1
|
Grigorescu RR, Husar-Sburlan IA, Gheorghe C. Pancreatic Cancer: A Review of Risk Factors. Life (Basel) 2024; 14:980. [PMID: 39202722 PMCID: PMC11355429 DOI: 10.3390/life14080980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/28/2024] [Accepted: 08/01/2024] [Indexed: 09/03/2024] Open
Abstract
Pancreatic adenocarcinoma is one of the most lethal types of gastrointestinal cancer despite the latest medical advances. Its incidence has continuously increased in recent years in developed countries. The location of the pancreas can result in the initial symptoms of neoplasia being overlooked, which can lead to a delayed diagnosis and a subsequent reduction in the spectrum of available therapeutic options. The role of modifiable risk factors in pancreatic cancer has been extensively studied in recent years, with smoking and alcohol consumption identified as key contributors. However, the few screening programs that have been developed focus exclusively on genetic factors, without considering the potential impact of modifiable factors on disease occurrence. Thus, fully understanding and detecting the risk factors for pancreatic cancer represents an important step in the prevention and early diagnosis of this type of neoplasia. This review reports the available evidence on different risk factors and identifies the areas that could benefit the most from additional studies.
Collapse
Affiliation(s)
- Raluca Roxana Grigorescu
- Gastroenterology Department, “Sfanta Maria” Hospital, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | | | - Cristian Gheorghe
- Center for Digestive Disease and Liver Transplantation, Fundeni Clinical Institute, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| |
Collapse
|
2
|
Zeng L, Wu Z, Yang J, Zhou Y, Chen R. Association of genetic risk and lifestyle with pancreatic cancer and their age dependency: a large prospective cohort study in the UK Biobank. BMC Med 2023; 21:489. [PMID: 38066552 PMCID: PMC10709905 DOI: 10.1186/s12916-023-03202-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 11/29/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Pancreatic cancer (PC) is influenced by both genetic and lifestyle factors. However, further research is still needed to comprehensively clarify the relationships among lifestyle, genetic factors, their combined effect on PC, and how these associations might be age-dependent. METHODS We included 340,631 participants from the UK Biobank. Three polygenic risk score (PRS) models for PC were applied, which were derived from the previous study and were categorized as low, intermediate, and high. Two healthy lifestyle scores (HLSs) were constructed using 9 lifestyle factors based on the World Cancer Research Fund/American Institute of Cancer Research (WCRF/AICR) lifestyle score and the American Cancer Society (ACS) guidelines and were categorized as unfavorable, intermediate, and favorable. Data were analyzed using Cox proportional hazards models. RESULTS There were 1,129 cases of incident PC during a median follow-up of 13.05 years. Higher PRS was significantly associated with an increased risk of PC (hazard ratio [HR], 1.58; 95% confidence intervals [CI], 1.47-1.71). Adhering to a favorable lifestyle was associated with a lower risk (HR, 0.48; 95% CI, 0.41-0.56). Participants with an unfavorable lifestyle and a high PRS had the highest risk of PC (HR, 2.84; 95% CI, 2.22-3.62). Additionally, when stratified by age, a favorable lifestyle was most pronounced associated with a lower risk of PC among participants aged ≤ 60 years (HR, 0.35; 95% CI, 0.23-0.54). However, the absolute risk reduction was more pronounced among those aged > 70 years (ARR, 0.19%, 95% CI, 0.13%-0.26%). A high PRS was more strongly associated with PC among participants aged ≤ 60 years (HR, 1.89; 95% CI, 1.30-2.73). Furthermore, we observed a significant multiplicative interaction and several significant additive interactions. CONCLUSIONS A healthy lifestyle was associated with a lower risk of PC, regardless of the participants' age, sex, or genetic risk. Importantly, our findings indicated the age-dependent association of lifestyle and genetic factors with PC, emphasizing the importance of early adoption for effective prevention and potentially providing invaluable guidance for setting the optimal age to start preventive measures.
Collapse
Affiliation(s)
- Liangtang Zeng
- School of Medicine, South China University of Technology, Guangzhou, Guangdong Province, China
- Department of Pancreatic Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
| | - Zhuo Wu
- School of Medicine, South China University of Technology, Guangzhou, Guangdong Province, China
- Department of Pancreatic Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
| | - Jiabin Yang
- School of Medicine, South China University of Technology, Guangzhou, Guangdong Province, China
- Department of Pancreatic Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
| | - Yu Zhou
- Department of Pancreatic Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China.
| | - Rufu Chen
- School of Medicine, South China University of Technology, Guangzhou, Guangdong Province, China.
- Department of Pancreatic Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
3
|
Kozłowska M, Śliwińska A. The Link between Diabetes, Pancreatic Tumors, and miRNAs-New Players for Diagnosis and Therapy? Int J Mol Sci 2023; 24:10252. [PMID: 37373398 DOI: 10.3390/ijms241210252] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/12/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Despite significant progress in medicine, pancreatic cancer is one of the most tardily diagnosed cancer and is consequently associated with a poor prognosis and a low survival rate. The asymptomatic clinical picture and the lack of relevant diagnostic markers for the early stages of pancreatic cancer are believed to be the major constraints behind an accurate diagnosis of this disease. Furthermore, underlying mechanisms of pancreatic cancer development are still poorly recognized. It is well accepted that diabetes increases the risk of pancreatic cancer development, however the precise mechanisms are weakly investigated. Recent studies are focused on microRNAs as a causative factor of pancreatic cancer. This review aims to provide an overview of the current knowledge of pancreatic cancer and diabetes-associated microRNAs, and their potential in diagnosis and therapy. miR-96, miR-124, miR-21, and miR-10a were identified as promising biomarkers for early pancreatic cancer prediction. miR-26a, miR-101, and miR-200b carry therapeutic potential, as they not only regulate significant biological pathways, including the TGF-β and PI3K/AKT, but their re-expression contributes to the improvement of the prognosis by reducing invasiveness or chemoresistance. In diabetes, there are also changes in the expression of microRNAs, such as in miR-145, miR-29c, and miR-143. These microRNAs are involved, among others, in insulin signaling, including IRS-1 and AKT (miR-145), glucose homeostasis (hsa-miR-21), and glucose reuptake and gluconeogenesis (miR-29c). Although, changes in the expression of the same microRNAs are observed in both pancreatic cancer and diabetes, they exert different molecular effects. For example, miR-181a is upregulated in both pancreatic cancer and diabetes mellitus, but in diabetes it contributes to insulin resistance, whereas in pancreatic cancer it promotes tumor cell migration, respectively. To conclude, dysregulated microRNAs in diabetes affect crucial cellular processes that are involved in pancreatic cancer development and progression.
Collapse
Affiliation(s)
- Małgorzata Kozłowska
- Student Scientific Society of Civilization Diseases, Medical University of Lodz, 251 Pomorska, 92-213 Lodz, Poland
| | - Agnieszka Śliwińska
- Department of Nucleic Acid Biochemistry, Medical University of Lodz, Pomorska 251, 92-213 Lodz, Poland
| |
Collapse
|
4
|
Hatch KS, Gao S, Ma Y, Russo A, Jahanshad N, Thompson PM, Adhikari BM, Bruce H, Van der Vaart A, Sotiras A, Kvarta MD, Nichols TE, Schmaal L, Hong LE, Kochunov P. Brain deficit patterns of metabolic illnesses overlap with those for major depressive disorder: A new metric of brain metabolic disease. Hum Brain Mapp 2023; 44:2636-2653. [PMID: 36799565 PMCID: PMC10028678 DOI: 10.1002/hbm.26235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/30/2023] [Accepted: 02/01/2023] [Indexed: 02/18/2023] Open
Abstract
Metabolic illnesses (MET) are detrimental to brain integrity and are common comorbidities in patients with mental illnesses, including major depressive disorder (MDD). We quantified effects of MET on standard regional brain morphometric measures from 3D brain MRI as well as diffusion MRI in a large sample of UK BioBank participants. The pattern of regional effect sizes of MET in non-psychiatric UKBB subjects was significantly correlated with the spatial profile of regional effects reported by the largest meta-analyses in MDD but not in bipolar disorder, schizophrenia or Alzheimer's disease. We used a regional vulnerability index (RVI) for MET (RVI-MET) to measure individual's brain similarity to the expected patterns in MET in the UK Biobank sample. Subjects with MET showed a higher effect size for RVI-MET than for any of the individual brain measures. We replicated elevation of RVI-MET in a sample of MDD participants with MET versus non-MET. RVI-MET scores were significantly correlated with the volume of white matter hyperintensities, a neurological consequence of MET and age, in both groups. Higher RVI-MET in both samples was associated with obesity, tobacco smoking and frequent alcohol use but was unrelated to antidepressant use. In summary, MET effects on the brain were regionally specific and individual similarity to the pattern was more strongly associated with MET than any regional brain structural metric. Effects of MET overlapped with the reported brain differences in MDD, likely due to higher incidence of MET, smoking and alcohol use in subjects with MDD.
Collapse
Affiliation(s)
- Kathryn S Hatch
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Si Gao
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Yizhou Ma
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Alessandro Russo
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Neda Jahanshad
- Imaging Genetics Center, Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, Marina del Rey, California, USA
| | - Paul M Thompson
- Imaging Genetics Center, Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, Marina del Rey, California, USA
| | - Bhim M Adhikari
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Heather Bruce
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Andrew Van der Vaart
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Aristeidis Sotiras
- Institute of Informatics, University of Washington, School of Medicine, St. Louis, Missouri, USA
- Department of Radiology, University of Washington, School of Medicine, St. Louis, Missouri, USA
| | - Mark D Kvarta
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Thomas E Nichols
- Nuffield Department of Population Health of the University of Oxford, Oxford, UK
| | - Lianne Schmaal
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
- Orygen, Parkville, Australia
| | - L Elliot Hong
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Peter Kochunov
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
5
|
Teglia F, Boffetta P. Association between trends of mortality and incidence, survival and stage at diagnosis for six digestive and respiratory cancers in United States (2009-2013). Eur J Cancer Prev 2023; 32:195-202. [PMID: 35881938 DOI: 10.1097/cej.0000000000000766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND A decrease in cancer mortality has been reported in the USA, possibly due to decreased incidence, downstaging and improved survival. The aim of the present study is to estimate the contribution of these factors on the trend in cancer mortality. METHODS Data on incidence, mortality, stage at diagnosis, and overall and stage-specific survival for six common digestive and respiratory cancers (esophagus, stomach, colorectal, liver, pancreas and lung) during 2009-2013 in the USA from the surveillance, epidemiology and end results (SEER) program, was analyzed using generalized linear models separately among men and women. RESULTS Our study showed a decrease in mortality for esophageal (-0.09/100 000/year and -0.03/100 000/year), stomach (-0.11/100 000/year and -0.05/100 000/year), colorectal (-0.45/100 000/year and -0.29/100 000/year) and lung cancer (-1.89/100 000/year in men and -0.78/100 000/year in women) in men and women, respectively: for all of them, except lung cancer in women, there was a decrease in the incidence of comparable or greater magnitude; stage distribution and survival also contributed to the decrease in mortality for lung and colorectal cancer. Mortality from pancreatic cancer was stable: an increase in incidence was counterbalanced by an improvement in survival. Mortality from liver cancer increased, driven by an increase in mortality that was not offset by favorable trends in stage distribution and survival. CONCLUSIONS Trends in mortality were primarily affected by changes in incidence; an increase in the proportion of local stage at diagnosis and improved survival, although evident for some cancers, played a lesser role in mortality trends.
Collapse
Affiliation(s)
- Federica Teglia
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Paolo Boffetta
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- Department of Family, Population & Preventive Medicine, Stony Brook Cancer Center, Stony Brook University, Stony Brook, New York, USA
| |
Collapse
|
6
|
Ali A, Manzoor MF, Ahmad N, Aadil RM, Qin H, Siddique R, Riaz S, Ahmad A, Korma SA, Khalid W, Aizhong L. The Burden of Cancer, Government Strategic Policies, and Challenges in Pakistan: A Comprehensive Review. Front Nutr 2022; 9:940514. [PMID: 35938114 PMCID: PMC9355152 DOI: 10.3389/fnut.2022.940514] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 06/23/2022] [Indexed: 01/09/2023] Open
Abstract
Cancer is a severe condition characterized by uncontrolled cell division and increasing reported mortality and diagnostic cases. In 2040, an estimated 28.4 million cancer cases are expected to happen globally. In 2020, an estimated 19.3 million new cancer cases (18.1 million excluding non-melanoma skin cancer) had been diagnosed worldwide, with around 10.0 million cancer deaths. Breast cancer cases have increased by 2.26 million, lung cancer by 2.21 million, stomach by 1.089 million, liver by 0.96 million, and colon cancer by 1.93 million. Cancer is becoming more prevalent in Pakistan, with 19 million new cancer cases recorded in 2020. Food adulteration, gutkha, paan, and nutritional deficiencies are major cancer risk factors that interplay with cancer pathogenesis in this country. Government policies and legislation, cancer treatment challenges, and prevention must be revised seriously. This review presents the current cancer epidemiology in Pakistan to better understand cancer basis. It summarizes current cancer risk factors, causes, and the strategies and policies of the country against cancer.
