1
|
Chang YM, Kang YR, Lee YG, Sung MK. Sex differences in colonic gene expression and fecal microbiota composition in a mouse model of obesity-associated colorectal cancer. Sci Rep 2024; 14:3576. [PMID: 38347027 PMCID: PMC10861586 DOI: 10.1038/s41598-024-53861-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 02/06/2024] [Indexed: 02/15/2024] Open
Abstract
This study investigated the sex-specific correlation between obesity and colorectal cancer emphasizing a more pronounced association in males. Estrogen, chromosomal genes, and gut bacteria were assessed in C57BL6/J male, female and ovariectomized (OVX) female mice, subjected to either a low-fat diet (LFD) or high-fat diet (HFD) for 14 weeks. Induction of colon tumor involved azoxymethane (10 mg/kg) administration, followed by three cycles of dextran sulfate sodium. Male mice on HFD exhibited higher final body weight and increased colon tumors compared to females. Colonic mucin 2 expression was significantly higher in females. HFD-modulated differentially expressed genes numbered 290 for males, 64 for females, and 137 for OVX females. Only one up-regulated gene (Gfra3) overlapped between females and OVX females, while two down-regulated genes (Thrsp and Gbp11) overlapped between males and OVX females. Genes up-regulated by HFD in males were linked to cytokine-cytokine interaction, HIF-1 signaling pathway, central carbon metabolism in cancer. Sex-specific changes in gut microbial composition in response to HFD were observed. These findings suggest a male-specific vulnerability to HFD-induced colon tumor formation, implicating key genes and colonic bacteria in colon tumorigenesis.
Collapse
Affiliation(s)
- Yoo-Mee Chang
- Department of Food and Nutrition, College of Human Ecology, Sookmyung Women's University, 100, Cheongpa-ro 47-gil, Yongsan-gu, Seoul, 04310, Republic of Korea
| | - Yoo-Ree Kang
- Department of Food and Nutrition, College of Human Ecology, Sookmyung Women's University, 100, Cheongpa-ro 47-gil, Yongsan-gu, Seoul, 04310, Republic of Korea
| | - Yu-Gyeong Lee
- Department of Food and Nutrition, College of Human Ecology, Sookmyung Women's University, 100, Cheongpa-ro 47-gil, Yongsan-gu, Seoul, 04310, Republic of Korea
| | - Mi-Kyung Sung
- Department of Food and Nutrition, College of Human Ecology, Sookmyung Women's University, 100, Cheongpa-ro 47-gil, Yongsan-gu, Seoul, 04310, Republic of Korea.
| |
Collapse
|
2
|
Lagou MK, Karagiannis GS. Obesity-induced thymic involution and cancer risk. Semin Cancer Biol 2023; 93:3-19. [PMID: 37088128 DOI: 10.1016/j.semcancer.2023.04.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/19/2023] [Accepted: 04/20/2023] [Indexed: 04/25/2023]
Abstract
Declining thymic functions associated either with old age (i.e., age-related thymic involution), or with acute involution as a result of stress, infectious disease, or cytoreductive therapies (e.g., chemotherapy/radiotherapy), have been associated with cancer development. A key mechanism underlying such increased cancer risk is the thymus-dependent debilitation of adaptive immunity, which is responsible for orchestrating immunoediting mechanisms and tumor immune surveillance. In the past few years, a blooming set of evidence has intriguingly linked obesity with cancer development and progression. The majority of such studies has focused on obesity-driven chronic inflammation, steroid/sex hormone and adipokine production, and hyperinsulinemia, as principal factors affecting the tumor microenvironment and driving the development of primary malignancy. However, experimental observations about the negative impact of obesity on T cell development and maturation have existed for more than half a century. Here, we critically discuss the molecular and cellular mechanisms of obesity-driven thymic involution as a previously underrepresented intermediary pathology leading to cancer development and progression. This knowledge could be especially relevant in the context of childhood obesity, because impaired thymic function in young individuals leads to immune system abnormalities, and predisposes to various pediatric cancers. A thorough understanding behind the molecular and cellular circuitries governing obesity-induced thymic involution could therefore help towards the rationalized development of targeted thymic regeneration strategies for obese individuals at high risk of cancer development.
Collapse
Affiliation(s)
- Maria K Lagou
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA; Tumor Microenvironment of Metastasis Program, Albert Einstein Cancer Center, Bronx, NY, USA
| | - George S Karagiannis
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA; Tumor Microenvironment of Metastasis Program, Albert Einstein Cancer Center, Bronx, NY, USA; Cancer Dormancy and Tumor Microenvironment Institute, Albert Einstein College of Medicine, Bronx, NY, USA; Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA; Integrated Imaging Program for Cancer Research, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
3
|
Iwama N, Matsuda M, Tsuruta M, Okabayashi K, Shigeta K, Kanai T, Kitagawa Y. Relationship between obesity-related colorectal tumors and the intestinal microbiome: an animal-based trial. J Cancer Res Clin Oncol 2023; 149:5265-5277. [PMID: 36399198 DOI: 10.1007/s00432-022-04477-1] [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: 08/17/2022] [Accepted: 11/09/2022] [Indexed: 11/19/2022]
Abstract
PURPOSE Obesity is a risk factor for colorectal cancer (CRC), and the intestinal microbiome is considered to contribute to CRC and obesity. Nonetheless, the role of the intestinal microbiome in obesity-related CRC is unclear. This study aimed to clarify the relationship between obesity-related CRC and the intestinal microbiome using a mouse model. METHODS We compared an obese and insulin-resistant type 2 diabetes mouse model [KKAy] to wild-type mice (WT) [C57BL/6 J]. Azoxymethane was intraperitoneally injected to develop a mouse model CRC. At 26 weeks, we compared the number of tumors and the intestinal microbiome. We also compared them across two models, namely, antibiotic cocktail and co-housing. RESULTS In all models, KKAy mice had a significantly greater number of tumors than WT mice. Analysis showed that the distribution of the intestinal microbiome changed in both models; however, no difference in tumor development was observed. Tumor expression was suppressed only in the antibiotic cocktail model of WT, whereas KKAy mice bore tumors (C57Bl/6 J: KKAy, 0/9:8/8; p < 0.001). KKAy mice remained predominantly tumor-bearing in all treatments. CONCLUSION Based on the results, the intestinal microbiome may not be associated with tumorigenesis in obesity-related CRC. It may be necessary to think of other facts linked to obesity-related CRC.
