1
|
Liang F, Wang M, Li J, Guo J. The evolution of S-nitrosylation detection methodology and the role of protein S-nitrosylation in various cancers. Cancer Cell Int 2024; 24:408. [PMID: 39702281 DOI: 10.1186/s12935-024-03568-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 11/07/2024] [Indexed: 12/21/2024] Open
Abstract
S-nitrosylation (SNO) modification, a nitric oxide (NO)-mediated post-translational modification (PTM) of proteins, plays an important role in protein microstructure, degradation, activity, and stability. Due to the presence of reducing agents, the SNO modification process mediated by NO derivatives is often reversible and unstable. This reversible transformation between SNO modification and denitrification often influences the structure, activity, and function of proteins. The reversibility of SNO modifications also poses a challenge when verifying changes in the biological functions of proteins. Moreover, SNO modification of key signaling pathway proteins, such as caspase-3, NF-κB, and Bcl-2, can affect tumor proliferation, invasion, and apoptosis. The SNO-modified proteins play important roles in both promoting and inhibiting cancer, which indirectly confirms the duality and complexity of SNO modification functions. This article reviews the biological significance of various SNO-modified proteins in different cancers, providing a theoretical basis for determining whether the related changes of SNO-modified proteins are universal in cancers. Additionally, this review presents a comprehensive and detailed summary of the evolution of detection methods for SNO-modified proteins, providing a possible methodological basis for future research on SNO-modified proteins.
Collapse
Affiliation(s)
- Feng Liang
- Department of General Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Min Wang
- Department of General Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Jiannan Li
- Department of General Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Jie Guo
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun, China.
| |
Collapse
|
2
|
Zhang B, Hong CQ, Lin YW, Luo Y, Ding TY, Xu YW, Peng YH, Wu FC. Association between IGFBP1 expression and cancer risk: A systematic review and meta-analysis. Heliyon 2023; 9:e16470. [PMID: 37251476 PMCID: PMC10220379 DOI: 10.1016/j.heliyon.2023.e16470] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 05/16/2023] [Accepted: 05/17/2023] [Indexed: 05/31/2023] Open
Abstract
BACKGROUND The results regarding the association between insulin-like growth factor binding protein 1 (IGFBP1) expression and cancer risk were controversial. We performed a meta-analysis to provide novel evidence on relationship between IGFBP1 expression and cancer risk. METHODS PubMed, Embase, Cochrane library and Web of science were searched for relevant cohort and case-control studies exploring the relationship between IGFBP1 expression and cancer risk. Odds ratios (ORs) were pooled in this meta-analysis using random model. Subgroup analyses were performed based on ethnicity, tumor types, publication year, study type, Newcastle-Ottawa Scale (NOS) score and sex. RESULTS A total of 27 studies including 16 cohort and 11 case-control studies were identified by literature search. No significant association was found between IGFBP1 expression and risk of various cancers [0.90, 95% confidence interval (CI): 0.79, 1.03]. The overall results showed that the pooled ORs were 0.71 (95% CI: 0.57, 0.88] for prostate cancer risk and 0.66 (95%CI: 0.44, 0.99) for colorectal cancer (CRC) risk. However, there is no significant association between IGFBP1 expression and risk for ovarian cancer (1.70, 95%CI: 0.41, 6.99), breast cancer (1.02, 95%CI: 0.85, 1.23), endometrial cancer (1.19, 95%CI: 0.64, 2.21), colorectal adenoma (0.93; 95%CI: 0.81, 1.07), lung cancer (0.81, 95%CI: 0.39, 1.68) or multiple myeloma (1.20, 95%CI: 0.98, 1.47). CONCLUSION In this study, compared with individuals at low IGFBP1 expression adjusted for age, smoking status, alcohol intake and so on, risk of the prostate cancer and CRC were decreased among individuals of high IGFBP1 expression. There needs further study to confirm this issue.
