pathology, McGill University, Montreal, Quebec, Canada
| | - Xian-Jun Qu
- Department of Pharmacology, Capital Medical University School of Basic Medical Sciences, Beijing, China
| |
Collapse
|
42
|
Nalbantoglu ILK, Blanc V, Davidson NO. Characterization of Colorectal Cancer Development in Apc (min/+) Mice. Methods Mol Biol 2017; 1422:309-27. [PMID: 27246043 DOI: 10.1007/978-1-4939-3603-8_27] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The Apc (min/+) mouse provides an excellent experimental model for studying genetic, environmental, and therapeutic aspects of intestinal neoplasia in humans. In this chapter, we will describe techniques for studying colon cancer development in Apc (min/+) mice on C57BL/6J (B6) background, focusing on the roles of environmental modifiers, including Dextran Sulfate Sodium (DSS), high fat diet, and bile acid supplementation in the context of experimental colorectal cancer. This chapter also includes protocols describing extraction and purification of DSS-contaminated RNA, as well as sampling, harvesting, and tissue processing. The common pathologic lesions encountered in these animals are described in detail.
Collapse
Affiliation(s)
- ILKe Nalbantoglu
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, 660 S. Euclid Ave, Box 8118, St. Louis, MO, 63110, USA.
| | - Valerie Blanc
- Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Nicholas O Davidson
- Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| |
Collapse
|
43
|
Liu L, Nishihara R, Qian ZR, Tabung FK, Nevo D, Zhang X, Song M, Cao Y, Mima K, Masugi Y, Shi Y, da Silva A, Twombly T, Gu M, Li W, Hamada T, Kosumi K, Inamura K, Nowak JA, Drew DA, Lochhead P, Nosho K, Wu K, Wang M, Garrett WS, Chan AT, Fuchs CS, Giovannucci EL, Ogino S. Association Between Inflammatory Diet Pattern and Risk of Colorectal Carcinoma Subtypes Classified by Immune Responses to Tumor. Gastroenterology 2017; 153:1517-1530.e14. [PMID: 28865736 PMCID: PMC5705461 DOI: 10.1053/j.gastro.2017.08.045] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 08/02/2017] [Accepted: 08/23/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Dietary patterns affect systemic and local intestinal inflammation, which have been linked to colorectal carcinogenesis. Chronic inflammation can interfere with the adaptive immune response. We investigated whether the association of a diet that promotes intestinal inflammation with risk of colorectal carcinoma was stronger for tumors with lower lymphocytic reactions than tumors with higher lymphocytic reactions. METHODS We collected data from the molecular pathological epidemiology databases of 2 prospective cohort studies: the Nurses' Health Study (since 1976) and the Health Professionals Follow-Up Study (since 1986). We used duplication-method time-varying Cox proportional cause-specific hazards regression to assess the association of empirical dietary inflammatory pattern (EDIP) score (derived from food frequency questionnaire data) with colorectal carcinoma subtype. Foods that contribute to high EDIP scores include red and processed meats, refined grains, carbonated beverages, and some vegetables; foods that contribute to low EDIP scores include beer, wine, coffee, tea, yellow and leafy vegetables, and fruit juice. Colorectal tissue samples were analyzed histologically for patterns of lymphocytic reactions (Crohn's-like lymphoid reaction, peritumoral lymphocytic reaction, intratumoral periglandular reaction, and tumor-infiltrating lymphocytes). RESULTS During follow-up of 124,433 participants, we documented 1311 incident colon and rectal cancer cases with available tissue data. The association between the EDIP and colorectal cancer risk was significant (Ptrend = .02), and varied with degree of peritumoral lymphocytic reaction (Pheterogeneity < .001). Higher EDIP scores were associated with increased risk of colorectal cancer with an absent or low peritumoral lymphocytic reaction (highest vs lowest EDIP score quintile hazard ratio, 2.60; 95% confidence interval, 1.60-4.23; Ptrend < .001), but not risk of tumors with intermediate or high peritumoral lymphocytic reaction (Ptrend > .80). CONCLUSIONS In 2 prospective cohort studies, we associated inflammatory diets with a higher risk of colorectal cancer subtype that contains little or no peritumoral lymphocytic reaction. These findings suggest that diet-related inflammation might contribute to development of colorectal cancer, by suppressing the adaptive anti-tumor immune response.