Collapse
Affiliation(s)
- Nozomi Iwama
- Department of Surgery, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Mutsuhito Matsuda
- Department of Surgery, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Masashi Tsuruta
- Department of Surgery, Keio University School of Medicine, Tokyo, 160-8582, Japan.
- Department of Hepato-Biliary-Pancreatic & Gastrointestinal Surgery, International University of Health and Welfare Narita Hospital, Chiba, 286-8520, Japan.
| | - Koji Okabayashi
- Department of Surgery, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Kohei Shigeta
- Department of Surgery, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Takanori Kanai
- Department of Gastroenterology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Yuko Kitagawa
- Department of Surgery, Keio University School of Medicine, Tokyo, 160-8582, Japan
| |
Collapse
|
4
|
Abstract
The epidemic of obesity, type 2 diabetes and nonalcoholic liver disease (NAFLD) favors drug consumption, which augments the risk of adverse events including liver injury. For more than 30 years, a series of experimental and clinical investigations reported or suggested that the common pain reliever acetaminophen (APAP) could be more hepatotoxic in obesity and related metabolic diseases, at least after an overdose. Nonetheless, several investigations did not reproduce these data. This discrepancy might come from the extent of obesity and steatosis, accumulation of specific lipid species, mitochondrial dysfunction and diabetes-related parameters such as ketonemia and hyperglycemia. Among these factors, some of them seem pivotal for the induction of cytochrome P450 2E1 (CYP2E1), which favors the conversion of APAP to the toxic metabolite N-acetyl-p-benzoquinone imine (NAPQI). In contrast, other factors might explain why obesity and NAFLD are not always associated with more frequent or more severe APAP-induced acute hepatotoxicity, such as increased volume of distribution in the body, higher hepatic glucuronidation and reduced CYP3A4 activity. Accordingly, the occurrence and outcome of APAP-induced liver injury in an obese individual with NAFLD would depend on a delicate balance between metabolic factors that augment the generation of NAPQI and others that can mitigate hepatotoxicity.
Collapse
|
5
|
Núñez-Ruiz A, Sánchez-Brena F, López-Pacheco C, Acevedo-Domínguez NA, Soldevila G. Obesity modulates the immune macroenvironment associated with breast cancer development. PLoS One 2022; 17:e0266827. [PMID: 35472214 PMCID: PMC9041840 DOI: 10.1371/journal.pone.0266827] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 03/28/2022] [Indexed: 11/19/2022] Open
Abstract
Growing evidence demonstrates a strong correlation between obesity and an increased risk of breast cancer, although the mechanisms involved have not been completely elucidated. Some reports have described a crosstalk between adipocytes, cancer cells, and immune cells within the tumor microenvironment, however, it is currently unknown whether obesity can promote tumor growth by inducing systemic alterations of the immune cell homeostasis in peripheral lymphoid organs and adipose tissue. Here, we used the E0771 breast cancer cell line in a mouse model of diet-induced obesity to analyze the immune subpopulations present in the tumors, visceral adipose tissue (VAT), and spleen of lean and obese mice. Our results showed a significant reduction in the frequency of infiltrating CD8+ T cells and a decreased M1/M2 macrophage ratio, indicative of the compromised anti-tumoral immune response reported in obesity. Despite not finding differences in the percentage or numbers of intratumoral Tregs, phenotypic analysis showed that they were enriched in CD39+, PD-1+ and CCR8+ cells, compared to the draining lymph nodes, confirming the highly immunosuppressive profile of infiltrating Tregs reported in established tumors. Analysis of peripheral T lymphocytes showed that tumor development in obese mice was associated to a significant increase in the percentage of peripheral Tregs, which supports the systemic immunosuppressive effect caused by the tumor. Interestingly, evaluation of immune subpopulations in the VAT showed that the characteristic increase in the M1/M2 macrophage ratio reported in obesity, was completely reversed in tumor-bearing mice, resembling the M2-polarized profile found in the microenvironment of the growing tumor. Importantly, VAT Tregs, which are commonly decreased in obese mice, were significantly increased in the presence of breast tumors and displayed significantly higher levels of Foxp3, indicating a regulatory feedback mechanism triggered by tumor growth. Altogether, our results identify a complex reciprocal relationship between adipocytes, immune cells, and the tumor, which may modulate the immune macroenvironment that promotes breast cancer development in obesity.
Collapse
Affiliation(s)
- Aleida Núñez-Ruiz
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, UNAM, México City, México
| | - Flor Sánchez-Brena
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, UNAM, México City, México
| | - Cynthia López-Pacheco
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, UNAM, México City, México
- Laboratorio Nacional de Citometría de Flujo, Instituto de Investigaciones Biomédicas, UNAM, México City, México
| | | | - Gloria Soldevila
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, UNAM, México City, México
- * E-mail:
| |
Collapse
|
6
|
Takahashi M, Hamoya T, Narita T, Fujii G, Totsuka Y, Hagio M, Tashiro K, Komiya M, Mutoh M. Complex Modulating Effects of Dietary Calcium Intake on Obese Mice. In Vivo 2021; 35:2107-2114. [PMID: 34182486 DOI: 10.21873/invivo.12480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/30/2021] [Accepted: 05/04/2021] [Indexed: 11/10/2022]
Abstract
BACKGROUND/AIM Οverweight and obesity are risk factors for chronic diseases. Dietary calcium has been reported to exert anti-obesity effects. However, the complex modulating effects of calcium intake on obese mice have not been clarified. MATERIALS AND METHODS The effects of calcium intake on body weight/visceral fat mass were examined in the obese mouse model, KK-Ay Results: Body weight gain decreased in mice fed a diet containing 0.4 to 3.2% calcium at the age of 11 and 13 weeks, but not at 12 weeks after normalization for food intake. Calcium intake also decreased serum insulin levels and increased the amount of feces excreted. Fecal deoxycholate levels were lower in the high-calcium group than in the normal diet control group. Furthermore, the ratio of the deoxycholate-producing microbiome in feces decreased. CONCLUSION Dietary calcium has anti-obesity effects in obese KK-Ay mice. Inhibition of insulin production and an increased amount of feces excreted with calcium intake may affect body weight.
