1
|
Molla MD, Symonds EL, Winter JM, Debie A, Wassie MM. Metabolic risk factors of colorectal cancer: Umbrella review. Crit Rev Oncol Hematol 2024; 204:104502. [PMID: 39245299 DOI: 10.1016/j.critrevonc.2024.104502] [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: 07/26/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/10/2024] Open
Abstract
BACKGROUND AND AIM The association between metabolic factors and colorectal cancer (CRC) risk is inconclusive. This umbrella review aimed to summarise and describe the association using existing systematic reviews and/or meta-analyses. METHOD Four databases (Medline, Scopus, Web of Science, and Cochrane Library) were searched for systematic reviews and/or meta-analyses of observational studies. Two independent authors extracted data on the summary estimated effect and heterogeneity of studies using I2 from the individual reviews. The Assessing the Methodological Quality of Systematic Reviews (AMSTAR 2) tool was used to evaluate the methodological quality. RESULTS 49 articles were included in this review. Although most included studies were graded with critically low methodological quality (81.6 %), we found a significant positive association between obesity (summary relative risk (SRR) range 1.19-1.49), diabetes mellitus (SRR range 1.20-1.37), hypertension (SRR range 1.07-1.62), metabolic syndrome (SRR range 1.25-1.36), non-alcoholic fatty liver disease (pooled odds ratio (POR) range 1.13-1.56), and risk of CRC. Higher serum high-density lipoprotein cholesterol levels were associated with a lower risk of CRC in 3/6 reviews, while others did not find any association. There was no clear association between high triglyceride levels, total cholesterol levels, low-density lipoprotein cholesterol levels, and risk of CRC. CONCLUSION This umbrella review identified that most metabolic factors are significantly associated with increased risk of CRC. Thus, people affected by metabolic factors may be benefited from CRC screening and surveillance.
Collapse
Affiliation(s)
- Meseret Derbew Molla
- Flinders University, College of Medicine and PublicHealth, Flinders Health and Medical Research Institute, Adelaide, South Australia, Australia; Department of Biochemistry, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia.
| | - Erin L Symonds
- Flinders University, College of Medicine and PublicHealth, Flinders Health and Medical Research Institute, Adelaide, South Australia, Australia; Gastroenterology and Hepatology Department, Flinders Medical Centre, Southern Adelaide Local Health Network, Bedford Park, South Australia, Australia
| | - Jean M Winter
- Flinders University, College of Medicine and PublicHealth, Flinders Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Ayal Debie
- Flinders University, College of Medicine and PublicHealth, Flinders Health and Medical Research Institute, Adelaide, South Australia, Australia; Department of Health Systems and Policy, Institute of Public Health, University of Gondar, Gondar, Ethiopia
| | - Molla M Wassie
- Flinders University, College of Medicine and PublicHealth, Flinders Health and Medical Research Institute, Adelaide, South Australia, Australia
| |
Collapse
|
2
|
Ruan X, Jin X, Sun F, Pi J, Jinghu Y, Lin X, Zhang N, Chen G. IGF signaling pathway in bone and cartilage development, homeostasis, and disease. FASEB J 2024; 38:e70031. [PMID: 39206513 DOI: 10.1096/fj.202401298r] [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: 06/08/2024] [Revised: 08/15/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
The skeleton plays a fundamental role in the maintenance of organ function and daily activities. The insulin-like growth factor (IGF) family is a group of polypeptide substances with a pronounced role in osteoblast differentiation, bone development, and metabolism. Disturbance of the IGFs and the IGF signaling pathway is inextricably linked with assorted developmental defects, growth irregularities, and jeopardized skeletal structure. Recent findings have illustrated the significance of the action of the IGF signaling pathway via growth factors and receptors and its interactions with dissimilar signaling pathways (Wnt/β-catenin, BMP, TGF-β, and Hh/PTH signaling pathways) in promoting the growth, survival, and differentiation of osteoblasts. IGF signaling also exhibits profound influences on cartilage and bone development and skeletal homeostasis via versatile cell-cell interactions in an autocrine, paracrine, and endocrine manner systemically and locally. Our review summarizes the role and regulatory function as well as a potentially integrated gene network of the IGF signaling pathway with other signaling pathways in bone and cartilage development and skeletal homeostasis, which in turn provides an enlightening insight into visualizing bright molecular targets to be eligible for designing effective drugs to handle bone diseases and maladies, such as osteoporosis, osteoarthritis, and dwarfism.
Collapse
Affiliation(s)
- Xinyi Ruan
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xiuhui Jin
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Fuju Sun
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Jiashun Pi
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yihan Jinghu
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xinyi Lin
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Nenghua Zhang
- Clinical Laboratory, Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing, China
| | - Guiqian Chen
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| |
Collapse
|
3
|
Marker PC, Unterberger CJ, Swanson SM. GH-dependent growth of experimentally induced carcinomas in vivo. Endocr Relat Cancer 2023; 30:e220403. [PMID: 36826838 PMCID: PMC10140676 DOI: 10.1530/erc-22-0403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/24/2023] [Indexed: 02/25/2023]
Abstract
Interest in investigating the role of the growth hormone (GH)/insulin-like growth factor 1 (IGF-1) axis in the initiation and progression of experimentally induced carcinomas has arisen due to several observations in the human population. First, subjects with Laron syndrome who lack GH signaling have significantly lower rates of cancer than people who have normal GH signaling. Second, epidemiologic studies have found strong associations between elevated circulating IGF-1 and the incidence of several common cancers. Third, women who bear children early in life have a dramatically reduced risk of developing breast cancer, which may be due to differences in hormone levels including GH. These observations have motivated multiple studies that have experimentally altered activity of the GH/IGF-1 axis in the context of experimental carcinoma models in mice and rats. Most of these studies have utilized carcinoma models for four organ systems that are also frequent sites of carcinomas in humans: the mammary gland, prostate gland, liver, and colon. This review focuses on these studies and describes some of the most common genetic models used to alter the activity of the GH/IGF-1 axis in experimentally induced carcinomas. A recurring theme that emerges from these studies is that manipulations that reduce the activity of GH or mediators of GH action also inhibit carcinogenesis in multiple model systems.
Collapse
Affiliation(s)
- Paul C. Marker
- School of Pharmacy, Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| | - Christopher J. Unterberger
- School of Pharmacy, Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| | - Steven M. Swanson
- School of Pharmacy, Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| |
Collapse
|
4
|
Xiao W, Huang J, Zhao C, Ding L, Wang X, Wu B. Diabetes and Risks of Right-Sided and Left-Sided Colon Cancer: A Meta-Analysis of Prospective Cohorts. Front Oncol 2022; 12:737330. [PMID: 35463382 PMCID: PMC9021717 DOI: 10.3389/fonc.2022.737330] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 03/17/2022] [Indexed: 11/22/2022] Open
Abstract
Background and Aims Diabetes is associated with an increased risk of colon cancer (CC). Epidemiologic studies previously reported a higher risk for right-sided colon cancer (RCC) compare to left-sided colon cancer (LCC), although data are conflicting. We performed a meta-analysis to investigate this issue. Methods We systematically searched the PubMed, EMBASE, Web of Science and Cochrane Library database for prospective cohort studies published up to June 2021. Studies were included if they reported site-specific estimates of the relative risk (RR) between diabetes and the risks of RCC and LCC. Random effects meta-analyses with inverse variance weighting were used to estimate the pooled site-specific RRs and the RCC-to-LCC ratio of RRs (RRRs). Results Data from 10 prospective cohort studies, representing 1,642,823 individuals (mainly white) and 17,624 CC patients, were included in the analysis. Diabetes was associated with an increased risk of both RCC (RR =1.35, 95% CI = 1.24-1.47) and LCC (RR = 1.18, 95% CI = 1.08-1.28). After adjusting for major risk factors, individuals with diabetes had a greater risk for RCC than for LCC (RRR = 1.13, 95% CI = 1.02-1.26), with no significant heterogeneity between studies (I2 = 0%). Conclusions This meta-analysis indicates that diabetes is associated with a higher risk for RCC than for LCC. Our findings suggest that colonoscopic surveillance in diabetic patients with careful examination of the right colon is warranted.