Collapse
Affiliation(s)
- Anwar Ali
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha, China
- Food and Nutrition Society, Gilgit Baltistan, Pakistan
| | | | - Nazir Ahmad
- Department of Nutritional Science, Government College University Faisalabad, Faisalabad, Pakistan
| | - Rana Muhammad Aadil
- National Institute of Food Science and Technology, University of Agriculture, Faisalabad, Pakistan
| | - Hong Qin
- School of Nutrition and Food Hygiene, Xiangya School of Public Health, Central South University, Changsha, China
| | - Rabia Siddique
- Department of Chemistry, Government College University Faisalabad, Faisalabad, Pakistan
| | - Sakhawat Riaz
- Department of Home Economics, Government College University Faisalabad, Faisalabad, Pakistan
| | - Arslan Ahmad
- Department of Home Economics, Government College University Faisalabad, Faisalabad, Pakistan
| | - Sameh A. Korma
- Department of Food Science, Faculty of Agriculture, Zagazig, Egypt
| | - Waseem Khalid
- Department of Food Sciences, Government College University, Faisalabad, Pakistan
| | - Liu Aizhong
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha, China
- *Correspondence: Liu Aizhong
| |
Collapse
|
7
|
Clement EJ, Law HCH, Qiao F, Noe D, Trevino JG, Woods NT. Combined Alcohol Exposure and KRAS Mutation in Human Pancreatic Ductal Epithelial Cells Induces Proliferation and Alters Subtype Signatures Determined by Multi-Omics Analysis. Cancers (Basel) 2022; 14:cancers14081968. [PMID: 35454872 PMCID: PMC9027648 DOI: 10.3390/cancers14081968] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/06/2022] [Accepted: 04/07/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Pancreatic ductal adenocarcinoma is a deadly disease wherein alcohol use increases the risk of developing this cancer. Mutations in the KRAS oncogene are required for alcohol to promote pancreatic cancer in mice, but little is known about the molecular events associated with the combined exposure of alcohol and mutant KRAS expression in pancreas cells. In this study, we use pancreas cell models with and without mutant KRAS to evaluate the impact of chronic alcohol exposure on transcription and protein expression. This study identifies numerous differentially expressed transcripts and proteins that could influence the emergence of oncogenic features, such as increased proliferation, in pancreas cells. Abstract Pancreatic Ductal adenocarcinoma (PDAC) is an aggressive cancer commonly exhibiting KRAS-activating mutations. Alcohol contributes to the risk of developing PDAC in humans, and murine models have shown alcohol consumption in the context of KRAS mutation in the pancreas promotes the development of PDAC. The molecular signatures in pancreas cells altered by alcohol exposure in the context of mutant KRAS could identify pathways related to the etiology of PDAC. In this study, we evaluated the combined effects of alcohol exposure and KRAS mutation status on the transcriptome and proteome of pancreatic HPNE cell models. These analyses identified alterations in transcription and translational processes in mutant KRAS cells exposed to alcohol. In addition, multi-omics analysis suggests an increase in the correlation between mRNA transcript and protein abundance in cells exposed to alcohol with an underlying KRAS mutation. Through differential co-expression, SERPINE1 was found to be influential for PDAC development in the context of mutant KRAS and ethanol. In terms of PDAC subtypes, alcohol conditioning of HPNE cells expressing mutant KRAS decreases the Inflammatory subtype signature and increases the Proliferative and Metabolic signatures, as we previously observed in patient samples. The alterations in molecular subtypes were associated with an increased sensitivity to chemotherapeutic agents gemcitabine, irinotecan, and oxaliplatin. These results provide a framework for distinguishing the molecular dysregulation associated with combined alcohol and mutant KRAS in a pancreatic cell line model.
Collapse
Affiliation(s)
- Emalie J. Clement
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (E.J.C.); (H.C.-H.L.); (F.Q.)
| | - Henry C.-H. Law
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (E.J.C.); (H.C.-H.L.); (F.Q.)
| | - Fangfang Qiao
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (E.J.C.); (H.C.-H.L.); (F.Q.)
| | - Dragana Noe
- Mass Spectrometry and Proteomics Core Facility, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Jose G. Trevino
- Department of Surgery, Virginia Commonwealth University, Richmond, VA 23298, USA;
| | - Nicholas T. Woods
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (E.J.C.); (H.C.-H.L.); (F.Q.)
- Correspondence:
| |
Collapse
|
8
|
Grigor’eva IN. Pancreatic cancer risk: alcoholic and non-alcoholic beverages. TERAPEVT ARKH 2022; 94:265-270. [DOI: 10.26442/00403660.2022.02.201375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 03/29/2022] [Indexed: 11/22/2022]
Abstract
This article provides an overview of the metaanalyzes (PubMed, 19952019) of alcohol and non-alcoholic (coffee, tea, dairy products) beverage consumption in relation to risk of pancreatic cancer PC (PubMed, 19952019). Increased the PC risk was associated with high alcohol intake. The increased risk for heavy drinking did not explained by residual confounding by history of pancreatitis or tobacco smoking or diabetes. Light-moderate alcohol intake may reduced the PC risk, probably due to the fasting insulin levels decrement, which leads to the diminished the РС risk. The association between alcohol and the PC was stronger in men than in women. Some metaanalyzes demonstrated that a small amount of coffee may reduce PC risk, and a large amount to increase PC risk. Another meta-analyzes have not confirmed any association between the PC risk and coffee or tea consumption. One meta-analysis revealed a direct association of the PC risk with the dairy products consumption, but most research showed no such connection. Nutrition is considered to be associated with the PC risk, but the degree of risk due to structure of beverages consumption (dose, duration, alcohol, coffee, tea, dairy products pattern) is still not clear.
Collapse
|
9
|
Petrick JL, Wilkinson JE, Michaud DS, Cai Q, Gerlovin H, Signorello LB, Wolpin BM, Ruiz-Narváez EA, Long J, Yang Y, Johnson WE, Shu XO, Huttenhower C, Palmer JR. The oral microbiome in relation to pancreatic cancer risk in African Americans. Br J Cancer 2022; 126:287-296. [PMID: 34718358 PMCID: PMC8770575 DOI: 10.1038/s41416-021-01578-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 09/14/2021] [Accepted: 10/01/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND African Americans have the highest pancreatic cancer incidence of any racial/ethnic group in the United States. The oral microbiome was associated with pancreatic cancer risk in a recent study, but no such studies have been conducted in African Americans. Poor oral health, which can be a cause or effect of microbial populations, was associated with an increased risk of pancreatic cancer in a single study of African Americans. METHODS We prospectively investigated the oral microbiome in relation to pancreatic cancer risk among 122 African-American pancreatic cancer cases and 354 controls. DNA was extracted from oral wash samples for metagenomic shotgun sequencing. Alpha and beta diversity of the microbial profiles were calculated. Multivariable conditional logistic regression was used to estimate odds ratios (ORs) and 95% confidence intervals (CIs) for associations between microbes and pancreatic cancer risk. RESULTS No associations were observed with alpha or beta diversity, and no individual microbial taxa were differentially abundant between cases and control, after accounting for multiple comparisons. Among never smokers, there were elevated ORs for known oral pathogens: Porphyromonas gingivalis (OR = 1.69, 95% CI: 0.80-3.56), Prevotella intermedia (OR = 1.40, 95% CI: 0.69-2.85), and Tannerella forsythia (OR = 1.36, 95% CI: 0.66-2.77). CONCLUSIONS Previously reported associations between oral taxa and pancreatic cancer were not present in this African-American population overall.
Collapse
Affiliation(s)
| | - Jeremy E Wilkinson
- Department of Biostatistics, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Dominique S Michaud
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA, USA
| | - Qiuyin Cai
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Hanna Gerlovin
- Slone Epidemiology Center, Boston University, Boston, MA, USA
| | - Lisa B Signorello
- Division of Cancer Prevention, National Cancer Institute, Bethesda, MD, USA
| | - Brian M Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Edward A Ruiz-Narváez
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Jirong Long
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Yaohua Yang
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - W Evan Johnson
- Department of Medicine, Division of Computational Biomedicine, Boston University, Boston, MA, USA
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Curtis Huttenhower
- Department of Biostatistics, Harvard TH Chan School of Public Health, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Julie R Palmer
- Slone Epidemiology Center, Boston University, Boston, MA, USA.
| |
Collapse
|
10
|
Koyanagi YN, Oze I, Kasugai Y, Kawakatsu Y, Taniyama Y, Hara K, Shimizu Y, Imoto I, Ito H, Matsuo K. New insights into the genetic contribution of ALDH2 rs671 in pancreatic carcinogenesis: evaluation by mediation analysis. Cancer Sci 2022; 113:1441-1450. [PMID: 35102643 PMCID: PMC8990728 DOI: 10.1111/cas.15286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/12/2022] [Accepted: 01/23/2022] [Indexed: 11/26/2022] Open
Abstract
A functional variant on ALDH2 rs671 (G>A) confers a protective effect against alcohol‐induced carcinogenesis through an indirect pathway mediated by decreased alcohol consumption. Conversely, this variant also contributes to the accumulation of carcinogenic agents, resulting in a direct carcinogenic effect. This study aimed to separately quantify these two opposing effects of the rs671 A allele on pancreatic cancer risk and explore the impact of the rs671 A allele and alcohol consumption on pancreatic carcinogenesis. We included 426 cases and 1456 age‐ and sex‐matched controls. Odds ratio (OR) and 95% confidence interval (CI) for alcohol consumption were estimated using a conditional logistic regression model. By defining rs671 A allele and alcohol consumption as exposure and mediator, respectively, we used mediation analysis to decompose the total‐effect OR of the rs671 A allele into direct‐ and indirect‐effect ORs. Alcohol consumption (10 g/d) was associated with pancreatic cancer risk (OR, 1.05; 95% CI, 1.01‐1.10), but tests for interaction between the rs671 A allele and alcohol consumption were nonsignificant, indicating that the effect of alcohol consumption did not vary by genotype. Mediation analysis showed that the nonsignificant total effect (OR, 1.15; 95% CI, 0.92‐1.44) can be decomposed into the carcinogenic direct (OR, 1.34; 95% CI, 1.04‐1.72) and protective indirect effect (OR, 0.86; 95% CI, 0.77‐0.95). This study supports the association between alcohol consumption and pancreatic cancer risk and indicates the potential contribution of the rs671 A allele to pancreatic carcinogenesis through impaired metabolism of known or unknown ALDH2 substrates.
Collapse
Affiliation(s)
- Yuriko N Koyanagi
- Division of Cancer Information and Control, Department of Preventive Medicine, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Isao Oze
- Division of Cancer Epidemiology and Prevention, Department of Preventive Medicine, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Yumiko Kasugai
- Division of Cancer Epidemiology and Prevention, Department of Preventive Medicine, Aichi Cancer Center Research Institute, Nagoya, Japan.,Department of Cancer Epidemiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yukino Kawakatsu
- Division of Cancer Epidemiology and Prevention, Department of Preventive Medicine, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Yukari Taniyama
- Division of Cancer Information and Control, Department of Preventive Medicine, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Kazuo Hara
- Department of Gastroenterology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Yasuhiro Shimizu
- Department of Gastroenterological Surgery, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Issei Imoto
- Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Hidemi Ito
- Division of Cancer Information and Control, Department of Preventive Medicine, Aichi Cancer Center Research Institute, Nagoya, Japan.,Department of Descriptive Cancer Epidemiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Keitaro Matsuo
- Division of Cancer Epidemiology and Prevention, Department of Preventive Medicine, Aichi Cancer Center Research Institute, Nagoya, Japan.,Department of Cancer Epidemiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
11
|
Yu W, Ma Y, Roy SK, Srivastava R, Shankar S, Srivastava RK. Ethanol exposure of human pancreatic normal ductal epithelial cells induces EMT phenotype and enhances pancreatic cancer development in KC (Pdx1-Cre and LSL-Kras G12D ) mice. J Cell Mol Med 2021; 26:399-409. [PMID: 34859959 PMCID: PMC8743655 DOI: 10.1111/jcmm.17092] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 11/17/2021] [Accepted: 11/19/2021] [Indexed: 12/20/2022] Open
Abstract
Alcohol is a risk factor for pancreatic cancer. However, the molecular mechanism by which chronic alcohol consumption influences pancreatic cancer development is not well understood. We have recently demonstrated that chronic ethanol exposure of pancreatic normal ductal epithelial cells (HPNE) induces cellular transformation by generating cancer stem cells (CSCs). Here, we examined whether chronic ethanol treatment induces epithelial–mesenchymal transition in HPNE cells and promotes pancreatic cancer development in KC (Pdx1‐Cre, and LSL‐KrasG12D) mice. Our data demonstrate that chronic ethanol exposure of HPNE cells induces SATB2 gene and those cells became highly motile. Ethanol treatment of HPNE cells results in downregulation of E‐Cadherin and upregulation of N‐Cadherin, Snail, Slug, Zeb1, Nanog and BMI‐1. Suppression of SATB2 expression in ethanol‐transformed HPNE cells inhibits EMT phenotypes. KC mice fed with an ethanol‐containing diet show enhanced pancreatic cancer growth and development than those fed with a control diet. Pancreas isolated from KC mice fed with an ethanol‐containing diet show higher expression of stem cell markers (CD133, CD44, CD24), pluripotency‐maintaining factors (cMyc, KLF4, SOX‐2, and Oct‐4), N‐Cadherin, EMT‐transcription factors (Snail, Slug, and Zeb1), and lower expression of E‐cadherin than those isolated from mice fed with a control diet. Furthermore, pancreas isolated from KC mice fed with an ethanol‐containing diet show higher expression of inflammatory cytokines (TNF‐α, IL‐6, and IL‐8) and PTGS‐2 (COX‐2) gene than those isolated from mice fed with a control diet. These data suggest that chronic alcohol consumption may contribute to pancreatic cancer development by generating inflammatory signals and CSCs.
Collapse
Affiliation(s)
- Wei Yu
- Kansas City VA Medical Center, Kansas City, Missouri, USA
| | - Yuming Ma
- Kansas City VA Medical Center, Kansas City, Missouri, USA
| | - Sanjit K Roy
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, Louisina, USA
| | - Rashmi Srivastava
- Department of Pharmacology, Louisiana State University Health Sciences Center, New Orleans, Louisina, USA
| | - Sharmila Shankar
- Kansas City VA Medical Center, Kansas City, Missouri, USA.,Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, Louisina, USA.,Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, Louisina, USA.,John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, Louisina, USA.,Southeast Louisiana Veterans Health Care System, New Orleans, Louisina, USA
| | - Rakesh K Srivastava
- Kansas City VA Medical Center, Kansas City, Missouri, USA.,Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, Louisina, USA.,Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, Louisina, USA.,Southeast Louisiana Veterans Health Care System, New Orleans, Louisina, USA
| |
Collapse
|
12
|
Rebelo R, Polónia B, Santos LL, Vasconcelos MH, Xavier CPR. Drug Repurposing Opportunities in Pancreatic Ductal Adenocarcinoma. Pharmaceuticals (Basel) 2021; 14:280. [PMID: 33804613 PMCID: PMC8003696 DOI: 10.3390/ph14030280] [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: 02/24/2021] [Revised: 03/15/2021] [Accepted: 03/17/2021] [Indexed: 12/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is considered one of the deadliest tumors worldwide. The diagnosis is often possible only in the latter stages of the disease, with patients already presenting an advanced or metastatic tumor. It is also one of the cancers with poorest prognosis, presenting a five-year survival rate of around 5%. Treatment of PDAC is still a major challenge, with cytotoxic chemotherapy remaining the basis of systemic therapy. However, no major advances have been made recently, and therapeutic options are limited and highly toxic. Thus, novel therapeutic options are urgently needed. Drug repurposing is a strategy for the development of novel treatments using approved or investigational drugs outside the scope of the original clinical indication. Since repurposed drugs have already completed several stages of the drug development process, a broad range of data is already available. Thus, when compared with de novo drug development, drug repurposing is time-efficient, inexpensive and has less risk of failure in future clinical trials. Several repurposing candidates have been investigated in the past years for the treatment of PDAC, as single agents or in combination with conventional chemotherapy. This review gives an overview of the main drugs that have been investigated as repurposing candidates, for the potential treatment of PDAC, in preclinical studies and clinical trials.
Collapse
Affiliation(s)
- Rita Rebelo
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal; (R.R.); (B.P.)
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Bárbara Polónia
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal; (R.R.); (B.P.)
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Lúcio Lara Santos
- Experimental Pathology and Therapeutics Group, IPO—Instituto Português de Oncologia, 4200-072 Porto, Portugal;
- ICBAS—Biomedical Sciences Institute Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - M. Helena Vasconcelos
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal; (R.R.); (B.P.)