Collapse
Affiliation(s)
- Maiko Takahashi
- Division of Prevention, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan
| | - Takahiro Hamoya
- Division of Prevention, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan.,Department of Molecular-Targeting Prevention, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takumi Narita
- Division of Prevention, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan.,Department of Molecular-Targeting Prevention, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Gen Fujii
- Central Radioisotope Division, National Cancer Center Research, Tokyo, Japan
| | - Yukari Totsuka
- Department of Animal Experimentation, National Cancer Center Research Institute, Tokyo, Japan
| | - Masahito Hagio
- Chemistry Division, Kanagawa Prefectural Institute of Public Health, Chigasaki, Japan
| | - Kosuke Tashiro
- Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushuu University, Fukuoka, Japan
| | - Masami Komiya
- Division of Prevention, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan
| | - Michihiro Mutoh
- Division of Prevention, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan; .,Department of Molecular-Targeting Prevention, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
7
|
Matsui S, Okabayashi K, Tsuruta M, Shigeta K, Seishima R, Ishida T, Kondo T, Suzuki Y, Hasegawa H, Shimoda M, Sugimoto S, Sato T, Kitagawa Y. Interleukin-13 and its signaling pathway is associated with obesity-related colorectal tumorigenesis. Cancer Sci 2019; 110:2156-2165. [PMID: 31099450 PMCID: PMC6609806 DOI: 10.1111/cas.14066] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 05/08/2019] [Accepted: 05/10/2019] [Indexed: 01/10/2023] Open
Abstract
The incidence of colorectal cancer (CRC) has been on the rise, which is linked to the increasing prevalence of obesity, based on global epidemiological evidence. Although chronic inflammation is implicated in tumor development, the mechanisms underlying obesity‐associated CRC remain unknown. Here, we sought to identify the inflammatory cytokines and their roles in obesity‐related colorectal tumorigenesis using cytokine array analyses in a mouse model. Colorectal tumorigenesis was induced through i.p. injection of azoxymethane once a week for 6 weeks in 6‐week‐old female WT C57Black/6J mice and the obesity diabetes model mouse KK/TaJcl, KK‐Ay/TaJcl. The formation of aberrant crypt foci and colorectal tumors were more frequent in obese mice compared with WT mice, and both serum interleukin (IL)‐13 and IL‐13 receptor (R) expression in the normal intestinal mucosal epithelium were significantly increased in the obese mice. Furthermore, addition of IL‐13 to a human CRC cell line and a human colon organoid culture altered the phenotype of intestinal epithelial cells. Knockdown experiments further revealed that IL‐13Rα1 dominantly induced mucosal proliferation. Collectively, These results suggest an association between anti‐inflammatory cytokines and colorectal carcinogenesis, and provide new research directions for cancer prevention strategies. In particular, inflammation provoked by obesity, notably by increased expression of the cytokine IL‐13, could play an important role in the carcinogenesis of obesity‐related CRC.
Collapse
Affiliation(s)
- Shimpei Matsui
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Koji Okabayashi
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Masashi Tsuruta
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Kohei Shigeta
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Ryo Seishima
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Takashi Ishida
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Takayuki Kondo
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Yoshiyuki Suzuki
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Hirotoshi Hasegawa
- Department of Surgery, Tokyo Dental College Ichikawa General Hospital, Chiba, Japan
| | - Masayuki Shimoda
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Shinya Sugimoto
- Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan
| | - Toshiro Sato
- Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan
| | - Yuko Kitagawa
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
8
|
Li YR, Ro V, Tchou JC. Obesity, Metabolic Syndrome, and Breast Cancer: From Prevention to Intervention. CURRENT SURGERY REPORTS 2018; 6:7. [PMID: 31293823 PMCID: PMC6619425 DOI: 10.1007/s40137-018-0204-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Obesity contributes to an estimated forty-percent, or 630,000 cases, of malignant neoplasms diagnosed in the United States[1] and higher body mass index (BMI) has been associated with at least seventeen types of solid tumors, including 9% of all breast cancer cases. In this review, we discuss the impact of obesity and consequences of obesity, including the metabolic syndrome and type 2 diabetes mellitus, on breast cancer risk and recurrence. Recent work has identified multiple molecular mechanisms that may underlie the association between obesity and breast cancer. In particular, insulin resistance, increased inflammatory cytokines, leptin signaling, and adipokine signaling have been shown to affect breast cancer risk and outcomes. While obesity is associated with higher breast cancer incidences and worse breast cancer outcomes, several risk reduction methods have been shown to attenuate these risks. Both metformin and statins have been shown to improve disease free survival and overall survival compared to non-users. Metformin also has been associated with lower risk of breast cancer incidence. Furthermore, increased physical activity and weight loss have been shown to decrease risk of breast cancer, especially in post-menopausal women. These studies have emphasized the potential impact that lifestyle changes can have on breast cancer risk and outcomes, and demonstrate the need for randomized control trials to evaluate the roles of metformin and statins for the treatment and chemoprevention of breast cancer.
Collapse
Affiliation(s)
- Yun Rose Li
- Department of Endocrine and Oncologic Surgery, Perelman School of Medicine, University of Pennsylvania,
Philadelphia, PA, USA, 19104
- Department of Radiation Oncology, Helen Diller Family Comprehensive Cancer Center, University of California
San Francisco, San Francisco, CA, 94143
| | - Vicky Ro
- Department of Endocrine and Oncologic Surgery, Perelman School of Medicine, University of Pennsylvania,
Philadelphia, PA, USA, 19104
| | - Julia C. Tchou
- Department of Endocrine and Oncologic Surgery, Perelman School of Medicine, University of Pennsylvania,
Philadelphia, PA, USA, 19104
| |
Collapse
|
9
|
Tanaka T, Kawabata K, Sugie S. 4-Nitroquinoline 1-Oxide-Induced Tongue and Esophagus Carcinogenesis in Obese and Diabetic TSOD Mice. World J Oncol 2017; 8:97-104. [PMID: 29147443 PMCID: PMC5650005 DOI: 10.14740/wjon1038w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 08/15/2017] [Indexed: 12/11/2022] Open
Abstract
Background Obesity and diabetes mellitus are associated with lifestyle-related carcinogenesis. They are also risk factors of esophageal adenocarcinoma, but there are only a few reports on association between obesity/diabetes and development of squamous cell carcinoma in the oral cavity and esophagus. In this study, we therefore aimed to determine whether obesity and diabetes affect oral and esophageal carcinogenesis using model mice of obesity and diabetes, the Tsumura Suzuki obese diabetes (TSOD) and Tsumura Suzuki non-obesity (TSNO) control mice, which were treated with 4-nitroquinoline 1-oxide (4-NQO) to produce tongue and esophageal carcinomas. Methods We used 28 each of the male TSOD and TSNO mice of 8 weeks of age. They were divided into the 4-NQO-treated group (n = 20) and untreated group (n = 8). 4-NQO was administered to mice in drinking water at a dose level of 20 ppm for 8 weeks. The untreated group was given distilled water without 4-NQO. At 28 experimental weeks, histopathological examination was performed on all organs including tongue and esophagus. We performed analysis of histopathology of all organs which included buccal capsule (a tongue)/esophagus after an experiment start in 28 weeks. Fasting plasma glucose (FPG) and lipid parameters including total cholesterol (T-Cho), triglyceride (TG), high-density lipoprotein (HDL)-cholesterol and low-density lipoprotein (LDL)-cholesterol were measured and all these parameters were compared between the two genotypes. Also, mRNA expression of eight cytokines including interleukin (IL)-1β, IL-6, IL-17, interferon (IFN)-γ, keratinocyte-derived cytokine (KC), macrophage inflammatory protein (MIP)-1α, MIP-2, and tumor necrosis factor (TNF)-α in the esophageal mucosa was assayed. Results 4-NQO treatment produced proliferative squamous cell lesions (dysplasia, papilloma and carcinoma) in the tongue and esophagus of both the TSOD and TSNO mice. The incidence and multiplicity of tongue tumors were 30% and 0.45 ± 0.83 in the TSOD mice and 30% and 0.40 ± 0.68 in the TSNO mice. The incidence and multiplicity of esophageal tumors were 70% and 2.25 ± 2.29 in the TSOD mice and 30% and 0.60 ± 1.14 (P < 0.01) in the TSNO mice. Conclusion Our findings indicate that the obese and diabetic TSOD mice were susceptible to 4-NQO-induced esophageal carcinogenesis, suggesting risk factors of obese and diabetes for esophageal squamous cell carcinoma. Additionally, the TSOD mice were useful as esophagus carcinogenic model. Our study first reported that 4-NQO induced esophageal cancer in mice.