Collapse
Affiliation(s)
- Biao Zhang
- Department of Clinical Laboratory Medicine, The Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Department of Preventive Medicine, Shantou University Medical College, Shantou China
| | - Chao-Qun Hong
- Esophageal Cancer Prevention and Control Research Center, The Cancer Hospital of Shantou University Medical College Shantou China
| | - Yi-Wei Lin
- Department of Clinical Laboratory Medicine, The Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Esophageal Cancer Prevention and Control Research Center, The Cancer Hospital of Shantou University Medical College Shantou China
| | - Yun Luo
- Department of Clinical Laboratory Medicine, The Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Esophageal Cancer Prevention and Control Research Center, The Cancer Hospital of Shantou University Medical College Shantou China
| | - Tian-Yan Ding
- Department of Clinical Laboratory Medicine, The Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Esophageal Cancer Prevention and Control Research Center, The Cancer Hospital of Shantou University Medical College Shantou China
| | - Yi-Wei Xu
- Department of Clinical Laboratory Medicine, The Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Esophageal Cancer Prevention and Control Research Center, The Cancer Hospital of Shantou University Medical College Shantou China
| | - Yu-Hui Peng
- Department of Clinical Laboratory Medicine, The Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Esophageal Cancer Prevention and Control Research Center, The Cancer Hospital of Shantou University Medical College Shantou China
| | - Fang-Cai Wu
- Esophageal Cancer Prevention and Control Research Center, The Cancer Hospital of Shantou University Medical College Shantou China
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
| |
Collapse
|
3
|
Koterov AN, Ushenkova LN. Causal Criteria in Medical and Biological Disciplines: History, Essence, and Radiation Aspects. Report 4, Part 1: The Post-Hill Criteria and Ecolgoical Criteria. BIOL BULL+ 2023; 49:2423-2466. [PMID: 36845199 PMCID: PMC9944838 DOI: 10.1134/s1062359022120068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 09/10/2021] [Accepted: 12/22/2021] [Indexed: 02/24/2023]
Abstract
Part 1 of Report 4 is focused on the development and modifications of causal criteria after A.B. Hill (1965). Criteria from B. MacMahon et al. (1970-1996), regarded as the first textbook for modern epidemiology, were considered, and it was found that the named researchers did not offer anything new despite the frequent mention of this source in relation to the theme. A similar situation emerged with the criteria of M. Susser: the three obligatory points of this author, "Association" (or "Probability" of causality), "Time order," and "Direction of effect," are trivial, and two more special criteria, which are the development of "Popperian Epidemiology," i.e., "Surviability" of the hypothesis when it is tested by different methods (included in the refinement in Hill's criterion "Consistency of association") and "Predictive performance" of the hypothesis are more theoretical and hardly applicable for the practice of epidemiology and public health. The same restrictions apply to the similar "Popperian" criteria of D.L. Weed, "Predictability" and "Testability" of the causal hypothesis. Although the universal postulates of A.S. Evans for infectious and noninfectious pathologies can be considered exhaustive, they are not used either in epidemiology or in any other discipline practice, except for the field of infectious pathologies, which is probably explained by the complication of the ten-point complex. The little-known criteria of P. Cole (1997) for medical and forensic practice are the most important. The three parts of Hill's criterion-based approaches are important in that they go from a single epidemiological study through a cycle of studies (coupled with the integration of data from other biomedical disciplines) to re-base Hill's criteria for assessing the individual causality of an effect. These constructs complement the earlier guidance from R.E. Gots (1986) on establishing probabilistic personal causation. The collection of causal criteria and the guidelines for environmental disciplines (ecology of biota, human ecoepidemiology, and human ecotoxicology) were considered. The total dominance of inductive causal criteria, both initial and in modifications and with additions, was revealed for an apparently complete base of sources (1979-2020). Adaptations of all known causal schemes based on guidelines have been found, from Henle-Koch postulates to Hill and Susser, including in the international programs and practice of the U.S. Environmental Protection Agency. The Hill Criteria are used by the WHO and other organizations on chemical safety (IPCS) to assess causality in animal experiments for subsequent extrapolation to humans. Data on the assessment of the causality of effects in ecology, ecoepidemiology, and ecotoxicology, together with the use of Hill's criteria for animal experiments, are of significant relevance not only for radiation ecology, but also for radiobiology.