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- Department of Biological Sciences, FFUP—Faculty of Pharmacy of the University of Porto, 4200-135 Porto, Portugal
| | - Cristina P. R. Xavier
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal; (R.R.); (B.P.)
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- Department of Biological Sciences, FFUP—Faculty of Pharmacy of the University of Porto, 4200-135 Porto, Portugal
| |
Collapse
|
13
|
Zanini S, Renzi S, Limongi AR, Bellavite P, Giovinazzo F, Bermano G. A review of lifestyle and environment risk factors for pancreatic cancer. Eur J Cancer 2021; 145:53-70. [PMID: 33423007 DOI: 10.1016/j.ejca.2020.11.040] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 11/04/2020] [Accepted: 11/24/2020] [Indexed: 12/12/2022]
Abstract
Pancreatic cancer (PaCa) is one of the deadliest cancers known and its incidence is increasing in the developed countries. Because of the lack of biomarkers that allow early detection and the tendency of the disease to be asymptomatic, the diagnosis comes often too late for effective surgical or chemotherapy intervention. Lifestyle factors, that may cause common genetic modifications occurring in the disease, interfere with pancreatic physiology or function, and play a role in PaCa development, have been of concern recently, since a strategy to prevent this severe cancer is needed. This review identifies the latest evidences related to increased risk of developing PaCa due to dietary habits such as high alcohol, fructose and red or processed meat intake, and pathological conditions such as diabetes, obesity and infections in addition to stress and smoking behaviour. It aims to highlight the importance of intervening on modifiable risk factors: the action on these factors could prevent a considerable number of new cases of PaCa.
Collapse
Affiliation(s)
- Sara Zanini
- Centre for Obesity Research and Education [CORE], School of Pharmacy & Life Sciences, Robert Gordon University, Aberdeen, UK
| | - Serena Renzi
- Centre for Obesity Research and Education [CORE], School of Pharmacy & Life Sciences, Robert Gordon University, Aberdeen, UK
| | - Antonina R Limongi
- Department of Science, University of Basilicata, Potenza, Italy; BioInnova Srl, Potenza, Italy
| | - Paolo Bellavite
- Department of Medicine, Section of General Pathology, University of Verona, Italy
| | | | - Giovanna Bermano
- Centre for Obesity Research and Education [CORE], School of Pharmacy & Life Sciences, Robert Gordon University, Aberdeen, UK.
| |
Collapse
|
14
|
Saha G, Singh R, Mandal A, Das S, Chattopadhyay E, Panja P, Roy P, DeSarkar N, Gulati S, Ghatak S, Ghosh S, Banerjee S, Roy B, Ghosh S, Chaudhuri D, Arora N, Biswas NK, Sikdar N. A novel hotspot and rare somatic mutation p.A138V, at TP53 is associated with poor survival of pancreatic ductal and periampullary adenocarcinoma patients. Mol Med 2020; 26:59. [PMID: 32552660 PMCID: PMC7302128 DOI: 10.1186/s10020-020-00183-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 06/03/2020] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Pancreatic Ductal Adenocarcinoma (PDAC) is a cancer of the exocrine pancreas and 5-year survival rates remain constant at 7%. Along with PDAC, Periampullary Adenocarcinoma (PAC) accounts for 0.5-2% of all gastrointestinal malignancies. Genomic observations were well concluded for PDAC and PACs in western countries but no reports are available from India till now. METHODS Targeted Next Generation Sequencing were performed in 8 (5 PDAC and 3 PAC) tumour normal pairs, using a panel of 412 cancer related genes. Primary findings were replicated in 85 tumour samples (31 PDAC and 54 PAC) using the Sanger sequencing. Mutations were also validated by ASPCR, RFLP, and Ion Torrent sequencing. IHC along with molecular dynamics and docking studies were performed for the p.A138V mutant of TP53. Key polymorphisms at TP53 and its associated genes were genotyped by PCR-RFLP method and association with somatic mutations were evaluated. All survival analysis was done using the Kaplan-Meier survival method which revealed that the survival rates varied significantly depending on the somatic mutations the patients harboured. RESULTS Among the total 114 detected somatic mutations, TP53 was the most frequently mutated (41%) gene, followed by KRAS, SMAD4, CTNNB1, and ERBB3. We identified a novel hotspot TP53 mutation (p.A138V, in 17% of all patients). Low frequency of KRAS mutation (33%) was detected in these samples compared to patients from Western counties. Molecular Dynamics (MD) simulation and DNA-protein docking analysis predicted p.A138V to have oncogenic characteristics. Patients with p.A138V mutation showed poorer overall survival (p = 0.01). So, our finding highlights elevated prevalence of the p53p.A138V somatic mutation in PDAC and pancreatobiliary PAC patients. CONCLUSION Detection of p.A138V somatic variant in TP53 might serve as a prognostic marker to classify patients. It might also have a role in determining treatment regimes. In addition, low frequency of KRAS hotspot mutation mostly in Indian PDAC patient cohort indicates presence of other early drivers in malignant transformation.
Collapse
Affiliation(s)
- Gourab Saha
- Human Genetics Unit, Indian Statistical Institute, 203, B. T. Road, Kolkata, 700108, India
| | - Richa Singh
- Human Genetics Unit, Indian Statistical Institute, 203, B. T. Road, Kolkata, 700108, India
| | - Argha Mandal
- Department of Biotechnology, Heritage Institute of Technology, Kolkata, India
| | - Subrata Das
- National Institute of Biomedical Genomics, Kalyani, West Bengal, India
| | - Esita Chattopadhyay
- Human Genetics Unit, Indian Statistical Institute, 203, B. T. Road, Kolkata, 700108, India
| | - Prasun Panja
- Human Genetics Unit, Indian Statistical Institute, 203, B. T. Road, Kolkata, 700108, India
| | - Paromita Roy
- Department of Pathology & Department of Gastrointestinal Surgery, Tata Medical Center, Rajarhat, Kolkata, India
| | - Navonil DeSarkar
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, USA
| | - Sumit Gulati
- Department of Surgical Gastroenterology, Calcutta Medical Research Institute, Kolkata, India
| | - Supriyo Ghatak
- Department of Surgical Gastroenterology, Calcutta Medical Research Institute, Kolkata, India
| | - Shibajyoti Ghosh
- Department of General Surgery, Medical College and Hospital, Kolkata, India
| | - Sudeep Banerjee
- Department of Pathology & Department of Gastrointestinal Surgery, Tata Medical Center, Rajarhat, Kolkata, India
| | - Bidyut Roy
- Human Genetics Unit, Indian Statistical Institute, 203, B. T. Road, Kolkata, 700108, India
| | - Saurabh Ghosh
- Human Genetics Unit, Indian Statistical Institute, 203, B. T. Road, Kolkata, 700108, India
| | - Dipankar Chaudhuri
- Department of Biotechnology, Heritage Institute of Technology, Kolkata, India
| | - Neeraj Arora
- Department of Pathology & Department of Gastrointestinal Surgery, Tata Medical Center, Rajarhat, Kolkata, India
| | - Nidhan K Biswas
- National Institute of Biomedical Genomics, Kalyani, West Bengal, India
| | - Nilabja Sikdar
- Human Genetics Unit, Indian Statistical Institute, 203, B. T. Road, Kolkata, 700108, India.
| |
Collapse
|
15
|
Differential methylation landscape of pancreatic ductal adenocarcinoma and its precancerous lesions. Hepatobiliary Pancreat Dis Int 2020; 19:205-217. [PMID: 32312637 DOI: 10.1016/j.hbpd.2020.03.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 03/18/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND Pancreatic cancer is one of the most lethal diseases with an incidence almost equal to the mortality. In addition to having genetic causes, cancer can also be considered an epigenetic disease. DNA methylation is the premier epigenetic modification and patterns of aberrant DNA methylation are recognized to be a common hallmark of human tumor. In the multistage carcinogenesis of pancreas starting from precancerous lesions to pancreatic ductal adenocarcinoma (PDAC), the epigenetic changes play a significant role. DATA SOURCES Relevant studies for this review were derived via an extensive literature search in PubMed via using various keywords such as pancreatic ductal adenocarcinoma, precancerous lesions, methylation profile, epigenetic biomarkers that are relevant directly or closely associated with the concerned area of our interest. The literature search was intensively done considering a time frame of 20 years (1998-2018). RESULT In this review we have highlighted the hypermethylation and hypomethylation of the precancerous PDAC lesions (pancreatic intra-epithelial neoplasia, intraductal papillary mucinous neoplasm, mucinous cystic neoplasm and chronic pancreatitis) and PDAC along with the potential biomarkers. We have also achieved the early epigenetic driver that leads to progression from precancerous lesions to PDAC. A bunch of epigenetic driver genes leads to progression of precancerous lesions to PDAC (ppENK, APC, p14/5/16/17, hMLH1 and MGMT) are also documented. We summarized the importance of these observations in therapeutics and diagnosis of PDAC hence identifying the potential use of epigenetic biomarkers in epigenetic targeted therapy. Epigenetic inactivation occurs by hypermethylation of CpG islands in the promoter regions of tumor suppressor genes. We listed all hyper- and hypomethylation of CpG islands of several genes in PDAC including its precancerous lesions. CONCLUSIONS The concept of the review would help to understand their biological effects, and to determine whether they may be successfully combined with other epigenetic drugs. However, we need to continue our research to develop more specific DNA-demethylating agents, which are the targets for hypermethylated CpG methylation sites.
Collapse
|
16
|
Han M, Li N, Li F, Wang H, Ma L. MiR-27b-3p exerts tumor suppressor effects in esophageal squamous cell carcinoma by targeting Nrf2. Hum Cell 2020; 33:641-651. [PMID: 32419118 DOI: 10.1007/s13577-020-00329-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 02/07/2020] [Indexed: 12/16/2022]
Abstract
MiR-27b-3p has been reported to function as tumor suppressor in several tumors, including breast cancer and lung cancer. Recently, miR-27b-3p has been identified to be significantly down-regulated in esophageal cancer. However, the clinical significance and biological role of miR-27b-3p in esophageal squamous cell carcinoma (ESCC) still remain unclear. In this study, the expression levels of miR-27b-3p were significantly reduced in ESCC clinical tissues and ESCC cell lines (EC97069 and TE-1). Moreover, down-regulated expression of miR-27b-3p was associated with poor cell differentiation, TNM stage and lymph node metastasis. Specially, overexpression of miR-27b-3p significantly suppressed cell proliferation, migration and invasion in vitro using CCK-8 and transwell assays. Targetscan bioinformatics predictions and luciferase reporter assay confirmed that nuclear factor erythroid 2-related factor 2 (NFE2L2, Nrf2) was a direct target gene of miR-27b-3p. Nrf2 expression was significantly increased in ESCC tissues compared with adjacent tissues. Up-regulated expression of Nrf2 was correlated with TNM stage and lymph node metastasis. Functionally, knockdown of Nrf2 exhibited similar effects to overexpression of miR-27b-3p. Higher expression of ZO-1, E-cadherin and lower expression of N-cadherin, Vimentin and Claudin-1 were observed after miR-27b-3p overexpression of Nrf2 knockdown. Rescue experiments proved that miR-27b-3p suppressed cell proliferation, migration, invasion and epithelial to mesenchymal transition (EMT) via suppression of Nrf2. Taken together, the newly identified miR-27b-3p/Nrf2 axis might represent a new candidate therapeutic target for ESCC treatment.
Collapse
Affiliation(s)
- Mei Han
- Department of Digestive System, Xinjiang Medical University Affiliated Tumor Hospital, No. 789, Suzhou East Street, Xinshi District, Urumqi, 830000, Xinjiang, China
| | - Na Li
- Department of Digestive System, Xinjiang Medical University Affiliated Tumor Hospital, No. 789, Suzhou East Street, Xinshi District, Urumqi, 830000, Xinjiang, China
| | - Fanzhou Li
- Department of Digestive System, Xinjiang Medical University Affiliated Tumor Hospital, No. 789, Suzhou East Street, Xinshi District, Urumqi, 830000, Xinjiang, China
| | - Hua Wang
- Basic Medical College of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Lanying Ma
- Department of Digestive System, Xinjiang Medical University Affiliated Tumor Hospital, No. 789, Suzhou East Street, Xinshi District, Urumqi, 830000, Xinjiang, China.
| |
Collapse
|
17
|
Abstract
Pancreatic cancer (PC) is an increasingly common disease worldwide. Having a better understanding of worldwide and regional epidemiologic features and risk factors of PC is essential to identify new approaches for prevention, early diagnosis, surveillance, and treatment. In this article, we review the epidemiologic features and risk factors for PC and discuss opportunities and challenges of PC future treatment.
Collapse
Affiliation(s)
- Wenhao Luo
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Jinxin Tao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Lianfang Zheng
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.,Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
18
|
Lifetime alcohol intake and pancreatic cancer incidence and survival: findings from the Melbourne Collaborative Cohort Study. Cancer Causes Control 2019; 30:323-331. [PMID: 30798509 DOI: 10.1007/s10552-019-01146-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 02/14/2019] [Indexed: 02/06/2023]
|
19
|
Luo G, Zhang Y, Guo P, Ji H, Xiao Y, Li K. Global Patterns and Trends in Pancreatic Cancer Incidence: Age, Period, and Birth Cohort Analysis. Pancreas 2019; 48:199-208. [PMID: 30589831 DOI: 10.1097/mpa.0000000000001230] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVES We aim to provide a global geographical picture of pancreatic cancer incidence and temporal trends from 1973 to 2015 for 41 countries. METHODS Joinpoint regression and age-period-cohort model was used. RESULTS In 2012, the highest age-adjusted rate was in Central and Eastern Europe for males and North America for females. Most regions showed sex disparities. During the recent 10 years, increasing trends were observed in North America, Western Europe, and Oceania. The greatest increase occurred in France. For recent birth cohorts, cohort-specific increases in risk were pronounced in Australia, Austria, Brazil, Canada, Costa Rica, Denmark, Estonia, France, Israel, Latvia, Norway, Philippines, Republic of Korea, Singapore, Spain, Sweden, the Netherlands, United States, and US white male populations and in Australia, Austria, Brazil, Bulgaria, Canada, China, Czech Republic, Finland, France, Italy, Japan, Lithuania, Norway, Republic of Korea, Singapore, Spain, The Netherlands, United Kingdom, United States, and US white female populations. CONCLUSIONS In contrast to the favorable effect of the decrease in smoking prevalence, other factors, including the increased prevalence of obesity and diabetes and increased physical inactivity, increased intake of red or processed meat and inadequate intake of fruits and vegetables are likely to have an unfavorable role in pancreatic cancer incidence worldwide.