Collapse
Affiliation(s)
- Takuji Tanaka
- Department of Diagnostic Pathology (DDP) & Research Center of Diagnostic Pathology (RC-DiP), Gifu Municipal Hospital, Gifu City, Gifu 500-8513, Japan.,Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu City, Gifu 501-1194, Japan
| | - Kunihiro Kawabata
- Palliative Care Center, Tokai Central Hospital, 4-6-2 Sohara-Higashijima-cho, Kakamigahara 504-8601, Japan
| | - Shigeyuki Sugie
- Department of Pathology, Murakami Memorial Hospital, Asahi University, School of Dentistry, 3-23 Hashimoto-cho, Gifu City, Gifu 500-8523, Japan
| |
Collapse
|
10
|
Onuma W, Asai D, Tomono S, Miyamoto S, Fujii G, Hamoya T, Nagano A, Takahashi S, Masumori S, Miyoshi N, Wakabayashi K, Mutoh M. Anticarcinogenic Effects of Dried Citrus Peel in Colon Carcinogenesis Due to Inhibition of Oxidative Stress. Nutr Cancer 2017; 69:855-861. [PMID: 28718722 DOI: 10.1080/01635581.2017.1339096] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Colorectal cancer is one of the leading causes of death worldwide. Reactive oxygen species produce oxidative stress and contribute to colorectal carcinogenesis. Because dietary citrus has been shown to reduce oxidative stress, we investigated the effects of citrus peel extract at dilutions of 1/200-1/500 on the activity of oxidative-stress-related transcription factors, including AP-1, NF-κB, NRF2, p53, and STAT3, in human colon cancer cell line HCT116 cells using a luciferase reporter gene assay. NRF2 transcriptional activities were 1.8- to 2.0-fold higher than the untreated control value. In addition, NF-κB, p53, and STAT3 transcriptional activities were 12-26% lower than the untreated control value. Administration of dried citrus peel in the diet of F344 rats at a dose of 1,000 ppm prevented the formation of azoxymethane-induced precancerous aberrant crypt foci (ACF) in the colon. The total number of ACF in rats fed with dried citrus peel was reduced to 75% of the control value. Moreover, the levels of oxidative-stress-related markers, reactive carbonyl species, in the serum of F344 rats were significantly reduced following the administration of dried citrus peel. These data suggest that citrus peel possesses an ability to suppress cellular oxidative stress through induction of NRF2, thereby preventing azoxymethane-induced colon carcinogenesis.
Collapse
Affiliation(s)
- Wakana Onuma
- a Epidemiology and Prevention Division , Research Center for Cancer Prevention and Screening, National Cancer Center , Tokyo , Japan
| | - Daichi Asai
- a Epidemiology and Prevention Division , Research Center for Cancer Prevention and Screening, National Cancer Center , Tokyo , Japan.,b Department of Experimental Pathology and Tumor Biology , Nagoya City University Graduate School of Medical Sciences , Nagoya , Japan
| | - Susumu Tomono
- c Graduate Division of Nutritional and Environmental Sciences , University of Shizuoka , Shizuoka , Japan
| | - Shingo Miyamoto
- a Epidemiology and Prevention Division , Research Center for Cancer Prevention and Screening, National Cancer Center , Tokyo , Japan
| | - Gen Fujii
- d Division of Carcinogenesis and Cancer Prevention , National Cancer Center Research Institute , Tokyo , Japan
| | - Takahiro Hamoya
- a Epidemiology and Prevention Division , Research Center for Cancer Prevention and Screening, National Cancer Center , Tokyo , Japan
| | - Aya Nagano
- a Epidemiology and Prevention Division , Research Center for Cancer Prevention and Screening, National Cancer Center , Tokyo , Japan.,b Department of Experimental Pathology and Tumor Biology , Nagoya City University Graduate School of Medical Sciences , Nagoya , Japan
| | - Satoru Takahashi
- b Department of Experimental Pathology and Tumor Biology , Nagoya City University Graduate School of Medical Sciences , Nagoya , Japan
| | | | - Noriyuki Miyoshi
- c Graduate Division of Nutritional and Environmental Sciences , University of Shizuoka , Shizuoka , Japan
| | - Keiji Wakabayashi
- c Graduate Division of Nutritional and Environmental Sciences , University of Shizuoka , Shizuoka , Japan
| | - Michihiro Mutoh
- a Epidemiology and Prevention Division , Research Center for Cancer Prevention and Screening, National Cancer Center , Tokyo , Japan.,d Division of Carcinogenesis and Cancer Prevention , National Cancer Center Research Institute , Tokyo , Japan
| |
Collapse
|
11
|
González N, Prieto I, del Puerto-Nevado L, Portal-Nuñez S, Ardura JA, Corton M, Fernández-Fernández B, Aguilera O, Gomez-Guerrero C, Mas S, Moreno JA, Ruiz-Ortega M, Sanz AB, Sanchez-Niño MD, Rojo F, Vivanco F, Esbrit P, Ayuso C, Alvarez-Llamas G, Egido J, García-Foncillas J, Ortiz A. 2017 update on the relationship between diabetes and colorectal cancer: epidemiology, potential molecular mechanisms and therapeutic implications. Oncotarget 2017; 8:18456-18485. [PMID: 28060743 PMCID: PMC5392343 DOI: 10.18632/oncotarget.14472] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 12/26/2016] [Indexed: 02/06/2023] Open
Abstract
Worldwide deaths from diabetes mellitus (DM) and colorectal cancer increased by 90% and 57%, respectively, over the past 20 years. The risk of colorectal cancer was estimated to be 27% higher in patients with type 2 DM than in non-diabetic controls. However, there are potential confounders, information from lower income countries is scarce, across the globe there is no correlation between DM prevalence and colorectal cancer incidence and the association has evolved over time, suggesting the impact of additional environmental factors. The clinical relevance of these associations depends on understanding the mechanism involved. Although evidence is limited, insulin use has been associated with increased and metformin with decreased incidence of colorectal cancer. In addition, colorectal cancer shares some cellular and molecular pathways with diabetes target organ damage, exemplified by diabetic kidney disease. These include epithelial cell injury, activation of inflammation and Wnt/β-catenin pathways and iron homeostasis defects, among others. Indeed, some drugs have undergone clinical trials for both cancer and diabetic kidney disease. Genome-wide association studies have identified diabetes-associated genes (e.g. TCF7L2) that may also contribute to colorectal cancer. We review the epidemiological evidence, potential pathophysiological mechanisms and therapeutic implications of the association between DM and colorectal cancer. Further studies should clarify the worldwide association between DM and colorectal cancer, strengthen the biological plausibility of a cause-and-effect relationship through characterization of the molecular pathways involved, search for specific molecular signatures of colorectal cancer under diabetic conditions, and eventually explore DM-specific strategies to prevent or treat colorectal cancer.