Collapse
Affiliation(s)
- Wenxuan Xiao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinglong Huang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chuanyi Zhao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lu Ding
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xuan Wang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bian Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
5
|
Obesity and aging: Molecular mechanisms and therapeutic approaches. Ageing Res Rev 2021; 67:101268. [PMID: 33556548 DOI: 10.1016/j.arr.2021.101268] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 01/19/2021] [Accepted: 02/02/2021] [Indexed: 02/08/2023]
Abstract
The epidemic of obesity is a major challenge for health policymakers due to its far-reaching effects on population health and potentially overwhelming financial burden on healthcare systems. Obesity is associated with an increased risk of developing acute and chronic diseases, including hypertension, stroke, myocardial infarction, cardiovascular disease, diabetes, and cancer. Interestingly, the metabolic dysregulation associated with obesity is similar to that observed in normal aging, and substantial evidence suggests the potential of obesity to accelerate aging. Therefore, understanding the mechanism of fat tissue dysfunction in obesity could provide insights into the processes that contribute to the metabolic dysfunction associated with the aging process. Here, we review the molecular and cellular mechanisms underlying both obesity and aging, and how obesity and aging can predispose individuals to chronic health complications. The potential of lifestyle and pharmacological interventions to counter obesity and obesity-related pathologies, as well as aging, is also addressed.
Collapse
|
6
|
Roslan NH, Makpol S, Mohd Yusof YA. A Review on Dietary Intervention in Obesity Associated Colon Cancer. Asian Pac J Cancer Prev 2019; 20:1309-1319. [PMID: 31127882 PMCID: PMC6857900 DOI: 10.31557/apjcp.2019.20.5.1309] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 04/22/2019] [Indexed: 12/13/2022] Open
Abstract
Background: Colorectal cancer (CRC) is one of the major causes of morbidity and mortality. According to National Cancer Registry, the incidence of colorectal cancer in Peninsular Malaysia increases with age. The incidence is highest among Chinese population but lower among Indians and Malays. Many reviews have suggested that obesity may be associated with a higher risk (>50%) of colorectal cancer. Methods: This study collects a comprehensive data from the literature review available from respective journals on dietary intervention and the chemo-protective mechanisms of a few natural resources in obesity -associated colon cancer based on previous and current studies. Results: In obesity-associated colon cancer, the genes of interest and pathways that are mainly involved include NFκB, P13K/Akt, and MAPK pathways, and FTO, leptin, Cyclin D, MMPs, and STAT3 genes. Dietary modification is one of the alternative steps in early prevention of colon cancer. It has been proposed that the components present in certain foods may have the ability to protect against many diseases including the prevention of cancer. Conclusion: There are many factors that lead to obesity-associated colon cancer and the mechanisms behind it is still undergoing intensive research. This review aims to scrutinize research as well as reviews that have been previously reported on obesity associated colorectal cancer and the beneficial effects of including antioxidants-rich foods such as vegetables and fruits in the diet to reduce the risk of obesity associated colorectal cancer.
Collapse
Affiliation(s)
- N H Roslan
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, Kuala Lumpur, Malaysia.
| | - S Makpol
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, Kuala Lumpur, Malaysia.
| | - Y A Mohd Yusof
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, Kuala Lumpur, Malaysia.
| |
Collapse
|
7
|
de Kort S, Simons CCJM, van den Brandt PA, Janssen-Heijnen MLG, Sanduleanu S, Masclee AAM, Weijenberg MP. Diabetes mellitus, genetic variants in the insulin-like growth factor pathway and colorectal cancer risk. Int J Cancer 2019; 145:1774-1781. [PMID: 31018241 PMCID: PMC6767781 DOI: 10.1002/ijc.32365] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 03/24/2019] [Accepted: 04/02/2019] [Indexed: 12/31/2022]
Abstract
Genetic variation in the insulin‐like growth factor (IGF) pathway may further increase the risk of colorectal cancer (CRC) associated with type 2 diabetes mellitus (T2DM). Joint effects of T2DM and genetic variation in the IGF pathway on CRC risk can increase mechanistic insights. Participants from the Netherlands Cohort Study (n = 120, 852) completed a baseline questionnaire in 1986 when 55–69 years old (case–cohort, nsubcohort = 5,000, ncases = 3,441 after 16.3 years follow‐up). Self‐reported DM at baseline with onset at ≥30 years was classified as T2DM. Eighteen single nucleotide polymorphisms (SNPs) from the IGF pathway were aggregated in a genetic risk score (GRS). Cox proportional hazard ratios (HRs) for CRC were estimated according to combinations of T2DM status with GRS tertiles and categories of an IGF1 19‐CA repeat polymorphism. Baseline T2DM prevalence was 3.1% in subcohort members and 3.8% in CRC cases. Comparison of combined categories with non‐T2DM individuals in the lowest GRS tertile as reference showed that those in the highest GRS tertiles with and without T2DM had significantly increased CRC risks, particularly those with T2DM (HR = 2.28, 95% CI: 1.11, 4.66). As compared to IGF1 19‐CA wild‐type carriers without T2DM, carrying two IGF1 19‐CA variant repeat alleles were associated with a significantly decreased CRC risk in those without T2DM (HR = 0.76, 95% CI: 0.63–0.91). This association was absent when T2DM was present. Our study of joint effects indicated that the presence of unfavorable alleles in the IGF pathway may further increase the risk of CRC associated with T2DM. What's new? Colorectal cancer (CRC) and type‐2 diabetes mellitus (T2DM) have several risk factors in common (high BMI, physical inactivity, etc.). In this study, the authors identified several SNPs in the insulin‐like growth factor (IGF) pathway that further increase CRC risk, particularly in people with T2DM. These results support the hypothesis that T2DM increases CRC risk by influencing the IGF pathway. Screening for these unfavorable genetic variations in the IGF pathway may also help to improve the assessment of personalized CRC risk.
Collapse
Affiliation(s)
- Sander de Kort
- Department of Epidemiology, GROW-School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands.,Division of Gastroenterology and Hepatology, Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, The Netherlands.,GROW-School for Oncology and Developmental Biology, NUTRIM, School for Nutrition, Toxicology and Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Colinda C J M Simons
- Department of Epidemiology, GROW-School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Piet A van den Brandt
- Department of Epidemiology, GROW-School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Maryska L G Janssen-Heijnen
- Department of Epidemiology, GROW-School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands.,Department of Clinical Epidemiology, VieCuri Medical Centre, Venlo, The Netherlands
| | - Silvia Sanduleanu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, The Netherlands.,GROW-School for Oncology and Developmental Biology, NUTRIM, School for Nutrition, Toxicology and Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Ad A M Masclee
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, The Netherlands.,GROW-School for Oncology and Developmental Biology, NUTRIM, School for Nutrition, Toxicology and Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Matty P Weijenberg
- Department of Epidemiology, GROW-School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
8
|
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: 2.1] [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
|
9
|
Distribution of a Single Nucleotide Polymorphism of Insulin-Like Growth Factor-1 in Colorectal Cancer Patients and Its Association with Mucinous Adenocarcinoma. Int J Biol Markers 2018. [DOI: 10.5301/jbm.2010.6119] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Purpose To analyze the difference in the distribution of an insulin growth factor-1 (IGF-1) polymorphism (-2995 C/A) between young and old colorectal cancer (CRC) patients. Methods Information from 950 CRC patients undergoing surgery at the Taipei Veterans General Hospital between 2000 and 2005 was collected. The IGF-1 polymorphism was analyzed in patients in extreme age ranges at the time of CRC onset (i.e., under the 20th and above the 80th percentiles, respectively). Associations between clinicopathological variables and the IGF-1 polymorphism were analyzed. Results Young CRC patients had a higher frequency of advanced disease (58.7%) and mucinous adenocarcinoma (20%) than old CRC patients. Among old CRC patients, the frequency of the AA genotype of IGF-1 was 12.7% (24/189), which was significantly higher than in young patients (4.2%). Other clinicopathological factors including tumor location, differentiation, lymphovascular invasion, and TNM stage were not associated with the AA genotype of IGF-1. Mucinous differentiation (but not the other clinicopathological factors) was significantly associated with the CA/AA genotype of IGF-1 (39/195). Conclusions Older patients had a higher frequency of the AA genotype of IGF-1(-2995 C/A), while younger patients more often had advanced disease and mucinous adenocarcinoma.