Collapse
Affiliation(s)
- Li Liu
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Department of Epidemiology and Biostatistics, and the Ministry of Education Key Lab of Environment and Health, School of Public Health, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Reiko Nishihara
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Zhi Rong Qian
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Fred K Tabung
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Daniel Nevo
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Xuehong Zhang
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Mingyang Song
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts; Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Yin Cao
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts; Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Kosuke Mima
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Yohei Masugi
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Yan Shi
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts; Medical Oncology Department 2, Chinese People's Liberation Army General Hospital, Beijing, People's Republic of China
| | - Annacarolina da Silva
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Tyler Twombly
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Mancang Gu
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts; College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Wanwan Li
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Tsuyoshi Hamada
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Keisuke Kosumi
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Kentaro Inamura
- Division of Pathology, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Jonathan A Nowak
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - David A Drew
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts; Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Paul Lochhead
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts; Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Katsuhiko Nosho
- Department of Gastroenterology and Hepatology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Kana Wu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Molin Wang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Wendy S Garrett
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Boston, Massachusetts
| | - Andrew T Chan
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts; Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts; Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Charles S Fuchs
- Yale Cancer Center, New Haven, Connecticut; Department of Medicine, Yale School of Medicine, New Haven, Connecticut; Smilow Cancer Hospital, New Haven, Connecticut
| | - Edward L Giovannucci
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Shuji Ogino
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts; Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
44
|
Joseph PV, Abey SK, Henderson WA. Emerging Role of Nutri-Epigenetics in Inflammation and Cancer. Oncol Nurs Forum 2017; 43:784-788. [PMID: 27768134 DOI: 10.1188/16.onf.784-788] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Nutrition is a factor involved in inflammation and a modulator of risk toward some cancers, and the complexity of linkages between dietary components and epigenetics mechanisms (e.g., DNA methylation, histone modification, chromatin remodeling) may affect the inflammation phenotype and the development of cancer. An increasing number of studies support the role of diet in cancer development, prevention, and treatment. Although current knowledge regarding nutri-epigenetics is expanding, more work is needed, and nurse scientists have the potential to significantly contribute to the expansion of this knowledge.
Collapse
|
45
|
Charepalli V, Reddivari L, Radhakrishnan S, Eriksson E, Xiao X, Kim SW, Shen F, Vijay-Kumar M, Li Q, Bhat VB, Knight R, Vanamala JKP. Pigs, Unlike Mice, Have Two Distinct Colonic Stem Cell Populations Similar to Humans That Respond to High-Calorie Diet prior to Insulin Resistance. Cancer Prev Res (Phila) 2017; 10:442-450. [PMID: 28576788 DOI: 10.1158/1940-6207.capr-17-0010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 04/10/2017] [Accepted: 05/30/2017] [Indexed: 12/12/2022]
Abstract
Basal colonic crypt stem cells are long lived and play a role in colon homeostasis. Previous evidence has shown that high-calorie diet (HCD) enhances colonic stem cell numbers and expansion of the proliferative zone, an important biomarker for colon cancer. However, it is not clear how HCD drives dysregulation of colon stem cell/colonocyte proliferative kinetics. We used a human-relevant pig model and developed an immunofluorescence technique to detect and quantify colonic stem cells. Pigs (n = 8/group) were provided either standard diet (SD; 5% fat) or HCD (23% fat) for 13 weeks. HCD- and SD-consuming pigs had similar total calorie intake, serum iron, insulin, and glucose levels. However, HCD elevated both colonic proliferative zone (KI-67) and stem cell zone (ASCL-2 and BMI-1). Proliferative zone correlated with elevated innate colonic inflammatory markers TLR-4, NF-κB, IL6, and lipocalin-2 (r ≥ 0.62, P = 0.02). Elevated gut bacterial phyla proteobacteria and firmicutes in HCD-consuming pigs correlated with proliferative and stem cell zone. Colonic proteome data revealed the upregulation of proteins involved in cell migration and proliferation and correlated with proliferative and stem cell zone expansion. Our study suggests that pig colon, unlike mice, has two distinct stem cells (ASCL-2 and BMI-1) similar to humans, and HCD increases expansion of colonic proliferative and stem cell zone. Thus, pig model can aid in the development of preventive strategies against gut bacterial dysbiosis and inflammation-promoted diseases, such as colon cancer. Cancer Prev Res; 10(8); 442-50. ©2017 AACR.