Collapse
Affiliation(s)
- Nieves González
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundacion Jimenez Diaz-UAM, Spanish Biomedical Research Network in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Isabel Prieto
- Radiation Oncology, Oncohealth Institute, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | - Laura del Puerto-Nevado
- Translational Oncology Division, Oncohealth Institute, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | - Sergio Portal-Nuñez
- Bone and Mineral Metabolism laboratory, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | - Juan Antonio Ardura
- Bone and Mineral Metabolism laboratory, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | - Marta Corton
- Genetics, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | | | - Oscar Aguilera
- Translational Oncology Division, Oncohealth Institute, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | | | - Sebastián Mas
- Nephrology, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | | | | | - Ana Belen Sanz
- Nephrology, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
- REDINREN, Madrid, Spain
| | | | - Federico Rojo
- Pathology, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | | | - Pedro Esbrit
- Bone and Mineral Metabolism laboratory, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | - Carmen Ayuso
- Genetics, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | | | - Jesús Egido
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundacion Jimenez Diaz-UAM, Spanish Biomedical Research Network in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
- Nephrology, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | - Jesús García-Foncillas
- Translational Oncology Division, Oncohealth Institute, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | - Alberto Ortiz
- Nephrology, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
- REDINREN, Madrid, Spain
| | | |
Collapse
|
12
|
Chen J, Huang XF. High fat diet-induced obesity increases the formation of colon polyps induced by azoxymethane in mice. ANNALS OF TRANSLATIONAL MEDICINE 2015; 3:79. [PMID: 25992378 DOI: 10.3978/j.issn.2305-5839.2015.03.46] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 03/02/2015] [Indexed: 02/04/2023]
Abstract
BACKGROUND Obesity has been found to be associated with colon cancer. However, the mechanism of this relationship is unclear and thus a good animal model is required. Our previous research showed that some mice developed diet-induced obesity (DIO) whilst others were diet-resistant (DR) when fed a high-fat diet. METHODS In the present study, we have tested the effects of a high-fat diet on the formation of colon polyps induced by azoxymethane (AOM) in both DIO and DR mice. RESULTS We found that the DIO mice have developed 2.5 times of polyps compared to the DR mice (P<0.05) and 3.4 times of polyps compared to the low fat fed mice (P<0.05). Although the DR mice tended to have more polyps than the low-fat diet fed mice, this was not statistically significant. The DIO mice could have an increased polyp formation due to obesity-related cancer risk factors and different gene expression from DR mice. CONCLUSIONS DIO mice could be used as an appropriate model for studying obesity-associated colon cancer; however DR mice are not suitable because they don't show any significant weight gains to indicate obesity.
Collapse
Affiliation(s)
- Jiezhong Chen
- 1 Illawarra Health and Medical Research Institute, University of Wollongong, Northfields Avenue, NSW 2522, Australia ; 2 School of Biomedical Sciences, University of Queensland, St Lucia, QLD 4072, Australia
| | - Xu-Feng Huang
- 1 Illawarra Health and Medical Research Institute, University of Wollongong, Northfields Avenue, NSW 2522, Australia ; 2 School of Biomedical Sciences, University of Queensland, St Lucia, QLD 4072, Australia
| |
Collapse
|
13
|
Dheer R, Davies JM, Abreu MT. Inflammation and Colorectal Cancer. INTESTINAL TUMORIGENESIS 2015:211-256. [DOI: 10.1007/978-3-319-19986-3_8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
14
|
Park J, Morley TS, Kim M, Clegg DJ, Scherer PE. Obesity and cancer--mechanisms underlying tumour progression and recurrence. Nat Rev Endocrinol 2014; 10:455-465. [PMID: 24935119 PMCID: PMC4374431 DOI: 10.1038/nrendo.2014.94] [Citation(s) in RCA: 551] [Impact Index Per Article: 50.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Over the past several years, the field of cancer research has directed increased interest towards subsets of obesity-associated tumours, which include mammary, renal, oesophageal, gastrointestinal and reproductive cancers in both men and women. The increased risk of breast cancer that is associated with obesity has been widely reported; this has drawn much attention and as such, warrants investigation of the key mechanisms that link the obese state with cancer aetiology. For instance, the obese setting provides a unique adipose tissue microenvironment with concomitant systemic endocrine alterations that favour both tumour initiation and progression. Major metabolic differences exist within tumours that distinguish them from non-transformed healthy tissues. Importantly, considerable metabolic differences are induced by tumour cells in the stromal vascular fraction that surrounds them. The precise mechanisms that underlie the association of obesity with cancer and the accompanying metabolic changes that occur in the surrounding microenvironment remain elusive. Nonetheless, specific therapeutic agents designed for patients with obesity who develop tumours are clearly needed. This Review discusses recent advances in understanding the contributions of obesity to cancer and their implications for tumour treatment.