Collapse
|
10
|
Demetz G, Oostendorp RA, Boxberg AM, Sitz W, Farrell E, Steppich B, Steinsiek AL, Rudelius M, Ott I. Overexpression of Insulin-Like Growth Factor-2 in Expanded Endothelial Progenitor Cells Improves Left Ventricular Function in Experimental Myocardial Infarction. J Vasc Res 2017; 54:321-328. [DOI: 10.1159/000479872] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 07/28/2017] [Indexed: 11/19/2022] Open
|
11
|
Jiang L, Hermeking H. miR-34a and miR-34b/c Suppress Intestinal Tumorigenesis. Cancer Res 2017; 77:2746-2758. [PMID: 28363996 DOI: 10.1158/0008-5472.can-16-2183] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 10/04/2016] [Accepted: 03/23/2017] [Indexed: 11/16/2022]
Abstract
The p53-inducible miR-34a and miR-34b/c genes are frequently silenced in colorectal cancer. To address the in vivo relevance of miR-34a/b/c function for suppression of intestinal tumor formation, we generated ApcMin/+ mice with deletions of the miR-34a and/or miR-34b/c genes separately or in combination. Combined deletion of miR-34a/b/c increased the number of intestinal stem cells as well as Paneth and Goblet cells, resulting in enlarged intestinal crypts. miR-34a/b/c-deficient ApcMin/+ mice displayed an increased tumor burden and grade and decreased survival. miR-34a/b/c-deficient adenomas showed elevated proliferation and decreased apoptosis and displayed pronounced bacterial infiltration, which may be due to an observed decrease in infiltrating immune cells and downregulation of barrier proteins. mRNA induction in miR-34a/b/c-deficient tumors was enriched for miR-34a/b/c seed-matching sites and for mRNAs encoding proteins related to epithelial-mesenchymal transition, stemness, and Wnt signaling. Accordingly, cells explanted from miR-34a/b/c-deficient adenomas formed tumor organoids at an increased rate. Several upregulated miR-34 targets displayed elevated expression in primary human colorectal cancers that was associated with lymph-node metastases (INHBB, AXL, FGFR1, and PDFGRB) and upregulation of INHBB and AXL in primary colorectal cancer was associated with poor patient survival. In conclusion, our results show that miR-34a/b/c suppress tumor formation caused by loss of Apc and control intestinal stem cell and secretory cell homeostasis by downregulation of multiple target mRNAs. Cancer Res; 77(10); 2746-58. ©2017 AACR.
Collapse
Affiliation(s)
- Longchang Jiang
- Experimental and Molecular Pathology, Institute of Pathology, Ludwig-Maximilians-University, München, Germany
| | - Heiko Hermeking
- Experimental and Molecular Pathology, Institute of Pathology, Ludwig-Maximilians-University, München, Germany. .,German Cancer Consortium (DKTK), Partner site Munich, Munich, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
12
|
Podlutsky A, Valcarcel-Ares MN, Yancey K, Podlutskaya V, Nagykaldi E, Gautam T, Miller RA, Sonntag WE, Csiszar A, Ungvari Z. The GH/IGF-1 axis in a critical period early in life determines cellular DNA repair capacity by altering transcriptional regulation of DNA repair-related genes: implications for the developmental origins of cancer. GeroScience 2017; 39:147-160. [PMID: 28233247 DOI: 10.1007/s11357-017-9966-x] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 02/14/2017] [Indexed: 12/31/2022] Open
Abstract
Experimental, clinical, and epidemiological findings support the concept of developmental origins of health and disease (DOHAD), suggesting that early-life hormonal influences during a sensitive period around adolescence have a powerful impact on cancer morbidity later in life. The endocrine changes that occur during puberty are highly conserved across mammalian species and include dramatic increases in circulating GH and IGF-1 levels. Importantly, patients with developmental IGF-1 deficiency due to GH insensitivity (Laron syndrome) do not develop cancer during aging. Rodents with developmental GH/IGF-1 deficiency also exhibit significantly decreased cancer incidence at old age, marked resistance to chemically induced carcinogenesis, and cellular resistance to genotoxic stressors. Early-life treatment of GH/IGF-1-deficient mice and rats with GH reverses the cancer resistance phenotype; however, the underlying molecular mechanisms remain elusive. The present study was designed to test the hypothesis that developmental GH/IGF-1 status impacts cellular DNA repair mechanisms. To achieve that goal, we assessed repair of γ-irradiation-induced DNA damage (single-cell gel electrophoresis/comet assay) and basal and post-irradiation expression of DNA repair-related genes (qPCR) in primary fibroblasts derived from control rats, Lewis dwarf rats (a model of developmental GH/IGF-1 deficiency), and GH-replete dwarf rats (GH administered beginning at 5 weeks of age, for 30 days). We found that developmental GH/IGF-1 deficiency resulted in persisting increases in cellular DNA repair capacity and upregulation of several DNA repair-related genes (e.g., Gadd45a, Bbc3). Peripubertal GH treatment reversed the radiation resistance phenotype. Fibroblasts of GH/IGF-1-deficient Snell dwarf mice also exhibited improved DNA repair capacity, showing that the persisting influence of peripubertal GH/IGF-1 status is not species-dependent. Collectively, GH/IGF-1 levels during a critical period during early life determine cellular DNA repair capacity in rodents, presumably by transcriptional control of genes involved in DNA repair. Because lifestyle factors (e.g., nutrition and childhood obesity) cause huge variation in peripubertal GH/IGF-1 levels in children, further studies are warranted to determine their persisting influence on cellular cancer resistance pathways.
Collapse
Affiliation(s)
- Andrej Podlutsky
- Department of Geriatric Medicine, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, 975 N. E. 10th Street-BRC 1303, Oklahoma City, OK, 73104, USA
- Department of Biology and Wildlife, Center for Alaska Native Health Research, University of Alaska Fairbanks, 902 N. Koyukuk, Fairbanks, AK, 99775, USA
| | - Marta Noa Valcarcel-Ares
- Department of Geriatric Medicine, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, 975 N. E. 10th Street-BRC 1303, Oklahoma City, OK, 73104, USA
| | - Krysta Yancey
- Department of Biology and Wildlife, Center for Alaska Native Health Research, University of Alaska Fairbanks, 902 N. Koyukuk, Fairbanks, AK, 99775, USA
| | - Viktorija Podlutskaya
- Department of Biology and Wildlife, Center for Alaska Native Health Research, University of Alaska Fairbanks, 902 N. Koyukuk, Fairbanks, AK, 99775, USA
| | - Eszter Nagykaldi
- Department of Geriatric Medicine, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, 975 N. E. 10th Street-BRC 1303, Oklahoma City, OK, 73104, USA
| | - Tripti Gautam
- Department of Geriatric Medicine, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, 975 N. E. 10th Street-BRC 1303, Oklahoma City, OK, 73104, USA
| | - Richard A Miller
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- University of Michigan Geriatrics Center, Ann Arbor, MI, USA
| | - William E Sonntag
- Department of Geriatric Medicine, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, 975 N. E. 10th Street-BRC 1303, Oklahoma City, OK, 73104, USA
| | - Anna Csiszar
- Department of Geriatric Medicine, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, 975 N. E. 10th Street-BRC 1303, Oklahoma City, OK, 73104, USA
- Department of Medical Physics and Informatics, University of Szeged, Szeged, Hungary
| | - Zoltan Ungvari
- Department of Geriatric Medicine, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, 975 N. E. 10th Street-BRC 1303, Oklahoma City, OK, 73104, USA.
- Department of Medical Physics and Informatics, University of Szeged, Szeged, Hungary.
| |
Collapse
|
13
|
Diabetes mellitus type 2 and subsite-specific colorectal cancer risk in men and women: results from the Netherlands Cohort Study on diet and cancer. Eur J Gastroenterol Hepatol 2016; 28:896-903. [PMID: 27097356 DOI: 10.1097/meg.0000000000000626] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) is associated with an increased risk of colorectal cancer (CRC); however, studies differentiating between subsites of CRC are limited. We investigated how diabetes mellitus (DM) was associated with subsite-specific CRC risk in men and women. METHODS The Netherlands Cohort Study on diet and cancer is a prospective study among 120 852 men and women aged 55-69 years old at baseline in 1986. Information on DM, anthropometric, dietary and lifestyle factors was self-reported at baseline. T2DM was defined as the diagnosis of DM after 30 years of age. Incident CRC cases were identified by record linkage with the Netherlands cancer registry and the Dutch pathology registry. After 17.3 years of follow-up, 1735 incident male CRC cases and 1321 female CRC cases were available for analyses. Subsite-specific hazard ratios (HRs) for CRC were estimated in case-cohort analyses using Cox regression. RESULTS At baseline, 3.1% of subcohort members reported T2DM, of whom 80% were diagnosed after 50 years of age. Multivariable-adjusted models showed that the risk of proximal colon cancer was significantly increased in women with T2DM versus women without T2DM (HR=1.80, 95% confidence interval: 1.10-2.94). There was no association between T2DM and the risk of overall CRC, distal colon cancer and rectal cancer in women. In men, T2DM was not associated with overall CRC (HR=0.98, 95% confidence interval: 0.64-1.50), or with risk at any subsite. CONCLUSIONS This prospective study showed an increased risk of proximal colon cancer in women with T2DM compared with non-T2DM women.