Collapse
Affiliation(s)
- Venkata Charepalli
- Department of Food Science, The Pennsylvania State University, University Park, Pennsylvania
| | - Lavanya Reddivari
- Department of Plant Science, The Pennsylvania State University, University Park, Pennsylvania
| | - Sridhar Radhakrishnan
- Department of Food Science, The Pennsylvania State University, University Park, Pennsylvania
| | - Elisabeth Eriksson
- Biotechnology, Applied Nutrition and Food Chemistry, Lund University, Lund, Sweden
| | - Xia Xiao
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania
| | - Sung Woo Kim
- Department of Animal Science, North Carolina State University, Raleigh, North Carolina
| | - Frank Shen
- Department of Statistics, The Pennsylvania State University, University Park, Pennsylvania
| | - Matam Vijay-Kumar
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania.,Department of Medicine, The Pennsylvania State University Medical Center, Hershey, Pennsylvania
| | - Qunhua Li
- Department of Statistics, The Pennsylvania State University, University Park, Pennsylvania
| | | | - Rob Knight
- Department of Pediatrics, University of California, San Diego, California.,Department of Computer Science and Engineering, University of California, La Jolla, California
| | - Jairam K P Vanamala
- Department of Food Science, The Pennsylvania State University, University Park, Pennsylvania. .,The Penn State Hershey Cancer Institute, Hershey, Pennsylvania.,Center for Molecular Immunology and Infectious Diseases, The Pennsylvania State University, University Park, Pennsylvania
| |
Collapse
|
46
|
Tabrizian T, Wang D, Guan F, Hu Z, Beck AP, Delahaye F, Huffman DM. Apc inactivation, but not obesity, synergizes with Pten deficiency to drive intestinal stem cell-derived tumorigenesis. Endocr Relat Cancer 2017; 24:253-265. [PMID: 28351943 PMCID: PMC5505256 DOI: 10.1530/erc-16-0536] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 03/28/2017] [Indexed: 12/13/2022]
Abstract
Obesity is a major risk factor for colorectal cancer and can accelerate Lgr5+ intestinal stem cell (ISC)-derived tumorigenesis after the inactivation of Apc However, whether non-canonical pathways involving PI3K-Akt signaling in ISCs can lead to tumor formation, and if this can be further exacerbated by obesity is unknown. Despite the synergy between Pten and Apc inactivation in epithelial cells on intestinal tumor formation, their combined role in Lgr5+-ISCs, which are the most rapidly dividing ISC population in the intestine, is unknown. Lgr5+-GFP mice were provided low-fat diet (LFD) or high-fat diet (HFD) for 8 months, and the transcriptome was evaluated in Lgr5+-ISCs. For tumor studies, Lgr5+-GFP and Lgr5+-GFP-Ptenflox/flox mice were tamoxifen treated to inactivate Pten in ISCs and provided LFD or HFD until 14-15 months of age. Finally, various combinations of Lgr5+-ISC-specific, Apc- and Pten-deleted mice were generated and evaluated for histopathology and survival. HFD did not overtly alter Akt signaling in ISCs, but did increase other metabolic pathways. Pten deficiency, but not HFD, increased BrdU-positive cells in the small intestine (P < 0.05). However, combining Pten and Apc deficiency synergistically increased proliferative markers, tumor pathology and mortality, in a dose-dependent fashion (P < 0.05). In summary, we show that HFD alone fails to drive Akt signaling in ISCs and that Pten deficiency is dispensable as a tumor suppressor in Lgr5+-ISCs. However, combining Pten and Apc deficiency in ISCs synergistically increases proliferation, tumor formation and mortality. Thus, aberrant Wnt/β-catenin, rather than PI3K-Akt signaling, is requisite for obesity to drive Lgr5+ ISC-derived tumorigenesis.