Collapse
Affiliation(s)
| | - Yanting Zhang
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, and
| | - Pi Guo
- Department of Public Health, Shantou University Medical College, Shantou, China
| | - Huanlin Ji
- Department of Public Health, Shantou University Medical College, Shantou, China
| | - Yuejiao Xiao
- Department of Public Health, Shantou University Medical College, Shantou, China
| | - Ke Li
- Department of Public Health, Shantou University Medical College, Shantou, China
| |
Collapse
|
20
|
Arriaga ME, Vajdic CM, MacInnis RJ, Canfell K, Magliano DJ, Shaw JE, Byles JE, Giles GG, Taylor AW, Gill TK, Hirani V, Cumming RG, Mitchell RP, Banks E, Marker J, Adelstein BA, Laaksonen MA. The burden of pancreatic cancer in Australia attributable to smoking. Med J Aust 2019; 210:213-220. [PMID: 30656698 DOI: 10.5694/mja2.12108] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 10/26/2018] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To estimate the burden of pancreatic cancer in Australia attributable to modifiable exposures, particularly smoking. DESIGN Prospective pooled cohort study. SETTING, PARTICIPANTS Seven prospective Australian study cohorts (total sample size, 365 084 adults); participant data linked to national registries to identify cases of pancreatic cancer and deaths. MAIN OUTCOME MEASURES Associations between exposures and incidence of pancreatic cancer, estimated in a proportional hazards model, adjusted for age, sex, study, and other exposures; future burden of pancreatic cancer avoidable by changes in exposure estimated as population attributable fractions (PAFs) for whole population and for specific population subgroups with a method accounting for competing risk of death. RESULTS There were 604 incident cases of pancreatic cancer during the first 10 years of follow-up. Current and recent smoking explained 21.7% (95% CI, 13.8-28.9%) and current smoking alone explained 15.3% (95% CI, 8.6-22.6%) of future pancreatic cancer burden. This proportion of the burden would be avoidable over 25 years were current smokers to quit and there were no new smokers. The burden attributable to current smoking is greater for men (23.9%; 95% CI, 13.3-33.3%) than for women (7.2%; 95% CI, -0.4% to 14.2%; P = 0.007) and for those under 65 (19.0%; 95% CI, 8.1-28.6%) than for older people (6.6%; 95% CI, 1.9-11.1%; P = 0.030). There were no independent relationships between body mass index or alcohol consumption and pancreatic cancer. CONCLUSIONS Strategies that reduce the uptake of smoking and encourage current smokers to quit could substantially reduce the future incidence of pancreatic cancer in Australia, particularly among men.
Collapse
Affiliation(s)
- Maria E Arriaga
- Centre for Big Data Research in Health, University of New South Wales, Sydney, NSW
| | - Claire M Vajdic
- Centre for Big Data Research in Health, University of New South Wales, Sydney, NSW
| | - Robert J MacInnis
- Cancer Council Victoria, Melbourne, VIC.,Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, VIC
| | - Karen Canfell
- Cancer Council NSW, Sydney, NSW.,University of Sydney, Sydney, NSW.,Prince of Wales Clinical School, University of New South Wales, Sydney, NSW
| | | | | | - Julie E Byles
- Research Centre for Gender, Health and Ageing, University of Newcastle, Newcastle, NSW
| | - Graham G Giles
- Cancer Council Victoria, Melbourne, VIC.,Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, VIC
| | - Anne W Taylor
- Adelaide Medical School, University of Adelaide, Adelaide, SA
| | - Tiffany K Gill
- Adelaide Medical School, University of Adelaide, Adelaide, SA
| | - Vasant Hirani
- University of Sydney, Sydney, NSW.,Charles Perkins Centre, University of Sydney, Sydney, NSW
| | - Robert G Cumming
- University of Sydney, Sydney, NSW.,ANZAC Research Institute, University of Sydney and Concord Hospital, Sydney, NSW
| | - R Paul Mitchell
- Centre for Vision Research, Westmead Institute for Medical Research, University of Sydney, Sydney, NSW
| | - Emily Banks
- Australian National University, Canberra, ACT
| | | | | | - Maarit A Laaksonen
- Centre for Big Data Research in Health, University of New South Wales, Sydney, NSW
| |
Collapse
|
21
|
Diaz KE, Lucas AL. Familial Pancreatic Ductal Adenocarcinoma. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:36-43. [PMID: 30558720 PMCID: PMC7073774 DOI: 10.1016/j.ajpath.2018.06.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 05/21/2018] [Accepted: 06/11/2018] [Indexed: 12/11/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC), although a rare disease, has a poor prognosis. With 5-year overall survival of 8%, there is a critical need to detect PDAC early or at a premalignant stage. Current screening methods are largely imaging based, but a more focused screening approach based on modifiable and nonmodifiable risk factors may improve the efficacy and likely outcomes of screening. In addition, the pathologic mechanisms that lead to the development of PDAC are discussed in an effort to further understand the targets of pancreatic cancer screening. The focus of this article will be inherited pancreatic cancer syndromes and familial pancreatic cancer, which together compose up to 10% of PDAC. Understanding the methods and targets of PDAC screening in high-risk individuals may translate to improved morbidity and mortality.
Collapse
Affiliation(s)
- Kelly E Diaz
- Samuel Bronfman Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Aimee L Lucas
- Samuel Bronfman Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York; Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
22
|
Chhoda A, Lu L, Clerkin BM, Risch H, Farrell JJ. Current Approaches to Pancreatic Cancer Screening. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:22-35. [PMID: 30558719 DOI: 10.1016/j.ajpath.2018.09.013] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 08/29/2018] [Accepted: 09/26/2018] [Indexed: 12/19/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a 5-year survival rate of only 8% and is estimated to be the second leading cause of cancer-related deaths by 2021. Prior convention held that screening for PDAC would not be beneficial; however, a deeper understanding of the carcinogenesis pathway supports a potential window of opportunity among the target population. Screening for PDAC is not a standard practice among the general population because of its low incidence. However, screening may be beneficial for individuals with familial history, chronic diseases with genetic predispositions, or inherited cancer syndromes, such as hereditary breast ovarian cancer syndrome, hereditary pancreatitis, Peutz-Jeghers syndrome, familial atypical multiple mole melanoma, Lynch syndrome (hereditary nonpolyposis colorectal cancer), ataxia telangiectasia, and Li-Fraumeni syndrome, all of which have been associated with an increased risk of developing PDAC. The screening strategies among these high-risk individuals are targeted to identify precursor lesions and PDAC at an early resectable stage. This review describes the risk factors for pancreatic cancer, especially the genetic risk factors in high-risk individuals and current screening strategies available for PDAC.
Collapse
Affiliation(s)
- Ankit Chhoda
- Yale Waterbury Internal Medicine Program, Yale School of Medicine, New Haven, Connecticut
| | - Lingeng Lu
- Chronic Disease Epidemiology, Yale School of Public Health, New Haven, Connecticut
| | - Barbara M Clerkin
- Pancreatic Disease Program, Yale School of Medicine, New Haven, Connecticut
| | - Harvey Risch
- Chronic Disease Epidemiology, Yale School of Public Health, New Haven, Connecticut
| | - James J Farrell
- Yale Center for Pancreatic Diseases, Yale School of Medicine, New Haven, Connecticut; Yale Center for Pancreatic Diseases, Department of Digestive Diseases, Yale School of Public Health, New Haven, Connecticut.
| |
Collapse
|
23
|
Ghosh J, Chowdhury AR, Srinivasan S, Chattopadhyay M, Bose M, Bhattacharya S, Raza H, Fuchs SY, Rustgi AK, Gonzalez FJ, Avadhani NG. Cigarette Smoke Toxins-Induced Mitochondrial Dysfunction and Pancreatitis Involves Aryl Hydrocarbon Receptor Mediated Cyp1 Gene Expression: Protective Effects of Resveratrol. Toxicol Sci 2018; 166:428-440. [PMID: 30165701 PMCID: PMC6260170 DOI: 10.1093/toxsci/kfy206] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
We previously reported that mitochondrial CYP1 enzymes participate in the metabolism of polycyclic aromatic hydrocarbons and other carcinogens leading to mitochondrial dysfunction. In this study, using Cyp1b1-/-, Cyp1a1/1a2-/-, and Cyp1a1/1a2/1b1-/- mice, we observed that cigarette and environmental toxins, namely benzo[a]pyrene (BaP) and 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), induce pancreatic mitochondrial respiratory dysfunction and pancreatitis. Our results suggest that aryl hydrocarbon receptor (AhR) activation and resultant mitochondrial dysfunction are associated with pancreatic pathology. BaP treatment markedly inhibits pancreatic mitochondrial oxygen consumption rate (OCR), ADP-dependent OCR, and also maximal respiration, in wild-type mice but not in Cyp1a1/1a2-/- and Cyp1a1/1a2/1b1-/- mice. In addition, both BaP and TCDD treatment markedly affected mitochondrial complex IV activity, in addition to causing marked reduction in mitochondrial DNA content. Interestingly, the AhR antagonist resveratrol, attenuated BaP-induced mitochondrial respiratory defects in the pancreas, and reversed pancreatitis, both histologically and biochemically in wild-type mice. These results reveal a novel role for AhR- and AhR-regulated CYP1 enzymes in eliciting mitochondrial dysfunction and cigarette toxin-mediated pancreatic pathology. We propose that increased mitochondrial respiratory dysfunction and oxidative stress are involved in polycyclic aromatic hydrocarbon associated pancreatitis. Resveratrol, a chemo preventive agent and AhR antagonist, and CH-223191, a potent and specific AhR inhibitor, confer protection against BaP-induced mitochondrial dysfunction and pancreatic pathology.
Collapse
Affiliation(s)
- Jyotirmoy Ghosh
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
- Department of Chemistry, Banwarilal Bhalotia College, Asansol, Ushagram, Asansol-713303, West Bengal, India
| | - Anindya Roy Chowdhury
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Satish Srinivasan
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
- Roche Molecular Systems, 1080, US-202, Branchburg, NJ 08876
| | - Mrittika Chattopadhyay
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Moumita Bose
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390
| | - Sabyasachi Bhattacharya
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426
| | - Haider Raza
- Department of Biochemistry, College of Medicine and Health Sciences, UAE University, Al-Ain, UAE
| | - Serge Y Fuchs
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Anil K Rustgi
- Division of Gastroenterology, Departments of Medicine and Genetics, and Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Frank J Gonzalez
- National Cancer Institute, Center for Cancer Research, Bethesda, Maryland 20892
| | - Narayan G Avadhani
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| |
Collapse
|
24
|
Yu W, Ma Y, Shankar S, Srivastava RK. Chronic ethanol exposure of human pancreatic normal ductal epithelial cells induces cancer stem cell phenotype through SATB2. J Cell Mol Med 2018; 22:3920-3928. [PMID: 29761897 PMCID: PMC6050497 DOI: 10.1111/jcmm.13666] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 03/31/2018] [Indexed: 12/13/2022] Open
Abstract
The incidence of pancreatic cancer is on the rise. Risk factors for pancreatic cancer include alcohol toxicity and metabolic conditions such as obesity, hypertension, dyslipidaemia, insulin resistance and type 2 diabetes. However, the molecular mechanism by which chronic alcohol consumption contributes to pancreatic cancer is not well understood. The purpose of the study was to demonstrate the effects of long-term chronic ethanol exposure on the transformation of human pancreatic normal ductal epithelial (HPNE) cells. Our data showed that ethanol-transformed HPNE cells were more progressively transformed exhibiting spheroids and colonies, and anchorage-independent growth. These transformed cells contained high levels of reactive oxygen species and induced SATB2 expression. Furthermore, during ethanol-induced cellular transformation, cells gained the phenotypes of cancer stem cells (CSCs) by expressing pluripotency maintaining factors (Oct4, Sox2, cMyc and KLF4) and stem cell markers (CD24, CD44 and CD133). Ethanol-induced SATB2 can bind to the promoters of KLF4, Oct4, cMyc, Sox2, Bcl-2 and XIAP genes. Suppression of SATB2 expression in ethanol-transformed HPNE cells inhibited cell proliferation, colony formation and markers of CSCs and pluripotency. These data suggest that chronic alcohol consumption may contribute toward the development of pancreatic cancer by converting HPNE cells to cancer stem-like cells.
Collapse
Affiliation(s)
- Wei Yu
- Kansas City VA Medical Center, Kansas City, MO, USA
| | - Yiming Ma
- Kansas City VA Medical Center, Kansas City, MO, USA
| | - Sharmila Shankar
- Kansas City VA Medical Center, Kansas City, MO, USA
- Department of Pathology, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Rakesh K Srivastava
- Kansas City VA Medical Center, Kansas City, MO, USA
- Department of Pharmaceutical Sciences, University of Missouri-Kansas City, Kansas City, MO, USA
| |
Collapse
|
25
|
Marks DL, Olson RL, Urrutia R, Billadeau DD, Roy N, Calin GA, Fabbri M, Koutsioumpa M, Iliopoulos D, Ordog T, Huebert R, Sarmento O, Bamidele AO, Faubion W, Lomberk GL, Siveke J, Ahuja N, Iovanna J, Hlady RA, Robertson K, Kisiel J, Pin CL, Fernandez-Zapico ME. Epigenetics of gastrointestinal diseases: notes from a workshop. Epigenetics 2018; 13:449-457. [PMID: 30056798 PMCID: PMC6140811 DOI: 10.1080/15592294.2018.1464351] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
International experts gathered at the Mayo Clinic (Rochester MN, USA) on February 27th-28th, 2017 for a meeting entitled ‘Basic and Translational Facets of the Epigenetics of GI Diseases’. This workshop summarized recent advances on the role of epigenetics in the pathobiology of gastrointestinal (GI) diseases. Highlights of the meeting included recent advances on the involvement of different epigenetic mechanisms in malignant and nonmalignant GI disorders and the epigenetic heterogeneity exhibited in these diseases. The translational value of epigenetic drugs, as well as the current and future use of epigenetic changes (i.e., DNA methylation patterns) as biomarkers for early detection tools or disease stratification were also important topics of discussion.