Collapse
|
14
|
Tracz AF, Szczylik C, Porta C, Czarnecka AM. Insulin-like growth factor-1 signaling in renal cell carcinoma. BMC Cancer 2016; 16:453. [PMID: 27405474 PMCID: PMC4942928 DOI: 10.1186/s12885-016-2437-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Accepted: 06/28/2016] [Indexed: 12/12/2022] Open
Abstract
Renal cell carcinoma (RCC) incidence is highest in highly developed countries and it is the seventh most common neoplasm diagnosed. RCC management include nephrectomy and targeted therapies. Type 1 insulin-like growth factor (IGF-1) pathway plays an important role in cell proliferation and apoptosis resistance. IGF-1 and insulin share overlapping downstream signaling pathways in normal and cancer cells. IGF-1 receptor (IGF1R) stimulation may promote malignant transformation promoting cell proliferation, dedifferentiation and inhibiting apoptosis. Clear cell renal cell carcinoma (ccRCC) patients with IGF1R overexpression have 70 % increased risk of death compared to patients who had tumors without IGF1R expression. IGF1R signaling deregulation may results in p53, WT, BRCA1, VHL loss of function. RCC cells with high expression of IGF1R are more resistant to chemotherapy than cells with low expression. Silencing of IGF1R increase the chemosensitivity of ccRCC cells and the effect is greater in VHL mutated cells. Understanding the role of IGF-1 signaling pathway in RCC may result in development of new targeted therapeutic interventions. First preclinical attempts with anti-IGF-1R monoclonal antibodies or fragment antigen-binding (Fab) fragments alone or in combination with an mTOR inhibitor were shown to inhibit in vitro growth and reduced the number of colonies formed by of RCC cells.
Collapse
Affiliation(s)
- Adam F Tracz
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, 04-141, Warsaw, Poland.,First Faculty of Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Cezary Szczylik
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, 04-141, Warsaw, Poland
| | - Camillo Porta
- Department of Medical Oncology, IRCCS San Matteo University Hospital Foundation, Pavia, Italy
| | - Anna M Czarnecka
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, 04-141, Warsaw, Poland.
| |
Collapse
|
15
|
Turner RT, Dube M, Branscum AJ, Wong CP, Olson DA, Zhong X, Kweh MF, Larkin IV, Wronski TJ, Rosen CJ, Kalra SP, Iwaniec UT. Hypothalamic leptin gene therapy reduces body weight without accelerating age-related bone loss. J Endocrinol 2015; 227:129-41. [PMID: 26487675 PMCID: PMC4917201 DOI: 10.1530/joe-15-0280] [Citation(s) in RCA: 11] [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] [Accepted: 09/14/2015] [Indexed: 02/04/2023]
Abstract
Excessive weight gain in adults is associated with a variety of negative health outcomes. Unfortunately, dieting, exercise, and pharmacological interventions have had limited long-term success in weight control and can result in detrimental side effects, including accelerating age-related cancellous bone loss. We investigated the efficacy of using hypothalamic leptin gene therapy as an alternative method for reducing weight in skeletally-mature (9 months old) female rats and determined the impact of leptin-induced weight loss on bone mass, density, and microarchitecture, and serum biomarkers of bone turnover (CTx and osteocalcin). Rats were implanted with cannulae in the 3rd ventricle of the hypothalamus and injected with either recombinant adeno-associated virus encoding the gene for rat leptin (rAAV-Leptin, n=7) or a control vector encoding green fluorescent protein (rAAV-GFP, n=10) and sacrificed 18 weeks later. A baseline control group (n=7) was sacrificed at vector administration. rAAV-Leptin-treated rats lost weight (-4±2%) while rAAV-GFP-treated rats gained weight (14±2%) during the study. At study termination, rAAV-Leptin-treated rats weighed 17% less than rAAV-GFP-treated rats and had lower abdominal white adipose tissue weight (-80%), serum leptin (-77%), and serum IGF1 (-34%). Cancellous bone volume fraction in distal femur metaphysis and epiphysis, and in lumbar vertebra tended to be lower (P<0.1) in rAAV-GFP-treated rats (13.5 months old) compared to baseline control rats (9 months old). Significant differences in cancellous bone or biomarkers of bone turnover were not detected between rAAV-Leptin and rAAV-GFP rats. In summary, rAAV-Leptin-treated rats maintained a lower body weight compared to baseline and rAAV-GFP-treated rats with minimal effects on bone mass, density, microarchitecture, or biochemical markers of bone turnover.
Collapse
Affiliation(s)
- Russell T Turner
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Michael Dube
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Adam J Branscum
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Carmen P Wong
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Dawn A Olson
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Xiaoying Zhong
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Mercedes F Kweh
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Iske V Larkin
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Thomas J Wronski
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Clifford J Rosen
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Satya P Kalra
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Urszula T Iwaniec
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| |
Collapse
|
16
|
Santoro MA, Blue RE, Andres SF, Mah AT, Van Landeghem L, Lund PK. Obesity and intestinal epithelial deletion of the insulin receptor, but not the IGF 1 receptor, affect radiation-induced apoptosis in colon. Am J Physiol Gastrointest Liver Physiol 2015; 309:G578-89. [PMID: 26251471 PMCID: PMC4593818 DOI: 10.1152/ajpgi.00189.2015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 07/31/2015] [Indexed: 01/31/2023]
Abstract
Current views suggest that apoptosis eliminates genetically damaged cells that may otherwise form tumors. Prior human studies link elevated insulin and reduced apoptosis to risk of colorectal adenomas. We hypothesized that hyperinsulinemia associated with obesity would lead to reduced colon epithelial cell (CEC) apoptosis after radiation and that this effect would be altered by deletion of the insulin-like growth factor (IGF) 1 receptor (IGF1R) or the insulin receptor (IR). Mice with villin-Cre-mediated IGF1R or IR deletion in CECs and floxed littermates were fed a high-fat diet to induce obesity and hyperinsulinemia or control low-fat chow. Mice were exposed to 5-Gy abdominal radiation to induce DNA damage and euthanized 4 h later for evaluation of apoptosis by localization of cleaved caspase-3. Obese mice exhibited decreased apoptosis of genetically damaged CECs. IGF1R deletion did not affect CEC apoptosis in lean or obese animals. In contrast, IR loss increased CEC apoptosis in both diet groups but did not prevent antiapoptotic effects of obesity. Levels of p53 protein were significantly reduced in CECs of obese mice with intact IR but increased in both lean and obese mice without IR. Levels of mRNAs encoding proapoptotic Perp and the cell cycle inhibitor Cdkn1b/p27 were reduced in CECs of obese mice and increased in lean mice lacking IR. Together, our studies provide novel evidence for antiapoptotic roles of obesity and IR, but not IGF1R, in colonic epithelium after DNA damage. However, neither IR nor IGF1R deletion prevented a reduction in radiation-induced CEC apoptosis during obesity and hyperinsulinemia.