Collapse
Affiliation(s)
- Tahmineh Tabrizian
- Department of Molecular PharmacologyAlbert Einstein College of Medicine, Bronx, New York, USA
- Institute for Aging ResearchAlbert Einstein College of Medicine, Bronx, New York, USA
| | - Donghai Wang
- Department of Molecular PharmacologyAlbert Einstein College of Medicine, Bronx, New York, USA
- Institute for Aging ResearchAlbert Einstein College of Medicine, Bronx, New York, USA
- Division of EndocrinologyDepartment of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Fangxia Guan
- Department of Molecular PharmacologyAlbert Einstein College of Medicine, Bronx, New York, USA
- Institute for Aging ResearchAlbert Einstein College of Medicine, Bronx, New York, USA
- Division of EndocrinologyDepartment of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Zunju Hu
- Department of Molecular PharmacologyAlbert Einstein College of Medicine, Bronx, New York, USA
- Institute for Aging ResearchAlbert Einstein College of Medicine, Bronx, New York, USA
- Division of EndocrinologyDepartment of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Amanda P Beck
- Department of Obstetrics & Gynecology and Women's HealthAlbert Einstein College of Medicine, Bronx, New York, USA
| | - Fabien Delahaye
- Department of GeneticsAlbert Einstein College of Medicine, Bronx, New York, USA
- Department of PathologyAlbert Einstein College of Medicine, Bronx, New York, USA
| | - Derek M Huffman
- Department of Molecular PharmacologyAlbert Einstein College of Medicine, Bronx, New York, USA
- Institute for Aging ResearchAlbert Einstein College of Medicine, Bronx, New York, USA
- Division of EndocrinologyDepartment of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
47
|
Kang M, Martin A. Microbiome and colorectal cancer: Unraveling host-microbiota interactions in colitis-associated colorectal cancer development. Semin Immunol 2017; 32:3-13. [PMID: 28465070 DOI: 10.1016/j.smim.2017.04.003] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 04/19/2017] [Indexed: 02/07/2023]
Abstract
Dysbiosis of gut microbiota occurs in many human chronic immune-mediated diseases, such as inflammatory bowel disease (IBD) and colitis-associated colorectal cancer (CAC). Reciprocally, uncontrolled immune responses, that may or may not be induced by dysbiosis, are central to the development of IBD and CAC. There has been a surge of interest in investigating the relationship between microbiota, inflammation and CAC. In this review, we discuss recent findings related to gut microbiota and chronic immune-mediated diseases, such as IBD and CAC. Moreover, the molecular mechanisms underlying the roles of chronic inflammation in CAC are examined. Finally, we discuss the development of novel microbiota-based therapeutics for IBD and colorectal cancer.
Collapse
Affiliation(s)
- Mingsong Kang
- University of Toronto, Department of Immunology, Toronto, Ontario, Canada
| | - Alberto Martin
- University of Toronto, Department of Immunology, Toronto, Ontario, Canada.