Collapse
Affiliation(s)
- David L Marks
- a Schulze Center for Novel Therapeutics, Division of Oncology Research , Mayo Clinic , Rochester , MN , USA
| | - Rachel L Olson
- a Schulze Center for Novel Therapeutics, Division of Oncology Research , Mayo Clinic , Rochester , MN , USA
| | - Raul Urrutia
- b Division of Research, Department of Surgery , Medical College of Wisconsin , Milwaukee , WI , USA
| | - Daniel D Billadeau
- a Schulze Center for Novel Therapeutics, Division of Oncology Research , Mayo Clinic , Rochester , MN , USA
| | - Nilotpal Roy
- c Diabetes Center , University of California at San Francisco , San Francisco , CA , USA
| | - George A Calin
- d Department of Experimental Therapeutics, Division of Cancer Medicine , MD Anderson Cancer Center , Houston , TX , USA
| | - Muller Fabbri
- e Children's Center for Cancer and Blood Diseases, Keck School of Medicine of USC , University of Southern California , Los Angeles , CA , USA
| | - Marina Koutsioumpa
- f Laboratory and the Center for Systems Biomedicine , University of California at Los Angeles , Los Angeles , CA , USA
| | - Dimitrios Iliopoulos
- f Laboratory and the Center for Systems Biomedicine , University of California at Los Angeles , Los Angeles , CA , USA
| | - Tamas Ordog
- g Division of Gastroenterology, Department of Medicine , Mayo Clinic , Rochester , MN , USA
| | - Robert Huebert
- g Division of Gastroenterology, Department of Medicine , Mayo Clinic , Rochester , MN , USA
| | - Olga Sarmento
- g Division of Gastroenterology, Department of Medicine , Mayo Clinic , Rochester , MN , USA
| | - Adebowale O Bamidele
- g Division of Gastroenterology, Department of Medicine , Mayo Clinic , Rochester , MN , USA
| | - William Faubion
- g Division of Gastroenterology, Department of Medicine , Mayo Clinic , Rochester , MN , USA
| | - Gwen L Lomberk
- b Division of Research, Department of Surgery , Medical College of Wisconsin , Milwaukee , WI , USA
| | - Jens Siveke
- h Division of Solid Tumor Translational Oncology, West German Cancer Center , University Hospital Essen , Essen , Germany
| | - Nita Ahuja
- i Department of Surgery , Yale School of Medicine , New Haven , CT , USA
| | - Juan Iovanna
- j Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258 , Institut Paoli-Calmettes , Aix Marseille , France
| | - Ryan A Hlady
- k Department of Molecular Pharmacology and Experimental Therapeutics , Mayo Clinic , Rochester , MN , USA
| | - Keith Robertson
- k Department of Molecular Pharmacology and Experimental Therapeutics , Mayo Clinic , Rochester , MN , USA
| | - John Kisiel
- g Division of Gastroenterology, Department of Medicine , Mayo Clinic , Rochester , MN , USA
| | - Christopher L Pin
- l Division of Genetics & Development, Children's Health Research Institute, Departments of Pediatrics, Physiology and Pharmacology, and Oncology , The University of Western Ontario , London , ON , Canada
| | - Martin E Fernandez-Zapico
- a Schulze Center for Novel Therapeutics, Division of Oncology Research , Mayo Clinic , Rochester , MN , USA
| |
Collapse
|
26
|
Relation between mortality trends of cardiovascular diseases and selected cancers in the European Union, in 1970-2017. Focus on cohort and period effects. Eur J Cancer 2018; 103:341-355. [PMID: 30029971 DOI: 10.1016/j.ejca.2018.06.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 05/29/2018] [Accepted: 06/05/2018] [Indexed: 01/01/2023]
Abstract
AIM To characterise mortality trends from major non-communicable diseases in the European Union (EU) analysing data from the World Health Organization (WHO) Mortality Database. METHODS We obtained EU population and death certification data for major non-communicable diseases, i.e. seven cancer sites (stomach, intestine, pancreas, lung, breast, prostate and haematopoietic), total cancers, coronary heart diseases (CHDs) and cerebrovascular diseases (CVDs) from the WHO Mortality Database over the 1970 and 2012 period. We computed age-standardised rates (world standard population) and applied joinpoint regression models to identify temporal trends and age period cohort (APC) models to disentangle the effects of age, period of death and cohort of birth on mortality. RESULTS In 2012, 2.4 million deaths were recorded in the EU (1.3 million from cancers and 1.1 million from CHD and CVD combined). Over the last decade, mortality from cancer fell by 14% in men and 8% in women, resulting in age-standardised rates of 144 and 88/100,000 persons, respectively, in 2012. The only exceptions to the general downward trends were pancreatic cancer and female lung cancer. Both cardiovascular diseases mortality fell over 35% in both sexes with rates of 60 and 28/100,000 for CHD, and of 30 and 23/100,000 for CVD, in men and women, respectively, in 2012. CONCLUSIONS Overall trends in mortality rates from non-communicable diseases in the EU were favourable, and the joinpoint and APC models indicated these trends are likely to continue in the near future. Lack of progress in tobacco-related mortality in women underlines the importance of female-specific anti-tobacco policies.
Collapse
|
27
|
Welinsky S, Lucas AL. Familial Pancreatic Cancer and the Future of Directed Screening. Gut Liver 2018; 11:761-770. [PMID: 28609837 PMCID: PMC5669591 DOI: 10.5009/gnl16414] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Revised: 11/01/2016] [Accepted: 11/01/2016] [Indexed: 12/11/2022] Open
Abstract
Pancreatic cancer (PC) is the third most common cause of cancer-related death in the United States and the 12th most common worldwide. Mortality is high, largely due to late stage of presentation and suboptimal treatment regimens. Approximately 10% of PC cases have a familial basis. The major genetic defect has yet to be identified but may be inherited by an autosomal dominant pattern with reduced penetrance. Several known hereditary syndromes or genes are associated with an increased risk of developing PC and account for approximately 2% of PCs. These syndromes include the hereditary breast-ovarian cancer syndrome, Peutz-Jeghers syndrome, familial atypical multiple mole melanoma, Lynch syndrome, familial polyposis, ataxia-telangiectasia, and hereditary pancreatitis. Appropriate screening using methods such as biomarkers or imaging, with endoscopic ultrasound and magnetic resonance imaging, may assist in the early detection of neoplastic lesions in the high-risk population. If these lesions are detected and treated before the development of invasive carcinoma, PC disease morbidity and mortality may be improved. This review will focus on familial PC and other hereditary syndromes implicated in the increased risk of PC; it will also highlight current screening methods and the future of new screening modalities.
Collapse
Affiliation(s)
- Sara Welinsky
- Samuel F. Bronfman Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Aimee L Lucas
- Samuel F. Bronfman Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
28
|
Pang Y, Holmes MV, Guo Y, Yang L, Bian Z, Chen Y, Iona A, Millwood IY, Bragg F, Chen J, Li L, Kartsonaki C, Chen Z. Smoking, alcohol, and diet in relation to risk of pancreatic cancer in China: a prospective study of 0.5 million people. Cancer Med 2017; 7:229-239. [PMID: 29271112 PMCID: PMC5773963 DOI: 10.1002/cam4.1261] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 08/24/2017] [Accepted: 10/13/2017] [Indexed: 12/11/2022] Open
Abstract
In China, the incidence of pancreatic cancer (PC) has increased in recent decades. However, little is known about the relevance to PC risk of lifestyle and behavioral factors such as smoking, alcohol drinking, and diet. The China Kadoorie Biobank prospective study recruited 512,891 adults (210,222 men, 302,669 women) aged 30–79 (mean 52) years from 10 diverse areas during 2004–08. During ~9 years of follow‐up, 688 incident cases of PC were recorded among those who had no prior history of cancer at baseline. Cox regression yielded adjusted hazard ratios (HR) for PC associated with smoking, alcohol and selected dietary factors. Overall, 74% of men were ever‐regular smokers and 33% of men drank at least weekly, compared with only 3% and 2% of women, respectively. Among men, current regular smoking was associated with an adjusted HR of 1.25 (95% CI 1.08–1.44) for PC, with greater excess risk in urban than rural areas (1.46 [1.19–1.79] vs 1.04 [0.86–1.26]). Heavy, but not light to moderate, alcohol drinking (i.e. ≥420 g/week) was associated with significant excess risk (1.69 [1.21–2.37]), again more extreme in urban than rural areas (1.93 [1.29–2.87] vs 1.35 [0.74–2.48]). Overall, regular consumption of certain foodstuffs was associated with PC risk, with adjusted daily vs never/rare consumption HRs of 0.66 (0.56–0.79) for fresh fruit and 1.16 (1.01–1.33) for red meat. In China, smoking and heavy alcohol drinking were independent risk factors for PC in men. Lower fresh fruit and higher red meat consumption were also associated with higher risk of PC.
Collapse
Affiliation(s)
- Yuanjie Pang
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Michael V Holmes
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom.,Medical Research Council Population Health Research Unit (MRC PHRU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom.,National Institute for Health Research Oxford Biomedical Research Centre, Oxford University Hospital, Old Road, Oxford OX3 7LE, UK
| | - Yu Guo
- Chinese Academy of Medical Sciences, 9 Dongdan San Tiao, Beijing, 100730, China
| | - Ling Yang
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom.,Medical Research Council Population Health Research Unit (MRC PHRU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Zheng Bian
- Chinese Academy of Medical Sciences, 9 Dongdan San Tiao, Beijing, 100730, China
| | - Yiping Chen
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom.,Medical Research Council Population Health Research Unit (MRC PHRU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Andri Iona
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom.,Medical Research Council Population Health Research Unit (MRC PHRU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Iona Y Millwood
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom.,Medical Research Council Population Health Research Unit (MRC PHRU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Fiona Bragg
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Junshi Chen
- National Center for Food Safety Risk Assessment, 37 Guangqu Road, Beijing, 100021, China
| | - Liming Li
- National Institute for Health Research Oxford Biomedical Research Centre, Oxford University Hospital, Old Road, Oxford OX3 7LE, UK.,School of Public Health, Peking University, Beijing, 100191, China
| | - Christiana Kartsonaki
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom.,Medical Research Council Population Health Research Unit (MRC PHRU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Zhengming Chen
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
29
|
Salem AA, Mackenzie GG. Pancreatic cancer: A critical review of dietary risk. Nutr Res 2017; 52:1-13. [PMID: 29764623 DOI: 10.1016/j.nutres.2017.12.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Revised: 10/31/2017] [Accepted: 12/08/2017] [Indexed: 12/14/2022]
Abstract
Pancreatic cancer is a deadly disease. It is estimated that about 90% of pancreatic cancer cases are due to environmental risk factors. Among these, approximately 50% of pancreatic cancer cases may be attributed to diet, which is largely modifiable. Given this large attribution to diet, there have been numerous epidemiological studies assessing the risk of various dietary factors on the incidence of pancreatic cancer. However, many of these studies present conflicting and/or inconclusive findings. The objective of this review is two-fold: (a) to summarize the current evidence on the association between various dietary factors and the risk of developing pancreatic cancer and (b) to discuss what additional studies are needed to better elucidate the role of diet as a potential risk factor for pancreatic cancer. We summarized the evidence by using data primarily from meta-analyses and pooled analysis when available, focusing on the most studied nutrients, foods, and dietary patterns. We observed that, while the association between individual nutrients and pancreatic cancer risk have been heavily studied, the evidence is mostly conflicting and inconclusive. In contrast, the evidence of certain associations among dietary patterns and pancreatic cancer risk is clearer, has more power, and is less conflicting. Therefore, we propose a shift in the focus of nutritional epidemiological research with regards to pancreatic cancer risk. We discourage further epidemiological research studies that focus on single nutrients, whereas we strongly encourage additional studies that investigate how a combination of diet and other lifestyle factors may promote or prevent pancreatic carcinogenesis.
Collapse
Affiliation(s)
- Asmaa A Salem
- Department of Nutrition, University of California, Davis, One Shields Ave, Davis, CA, 95616
| | - Gerardo G Mackenzie
- Department of Nutrition, University of California, Davis, One Shields Ave, Davis, CA, 95616.
| |
Collapse
|
30
|
Ransome Y, Slopen N, Karlsson O, Williams DR. Elevated inflammation in association with alcohol abuse among Blacks but not Whites: results from the MIDUS biomarker study. J Behav Med 2017; 41:374-384. [PMID: 29230616 DOI: 10.1007/s10865-017-9905-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 11/23/2017] [Indexed: 12/14/2022]
Abstract
Some studies document racial disparities in self-reported health associated with alcohol use and abuse. However, few studies examined biomarkers that underlie the onset of alcohol-related chronic diseases. We investigated whether the association between alcohol abuse and five biomarkers of inflammation (CRP, IL-6, fibrinogen, E-selectin, sICAM-1) vary between Black and White Americans aged 35 to 84 (n = 1173) from the Midlife in the United States Biomarker Study. Multivariable Ordinary Least Squares regressions were used to assess Black-White differences in the association between alcohol abuse and the biomarkers. Race moderated the association between alcohol abuse and CRP (b = 0.56, SE = 0.28, p = 0.048), IL-6 (b = 0.65, SE = 0.22, p = 0.004), and a composite inflammation score (b = 0.014, SE = 0.07, p = 0.041). These findings potentially shed light for why alcohol has a stronger negative association with poorer health for Blacks compared to Whites. Analysis should be replicated in larger prospective cohorts.
Collapse
Affiliation(s)
- Yusuf Ransome
- Department of Social and Behavioral Sciences, Yale School of Public Health, 60 College Street, 403, New Haven, CT, 06510, USA.
| | - Natalie Slopen
- Department of Epidemiology and Biostatistics, University of Maryland School of Public Health, 4200 Valley Drive, College Park, MD, 20742, USA
| | - Oskar Karlsson
- Center for Molecular Medicine, Karolinska Institute, Solna, Sweden.,Department of Pharmaceutical Biosciences, Uppsala University, Box 591, 751 24, Uppsala, Sweden
| | - David R Williams
- Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, 02115, USA
| |
Collapse
|
31
|
Moutinho-Ribeiro P, Coelho R, Giovannini M, Macedo G. Pancreatic cancer screening: Still a delusion? Pancreatology 2017; 17:754-765. [PMID: 28739291 DOI: 10.1016/j.pan.2017.07.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 06/17/2017] [Accepted: 07/05/2017] [Indexed: 12/11/2022]
Abstract
Pancreatic adenocarcinoma represents the fourth most common cause of cancer mortality and death due to pancreatic cancer (PC) have increased since 2003. Its incidence has also raised about 30% in the past decade and it is expected to become the second cause of cancer mortality by 2020 in the USA. Most PC present with metastatic disease and improvements in treatment outcomes for this group have been disappointing. These observations support the idea that screening to identify patients at an earlier stage might be an important strategy in improving overall PC outcomes. Many protocols have been tested, nevertheless, by now there is no effective screening program. Given the overall low incidence of disease and the current lack of accurate, inexpensive and noninvasive screening tests, the consensus is that widespread population-based screening for PC in the general population or in patients with only one affected first-degree relative is neither practicable nor indicated in most countries. However, a different scenario is screening patients with higher risk for PC, most of them with hereditary conditions predisposing the development of this neoplasia. In fact, some guidelines are now available helping to select these individuals at risk and to screen them, in order to achieve early detection of PC.