Collapse
Affiliation(s)
- Jiyoung Park
- Department of Biological Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology, 50 UNIST Street, Ulsan 689-798, South Korea (J.P.). Touchstone Diabetes Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA (T.S.M., M.K., D.J.C., P.E.S.)
| | - Thomas S Morley
- Department of Biological Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology, 50 UNIST Street, Ulsan 689-798, South Korea (J.P.). Touchstone Diabetes Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA (T.S.M., M.K., D.J.C., P.E.S.)
| | - Min Kim
- Department of Biological Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology, 50 UNIST Street, Ulsan 689-798, South Korea (J.P.). Touchstone Diabetes Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA (T.S.M., M.K., D.J.C., P.E.S.)
| | - Deborah J Clegg
- Department of Biological Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology, 50 UNIST Street, Ulsan 689-798, South Korea (J.P.). Touchstone Diabetes Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA (T.S.M., M.K., D.J.C., P.E.S.)
| | - Philipp E Scherer
- Department of Biological Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology, 50 UNIST Street, Ulsan 689-798, South Korea (J.P.). Touchstone Diabetes Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA (T.S.M., M.K., D.J.C., P.E.S.)
| |
Collapse
|
15
|
Yamamoto M, Yanagisawa R, Motomura E, Nakamura M, Sakamoto M, Takeya M, Eto K. Increased methylmercury toxicity related to obesity in diabetic KK-Ay mice. J Appl Toxicol 2013; 34:914-23. [PMID: 24243536 DOI: 10.1002/jat.2954] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Revised: 09/23/2013] [Accepted: 09/23/2013] [Indexed: 11/07/2022]
Abstract
We examined the toxic effects of methylmercury (MeHg) in KK-Ay type 2 diabetic mice to clarify how metabolic changes associated with type 2 diabetes mellitus affect MeHg toxicity. MeHg (5 mg Hg kg (-1) day(-1) p.o.) was given to 4-week-old male KK-Ay and C57BL/6J (BL/6) mice three times per week for 6 weeks. Average body weights (BW) of vehicle-treated BL/6 and KK-Ay mice were 16.3 and 16.4 g respectively on the first day, and 24.8 and 42.3 g respectively on the last day of the experiment. MeHg-treated KK-Ay mice began to lose weight about 5 weeks after MeHg administration. Six of seven MeHg-treated KK-Ay mice showed hind-limb clasping in the final stage of the experiment. The mean blood mercury level of MeHg-treated KK-Ay mice reached a maximum of 9.8 µg ml(-1) , whereas that of the MeHg-treated BL/6 mice was 2.8 µg ml(-1) after 10 days of treatment. The average total mercury concentrations in the cerebrum and epididymal fat pad were 7.4 and 0.57 µg g(-1) , respectively, for BL/6 mice and 27 and 1.6 µg g(-1) , respectively, for KK-Ay mice. In MeHg-treated KK-Ay mice with neurological symptoms, CD204-positive macrophages were observed in the brain, kidney and spleen, indicating CD204 could be a marker for injured tissues. BW loss and significant pathological changes were not observed in other groups of mice. These results indicate that body fat gain in type 2 diabetes mellitus and low mercury accumulation in adipose tissue increased MeHg concentrations in organs and enhanced toxicity in KK-Ay mice at the same dose of MeHg per BW.
Collapse
Affiliation(s)
- Megumi Yamamoto
- Integrated Physiology Section, Department of Basic Medical Science, National Institute for Minamata Disease, Minamata, Kumamoto, Japan
| | | | | | | | | | | | | |
Collapse
|
16
|
Ito K, Ishigamori R, Mutoh M, Ohta T, Imai T, Takahashi M. Ay allele promotes azoxymethane-induced colorectal carcinogenesis by macrophage migration in hyperlipidemic/diabetic KK mice. Cancer Sci 2013; 104:835-43. [PMID: 23551905 PMCID: PMC7657178 DOI: 10.1111/cas.12162] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Revised: 03/18/2013] [Accepted: 03/19/2013] [Indexed: 01/17/2023] Open
Abstract
The incidence of colorectal cancer has been increasing and is associated with obesity and diabetes. We have found that type 2 diabetes model KK-Ay/TaJcl (KK-Ay) mice develop tumors within a short period after treatment with azoxymethane (AOM). However, factors that contribute to the promotion of carcinogenesis have not been clarified. Therefore, we looked at the genetic background of KK-Ay, including two genetic characteristics of KK/TaJcl (KK) mice and C57BL/6J-Ham-Ay/+ (Ay) mice, compared with other non-obese and non-diabetic mouse strains C57BL/6J and ICR, and induced colorectal premalignant lesions, aberrant crypt foci (ACF), and tumors using AOM (150 μg/mouse/week for 4 weeks and 200 μg/mouse/week for 6 weeks, respectively). The mice with a diabetes feature, KK-Ay and KK, developed significantly more ACF, 67 and 61 per mouse, respectively, whereas ICR, Ay, and C57BL/6J mice developed 42, 24, and 18 ACF/mouse, respectively, at 17 weeks of age. Serum insulin and triglyceride levels in KK-Ay and KK mice were quite high compared with other non-diabetic mouse strains. Interestingly, KK-Ay mice developed more colorectal tumors (2.7 ± 2.3 tumor/mouse) than KK mice (1.2 ± 1.1 tumor/mouse) at 25 weeks of age, in spite of similar diabetic conditions. The colon cancers that developed in both KK-Ay and KK mice showed similar activation of β-catenin signaling. However, mRNA levels of inflammatory factors related to the activation of macrophages were significantly higher in colorectal cancer of KK-Ay mice than in KK. These data indicate that factors such as insulin resistance and dyslipidemia observed in obese and diabetic patients could be involved in susceptibility to colorectal carcinogenesis. In addition, increase of tumor-associated macrophages may play important roles in the stages of promotion of colorectal cancer.