Collapse
Affiliation(s)
- M. Agostina Santoro
- 1Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and
| | - R. Eric Blue
- 1Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and
| | - Sarah F. Andres
- 1Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and
| | - Amanda T. Mah
- 2Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Laurianne Van Landeghem
- 1Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and
| | - P. Kay Lund
- 1Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and
| |
Collapse
|
17
|
Kasdagly M, Radhakrishnan S, Reddivari L, Veeramachaneni DR, Vanamala J. Colon carcinogenesis: Influence of Western diet-induced obesity and targeting stem cells using dietary bioactive compounds. Nutrition 2014; 30:1242-56. [DOI: 10.1016/j.nut.2014.02.016] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 02/07/2014] [Accepted: 02/12/2014] [Indexed: 02/07/2023]
|
18
|
Harvey AE, Lashinger LM, Hays D, Harrison LM, Lewis K, Fischer SM, Hursting SD. Calorie restriction decreases murine and human pancreatic tumor cell growth, nuclear factor-κB activation, and inflammation-related gene expression in an insulin-like growth factor-1-dependent manner. PLoS One 2014; 9:e94151. [PMID: 24804677 PMCID: PMC4013119 DOI: 10.1371/journal.pone.0094151] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2013] [Accepted: 03/11/2014] [Indexed: 12/31/2022] Open
Abstract
Calorie restriction (CR) prevents obesity and has potent anticancer effects that may be mediated through its ability to reduce serum growth and inflammatory factors, particularly insulin-like growth factor (IGF)-1 and protumorigenic cytokines. IGF-1 is a nutrient-responsive growth factor that activates the inflammatory regulator nuclear factor (NF)-κB, which is linked to many types of cancers, including pancreatic cancer. We hypothesized that CR would inhibit pancreatic tumor growth through modulation of IGF-1-stimulated NF-κB activation and protumorigenic gene expression. To test this, 30 male C57BL/6 mice were randomized to either a control diet consumed ad libitum or a 30% CR diet administered in daily aliquots for 21 weeks, then were subcutaneously injected with syngeneic mouse pancreatic cancer cells (Panc02) and tumor growth was monitored for 5 weeks. Relative to controls, CR mice weighed less and had decreased serum IGF-1 levels and smaller tumors. Also, CR tumors demonstrated a 70% decrease in the expression of genes encoding the pro-inflammatory factors S100a9 and F4/80, and a 56% decrease in the macrophage chemoattractant, Ccl2. Similar CR effects on tumor growth and NF-κB-related gene expression were observed in a separate study of transplanted MiaPaCa-2 human pancreatic tumor cell growth in nude mice. In vitro analyses in Panc02 cells showed that IGF-1 treatment promoted NF-κB nuclear localization, increased DNA-binding of p65 and transcriptional activation, and increased expression of NF-κB downstream genes. Finally, the IGF-1-induced increase in expression of genes downstream of NF-κB (Ccdn1, Vegf, Birc5, and Ptgs2) was decreased significantly in the context of silenced p65. These findings suggest that the inhibitory effects of CR on Panc02 pancreatic tumor growth are associated with reduced IGF-1-dependent NF-κB activation.
Collapse
Affiliation(s)
- Alison E. Harvey
- Department of Nutritional Sciences, University of Texas, Austin, Austin, Texas, United States of America
| | - Laura M. Lashinger
- Department of Nutritional Sciences, University of Texas, Austin, Austin, Texas, United States of America
| | - Drew Hays
- Department of Nutritional Sciences, University of Texas, Austin, Austin, Texas, United States of America
| | - Lauren M. Harrison
- Department of Nutritional Sciences, University of Texas, Austin, Austin, Texas, United States of America
| | - Kimberly Lewis
- Department of Nutritional Sciences, University of Texas, Austin, Austin, Texas, United States of America
| | - Susan M. Fischer
- Department of Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Smithville, Texas, United States of America
| | - Stephen D. Hursting
- Department of Nutritional Sciences, University of Texas, Austin, Austin, Texas, United States of America
- Department of Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Smithville, Texas, United States of America
- * E-mail:
| |
Collapse
|
19
|
Olivo-Marston SE, Hursting SD, Perkins SN, Schetter A, Khan M, Croce C, Harris CC, Lavigne J. Effects of calorie restriction and diet-induced obesity on murine colon carcinogenesis, growth and inflammatory factors, and microRNA expression. PLoS One 2014; 9:e94765. [PMID: 24732966 PMCID: PMC3986228 DOI: 10.1371/journal.pone.0094765] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 05/28/2013] [Indexed: 12/17/2022] Open
Abstract
Obesity is an established colon cancer risk factor, while preventing or reversing obesity via a calorie restriction (CR) diet regimen decreases colon cancer risk. Unfortunately, the biological mechanisms underlying these associations are poorly understood, hampering development of mechanism-based approaches for preventing obesity-related colon cancer. We tested the hypotheses that diet-induced obesity (DIO) would increase (and CR would decrease) colon tumorigenesis in the mouse azoxymethane (AOM) model. In addition, we established that changes in inflammatory cytokines, growth factors, and microRNAs are associated with these energy balance-colon cancer links, and thus represent mechanism-based targets for colon cancer prevention. Mice were injected with AOM once a week for 5 weeks and randomized to: 1) control diet; 2) 30% CR diet; or 3) DIO diet. Mice were euthanized at week 5 (n = 12/group), 10 (n = 12/group), and 20 (n = 20/group) after the last AOM injection. Colon tumors were counted, and cytokines, insulin-like growth factor 1 (IGF-1), IGF binding protein 3 (IGFBP-3), adipokines, proliferation, apoptosis, and expression of microRNAs (miRs) were measured. The DIO diet regimen induced an obese phenotype (∼36% body fat), while CR induced a lean phenotype (∼14% body fat); controls were intermediate (∼26% body fat). Relative to controls, DIO increased (and CR decreased) the number of colon tumors (p = 0.01), cytokines (p<0.001), IGF-1 (p = 0.01), and proliferation (p<0.001). DIO decreased (and CR increased) IGFBP-3 and apoptosis (p<0.001). miRs including mir-425, mir-196, mir-155, mir-150, mir-351, mir-16, let-7, mir34, and mir-138 were differentially expressed between the dietary groups. We conclude that the enhancing effects of DIO and suppressive effects of CR on colon carcinogenesis are associated with alterations in several biological pathways, including inflammation, IGF-1, and microRNAs.
Collapse
Affiliation(s)
- Susan E. Olivo-Marston
- Laboratory of Human Carcinogenesis, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland, United States of America
- Division of Epidemioogy, The Ohio State University College of Public Health, Columbus, Ohio, United States of America
- * E-mail:
| | - Stephen D. Hursting
- Department of Nutritional Sciences, University of Texas-Austin, Austin, Texas, United States of America
- Department of Molecular Carcinogenesis, University of Texas-MD Anderson Cancer Center, Smithville, Texas, United States of America
| | - Susan N. Perkins
- Center for Cancer Training, The National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Aaron Schetter
- Laboratory of Human Carcinogenesis, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Mohammed Khan
- Laboratory of Human Carcinogenesis, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Carlo Croce
- Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University College of Medicine, Columbus, Ohio, United States of America
| | - Curtis C. Harris
- Laboratory of Human Carcinogenesis, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jackie Lavigne
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, Maryland, United States of America
| |
Collapse
|
20
|
Lashinger LM, Ford NA, Hursting SD. Interacting inflammatory and growth factor signals underlie the obesity-cancer link. J Nutr 2014; 144:109-13. [PMID: 24285690 PMCID: PMC3901418 DOI: 10.3945/jn.113.178533] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The prevalence of obesity, an established risk factor for many chronic diseases (including diabetes, cardiovascular disease, stroke, and several types of cancer), has risen steadily for the past several decades in the United States and many parts of the world. Today, ∼70% of U.S. adults and 30% of children are at an unhealthy weight. The evidence on key biologic mechanisms underlying the obesity-cancer link, with an emphasis on local and systemic inflammatory processes and their crosstalk with energy-sensing growth factor signaling pathways, will be discussed. Understanding the influence and underlying mechanisms of obesity on chronic inflammation and cancer will identify promising mechanistic targets and strategies for disrupting the obesity-cancer link and provide important lessons regarding the associations between obesity, inflammation, and other chronic diseases.
Collapse
Affiliation(s)
- Laura M. Lashinger
- Department of Nutritional Sciences, University of Texas, Austin, TX; and
| | - Nikki A. Ford
- Department of Nutritional Sciences, University of Texas, Austin, TX; and
| | - Stephen D. Hursting
- Department of Nutritional Sciences, University of Texas, Austin, TX; and,Department of Molecular Carcinogenesis, University of Texas–MD Anderson Cancer Center, Smithville, TX,To whom correspondence should be addressed. E-mail:
| |
Collapse
|
21
|
Limesand KH, Chibly AM, Fribley A. Impact of targeting insulin-like growth factor signaling in head and neck cancers. Growth Horm IGF Res 2013; 23:135-140. [PMID: 23816396 PMCID: PMC3755042 DOI: 10.1016/j.ghir.2013.06.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Revised: 06/04/2013] [Accepted: 06/10/2013] [Indexed: 12/23/2022]
Abstract
The IGF system has been shown to have either negative or negligible impact on clinical outcomes of tumor development depending on specific tumor sites or stages. This review focuses on the clinical impact of IGF signaling in head and neck cancer, the effects of IGF targeted therapies, and the multi-dimensional role of IRS 1/2 signaling as a potential mechanism in resistance to targeted therapies. Similar to other tumor sites, both negative and positive correlations between levels of IGF-1/IGF-1-R and clinical outcomes in head and neck cancer have been reported. In addition, utilization of IGF targeted therapies has not demonstrated significant clinical benefit; therefore the prognostic impact of the IGF system on head and neck cancer remains uncertain.