| |
Collapse
|
48
|
Cao H, Xu M, Dong W, Deng B, Wang S, Zhang Y, Wang S, Luo S, Wang W, Qi Y, Gao J, Cao X, Yan F, Wang B. Secondary bile acid-induced dysbiosis promotes intestinal carcinogenesis. Int J Cancer 2017; 140:2545-2556. [PMID: 28187526 DOI: 10.1002/ijc.30643] [Citation(s) in RCA: 146] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 01/08/2017] [Accepted: 01/30/2017] [Indexed: 12/13/2022]
Abstract
The gut microbiota plays an important role in maintaining intestinal homeostasis. Dysbiosis is associated with intestinal tumorigenesis. Deoxycholic acid (DCA), a secondary bile acid increased by a western diet, correlates with intestinal carcinogenesis. However, evidence relating bile acids, intestinal microbiota and tumorigenesis are limited. In our study, we investigated the effect of DCA on induction of intestinal dysbiosis and its roles in intestinal carcinogenesis. Alteration of the composition of the intestinal microbiota was induced in DCA-treated APCmin/+ mice, which was accompanied by impaired intestinal barrier, gut low grade inflammation and tumor progression. The transfer of fecal microbiota from DCA-treated mice to another group of Apcmin/+ mice increased tumor multiplicity, induced inflammation and recruited M2 phenotype tumor-associated macrophages. Importantly, the fecal microbiota transplantation activated the tumor-associated Wnt/β-catenin signaling pathway. Moreover, microbiota depletion by a cocktail of antibiotics was sufficient to block DCA-induced intestinal carcinogenesis, further suggesting the role of dysbiosis in tumor development. Our study demonstrated that alteration of the microbial community induced by DCA promoted intestinal carcinogenesis.
Collapse
Affiliation(s)
- Hailong Cao
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, People's Republic of China.,Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
| | - Mengque Xu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, People's Republic of China.,Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Wenxiao Dong
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, People's Republic of China
| | - Baoru Deng
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, People's Republic of China
| | - Sinan Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, People's Republic of China
| | - Yujie Zhang
- Department of Pathology, General Hospital, Tianjin Medical University, Tianjin, People's Republic of China
| | - Shan Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, People's Republic of China
| | - Shenhui Luo
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, People's Republic of China
| | - Weiqiang Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, People's Republic of China
| | - Yanrong Qi
- Department of Gastroenterology and Hepatology, Tianjin Haibin People's Hospital, Tianjin, People's Republic of China
| | - Jianxin Gao
- Department of Gastroenterology and Hepatology, Tianjin Haibin People's Hospital, Tianjin, People's Republic of China
| | - Xiaocang Cao
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, People's Republic of China
| | - Fang Yan
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, People's Republic of China.,Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, People's Republic of China
| |
Collapse
|
49
|
Cao HL, Chen X, Du SC, Song WJ, Wang WQ, Xu MQ, Wang SN, Piao MY, Cao XC, Wang BM. Detection Rate, Distribution, Clinical and Pathological Features of Colorectal Serrated Polyps. Chin Med J (Engl) 2017; 129:2427-2433. [PMID: 27748334 PMCID: PMC5072254 DOI: 10.4103/0366-6999.191759] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background: Colorectal serrated polyp is considered as histologically heterogeneous lesions with malignant potential in western countries. However, few Asian studies have investigated the comprehensive clinical features of serrated polyps in symptomatic populations. The aim of the study was to evaluate the features of colorectal serrated polyps in a Chinese symptomatic population. Methods: Data from all consecutive symptomatic patients were documented from a large colonoscopy database and were analyzed. Chi-square test or Fisher's exact test and logistic regression analysis were used for the data processing. Results: A total of 9191 (31.7%) patients were detected with at least one colorectal polyp. The prevalence of serrated polyps was 0.53% (153/28,981). The proportions of hyperplastic polyp (HP), sessile serrated adenoma/polyp (SSA/P), and traditional serrated adenoma (TSA) of all serrated polyps were 41.2%, 7.2%, and 51.6%, respectively, which showed a lower proportion of HP and SSA/P and a higher proportion of TSA. Serrated polyps appeared more in males and elder patients while there was no significant difference in the subtype distribution in gender and age. The proportions of large and proximal serrated polyps were 13.7% (21/153) and 46.4% (71/153), respectively. In total, 98.9% (89/90) serrated adenomas were found with dysplasia. Moreover, 14 patients with serrated polyps were found with synchronous advanced colorectal neoplasia, and large serrated polyps (LSPs) (odds ratio: 3.446, 95% confidence interval: 1.010–11.750, P < 0.05), especially large HPs, might have an association with synchronous advanced neoplasia (AN). Conclusions: The overall detection rate of colorectal serrated polyps in Chinese symptomatic patient population was low, and distribution pattern of three subtypes is different from previous reports. Moreover, LSPs, especially large HPs, might be associated with an increased risk of synchronous AN.