Collapse
Affiliation(s)
- A. N. Koterov
- Burnasyan Federal Medical Biophysical Center, Federal Medical Biological Agency, Moscow, Russia
| | - L. N. Ushenkova
- Burnasyan Federal Medical Biophysical Center, Federal Medical Biological Agency, Moscow, Russia
| |
Collapse
|
4
|
Delgado AB, Tylden ES, Lukic M, Moi L, Busund LTR, Lund E, Olsen KS. Cohort profile: The Clinical and Multi-omic (CAMO) cohort, part of the Norwegian Women and Cancer (NOWAC) study. PLoS One 2023; 18:e0281218. [PMID: 36745618 PMCID: PMC9901780 DOI: 10.1371/journal.pone.0281218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 01/18/2023] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Breast cancer is the most common cancer worldwide and the leading cause of cancer related deaths among women. The high incidence and mortality of breast cancer calls for improved prevention, diagnostics, and treatment, including identification of new prognostic and predictive biomarkers for use in precision medicine. MATERIAL AND METHODS With the aim of compiling a cohort amenable to integrative study designs, we collected detailed epidemiological and clinical data, blood samples, and tumor tissue from a subset of participants from the prospective, population-based Norwegian Women and Cancer (NOWAC) study. These study participants were diagnosed with invasive breast cancer in North Norway before 2013 according to the Cancer Registry of Norway and constitute the Clinical and Multi-omic (CAMO) cohort. Prospectively collected questionnaire data on lifestyle and reproductive factors and blood samples were extracted from the NOWAC study, clinical and histopathological data were manually curated from medical records, and archived tumor tissue collected. RESULTS The lifestyle and reproductive characteristics of the study participants in the CAMO cohort (n = 388) were largely similar to those of the breast cancer patients in NOWAC (n = 10 356). The majority of the cancers in the CAMO cohort were tumor grade 2 and of the luminal A subtype. Approx. 80% were estrogen receptor positive, 13% were HER2 positive, and 12% were triple negative breast cancers. Lymph node metastases were present in 31% at diagnosis. The epidemiological dataset in the CAMO cohort is complemented by mRNA, miRNA, and metabolomics analyses in plasma, as well as miRNA profiling in tumor tissue. Additionally, histological analyses at the level of proteins and miRNAs in tumor tissue are currently ongoing. CONCLUSION The CAMO cohort provides data suitable for epidemiological, clinical, molecular, and multi-omics investigations, thereby enabling a systems epidemiology approach to translational breast cancer research.
Collapse
Affiliation(s)
- André Berli Delgado
- Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway
- * E-mail:
| | - Eline Sol Tylden
- Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Marko Lukic
- Department of Community Medicine, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Line Moi
- Department of Clinical Pathology, University Hospital of North Norway, Tromsø, Norway
| | - Lill-Tove Rasmussen Busund
- Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway
- Department of Clinical Pathology, University Hospital of North Norway, Tromsø, Norway
| | - Eiliv Lund
- Department of Community Medicine, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Karina Standahl Olsen
- Department of Community Medicine, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
5
|
Barajas R, Hair B, Lai G, Rotunno M, Shams-White MM, Gillanders EM, Mechanic LE. Facilitating cancer systems epidemiology research. PLoS One 2022; 16:e0255328. [PMID: 34972102 PMCID: PMC8719747 DOI: 10.1371/journal.pone.0255328] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Systems epidemiology offers a more comprehensive and holistic approach to studies of cancer in populations by considering high dimensionality measures from multiple domains, assessing the inter-relationships among risk factors, and considering changes over time. These approaches offer a framework to account for the complexity of cancer and contribute to a broader understanding of the disease. Therefore, NCI sponsored a workshop in February 2019 to facilitate discussion about the opportunities and challenges of the application of systems epidemiology approaches for cancer research. Eight key themes emerged from the discussion: transdisciplinary collaboration and a problem-based approach; methods and modeling considerations; interpretation, validation, and evaluation of models; data needs and opportunities; sharing of data and models; enhanced training practices; dissemination of systems models; and building a systems epidemiology community. This manuscript summarizes these themes, highlights opportunities for cancer systems epidemiology research, outlines ways to foster this research area, and introduces a collection of papers, "Cancer System Epidemiology Insights and Future Opportunities" that highlight findings based on systems epidemiology approaches.
Collapse
Affiliation(s)
- Rolando Barajas
- Epidemiology and Genomics Research Program, Division of Cancer Control and Population Sciences (DCCPS), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Brionna Hair
- DCCPS, NCI, NIH, Bethesda, Maryland, United States of America
| | - Gabriel Lai
- Epidemiology and Genomics Research Program, Division of Cancer Control and Population Sciences (DCCPS), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Melissa Rotunno
- Epidemiology and Genomics Research Program, Division of Cancer Control and Population Sciences (DCCPS), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Marissa M. Shams-White
- Epidemiology and Genomics Research Program, Division of Cancer Control and Population Sciences (DCCPS), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Elizabeth M. Gillanders
- Epidemiology and Genomics Research Program, Division of Cancer Control and Population Sciences (DCCPS), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Leah E. Mechanic
- Epidemiology and Genomics Research Program, Division of Cancer Control and Population Sciences (DCCPS), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
6
|
Rios Guzman E, Hultquist JF. Clinical and biological consequences of respiratory syncytial virus genetic diversity. Ther Adv Infect Dis 2022; 9:20499361221128091. [PMID: 36225856 PMCID: PMC9549189 DOI: 10.1177/20499361221128091] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 08/30/2022] [Indexed: 11/17/2022] Open
Abstract
Respiratory syncytial virus (RSV) is one of the most common etiological agents of global acute respiratory tract infections with a disproportionate burden among infants, individuals over the age of 65, and immunocompromised populations. The two major subtypes of RSV (A and B) co-circulate with a predominance of either group during different epidemic seasons, with frequently emerging genotypes due to RSV's high genetic variability. Global surveillance systems have improved our understanding of seasonality, disease burden, and genomic evolution of RSV through genotyping by sequencing of attachment (G) glycoprotein. However, the integration of these systems into international infrastructures is in its infancy, resulting in a relatively low number (~2200) of publicly available RSV genomes. These limitations in surveillance hinder our ability to contextualize RSV evolution past current canonical attachment glycoprotein (G)-oriented understanding, thus resulting in gaps in understanding of how genetic diversity can play a role in clinical outcome, therapeutic efficacy, and the host immune response. Furthermore, utilizing emerging RSV genotype information from surveillance and testing the impact of viral evolution using molecular techniques allows us to establish causation between the clinical and biological consequences of arising genotypes, which subsequently aids in informed vaccine design and future vaccination strategy. In this review, we aim to discuss the findings from current molecular surveillance efforts and the gaps in knowledge surrounding the consequence of RSV genetic diversity on disease severity, therapeutic efficacy, and RSV-host interactions.