Collapse
Affiliation(s)
- Kirsten H Limesand
- Department of Nutritional Sciences, University of Arizona, Tucson, AZ 85721, USA.
| | | | | |
Collapse
|
22
|
Poh TW, Madsen CS, Gorman JE, Marler RJ, Leighton JA, Cohen PA, Gendler SJ. Downregulation of hematopoietic MUC1 during experimental colitis increases tumor-promoting myeloid-derived suppressor cells. Clin Cancer Res 2013; 19:5039-52. [PMID: 23873692 DOI: 10.1158/1078-0432.ccr-13-0278] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
PURPOSE MUC1 is a tumor-associated antigen that is aberrantly expressed in cancer and inflammatory bowel disease (IBD). Even though immune cells express low MUC1 levels, their modulations of MUC1 are important in tumor progression. Consistent with previous clinical data that show increased myeloid-derived suppressor cells (MDSCs) in IBD, we now show that downregulation of MUC1 on hematopoietic cells increases MDSCs in IBD, similar to our data in tumor-bearing mice. We hypothesize that MDSC expansion in IBD is critical for tumor progression. EXPERIMENTAL DESIGN To mechanistically confirm the linkage between Muc1 downregulation and MDSC expansion, we generated chimeric mice that did not express Muc1 in the hematopoietic compartment (KO→WT). These mice were used in two models of colitis and colitis-associated cancer (CAC) and their responses were compared with wild-type (WT) chimeras (WT→WT). RESULTS KO→WT mice show increased levels of MDSCs during colitis and increased protumorigenic signaling in the colon during CAC, resulting in larger colon tumors. RNA and protein analysis show increased upregulation of metalloproteinases, collagenases, defensins, complements, growth factors, cytokines, and chemokines in KO→WT mice as compared with WT→WT mice. Antibody-mediated depletion of MDSCs in mice during colitis reduced colon tumor formation during CAC. CONCLUSION Development of CAC is a serious complication of colitis and our data highlight MDSCs as a targetable link between inflammation and cancer. In addition, the lack of MUC1 expression on MDSCs can be a novel marker for MDSCs, given that MDSCs are still not well characterized in human cancers.
Collapse
Affiliation(s)
- Tze Wei Poh
- Authors' Affiliations: Departments of Biochemistry and Molecular Biology, Comparative Medicine, Gastroenterology and Hepatology, and Immunology, Mayo Clinic Arizona, Scottsdale, Arizona
| | | | | | | | | | | | | |
Collapse
|
23
|
Ford NA, Nunez NP, Holcomb VB, Hursting SD. IGF1 dependence of dietary energy balance effects on murine Met1 mammary tumor progression, epithelial-to-mesenchymal transition, and chemokine expression. Endocr Relat Cancer 2013; 20:39-51. [PMID: 23152442 DOI: 10.1530/erc-12-0329] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Luminal breast tumors with little or no estrogen receptor α expression confer poor prognosis. Using the Met1 murine model of luminal breast cancer, we characterized the IGF1-dependency of diet-induced obesity (DIO) and calorie restriction (CR) effects on tumor growth, growth factor signaling, epithelial-to-mesenchymal transition (EMT), and chemokine expression. Liver-specific IGF1-deficient (LID) and littermate control (LC) mice were administered control, DIO, or 30% CR diets for 3 months before orthotopic injection of Met1 cells. Tumors grew for 1 month and then were assessed for Akt pathway activation and mRNA expression of chemokine and EMT constituents. LID mice, regardless of diet, displayed reduced Met1 tumor growth and downregulated Akt, EMT, and chemokine pathways. CR, relative to control, reduced serum IGF1 and Met1 tumor growth in LC (but not LID) mice. DIO, relative to control, increased Met1 tumor growth and chemokine expression in LID mice, and had no effect on serum IGF1 or pAkt or cyclin D1 expression in either genotype. Thus, circulating IGF1 (in association with Akt, EMT, and chemokines) regulated Met1 tumor growth. While the anticancer effects of CR were largely IGF1-dependent, the procancer effects of DIO manifested only when circulating IGF1 levels were low. Thus, in a murine model of luminal breast cancer, IGF1 and its downstream signaling pathway, EMT, and chemokines present possible mechanistic regulatory targets. Transplanted MMTV1 Wnt1 mammary tumor growth was also reduced in LID mice, relative to LC mice, suggesting that the IGF1 effects on mammary tumor growth are not limited to Met1 tumors.
Collapse
Affiliation(s)
- Nikki A Ford
- Department of Nutritional Sciences, Dell Pediatric Research Institute, University of Texas, Austin, 78722, USA
| | | | | | | |
Collapse
|
24
|
Abstract
Obesity, a growing health problem worldwide, has been associated with the metabolic syndrome, diabetes, cardiovascular disease, hypertension, and other chronic diseases. Recently, the obesity–cancer link has received much attention. Epidemiological studies have shown that obesity is also associated with increased risk of several cancer types, including colon, breast, endometrium, liver, kidney, esophagus, gastric, pancreatic, gallbladder, and leukemia, and can also lead to poorer treatment and increased cancer-related mortality. Biological mechanisms underlying the relationship between obesity and cancer are not well understood. They include modulation of energy balance and calorie restriction, growth factors, multiple signaling pathways, and inflammatory processes. Key among the signaling pathways linking obesity and cancer is the PI3K/Akt/mTOR cascade, which is a target of many of the obesity-associated factors and regulates cell proliferation and survival. Understanding the molecular and cellular mechanisms of the obesity–cancer connection is important in developing potential therapeutics. The link between obesity and cancer underscores the recommendation to maintain a healthy body weight throughout life as one of the most important ways to protect against cancer.
Collapse
Affiliation(s)
- Ivana Vucenik
- Department of Medical and Research Technology, University of Maryland School of Medicine, Baltimore, Maryland, USA.
| | | |
Collapse
|
25
|
Nogueira LM, Lavigne JA, Chandramouli GVR, Lui H, Barrett JC, Hursting SD. Dose-dependent effects of calorie restriction on gene expression, metabolism, and tumor progression are partially mediated by insulin-like growth factor-1. Cancer Med 2012; 1:275-88. [PMID: 23342276 PMCID: PMC3544443 DOI: 10.1002/cam4.23] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Revised: 06/18/2012] [Accepted: 07/05/2012] [Indexed: 01/19/2023] Open
Abstract
The prevalence of obesity, an established risk and progression factor for breast and many other cancer types, remains very high in the United States and throughout the world. Calorie restriction (CR), a reduced-calorie dietary regimen typically involving a 20-40% reduction in calorie consumption, prevents or reverses obesity, and inhibits mammary and other types of cancer in multiple tumor model systems. Unfortunately, the mechanisms underlying the tumor inhibitory effects of CR are poorly understood, and a better understanding of these mechanisms may lead to new intervention targets and strategies for preventing or controlling cancer. We have previously shown that the anticancer effects of CR are associated with decreased systemic levels of insulin-like growth factor-1 (IGF-1), the primary source of which is liver. We have also reported that CR strongly suppresses tumor development and growth in multiple mammary cancer models. To identify CR-responsive genes and pathways, and to further characterize the role of IGF-1 as a mediator of the anticancer effects of CR, we assessed hepatic and mammary gland gene expression, hormone levels and growth of orthotopically transplanted mammary tumors in control and CR mice with and without exogenous IGF-1. C57BL/6 mice were fed either control AIN-76A diet ad libitum (AL), subjected to 20%, 30%, or 40% CR plus placebo timed-release pellets, or subjected to 30% or 40% CR plus timed-release pellets delivering murine IGF-1 (mIGF-1, 20 μg/day). Compared with AL-fed controls, body weights were decreased 14.3% in the 20% CR group, 18.5% in the 30% CR group, and 38% in the 40% CR group; IGF-1 infusion had no effect on body weight. Hepatic transcriptome analyses indicated that compared with 20% CR, 30% CR significantly modulated more than twice the number of genes and 40% CR more than seven times the number of genes. Many of the genes specific to the 40% CR regimen were hepatic stress-related and/or DNA damage-related genes. Exogenous IGF-1 rescued the hepatic expression of several metabolic genes and pathways affected by CR. Exogenous IGF-1 also rescued the expression of several metabolism- and cancer-related genes affected by CR in the mammary gland. Furthermore, exogenous IGF-1 partially reversed the mammary tumor inhibitory effects of 30% CR. We conclude that several genes and pathways, particularly those associated with macronutrient and steroid hormone metabolism, are associated with the anticancer effects of CR, and that reduced IGF-1 levels can account, at least in part, for many of the effects of CR on gene expression and mammary tumor burden.