Collapse
Affiliation(s)
- Pedro Moutinho-Ribeiro
- Department of Gastroenterology, Centro Hospitalar São João, Porto, Portugal; Faculty of Medicine, University of Porto, Portugal.
| | - Rosa Coelho
- Department of Gastroenterology, Centro Hospitalar São João, Porto, Portugal
| | - Marc Giovannini
- Endoscopic Unit, Paoli-Calmettes Institute, Marseilles, France
| | - Guilherme Macedo
- Department of Gastroenterology, Centro Hospitalar São João, Porto, Portugal; Faculty of Medicine, University of Porto, Portugal
| |
Collapse
|
32
|
Gorelick FS, Lerch MM. Do Animal Models of Acute Pancreatitis Reproduce Human Disease? Cell Mol Gastroenterol Hepatol 2017; 4:251-262. [PMID: 28752114 PMCID: PMC5518169 DOI: 10.1016/j.jcmgh.2017.05.007] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 05/26/2017] [Indexed: 12/10/2022]
Abstract
Acute pancreatitis is currently the most common cause of hospital admission among all nonmalignant gastrointestinal diseases. To understand the pathophysiology of the disease and as a potential step toward developing targeted therapies, attempts to induce the disease experimentally began more than 100 years ago. Recent decades have seen progress in developing new experimental pancreatitis models as well as elucidating many underlying cell biological and pathophysiological disease mechanisms. Some models have been developed to reflect specific causes of acute pancreatitis in human beings. However, the paucity of data relating to the molecular mechanisms of human disease, the likelihood that multiple genetic and environmental factors affect the risk of disease development and its severity, and the limited information regarding the natural history of disease in human beings make it difficult to evaluate the value of disease models. Here, we provide an overview of key models and discuss our views on their strengths for characterizing cell biological disease mechanisms or for identifying potential therapeutic targets. We also acknowledge their limitations.
Collapse
Affiliation(s)
- Fred S. Gorelick
- Yale University Medical School and Veterans Affairs Medical Center, West Haven, Connecticut
- Correspondence Address correspondence to: Fred S. Gorelick, MD, VA Connecticut Healthcare System/Yale University Medical School, 950 Campbell Avenue, West Haven, Connecticut 06516. fax: (203) 937-3852.VA Connecticut Healthcare System/Yale University Medical School950 Campbell AvenueWest HavenConnecticut 06516
| | - Markus M. Lerch
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
33
|
Lin X, Zheng L, Song H, Xiao J, Pan B, Chen H, Jin X, Yu H. Effects of microRNA-183 on epithelial-mesenchymal transition, proliferation, migration, invasion and apoptosis in human pancreatic cancer SW1900 cells by targeting MTA1. Exp Mol Pathol 2017; 102:522-532. [PMID: 28506766 DOI: 10.1016/j.yexmp.2017.05.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 05/11/2017] [Indexed: 10/19/2022]
Abstract
OBJECTIVE This study aims to explore effects of miR-183 on epithelial-mesenchymal transition (EMT) and invasion by targeting MTA1 in human pancreatic cancer (PC) cells. METHODS Totally, 108 PC patients admitted in Wenzhou Central Hospital and Wenzhou People's Hospital, The Dingli Clinical Institute of Wenzhou Medical University from March 2010 to March 2014 were enrolled. qRT-PCR and immunohistochemistry were applied to examine expression of MTA1 mRNA and protein. Samples were divided into 6 groups: blank, NC, miR-183 mimics, miR-183 inhibitors, MTA1-siRNA and miR-183 inhibitors +MTA1-siRNA groups. CCK8 method was employed for determining cell proliferation rate, flow cytometry for cell apoptosis rate, scratch test for cell migration and Transwell assay for cell invasion. qRT-PCR and Western blotting were used to determine expression of MTA1, E-cadherin and Vimentin mRNA and protein. RESULTS Positive expression rate of MTA1 was upregulated in PC tissues, and expression of miR-183 and MTA1 was associated with differentiation, migration, tumor size, TNM. The miR-183 mimics and MTA1-siRNA groups showed a decrease in proliferation, migration and invasion, whereas increased apoptosis, in comparison with blank and NC groups, as expression of MTA1 and Vimentin mRNA and protein were reduced, expression of E-cadherin mRNA and protein was elevated. Compared to blank and NC groups, the miR-183 inhibitors group exhibited enhanced proliferation, migration and invasion and inhibited apoptosis; increased expressions of MTA1 and Vimentin mRNA and protein and decreased expressions of E-cadherin mRNA and protein. CONCLUSION Our study supported that miR-183 could repress EMT and invasion of human PC cells through inhibition of MTA1 expression.
Collapse
Affiliation(s)
- Xizhou Lin
- Department of Digestive Diseases, Wenzhou People's Hospital, The Third Clinical College of Wenzhou Medical University, Wenzhou 325000, PR China
| | - Liang Zheng
- Department of Digestive Diseases, Wenzhou People's Hospital, The Third Clinical College of Wenzhou Medical University, Wenzhou 325000, PR China
| | - Hongliang Song
- Department of Surgery, Wenzhou Central Hospital, The Dingli Clinical Institute of Wenzhou Medical University, Wenzhou 325000, PR China
| | - Jun Xiao
- Department of Surgery, Wenzhou Central Hospital, The Dingli Clinical Institute of Wenzhou Medical University, Wenzhou 325000, PR China
| | - Bujian Pan
- Department of Surgery, Wenzhou Central Hospital, The Dingli Clinical Institute of Wenzhou Medical University, Wenzhou 325000, PR China
| | - Haichuan Chen
- Department of Surgery, Wenzhou Central Hospital, The Dingli Clinical Institute of Wenzhou Medical University, Wenzhou 325000, PR China
| | - Xiaodan Jin
- Department of Surgery, Wenzhou Central Hospital, The Dingli Clinical Institute of Wenzhou Medical University, Wenzhou 325000, PR China
| | - Haibo Yu
- Department of Surgery, Wenzhou Central Hospital, The Dingli Clinical Institute of Wenzhou Medical University, Wenzhou 325000, PR China.
| |
Collapse
|
34
|
Nogueira LM, Newton CC, Pollak M, Silverman DT, Albanes D, Männistö S, Weinstein SJ, Jacobs EJ, Stolzenberg-Solomon RZ. Serum C-peptide, Total and High Molecular Weight Adiponectin, and Pancreatic Cancer: Do Associations Differ by Smoking? Cancer Epidemiol Biomarkers Prev 2017; 26:914-922. [PMID: 28096201 PMCID: PMC5457333 DOI: 10.1158/1055-9965.epi-16-0891] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 01/09/2017] [Accepted: 01/12/2017] [Indexed: 12/16/2022] Open
Abstract
Background: Studies examining associations between circulating concentrations of C-peptide and total adiponectin, two biomarkers related to obesity and insulin secretion and sensitivity and pancreatic ductal adenocarcinoma (PDA) risk have shown inconsistent results and included limited numbers of smokers.Methods: We examined associations of these biomarkers and high molecular weight (HMW) adiponectin with PDA, overall, and by smoking status. We conducted a pooled nested case-control analysis in 3 cohorts (Prostate, Lung, Colorectal, and Ovarian Cancer Trial, Alpha-Tocopherol, Beta-Carotene Cancer Prevention Study, and Cancer Prevention Study-II), with 758 cases (435 current smokers) and 1,052 controls (531 smokers) matched by cohort, age, sex, race, blood draw date and follow-up time. We used conditional logistic regression adjusted for age, smoking, diabetes, and body mass index to calculate ORs and 95% confidence intervals (CI).Results: Circulating C-peptide concentration was not associated with PDA in never or former smokers, but was inversely associated with PDA in current smokers (per SD OR = 0.67; 95% CI, 0.54-0.84; Pinteraction = 0.005). HMW adiponectin was inversely associated with PDA in never smokers (OR = 0.43; 95% CI, 0.23-0.81), not associated in former smokers, and positively associated in smokers (OR = 1.23; 95% CI, 1.04-1.45; Pinteraction = 0.009). Total adiponectin was not associated with PDA in nonsmokers or current smokers.Conclusions: Associations of biomarkers of insulin secretion and sensitivity with PDA differ by smoking status. Smoking-induced pancreatic damage may explain the associations in smokers while mechanisms related to insulin resistance associations in nonsmokers.Impact: Future studies of these biomarkers and PDA should examine results by smoking status. Cancer Epidemiol Biomarkers Prev; 26(6); 914-22. ©2017 AACR.
Collapse
Affiliation(s)
- Leticia M Nogueira
- Texas Cancer Registry, Department of State Health Services, Austin, Texas
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, Rockville, Maryland
| | | | - Michael Pollak
- Department of Oncology, Lady Davis Research Institute of the Jewish General Hospital and McGill University, Montreal, Quebec, Canada
| | - Debra T Silverman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, Rockville, Maryland
| | - Demetrius Albanes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, Rockville, Maryland
| | - Satu Männistö
- Department of Health, National Institute for Health and Welfare, Helsinki, Finland
| | - Stephanie J Weinstein
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, Rockville, Maryland
| | - Eric J Jacobs
- Epidemiology Research Program, American Cancer Society, Atlanta Georgia.
| | | |
Collapse
|
35
|
Maisonneuve P, Amar S, Lowenfels A. Periodontal disease, edentulism, and pancreatic cancer: a meta-analysis. Ann Oncol 2017; 28:985-995. [DOI: 10.1093/annonc/mdx019] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
36
|
Abstract
OBJECTIVES Tobacco exposure is an established risk factor for pancreatic cancer and chronic pancreatitis; however, its role in pancreatic insufficiency is not clear. METHODS This controlled, cross-sectional study examined smokers and nonsmokers with no history of pancreatic disease. Histories and validated inventories of alcohol and tobacco use were obtained, and pancreatic insufficiency was assessed using the fecal elastase-1 assay. RESULTS Of 7854 patients approached, 226 were interviewed and 200 enrolled. The rates of pancreatic insufficiency [18% (18/100)] and severe pancreatic insufficiency [10% (10/100)] were significantly higher in smokers than in controls [6% (6/100), P = 0.009 and 1% (1/100), P = 0.010, respectively]. On multivariate logistic regression, the risk of pancreatic insufficiency in smokers was significantly increased [odds ratio, 4.34 (1.37-13.75); P = 0.012], controlling for alcohol use and relevant covariates. Tobacco exposure was associated with the highest odds ratio for pancreatic insufficiency. Alcohol consumption was strongly associated with tobacco exposure (P < 0.001), but not with pancreatic insufficiency by multivariate analysis (P = 0.792). CONCLUSIONS This study suggests that tobacco exposure is independently associated with pancreatic exocrine insufficiency in patients without a prior diagnosis of pancreatic disease. Tobacco exposure seems to have greater detrimental effects on pancreatic function than alcohol in this population.
Collapse
|
37
|
Lu PY, Shu L, Shen SS, Chen XJ, Zhang XY. Dietary Patterns and Pancreatic Cancer Risk: A Meta-Analysis. Nutrients 2017; 9:nu9010038. [PMID: 28067765 PMCID: PMC5295082 DOI: 10.3390/nu9010038] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Revised: 12/13/2016] [Accepted: 12/23/2016] [Indexed: 12/13/2022] Open
Abstract
A number of studies have examined the associations between dietary patterns and pancreatic cancer risk, but the findings have been inconclusive. Herein, we conducted this meta-analysis to assess the associations between dietary patterns and the risk of pancreatic cancer. MEDLINE (provided by the National Library of Medicine) and EBSCO (Elton B. Stephens Company) databases were searched for relevant articles published up to May 2016 that identified common dietary patterns. Thirty-two studies met the inclusion criteria and were finally included in this meta-analysis. A reduced risk of pancreatic cancer was shown for the highest compared with the lowest categories of healthy patterns (odds ratio, OR = 0.86; 95% confidence interval, CI: 0.77–0.95; p = 0.004) and light–moderate drinking patterns (OR = 0.90; 95% CI: 0.83–0.98; p = 0.02). There was evidence of an increased risk for pancreatic cancer in the highest compared with the lowest categories of western-type pattern (OR = 1.24; 95% CI: 1.06–1.45; p = 0.008) and heavy drinking pattern (OR = 1.29; 95% CI: 1.10–1.48; p = 0.002). The results of this meta-analysis demonstrate that healthy and light–moderate drinking patterns may decrease the risk of pancreatic cancer, whereas western-type and heavy drinking patterns may increase the risk of pancreatic cancer. Additional prospective studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Pei-Ying Lu
- Department of Geriatrics, Zhejiang Hospital, Xihu District, Hangzhou 310013, China.
| | - Long Shu
- Department of Nutrition, Zhejiang Hospital, Xihu District, Hangzhou 310013, China.
| | - Shan-Shan Shen
- Department of Geriatrics, Zhejiang Hospital, Xihu District, Hangzhou 310013, China.
| | - Xu-Jiao Chen
- Department of Geriatrics, Zhejiang Hospital, Xihu District, Hangzhou 310013, China.
| | - Xiao-Yan Zhang
- Department of Nutrition, Zhejiang Hospital, Xihu District, Hangzhou 310013, China.
| |
Collapse
|
38
|
Todoric J, Antonucci L, Karin M. Targeting Inflammation in Cancer Prevention and Therapy. Cancer Prev Res (Phila) 2016; 9:895-905. [PMID: 27913448 DOI: 10.1158/1940-6207.capr-16-0209] [Citation(s) in RCA: 258] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 10/03/2016] [Indexed: 12/14/2022]
Abstract
Inflammation is associated with the development and malignant progression of most cancers. As most of the cell types involved in cancer-associated inflammation are genetically stable and thus are not subjected to rapid emergence of drug resistance, the targeting of inflammation represents an attractive strategy both for cancer prevention and for cancer therapy. Tumor-extrinsic inflammation is caused by many factors, including bacterial and viral infections, autoimmune diseases, obesity, tobacco smoking, asbestos exposure, and excessive alcohol consumption, all of which increase cancer risk and stimulate malignant progression. In contrast, cancer-intrinsic or cancer-elicited inflammation can be triggered by cancer-initiating mutations and can contribute to malignant progression through the recruitment and activation of inflammatory cells. Both extrinsic and intrinsic inflammation can result in immunosuppression, thereby providing a preferred background for tumor development. In clinical trials, lifestyle modifications including healthy diet, exercise, alcohol, and smoking cessation have proven effective in ameliorating inflammation and reducing the risk of cancer-related deaths. In addition, consumption of certain anti-inflammatory drugs, including aspirin, can significantly reduce cancer risk, suggesting that common nonsteroidal anti-inflammatory drugs (NSAID) and more specific COX2 inhibitors can be used in cancer prevention. In addition to being examined for their preventative potential, both NSAIDs and more potent anti-inflammatory antibody-based drugs need to be tested for their ability to augment the efficacy of more conventional therapeutic approaches on the basis of tumor resection, radiation, and cytotoxic chemicals. Cancer Prev Res; 9(12); 895-905. ©2016 AACR.