Collapse
Affiliation(s)
- Kumiko Ito
- Division of Cancer Prevention Research, National Cancer Center Research Institute, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
17
|
NAG-1/GDF15 transgenic mouse has less white adipose tissue and a reduced inflammatory response. Mediators Inflamm 2013; 2013:641851. [PMID: 23737651 PMCID: PMC3662179 DOI: 10.1155/2013/641851] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 03/18/2013] [Accepted: 04/03/2013] [Indexed: 02/07/2023] Open
Abstract
NAG-1/GDF15 is a TGF-β superfamily member with poorly characterized biological activity proposed to inhibit inflammatory cytokine production. Transgenic mice expressing human NAG-1/GDF15 (NAG-1Tg/Lox) are leaner with lower body weight and are resistant to chemically or genetically induced intestinal tumors. Because of the link between obesity, inflammation, and cancer, we examined whether these mice exhibit a reduced response to inflammatory stimuli. The NAG-1Tg/Lox mice had a reduced inflammatory response to LPS based on the serum levels of cytokines KC, IL-6, MCP-1, and TNFα. In contrast to literature reports and our in vivo results, NAG-1 did not inhibit LPS-induced cytokine expression in vitro in RAW264.7 cells, mouse peritoneal macrophages, or mouse liver Kupffer cells, suggesting that NAG-1/GDF15 does not directly inhibit LPS-induced inflammatory cytokine production. However, NAG-1Tg/Lox mice have less white adipose tissue, the major source of inflammatory adipokines including leptin. Basal and LPS-treated serum leptin and mRNA levels in the adipose tissue of NAG-1Tg/Lox mice were lower than those in WT mice. We propose that the reduced white adipose tissue and reduced leptin expression may be responsible, in part, for the reduced inflammatory response to LPS and the decrease in intestinal tumors observed in NAG-1Tg/Lox mice.
Collapse
|
18
|
Diet-induced obesity promotes colon tumor development in azoxymethane-treated mice. PLoS One 2013; 8:e60939. [PMID: 23560112 PMCID: PMC3616169 DOI: 10.1371/journal.pone.0060939] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 03/05/2013] [Indexed: 12/27/2022] Open
Abstract
Obesity is an important risk factor for colon cancer in humans, and numerous studies have shown that a high fat diet enhances colon cancer development. As both increased adiposity and high fat diet can promote tumorigenesis, we examined the effect of diet-induced obesity, without ongoing high fat diet, on colon tumor development. C57BL/6J male mice were fed regular chow or high fat diet for 8 weeks. Diets were either maintained or switched resulting in four experimental groups: regular chow (R), high fat diet (H), regular chow switched to high fat diet (RH), and high fat diet switched to regular chow (HR). Mice were then administered azoxymethane to induce colon tumors. Tumor incidence and multiplicity were dramatically smaller in the R group relative to all groups that received high fat diet at any point. The effect of obesity on colon tumors could not be explained by differences in aberrant crypt foci number. Moreover, diet did not alter colonic expression of pro-inflammatory cytokines tumor necrosis factor-α, interleukin-6, interleukin-1β, and interferon-γ, which were measured immediately after azoxymethane treatment. Crypt apoptosis and proliferation, which were measured at the same time, were increased in the HR relative to all other groups. Our results suggest that factors associated with obesity – independently of ongoing high fat diet and obesity – promote tumor development because HR group animals had significantly more tumors than R group, and these mice were fed the same regular chow throughout the entire carcinogenic period. Moreover, there was no difference in the number of aberrant crypt foci between these groups, and thus the effect of obesity appears to be on subsequent stages of tumor development when early preneoplastic lesions transition into adenomas.
Collapse
|
19
|
Metabolic syndrome: a novel high-risk state for colorectal cancer. Cancer Lett 2012; 334:56-61. [PMID: 23085010 DOI: 10.1016/j.canlet.2012.10.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Revised: 10/10/2012] [Accepted: 10/10/2012] [Indexed: 12/17/2022]
Abstract
Metabolic syndrome (MS) and related disorders, including cancer, are steadily increasing in most countries of the world. However, mechanisms underlying the link between MS and colon carcinogenesis have yet to be fully elucidated. In this review article we focus on the relationships between various individual associated conditions (obesity, dyslipidemia, diabetes mellitus type 2 and hypertension) and colon cancer development, and demonstrate probable related factors revealed by in vivo and in vitro studies. Furthermore, molecules suggested to be involved in cancer promotion are addressed, and the potential for cancer prevention by targeting these molecules is discussed.
Collapse
|
20
|
Qin B, Anderson RA, Kuzuya T, Kitaura Y, Shimomura Y. Multiple factors and pathways involved in hepatic very low density lipoprotein-apoB100 overproduction in Otsuka Long-Evans Tokushima Fatty rats. Atherosclerosis 2012; 222:409-16. [PMID: 22546076 DOI: 10.1016/j.atherosclerosis.2012.03.033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2011] [Revised: 03/02/2012] [Accepted: 03/29/2012] [Indexed: 01/04/2023]
Abstract
AIMS Overproduction of hepatic very low-density lipoprotein (VLDL) particles is a major abnormality of lipoprotein dysregulation in type 2 diabetes (T2D). We sought to examine the relationship between systemic/hepatic inflammation associated with insulin resistance and apolipoprotein (apo)B100-containing VLDL production. METHODS AND RESULTS At the age of 19 wks, Otsuka Long-Evans Tokushima Fatty (OLETF) rats showed systemic inflammation (plasma TNF-α and interleukin (IL)-6 levels increased), insulin resistance (plasma retinol binding protein 4 and soluble CD36 levels were higher), dyslipidemia and fatty liver (plasma and liver triglyceride and cholesterol levels were higher as well as total VLDL-, VLDL(1)-, VLDL(2)-apoB100 and VLDL-triglycerides were overproduced), compared with the control rats. In livers of OLETF rats, mRNA levels of tnf, il1b and il6 were increased, but an anti-inflammatory protein, zinc finger protein 36, and its mRNA expression were decreased. We also found that the liver mRNA, protein levels, and tyrosine phosphorylation (pY) of insulin receptor (InsR) substrate (IRS) 2, but not IRS1, were decreased in OLETF rats; pY of InsR and Akt protein and phospho-Akt (ser437) were also reduced; but protein tyrosine phosphatase-1B protein was overexpressed. The gene expressions of glucose transporters 1 and 2, and glycogen synthase were decreased, but phosphatase and tensin homolog deleted on chromosome ten and glycogen synthase kinase 3β mRNAs were overexpressed, compared with the controls. Sterol regulatory element binding protein-1c mRNA, ATP-binding cassette transporter A1 mRNA, microsomal triglyceride transfer protein mRNA/protein, and CD36 mRNA/protein levels were increased and lipoprotein lipase and Niemann-Pick c1-like1 mRNA levels were decreased, which are all involved in lipogenesis. Decreased sirtuins1-3 mRNA levels were also observed in OLETF rats. CONCLUSIONS These abnormal genes, proteins expression and phosphorylation of multiple pathways related to inflammatory, insulin signaling and lipogenesis may be important underlying factors in VLDL-apoB100 particles overproduction observed in T2D. Our data contribute to the further understanding of an association of dyslipoproteinemia with systemic metabolic disorders, fatty liver and dysregulated hepatic metabolic pathways.