Collapse
Affiliation(s)
- Leticia M Nogueira
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, Maryland, 20892 USA
| | | | | | | | | | | |
Collapse
|
26
|
Harvey AE, Lashinger LM, Otto G, Nunez NP, Hursting SD. Decreased systemic IGF-1 in response to calorie restriction modulates murine tumor cell growth, nuclear factor-κB activation, and inflammation-related gene expression. Mol Carcinog 2012; 52:997-1006. [PMID: 22778026 DOI: 10.1002/mc.21940] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Revised: 06/01/2012] [Accepted: 06/04/2012] [Indexed: 12/15/2022]
Abstract
Calorie restriction (CR) prevents obesity and has potent anticancer effects associated with altered hormones and cytokines. We tested the hypothesis that CR inhibits MC38 mouse colon tumor cell growth through modulation of hormone-stimulated nuclear factor (NF)-κB activation and protumorigenic gene expression. Female C57BL/6 mice were randomized (n = 30/group) to receive control diet or 30% CR diet. At 20 wk, 15 mice/group were killed for body composition analysis. At 21 wk, serum was obtained for hormone analysis. At 22 wk, mice were injected with MC38 cells; tumor growth was monitored for 24 d. Gene expression in excised tumors and MC38 cells was analyzed using real-time RT-PCR. In vitro MC38 NF-κB activation (by p65 ELISA and immunofluorescence) were measured in response to varying IGF-1 concentrations (1-400 ng/mL). Relative to controls, CR mice had decreased tumor volume, body weight, body fat, serum IGF-1, serum leptin, and serum insulin, and increased serum adiponectin (P < 0.05, each). Tumors from CR mice, versus controls, had downregulated inflammation- and/or cancer-related gene expression, including interleukin (IL)-6, IL-1β, tumor necrosis factor-α, cyclooxygenase-2, chemokine (C-C motif) ligand-2, S100A9, and F4/80, and upregulated 15-hydroxyprostaglandin dehydrogenase expression. In MC38 cells in vitro, IGF-1 increased NF-κB activation and NF-κB downstream gene expression (P < 0.05, each). We conclude that CR, in association with reduced systemic IGF-1, modulates MC38 tumor growth, NF-κB activation, and inflammation-related gene expression. Thus, IGF-1 and/or NF-κB inhibition may pharmacologically mimic the anticancer effects of CR to break the obesity-colon cancer link.
Collapse
Affiliation(s)
- Alison E Harvey
- Department of Nutritional Sciences, University of Texas at Austin, Austin, Texas
| | | | | | | | | |
Collapse
|
27
|
Doyle SL, Donohoe CL, Finn SP, Howard JM, Lithander FE, Reynolds JV, Pidgeon GP, Lysaght J. IGF-1 and its receptor in esophageal cancer: association with adenocarcinoma and visceral obesity. Am J Gastroenterol 2012; 107:196-204. [PMID: 22146489 DOI: 10.1038/ajg.2011.417] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVES The insulin-like growth factor (IGF) pathway and visceral obesity have been independently linked with esophageal cancer. This study aimed to delineate the differential and interlinked role of visceral obesity and the IGF-1 system in esophageal adenocarcinoma and esophageal squamous-cell carcinoma (SCC). METHODS IGF-1 receptor (IGF-1R) mRNA and protein were examined in esophageal SCC (KYSE 410, OE21) and esophageal adenocarcinoma (OE19, OE33) cell lines by western blotting. Tumor cell proliferation in response to IGF-1 was assessed by bromodeoxyuridine incorporation assay. In esophageal tumor sections, expression of IGF-1R and CD68(+) cell numbers were assessed by immunohistochemistry. IGF-1 was measured in serum from esophageal cancer patients, Barrett's esophagus patients, and healthy controls by enzyme-linked immunosorbent assay. RESULTS Higher IGF-1R protein expressions were observed in SCC cells compared with esophageal adenocarcinoma cells however only adenocarcinoma cell lines significantly increased proliferation in response to IGF-1 (P<0.01). Serum IGF-1 levels were highest in esophageal adenocarcinoma patients (P<0.01) and higher in viscerally obese vs. nonobese (P<0.05) patients. In resected esophageal cancer, increased expression of IGF-1R was observed in the tumor and invasive edge compared with tumor-associated stroma (P<0.05), which coincided with increased CD68(+) cells in stromal tissue surrounding invasive tumor edge (P<0.01). CONCLUSIONS This novel study examined the differential role of the IGF system in esophageal adenocarcinoma and SCC, and its association with visceral obesity. These results indicate that the IGF-1 axis has a key role in malignant progression of esophageal cancer, and represents a plausible mechanism through which visceral obesity impacts on esophageal adenocarcinoma risk and tumor biology.
Collapse
Affiliation(s)
- Suzanne L Doyle
- Department of Surgery, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St James's Hospital and University of Dublin, Trinity College, Dublin, Ireland
| | | | | | | | | | | | | | | |
Collapse
|
28
|
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
|
29
|
Abstract
This paper presents emerging evidence linking visceral adiposity and the metabolic syndrome (MetSyn) with carcinogenesis. The link between obesity and cancer has been clearly identified in a multitude of robust epidemiological studies. Research is now focusing on the role of visceral adipose tissue in carcinogenesis; as it is recognised as an important metabolic tissue that secretes factors that systemically alter the immunological, metabolic and endocrine milieu. Excess visceral adipose tissue gives rise to a state of chronic systemic inflammation with associated insulin resistance and dysmetabolism, collectively known as the MetSyn. Prospective cohort studies have shown associations between visceral adiposity, the MetSyn and increased risk of breast cancer, colorectal cancer and oesophageal adenocarcinoma. Furthermore, visceral adiposity and the MetSyn have been associated with increased tumour progression and reduced survival. The mechanisms by which visceral adiposity and the MetSyn are thought to promote tumorigenesis are manifold. These include alterations in adipokine secretion and cell signalling pathways. In addition, hyperinsulinaemia, subsequent insulin resistance and stimulation of the insulin-like growth factor-1 axis have all been linked with visceral adiposity and promote tumour progression. Furthermore, the abundance of inflammatory cells in visceral adipose tissue, including macrophages and T-cells, create systemic inflammation and a pro-tumorigenic environment. It is clear from current research that excess visceral adiposity and associated dysmetabolism play a central role in the pathogenesis of certain cancer types. Further research is required to elucidate the exact mechanisms at play and identify potential targets for intervention.
Collapse
|
30
|
Harvey AE, Lashinger LM, Hursting SD. The growing challenge of obesity and cancer: an inflammatory issue. Ann N Y Acad Sci 2011; 1229:45-52. [PMID: 21793838 DOI: 10.1111/j.1749-6632.2011.06096.x] [Citation(s) in RCA: 186] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The prevalence of obesity, an established risk factor for many cancers, has risen steadily for the past several decades in the United States and in many parts of the world. This review synthesizes the evidence on key biological mechanisms underlying the obesity-cancer link, with particular emphasis on the impact of energy balance modulation, such as diet-induced obesity and calorie restriction, on growth factor signaling pathways and inflammatory processes. Particular attention is placed on the proinflammatory environment associated with the obese state, specifically highlighting the involvement of obesity-associated hormones/growth factors in crosstalk between macrophages, adipocytes, and epithelial cells in many cancers. Understanding the contribution of obesity to growth factor signaling and chronic inflammation provides mechanistic targets for disrupting the obesity-cancer link.