Collapse
Affiliation(s)
- Jelena Todoric
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California San Diego, La Jolla, California.,Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Laura Antonucci
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California San Diego, La Jolla, California
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California San Diego, La Jolla, California. .,Department of Pathology, School of Medicine, University of California San Diego, La Jolla, California
| |
Collapse
|
39
|
Lucas AL, Malvezzi M, Carioli G, Negri E, La Vecchia C, Boffetta P, Bosetti C. Global Trends in Pancreatic Cancer Mortality From 1980 Through 2013 and Predictions for 2017. Clin Gastroenterol Hepatol 2016; 14:1452-1462.e4. [PMID: 27266982 PMCID: PMC5028258 DOI: 10.1016/j.cgh.2016.05.034] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 05/10/2016] [Accepted: 05/12/2016] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Pancreatic cancer is a leading cause of cancer mortality, and its mortality has not decreased in recent years. We sought to determine global trends in pancreatic cancer mortality. METHODS We derived data on deaths from pancreatic cancer from the World Health Organization database for 59 countries from 1980 through 2013. Age-standardized mortalities were computed for persons of all ages and for persons 35-64 years old; for selected countries, they were computed for persons 25-49 years old. Joinpoint regression models were used to identify significant changes in mortality. For selected larger countries, we predicted number of deaths and mortality for 2017. RESULTS Between 1980 and 2013, overall pancreatic cancer mortality in men increased in the European Union (EU) as well as in Southern and Eastern Europe, Brazil, Japan, and Republic of Korea. Overall pancreatic cancer mortality decreased in most Northern European countries, Australia, Canada, Mexico, and the United States (US). In women, mortality increased in the EU, Brazil, US, Japan, and Republic of Korea but decreased in Canada and Mexico. In 2012, Eastern Europe and Japan had the highest pancreatic cancer mortality for both sexes. In men 25-49 years old, mortality decreased in the EU, US, Japan, and most large European countries. On the basis of our data, we predict overall pancreatic cancer mortality in 2017 to level off in men in the EU and US but increase in Japan. In women, mortality will continue to increase in most countries except the US; the greatest increase is predicted to occur in Japan. CONCLUSIONS Mortality from pancreatic cancer has not decreased as it has for other cancers in recent years. A notable exception is a decrease in mortality in men 25-49 years old, which could indicate a reversal in the current increasing global trends.
Collapse
Affiliation(s)
- Aimee L. Lucas
- Henry D. Janowitz Division of Gastroenterology, Samuel Bronfman Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, US
| | - Matteo Malvezzi
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy,Department of Epidemiology, IRCCS-Istituto di Ricerche Farmacologiche “Mario Negri”, Milan, Italy
| | - Greta Carioli
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Eva Negri
- Department of Epidemiology, IRCCS-Istituto di Ricerche Farmacologiche “Mario Negri”, Milan, Italy
| | - Carlo La Vecchia
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Paolo Boffetta
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, US
| | - Cristina Bosetti
- Department of Epidemiology, IRCCS-Istituto di Ricerche Farmacologiche “Mario Negri”, Milan, Italy
| |
Collapse
|
40
|
Pierce KJ, de Abreu FB, Peterson JD, Suriawinata AA, Tsongalis GJ, Liu X. The genomic profile of pancreatic adenocarcinoma and its relationship to metastatic disease. Exp Mol Pathol 2016; 101:172-175. [PMID: 27498048 DOI: 10.1016/j.yexmp.2016.07.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 07/27/2016] [Indexed: 01/05/2023]
Affiliation(s)
- K J Pierce
- Department of Pathology and Laboratory Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH, United States; Norris Cotton Cancer Center, One Medical Center Drive, Lebanon, NH, United States; The Theodore and Audrey Geisel School of Medicine at Dartmouth, Hanover, NH, United States.
| | - F B de Abreu
- Department of Pathology and Laboratory Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH, United States; Norris Cotton Cancer Center, One Medical Center Drive, Lebanon, NH, United States; The Theodore and Audrey Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - J D Peterson
- Department of Pathology and Laboratory Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH, United States; Norris Cotton Cancer Center, One Medical Center Drive, Lebanon, NH, United States; The Theodore and Audrey Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - A A Suriawinata
- Department of Pathology and Laboratory Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH, United States; Norris Cotton Cancer Center, One Medical Center Drive, Lebanon, NH, United States; The Theodore and Audrey Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - G J Tsongalis
- Department of Pathology and Laboratory Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH, United States; Norris Cotton Cancer Center, One Medical Center Drive, Lebanon, NH, United States; The Theodore and Audrey Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - X Liu
- Department of Pathology and Laboratory Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH, United States; Norris Cotton Cancer Center, One Medical Center Drive, Lebanon, NH, United States; The Theodore and Audrey Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| |
Collapse
|
41
|
Tong M, Andreani T, Krotow A, Gundogan F, de la Monte SM. Potential Contributions of the Tobacco Nicotine-Derived Nitrosamine Ketone to White Matter Molecular Pathology in Fetal Alcohol Spectrum Disorder. ACTA ACUST UNITED AC 2016; 3. [PMID: 28868525 PMCID: PMC5575815 DOI: 10.15436/2377-1348.16.729] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background Fetal alcohol spectrum disorder (FASD) is associated with long-term
deficits in cognitive and motor functions. Previous studies linked
neurodevelopmental abnormalities to increased oxidative stress and white
matter hypotrophy. However, similar effects occur with low-dose nitrosamine
exposures, alcohol abuse correlates with cigarette smoking, and tobacco
smoke contains tobacco-specific nitrosamines, including NNK. Hypothesis Tobacco smoke exposure is a co-factor in FASD. Design Long Evans rat pups were i.p. administered ethanol (2 g/kg) on
postnatal days (P) 2, 4, 6 and/or NNK (2 mg/kg) on P3, P5, and P7 to
simulate third trimester human exposures. Oligodendroglial
myelin-associated, neuroglial, and relevant transcription factor mRNA
transcripts were measured using targeted PCR arrays. Results Ethanol and NNK differentially altered the expression of immature and
mature oligodendroglial, neuronal and astrocytic structural and
plasticity-associated, and various transcription factor genes. NNK’s
effects were broader and more pronounced than ethanol’s, and
additive or synergistic effects of dual exposures impacted expression of all
four categories of genes investigated. Conclusion Developmental exposures to alcohol and NNK (via tobacco smoke)
contribute to sustained abnormalities in brain white matter structure and
function via distinct but overlapping alterations in the expression of genes
that regulate oligodendrocyte survival, maturation and function, neuroglial
structural integrity, and synaptic plasticity. The results support the
hypothesis that smoking may contribute to brain abnormalities associated
with FASD.
Collapse
Affiliation(s)
- Ming Tong
- Department of Medicine, Division of Gastroenterology, and the Liver Research Center Rhode Island Hospital, Providence, RI.,Warren Alpert Medical School of Brown University, Providence, RI
| | - Tomas Andreani
- Department of Medicine, Division of Gastroenterology, and the Liver Research Center Rhode Island Hospital, Providence, RI
| | | | - Fusun Gundogan
- Warren Alpert Medical School of Brown University, Providence, RI.,Department of Pathology, Women and Infants Hospital of Rhode Island, Providence, RI
| | - Suzanne M de la Monte
- Department of Medicine, Division of Gastroenterology, and the Liver Research Center Rhode Island Hospital, Providence, RI.,Warren Alpert Medical School of Brown University, Providence, RI.,Pathobiology Graduate Program, Brown University, Providence, RI.,Departments of Pathology and Neurology, and the Division of Neuropathology, Rhode Island Hospital, Providence, RI
| |
Collapse
|
42
|
Re E, Tong M, de la Monte SM. Tobacco Nitrosamine Exposures Contribute to Fetal Alcohol Spectrum Disorder Associated Cerebellar Dysgenesis. ACTA ACUST UNITED AC 2016; 8:10-21. [PMID: 29201262 PMCID: PMC5711469 DOI: 10.5539/ijb.v8n3p10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Variability in the phenotypic features and severity of fetal alcohol spectrum disorder (FASD) is not fully linked to alcohol dose. We hypothesize that FASD-type neurodevelopmental abnormalities may be caused by exposures to the tobacco-specific nitrosamine, NNK, since a high percentage of pregnant women who drink also smoke. In vitro experiments using PNET2 human cerebellar neuronal cultures examined ethanol and NNK effects on viability and mitochondrial function. Early postnatal rat cerebellar slice cultures were used to examine effects of ethanol and NNK on cerebellar histology and neuroglial and stress protein expression. Ethanol (50 mM) decreased viability and ATP content and increased mitochondrial mass, while NNK (100 μM or higher) selectively inhibited mitochondrial function. The slice culture studies demonstrated striking adverse effects of ethanol, NNK and ethanol+NNK exposures manifested by architectural disorganization of the cortex with relative reductions of internal granule cells, increases in external granule cells, and loss of Purkinje cells. Ethanol, NNK, and ethanol+NNK inhibited expression of choline acetyltransferase (ChAT) and acetylcholinesterase (AChE), and increased levels of 4-hydroxynonenal (HNE). In addition, ethanol increased activated Caspase 3, NNK decreased tau and phospho-tau, and ethanol+NNK inhibited expression of Aspartyl-β-hydroxylase (ASPH), which mediates neuronal migration. In conclusion, ethanol and NNK were shown to exert independent but overlapping adverse effects on cerebellar cortical development, neuronal viability, function, and neuroglial protein expression. These findings support our hypothesis that NNK exposures via tobacco smoking in pregnancy can contribute to FASD-associated neurodevelopmental abnormalities.
Collapse
Affiliation(s)
- Edward Re
- Warren Alpert Medical School of Brown University, Providence, RI
| | - Ming Tong
- Department of Medicine, Division of Gastroenterology, and Liver Research Center Rhode Island Hospital, Providence, RI.,Warren Alpert Medical School of Brown University, Providence, RI
| | - Suzanne M de la Monte
- Department of Medicine, Division of Gastroenterology, and Liver Research Center Rhode Island Hospital, Providence, RI.,Departments of Pathology, Neurology, and Neurosurgery, and the Division of Neuropathology, Rhode Island Hospital, Providence, RI.,Warren Alpert Medical School of Brown University, Providence, RI
| |
Collapse
|
43
|
Increased Incidence of Second Primary Pancreatic Cancer in Patients with Prior Colorectal Cancer: A Population-Based US Study. Dig Dis Sci 2016; 61:1652-60. [PMID: 27107866 DOI: 10.1007/s10620-016-4170-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 04/15/2016] [Indexed: 12/12/2022]
Abstract
BACKGROUND Identifying high-risk groups for pancreatic cancer (PC) may lead to earlier detection. We determined the risk of subsequent PC among survivors of sporadic colorectal cancer (CRC). METHODS We evaluated data from the US Surveillance, Epidemiology, and End Results registry to identify individuals with primary CRC between the years 1973-2006. Standardized incidence ratios (SIRs) and 95 % confidence interval (95 % CI) were calculated to compare the risk of subsequent primary PC in the study cohort to that of the standard population. Analysis was stratified by age at diagnosis of CRC and sex. CRC characteristics were compared among CRC survivors with and without PC. Multivariate sub-hazard ratios were calculated to identify factors associated with subsequent primary PC, using death from non-PC causes as a competing event. RESULTS Of the 273,144 patients with first primary CRC, 657 (0.24 %) developed subsequent PC. CRC survivors were more likely to develop PC (SIR 1.22; 95 % CI 1.09-1.35). Mean latency period (time between CRC and PC diagnosis) was 1, 3, and 5 years from index age of CRC 20-49, 50-64, and >65 years, respectively. Multivariate analysis showed CRC survivors >50 years had 3.5-fold, those with right-sided CRC had 1.2-fold, and those with localized and regional CRCs had sixfold and fivefold increased risk of PC, respectively. CONCLUSION This study suggests that CRC survivors have an increased risk of developing subsequent PC within 1-5 years. CRC survivors age >50 with localized/regional stage, and right-sided CRC have higher predisposition to PC.
Collapse
|
44
|
de la Monte SM, Tong M, Agarwal AR, Cadenas E. Tobacco Smoke-Induced Hepatic Injury with Steatosis, Inflammation, and Impairments in Insulin and Insulin-Like Growth Factor Signaling. ACTA ACUST UNITED AC 2016; 6. [PMID: 27525191 PMCID: PMC4979551 DOI: 10.4172/2161-0681.1000269] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background Alcoholic liver disease (ALD) is associated with impairments in hepatic insulin and insulin-like growth factor (IGF) signaling through cell growth, survival, and metabolic pathways. Since not all heavy drinkers develop ALD, co-factors may be important. Epidemiologic data indicate that most heavy drinkers smoke tobacco and experimental data revealed that low-level nitrosamine exposures, including those from tobacco, can cause steatohepatitis with hepatic insulin/IGF resistance and exacerbate ALD. We hypothesize that cigarette smoke (CS) exposures also cause liver injury with impaired hepatic insulin/IGF signaling, and thereby contribute to ALD. Methods Adult male A/J mice were exposed to air for 8 weeks (A8), CS for 4 (CS4) or 8 (CS8) weeks, or CS for 8 weeks with 2 weeks recovery (CS8+R). Results CS exposures caused progressive liver injury with disruption of the normal hepatic chord architecture, lobular inflammation, apoptosis or necrosis, micro-steatosis, sinusoidal dilatation, and nuclear pleomorphism. Histopathological liver injury scores increased significantly from A8 to CS4 and then further to CS8 (P<0.0001). The mean histological grade was also higher in CS8+R relative to A8 (P<0.0001) but lower than in CS4, reflecting partial resolution of injury by CS withdrawal. CS exposures impaired insulin and IGF-1 signaling through IRS-1, Akt, GSK-3β, and PRAS40. Livers from CS8+R mice had normalized or elevated levels of insulin receptor, pYpY-Insulin-R, 312S-IRS-1, 473S-Akt, S9-GSK-3β, and pT246-PRAS40 relative to A8, CS4, or CS8, reflecting partial recovery. Conclusion CS-mediated liver injury and steatohepatitis with impairments in insulin/IGF signalling are reminiscent of the findings in ALD. Therefore, CS exposures (either first or second-hand) may serve as a co-factor in ALD. The persistence of several abnormalities following CS exposure cessation suggests that some aspects of CS-mediated hepatic metabolic dysfunction are not readily reversible.