Collapse
Affiliation(s)
- Hai-Long Cao
- Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Xue Chen
- Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Shao-Chun Du
- Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Wen-Jing Song
- Department of Pathology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Wei-Qiang Wang
- Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Meng-Que Xu
- Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Si-Nan Wang
- Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Mei-Yu Piao
- Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Xiao-Cang Cao
- Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Bang-Mao Wang
- Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
50
|
Sido A, Radhakrishnan S, Kim SW, Eriksson E, Shen F, Li Q, Bhat V, Reddivari L, Vanamala JKP. A food-based approach that targets interleukin-6, a key regulator of chronic intestinal inflammation and colon carcinogenesis. J Nutr Biochem 2017; 43:11-17. [PMID: 28193578 DOI: 10.1016/j.jnutbio.2017.01.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Revised: 12/15/2016] [Accepted: 01/19/2017] [Indexed: 12/31/2022]
Abstract
Studies have shown a causal link between high-calorie diet (HCD) and colon cancer. However, molecular mechanisms are not fully elucidated. To understand etiology of HCD-induced colon carcinogenesis, we screened 10 pathways linked to elevated colonic cell proliferation and chronic inflammation in an HCD-consuming human-relevant pig model. We observed elevated colonic mucosal interleukin-6 (IL-6) expression in HCD-consuming pigs compared to standard diet controls (SD, P=.04), and IL-6 strongly correlated with Ki-67 proliferative index and zone, early biomarkers of colon cancer risk (r=0.604 and 0.743 and P=.017 and .002, respectively). Liquid chromatography-tandem mass spectrometry-based proteomic analysis and Ingenuity Pathway Analysis showed that HCD consumption altered IL-6 signaling pathway proteins (PI3KR4, IL-1α, Mapk10, Akt3, PIK3CG, PIK3R5, Map2k2). Furthermore, these proteins also correlated with Ki-67 proliferative index/zone. Anti-IL-6 therapeutics are available for treating colon cancer; however, they are expensive and induce negative side effects. Thus, whole foods could be a better way to combat low-grade chronic colonic inflammation and colon cancer. Whole plant foods have been shown to decrease chronic diseases due to the potential of anti-inflammatory dietary compounds acting synergistically. We observed that supplementation of HCD with anthocyanin-containing purple-fleshed potatoes (10% w/w), even after baking, suppressed HCD-induced IL-6 expression (P=.03) and the IL-6-related proteins IL-1α and Map2k1 (P≤.1). Our results highlight the importance of IL-6 signaling in diet-linked induction/prevention of colonic inflammation/cancer and demonstrate the potential of a food-based approach to target IL-6 signaling.
Collapse
Affiliation(s)
- Abigail Sido
- Department of Food Science, The Pennsylvania State University, University Park, PA, USA
| | - Sridhar Radhakrishnan
- Department of Food Science, The Pennsylvania State University, University Park, PA, USA
| | - Sung Woo Kim
- Department of Animal Science, North Carolina State University, Raleigh, North Carolina, USA
| | | | - Frank Shen
- Department of Statistics, The Pennsylvania State University, University Park, PA, USA
| | - Qunhua Li
- Agilent Technologies, Wilmington, DE, USA
| | | | - Lavanya Reddivari
- Department of Plant Science, The Pennsylvania State University, University Park, PA, USA
| | - Jairam K P Vanamala
- Department of Food Science, The Pennsylvania State University, University Park, PA, USA; The Pennsylvania State University Milton S. Hershey College of Medicine, Hershey, PA, USA; Center for Molecular Immunology and Infectious Diseases, The Pennsylvania State University, University Park, PA, USA.
| |
Collapse
|