Collapse
Affiliation(s)
- Bolin Qin
- United States Department of Agriculture, Agricultural Research Service, Beltsville Human Nutrition Research Center, Diet, Genomics and Immunology Laboratory, Beltsville, MD 20705, USA.
| | | | | | | | | |
Collapse
|
21
|
Abstract
Obesity is increasing worldwide and reaches to a large proportion of the population in developed countries. Thus, obesity-associated cancer has become a major health problem. Multiple cancer risk factors in obesity have been identified including insulin/insulin-like growth factor axis, adipokines and cytokines; and multiple intracellular signal pathways have been studied. However, the role of each signal pathway in obesity-associated cancer is controversial. In this review, the recent studies on signal pathways in obesity-associated cancer are summarized and a unified explanation is provided. Multiple risk factors could initially activate phosphoinositide 3-kinase (PI3K/Akt), mitogen-activated protein kinase (MAPK) and signal transducer and activator of transcription 3 (STAT3) pathways. With increased severity of obesity, mammalian target of rapamycin (mTOR), which is down-stream of both PI3K/Akt and MAPK, is highly activated. Activated mTOR in turn inhibits the PI3K/Akt pathway and further activates the STAT3 pathway. This may explain the activation of the PI3K/Akt pathway at the early stage of obesity and its inhibition at the later stage. mTOR inhibition may be used for cancer therapy, but it may be necessary to be combined with the PI3K/Akt inhibitor as decreased mTOR activity will release its feedback inhibition on the PI3K/Akt pathway, which is under the influence of multiple cancer risk factors in obesity. Thus, dual inhibitors of PI3K and mTOR may provide a novel approach.
Collapse
Affiliation(s)
- J Chen
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, New South Wales, Australia.
| |
Collapse
|
22
|
Abstract
The increasing incidence of obesity and its co-morbid conditions poses a great challenge to global health. In addition to cardiovascular disease and diabetes, epidemiological data demonstrate a link between obesity and multiple types of cancer. The molecular mechanisms underlying how obesity causes an increased risk of cancer are poorly understood. Obesity disrupts the dynamic role of the adipocyte in energy homeostasis, resulting in inflammation and alteration of adipokine (for example, leptin and adiponectin) signalling. Additionally, obesity causes secondary changes that are related to insulin signalling and lipid deregulation that may also foster cancer development. Understanding these molecular links may provide an avenue for preventive and therapeutic strategies to reduce cancer risk and mortality in an increasingly obese population.
Collapse
Affiliation(s)
- Melin J Khandekar
- Dana-Farber Cancer Institute and Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | |
Collapse
|
23
|
Molecular mechanisms linking adipokines to obesity-related colon cancer: focus on leptin. Proc Nutr Soc 2011; 71:175-80. [PMID: 22014041 DOI: 10.1017/s0029665111003259] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Obesity is linked to increased risk of colon cancer, currently the third most common cancer. Consequently rising levels of obesity worldwide are likely to significantly impact on obesity-related colon cancers in the decades to come. Understanding the molecular mechanisms whereby obesity increases colon cancer risk is thus a focus for research to inform strategies to prevent the increasing trend in obesity-related cancers. This review will consider research on deregulation of adipokine signalling, a consequence of altered adipokine hormone secretion from excess adipose tissue, with a focus on leptin, which has been studied extensively as a potential mediator of obesity-related colon cancer. Numerous investigations using colon cell lines in vitro, in vivo studies in rodents and investigations of colon cancer patients illuminate the complexity of the interactions of leptin with colon tissues via leptin receptors expressed by the colon epithelium. Although evidence indicates a role for leptin in proliferation of colon epithelial cells in vitro, this has been contradicted by studies in rodent models. However, recent studies have indicated that leptin may influence inflammatory mediators linked with colon cancer and also promote cell growth dependent on genotype and is implicated in growth promotion of colon cancer cells. Studies in human cancer patients indicate that there may be different tumour sub-types with varying levels of leptin receptor expression, indicating the potential for leptin to induce variable responses in the different tumour types. These studies have provided insights into the complex interplay of adipokines with responsive tissues prone to obesity-related colon cancer. Deregulation of adipokine signalling via adipokine receptors located in the colon appears to be a significant factor in obesity-related colon cancer. Molecular profiling of colon tumours will be a useful tool in future strategies to characterise the influence that adipokines may have on tumour development and subsequent therapeutic intervention. Study of the molecular mechanisms linking obesity with cancer also supports recommendations to maintain a normal body weight to reduce the risk of colon cancer.
Collapse
|
24
|
Takasu S, Mutoh M, Takahashi M, Nakagama H. Lipoprotein lipase as a candidate target for cancer prevention/therapy. Biochem Res Int 2011; 2012:398697. [PMID: 22028972 PMCID: PMC3199119 DOI: 10.1155/2012/398697] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Accepted: 08/17/2011] [Indexed: 12/30/2022] Open
Abstract
Epidemiological studies have shown that serum triglyceride (TG) levels are linked with risk of development of cancer, including colorectal and pancreatic cancers, and their precancerous lesions. Thus, it is assumed that serum TG plays an important role in carcinogenesis, and the key enzyme lipoprotein lipase (LPL), which catalyzes the hydrolysis of plasma TG, may therefore be involved. Dysregulation of LPL has been reported to contribute to many human diseases, such as atherosclerosis, chylomicronaemia, obesity, and type 2 diabetes. Recently, it has been reported that LPL gene deficiency, such as due to chromosome 8p22 loss, LPL gene polymorphism, and epigenetic changes in its promoter region gene, increases cancer risk, especially in the prostate. In animal experiments, high serum TG levels seem to promote sporadic/carcinogen-induced genesis of colorectal and pancreatic cancers. Interestingly, tumor suppressive effects of LPL inducers, such as PPAR ligands, NO-1886, and indomethacin, have been demonstrated in animal models. Moreover, recent evidence that LPL plays important roles in inflammation and obesity implies that it is an appropriate general target for chemopreventive and chemotherapeutic agents.
Collapse
Affiliation(s)
- Shinji Takasu
- Division of Cancer Development System, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Michihiro Mutoh
- Division of Cancer Development System, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Mami Takahashi
- Division of Cancer Development System, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Hitoshi Nakagama
- Division of Cancer Development System, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| |
Collapse
|