Collapse
Affiliation(s)
- Suzanne M de la Monte
- Liver Research Center, Department of Medicine, Rhode Island Hospital and the Alpert Medical School of Brown University, USA; Division of Neuropathology and Departments of Pathology, Neurology, and Neurosurgery, Rhode Island Hospital and the Alpert Medical School of Brown University, USA
| | - M Tong
- Liver Research Center, Department of Medicine, Rhode Island Hospital and the Alpert Medical School of Brown University, USA
| | - A R Agarwal
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | - E Cadenas
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
45
|
Zabala V, Silbermann E, Re E, Andreani T, Tong M, Ramirez T, Gundogan F, de la Monte SM. Potential Co-Factor Role of Tobacco Specific Nitrosamine Exposures in the Pathogenesis of Fetal Alcohol Spectrum Disorder. GYNECOLOGY AND OBSTETRICS RESEARCH : OPEN JOURNAL 2016; 2:112-125. [PMID: 28845454 PMCID: PMC5570438 DOI: 10.17140/goroj-2-125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND Cerebellar developmental abnormalities in Fetal Alcohol Spectrum Disorder (FASD) are linked to impairments in insulin signaling. However, co-morbid alcohol and tobacco abuses during pregnancy are common. Since smoking leads to tobacco specific Nitrosamine (NNK) exposures which have been shown to cause brain insulin resistance, we hypothesized that neurodevelopmental abnormalities in FASD could be mediated by ethanol and/or NNK. METHODS Long Evans rat pups were intraperitoneal (IP) administered ethanol (2 g/kg) on postnatal days (P) 2, 4, 6 and/or NNK (2 mg/kg) on P3, P5, and P7 to simulate third trimester human exposures. The Cerebellar function, histology, insulin and Insulin-like Growth Factor (IGF) signaling, and neuroglial protein expression were assessed. RESULTS Ethanol, NNK and ethanol+NNK groups had significant impairments in motor function (rotarod tests), abnormalities in cerebellar structure (Purkinje cell loss, simplification and irregularity of folia, and altered white matter), signaling through the insulin and IGF-1 receptors, IRS-1, Akt and GSK-3β, and reduced expression of several important neuroglial proteins. Despite similar functional effects, the mechanisms and severity of NNK and ethanol+NNK induced alterations in cerebellar protein expression differed from those of ethanol. CONCLUSIONS Ethanol and NNK exert independent but overlapping adverse effects on cerebellar development, function, insulin signaling through cell survival, plasticity, metabolic pathways, and neuroglial protein expression. The results support the hypothesis that tobacco smoke exposure can serve as a co-factor mediating long-term effects on brain structure and function in FASD.
Collapse
Affiliation(s)
- Valerie Zabala
- Molecular Pharmacology and Physiology Graduate Program, Brown University, Providence, RI, USA
| | | | - Edward Re
- Alpert Medical School of Brown University, Providence, RI, USA
| | - Tomas Andreani
- Graduate Program in Neuroscience, Northwestern University, Chicago, IL, USA
| | - Ming Tong
- Liver Research Center, Division of Gastroenterology and Department of Medicine, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| | | | - Fusun Gundogan
- Department of Pathology, Women and Infants Hospital of Rhode Island, Alpert Medical School of Brown University, Providence, RI, USA
| | - Suzanne M. de la Monte
- Departments of Neurology, Neurosurgery, and Pathology, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
46
|
Andreani T, Tong M, Gundogan F, Silbermann E, de la Monte SM. Differential Effects of 3rd Trimester-Equivalent Binge Ethanol and Tobacco-Specific Nitrosamine Ketone Exposures on Brain Insulin Signaling in Adolescence. JOURNAL OF DIABETES AND RELATED DISORDERS 2016; 1:105. [PMID: 29242853 PMCID: PMC5726776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
BACKGROUND Fetal alcohol spectrum disorder (FASD) is associated with impairments in insulin and insulin-like growth factor (IGF) signaling through Akt pathways and altered expression of neuro-glial proteins needed for structural and functional integrity of the brain. However, alcohol abuse correlates with smoking, and tobacco smoke contains 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK), which like other nitrosamines, impairs insulin and IGF signaling. HYPOTHESIS NNK exposure can serve as a co-factor in mediating long-term neuro-developmental abnormalities associated with FASD. DESIGN Long Evans rat pups were IP administered ethanol (2 g/kg) on postnatal days (P) 2, 4, 6 and/or NNK (2 mg/kg) on P3, P5, and P7, simulating third trimester human exposures. Temporal lobes from P30 rats (young adolescent) were used to measure signaling through the insulin/IGF-1/Akt pathways by multiplex ELISAs, and expression of neuroglial proteins by duplex ELISAs. RESULTS Ethanol, NNK, and ethanol + NNK exposures significantly inhibited insulin receptor tyrosine phosphorylation, and IRS-1 and myelin-associated glycoprotein expression. However, the major long-term adverse effects on Akt pathway downstream signaling and its targeted proteins including choline acetyltransferase, Tau, pTau, ubiquitin, and aspartate-β-hydroxylase were due to NNK rather than ethanol. CONCLUSION Alcohol and tobacco exposures can both contribute to long-term brain abnormalities currently regarded fetal ethanol effects. However, the findings suggest that many of the adverse effects on brain function are attributable to smoking, including impairments in signaling through survival and metabolic pathways, and altered expression of genes that regulate myelin synthesis, maturation and integrity and synaptic plasticity. Therefore, public health measures should address both substances of abuse to prevent "FASD".
Collapse
Affiliation(s)
- Tomas Andreani
- Department of Medicine, Division of Gastroenterology, and the Liver
Research Center Rhode Island Hospital, Providence, RI, USA
| | - Ming Tong
- Department of Medicine, Division of Gastroenterology, and the Liver
Research Center Rhode Island Hospital, Providence, RI, USA
- Warren Alpert Medical School of Brown University, Providence, RI,
USA
| | - Fusun Gundogan
- Department of Pathology, Women and Infants Hospital of Rhode Island,
Providence, RI, USA
- Warren Alpert Medical School of Brown University, Providence, RI,
USA
| | | | - Suzanne M. de la Monte
- Department of Medicine, Division of Gastroenterology, and the Liver
Research Center Rhode Island Hospital, Providence, RI, USA
- Departments of Pathology and Neurology, and the Division of
Neuropathology, Rhode Island Hospital, Providence, RI, USA
- Warren Alpert Medical School of Brown University, Providence, RI,
USA
| |
Collapse
|
47
|
Variation in KRAS driver substitution distributions between tumor types is determined by both mutation and natural selection. Sci Rep 2016; 6:21927. [PMID: 26902163 PMCID: PMC4763303 DOI: 10.1038/srep21927] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 02/02/2016] [Indexed: 02/06/2023] Open
Abstract
Different tumor types vary greatly in their distribution of driver substitutions. Here, we analyzed how mutation and natural selection contribute to differences in the distribution of KRAS driver substitutions between lung, colon and pancreatic adenocarcinomas. We were able to demonstrate that both differences in mutation and differences in selection drive variation in the distribution of KRAS driver substitutions between tumor types. By accounting for the effects of mutation on the distribution of KRAS driver substitutions, we could identify specific KRAS driver substitutions that are more favored by selection in specific tumor types. Such driver substitutions likely improve fitness most when they occur within the context of the tumor type in which they are preferentially favored. Fitting with this, we found that driver substitutions that are more favored by natural selection in a specific type of tumor tend to associate with worse clinical outcomes specifically in that type of tumor.
Collapse
|
48
|
Malvezzi M, Carioli G, Bertuccio P, Rosso T, Boffetta P, Levi F, La Vecchia C, Negri E. European cancer mortality predictions for the year 2016 with focus on leukaemias. Ann Oncol 2016; 27:725-31. [PMID: 26812903 DOI: 10.1093/annonc/mdw022] [Citation(s) in RCA: 133] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 01/11/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Current cancer mortality statistics are important for public health decision-making and resource allocation. Age-standardized rates and numbers of deaths are predicted for 2016 in the European Union (EU). PATIENTS AND METHODS Population and death certification data for stomach, colorectum, pancreas, lung, breast, uterus, prostate, leukaemias and total cancers were obtained from the World Health Organization database and Eurostat. Figures were derived for the EU, France, Germany, Italy, Poland, Spain and the UK. Projected numbers of deaths by age group were obtained for 2016 by linear regression on estimated numbers of deaths over the most recent time period identified by a joinpoint regression model. RESULTS Projected total cancer mortality trends for 2016 in the EU are favourable in both sexes with rates of 133.5/100 000 men and 85.2/100 000 women (8% and 3% falls since 2011) corresponding to 753 600 and 605 900 deaths in men and women for a total number of 1 359 500 projected cancer deaths (+3% compared with 2011, due to population ageing). In men, lung, colorectal and prostate cancer have fallen 11%, 5% and 8%, respectively, since 2011. Breast and colorectal cancer trends in women are favourable (8% and 7% falls, respectively), but lung and pancreatic cancer rates have risen 5% and 4% since 2011 reaching rates of 14.4 and 5.6/100 000 women. Leukaemias show favourable projected mortality for both sexes and all age groups, with stronger falls in the younger age groups. All ages rates are 4.0/100 000 men and 2.5/100 000 women, with falls of 14% and 12% respectively. CONCLUSION The 2016 predictions for EU cancer mortality confirm the favourable trends in rates particularly for men. Lung cancer is likely to be the leading site for female cancer rates. Continuing falls in mortality, larger in children and young adults, are predicted in leukaemias, essentially due to advancements in management and therapy, and their subsequent adoption across Europe.
Collapse
Affiliation(s)
- M Malvezzi
- Department of Epidemiology, IRCCS-Istituto di Ricerche Farmacologiche 'Mario Negri', Milan Department of Clinical Sciences and Community Health, Universitá degli Studi di Milano, Milan, Italy
| | - G Carioli
- Department of Clinical Sciences and Community Health, Universitá degli Studi di Milano, Milan, Italy
| | - P Bertuccio
- Department of Clinical Sciences and Community Health, Universitá degli Studi di Milano, Milan, Italy
| | - T Rosso
- Department of Clinical Sciences and Community Health, Universitá degli Studi di Milano, Milan, Italy
| | - P Boffetta
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - F Levi
- Institute of Social and Preventive Medicine (IUMSP), Lausanne University Hospital, Lausanne, Switzerland
| | - C La Vecchia
- Department of Clinical Sciences and Community Health, Universitá degli Studi di Milano, Milan, Italy
| | - E Negri
- Department of Epidemiology, IRCCS-Istituto di Ricerche Farmacologiche 'Mario Negri', Milan
| |
Collapse
|
49
|
Lucas AL, Bravi F, Boffetta P, Polesel J, Serraino D, La Vecchia C, Bosetti C. Adherence to World Cancer Research Fund/American Institute for Cancer Research recommendations and pancreatic cancer risk. Cancer Epidemiol 2015; 40:15-21. [PMID: 26605429 DOI: 10.1016/j.canep.2015.10.026] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 10/22/2015] [Accepted: 10/27/2015] [Indexed: 01/02/2023]
Abstract
BACKGROUND Pancreatic cancer is a leading cause of cancer death. A role of dietary factors in pancreatic carcinogenesis has been suggested. The World Cancer Research Fund (WCRF) and the American Institute for Cancer Research (AICR) published 8 recommendations for cancer prevention. We evaluated the effect of adherence to the WCRF/AICR recommendations on pancreatic cancer risk. METHODS We operationalized 7 of the 8 WCRF/AICR recommendations to generate a WCRF/AICR score. We examined the association of WCRF/AICR score with pancreatic cancer in data from an Italian case-control study of 326 incident cases and 652 controls. RESULTS Adherence to WCRF/AICR recommendations was associated with a significantly decreased risk of pancreatic cancer. Using a WCRF/AICR score <3.5 as a reference, the adjusted odds ratio (OR) for a score 3.5-<4 was 0.80 (95% CI 0.49, 1.28), for a score 4-<5 0.54 (95% CI 0.35, 0.82), and for score 5 or more 0.41 (95% CI 0.24, 0.68; p-value for trend 0.0002). The OR for a continuous increment of one unit of the WCRF/AICR score was 0.72 (95% CI 0.60, 0.87). CONCLUSION Adherence to the WCRF/AICR recommendations may reduce pancreatic cancer risk.
Collapse
Affiliation(s)
- Aimee L Lucas
- Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, NY, USA.
| | - Francesca Bravi
- Department of Epidemiology, IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri"-IRCCS, Milan, Italy
| | - Paolo Boffetta
- Tisch Cancer Institute and Institute of Translational Epidemiology, Icahn School of Medicine at Mount Sinai, NY, USA
| | - Jerry Polesel
- Unit of Epidemiology and Biostatistics, CRO Aviano National Cancer Institute, Aviano (PN), Italy
| | - Diego Serraino
- Unit of Epidemiology and Biostatistics, CRO Aviano National Cancer Institute, Aviano (PN), Italy
| | - Carlo La Vecchia
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Cristina Bosetti
- Department of Epidemiology, IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri"-IRCCS, Milan, Italy
| |
Collapse
|
50
|
Tong M, Yu R, Silbermann E, Zabala V, Deochand C, de la Monte SM. Differential Contributions of Alcohol and Nicotine-Derived Nitrosamine Ketone (NNK) to White Matter Pathology in the Adolescent Rat Brain. Alcohol Alcohol 2015; 50:680-9. [PMID: 26373813 DOI: 10.1093/alcalc/agv102] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 08/17/2015] [Indexed: 12/30/2022] Open
Abstract
AIM Epidemiologic studies have demonstrated high rates of smoking among alcoholics, and neuroimaging studies have detected white matter atrophy and degeneration in both smokers and individuals with alcohol-related brain disease (ARBD). These findings suggest that tobacco smoke exposure may be a co-factor in ARBD. The present study examines the differential and additive effects of tobacco-specific nitrosamine (NNK) and ethanol exposures on the structural and functional integrity of white matter in an experimental model. METHODS Adolescent Long Evans rats were fed liquid diets containing 0 or 26% ethanol for 8 weeks. In weeks 3-8, rats were treated with nicotine-derived nitrosamine ketone (NNK) (2 mg/kg, 3×/week) or saline by i.p. injection. In weeks 7-8, the ethanol group was binge-administered ethanol (2 g/kg; 3×/week). RESULTS Ethanol, NNK and ethanol + NNK caused striking degenerative abnormalities in white matter myelin and axons, with accompanying reductions in myelin-associated glycoprotein expression. Quantitative RT-PCR targeted array and heatmap analyses demonstrated that ethanol modestly increased, whereas ethanol + NNK sharply increased expression of immature and mature oligodendroglial genes, and that NNK increased immature but inhibited mature oligodendroglial genes. In addition, NNK modulated expression of neuroglial genes in favor of growth cone collapse and synaptic disconnection. Ethanol- and NNK-associated increases in FOXO1, FOXO4 and NKX2-2 transcription factor gene expression could reflect compensatory responses to brain insulin resistance in this model. CONCLUSION Alcohol and tobacco exposures promote ARBD by impairing myelin synthesis, maturation and integrity via distinct but overlapping mechanisms. Public health measures to reduce ARBD should target both alcohol and tobacco abuses.
Collapse
Affiliation(s)
- Ming Tong
- Department of Medicine, Division of Gastroenterology, and the Liver Research Center, Rhode Island Hospital, Providence, RI, USA Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Rosa Yu
- Departments of Pathology and Neurology, and the Division of Neuropathology, Rhode Island Hospital, Providence, RI, USA
| | | | - Valerie Zabala
- Molecular Pharmacology and Physiology Graduate Program, Brown University, Providence, RI, USA
| | - Chetram Deochand
- Biotechnology Graduate Program, Brown University, Providence, RI, USA
| | - Suzanne M de la Monte
- Department of Medicine, Division of Gastroenterology, and the Liver Research Center, Rhode Island Hospital, Providence, RI, USA Warren Alpert Medical School of Brown University, Providence, RI, USA Departments of Pathology and Neurology, and the Division of Neuropathology, Rhode Island Hospital, Providence, RI, USA
| |
Collapse
|