Collapse
Affiliation(s)
- Alison E Harvey
- Department of Nutritional Sciences, University of Texas, Austin, Texas, USA
| | | | | |
Collapse
|
31
|
Abstract
Obesity is associated with increased risk and worsened prognosis for postmenopausal breast cancer, but the underlying mechanisms remain unclear. Elegant work reported by Subbaramaiah and colleagues in this issue of the journal (beginning on page 329) adds important insights, particularly into the connections between obesity, inflammation, and aromatase via cross-talk among adipocytes, macrophages, and epithelial cells. This work provides several new molecular targets and strategies to test in model systems for preventing or controlling obesity-related breast cancer and provides a framework for studying the linkages among the complex mechanistic pathways underlying the obesity-cancer relationship.
Collapse
Affiliation(s)
- Stephen D Hursting
- Department of Nutritional Sciences, University of Texas at Austin, Dell Pediatric Research Institute, Austin, TX 78723, USA.
| |
Collapse
|
32
|
Lashinger LM, Malone LM, McArthur MJ, Goldberg JA, Daniels EA, Pavone A, Colby JK, Smith NC, Perkins SN, Fischer SM, Hursting SD. Genetic reduction of insulin-like growth factor-1 mimics the anticancer effects of calorie restriction on cyclooxygenase-2-driven pancreatic neoplasia. Cancer Prev Res (Phila) 2011; 4:1030-40. [PMID: 21593196 DOI: 10.1158/1940-6207.capr-11-0027] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Risk of pancreatic cancer, the fourth deadliest cancer in the United States, is increased by obesity. Calorie restriction (CR) prevents obesity, suppresses carcinogenesis in many models, and reduces serum levels of IGF-1. In the present study, we examined the impact of CR on a model of inflammation-associated pancreatitis and pancreatic dysplasia, with a focus on the mechanistic contribution of systemic IGF-1. Administration of a 30% CR diet for 14 weeks decreased serum IGF-1 levels and hindered pancreatic ductal lesion formation and dysplastic severity, relative to a higher calorie control diet, in transgenic mice overexpressing COX-2 [bovine keratin-5 promoter (BK5.COX-2)]. These findings in CR mice correlated with reductions in Ki-67-positive cells, vascular luminal size, VEGF expression, and phosphorylation and total expression of downstream mediators of the IGF-1 pathway. Cell lines derived from BK5.COX-2 ductal lesions (JC101 cells) formed pancreatic tumors in wild-type FVB mice that were significantly reduced in size by a 14-week CR regimen, relative to the control diet. To further understand the impact of circulating levels of IGF-1 on tumor growth in this model, we orthotopically injected JC101 cells into liver-specific IGF-1-deficient (LID) mice. The approximate 65% reduction of serum IGF-1 levels in LID mice resulted in significantly decreased burden of JC101 tumors, despite modestly elevated levels of circulating insulin and leptin. These data show that CR prevents development of dysplasia and growth of pancreatic cancer through alterations in IGF-1, suggesting that modulation of this pathway with dietary and/or pharmacologic interventions is a promising pancreatic cancer prevention strategy.
Collapse
Affiliation(s)
- Laura M Lashinger
- Department of Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Smithville, Texas, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Migliore L, Migheli F, Spisni R, Coppedè F. Genetics, cytogenetics, and epigenetics of colorectal cancer. J Biomed Biotechnol 2011; 2011:792362. [PMID: 21490705 PMCID: PMC3070260 DOI: 10.1155/2011/792362] [Citation(s) in RCA: 172] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2010] [Accepted: 12/14/2010] [Indexed: 12/17/2022] Open
Abstract
Most of the colorectal cancer (CRC) cases are sporadic, only 25% of the patients have a family history of the disease, and major genes causing syndromes predisposing to CRC only account for 5-6% of the total cases. The following subtypes can be recognized: MIN (microsatellite instability), CIN (chromosomal instability), and CIMP (CpG island methylator phenotype). CIN occurs in 80-85% of CRC. Chromosomal instability proceeds through two major mechanisms, missegregation that results in aneuploidy through the gain or loss of whole chromosomes, and unbalanced structural rearrangements that lead to the loss and/or gain of chromosomal regions. The loss of heterozygosity that occur in the first phases of the CRC cancerogenesis (in particular for the genes on 18q) as well as the alteration of methylation pattern of multiple key genes can drive the development of colorectal cancer by facilitating the acquisition of multiple tumor-associated mutations and the instability phenotype.
Collapse
Affiliation(s)
- Lucia Migliore
- Department of Human and Environmental Sciences, University of Pisa, Street S. Giuseppe 22, 56126 Pisa, Italy.
| | | | | | | |
Collapse
|
34
|
Abstract
A substantial body of evidence supports a role for the growth hormone (GH)-IGF-1 axis in cancer incidence and progression. This includes epidemiological evidence relating elevated plasma IGF-1 to cancer incidence as well as a lack of cancers in GH/IGF-1 deficiency. Rodent models lacking GH or its receptor are strikingly resistant to the induction of a wide range of cancers, and treatment with the GH antagonist pegvisomant slows tumor progression. While GH receptor expression is elevated in many cancers, autocrine GH is present in several types, and overexpression of autocrine GH can induce cell transformation. While the mechanism of autocrine action is not clear, it does involve both STAT5 and STAT3 activation, and probably nuclear translocation of the GH receptor. Development of a more potent GH receptor antagonist or secretion inhibitor is warranted for cancer therapy.
Collapse
Affiliation(s)
- Yash Chhabra
- a The University of Queensland, Institute for Molecular Bioscience, Brisbane, Qld 4072, Australia
| | - Michael J Waters
- a The University of Queensland, Institute for Molecular Bioscience, Brisbane, Qld 4072, Australia
- b
| | - Andrew J Brooks
- a The University of Queensland, Institute for Molecular Bioscience, Brisbane, Qld 4072, Australia
| |
Collapse
|
35
|
Clayton PE, Banerjee I, Murray PG, Renehan AG. Growth hormone, the insulin-like growth factor axis, insulin and cancer risk. Nat Rev Endocrinol 2011; 7:11-24. [PMID: 20956999 DOI: 10.1038/nrendo.2010.171] [Citation(s) in RCA: 249] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Growth hormone (GH), insulin-like growth factor (IGF)-I and insulin have potent growth-promoting and anabolic actions. Their potential involvement in tumor promotion and progression has been of concern for several decades. The evidence that GH, IGF-I and insulin can promote and contribute to cancer progression comes from various sources, including transgenic and knockout mouse models and animal and human cell lines derived from cancers. Assessments of the GH-IGF axis in healthy individuals followed up to assess cancer incidence provide direct evidence of this risk; raised IGF-I levels in blood are associated with a slightly increased risk of some cancers. Studies of human diseases characterized by excess growth factor secretion or treated with growth factors have produced reassuring data, with no notable increases in de novo cancers in children treated with GH. Although follow-up for the vast majority of these children does not yet extend beyond young adulthood, a slight increase in cancers in those with long-standing excess GH secretion (as seen in patients with acromegaly) and no overall increase in cancer with insulin treatment, have been observed. Nevertheless, long-term surveillance for cancer incidence in all populations exposed to increased levels of GH is vitally important.
Collapse
Affiliation(s)
- Peter E Clayton
- Manchester Academic Health Sciences Centre, University of Manchester, Paediatric Endocrinology, Royal Manchester Children's Hospital, Oxford Road, Manchester, UK.
| | | | | | | |
Collapse
|
36
|
Hursting SD, Berger NA. Energy balance, host-related factors, and cancer progression. J Clin Oncol 2010; 28:4058-65. [PMID: 20697088 DOI: 10.1200/jco.2010.27.9935] [Citation(s) in RCA: 186] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Obesity is associated with an increased risk and worsened prognosis for many types of cancer, but the mechanisms underlying the obesity-cancer progression link are poorly understood. Several energy balance-related host factors are known to influence tumor progression and/or treatment responsiveness after cancer develops, and these have been implicated as key contributors to the complex effects of obesity on cancer outcome. These host factors include leptin, adiponectin, steroid hormones, reactive oxygen species associated with inflammation, insulin, insulin-like growth factor-1, and sirtuins. Each of these host factors is considered in this article in the context of energy balance and cancer progression. In addition, future research directions in this field are discussed, including the importance of study designs addressing energy balance across the life course, the development and application of highly relevant animal models, potential roles of cancer stem cells in the response to energy balance modulation, and emerging pharmacologic approaches that target energy balance-related pathways.
Collapse
Affiliation(s)
- Stephen D Hursting
- The University of Texas, 1 University Station A2700, Painter Hall, Room 5.32, Austin, TX 78712, USA
| | | |
Collapse
|