Collapse
Affiliation(s)
- Estefany Rios Guzman
- Department of Medicine, Division of Infectious
Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL,
USA
- Center for Pathogen Genomics and Microbial
Evolution, Institute for Global Health, Northwestern University Feinberg
School of Medicine, Chicago, IL, USA
| | - Judd F. Hultquist
- Robert H. Lurie Medical Research Center,
Northwestern University, 9-141, 303 E. Superior St., Chicago, IL 60611,
USA
- Department of Medicine, Division of Infectious
Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL,
USA
- Center for Pathogen Genomics and Microbial
Evolution, Institute for Global Health, Northwestern University Feinberg
School of Medicine, Chicago, IL, USA
| |
Collapse
|
7
|
Park J, Choi J, Choi JY. Network Analysis in Systems Epidemiology. J Prev Med Public Health 2021; 54:259-264. [PMID: 34370939 PMCID: PMC8357545 DOI: 10.3961/jpmph.21.190] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/28/2021] [Accepted: 06/21/2021] [Indexed: 11/23/2022] Open
Abstract
Traditional epidemiological studies have identified a number of risk factors for various diseases using regression-based methods that examine the association between an exposure and an outcome (i.e., one-to-one correspondences). One of the major limitations of this approach is the "black-box" aspect of the analysis, in the sense that this approach cannot fully explain complex relationships such as biological pathways. With high-throughput data in current epidemiology, comprehensive analyses are needed. The network approach can help to integrate multi-omics data, visualize their interactions or relationships, and make inferences in the context of biological mechanisms. This review aims to introduce network analysis for systems epidemiology, its procedures, and how to interpret its findings.
Collapse
Affiliation(s)
- JooYong Park
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea
| | - Jaesung Choi
- Institute of Health Policy and Management, Seoul National University Medical Research Center, Seoul, Korea
| | - Ji-Yeob Choi
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea
- Institute of Health Policy and Management, Seoul National University Medical Research Center, Seoul, Korea
- Cancer Research Institute, Seoul National University, Seoul, Korea
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
8
|
Shams-White MM, Barajas R, Jensen RE, Rotunno M, Dueck H, Ginexi EM, Rogers SD, Gillanders EM, Mechanic LE. Systems epidemiology and cancer: A review of the National Institutes of Health extramural grant portfolio 2013-2018. PLoS One 2021; 16:e0250061. [PMID: 33857240 PMCID: PMC8049352 DOI: 10.1371/journal.pone.0250061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVES Systems epidemiology approaches may lead to a better understanding of the complex and dynamic multi-level constellation of contributors to cancer risk and outcomes and help target interventions. This grant portfolio analysis aimed to describe the National Institutes of Health (NIH) and the National Cancer Institute (NCI) investments in systems epidemiology and to identify gaps in the cancer systems epidemiology portfolio. METHODS The analysis examined grants funded (2013-2018) through seven NIH systems science Funding Opportunity Announcements (FOAs) as well as cancer-specific systems epidemiology grants funded by NCI during that same time. Study characteristics were extracted from the grant abstracts and specific aims and coded. RESULTS Of the 137 grants awarded under the NIH FOAs, 52 (38%) included systems epidemiology. Only five (4%) were focused on cancer systems epidemiology. The NCI-wide search (N = 453 grants) identified 35 grants (8%) that included cancer systems epidemiology in their specific aims. Most of these grants examined epidemiology and surveillance-based questions (60%); fewer addressed clinical care or clinical trials (37%). Fifty-four percent looked at multiple scales within the individual (e.g., cell, tissue, organ), 49% looked beyond the individual (e.g., individual, community, population), and few (9%) included both. Across all grants examined, the systems epidemiology grants primarily focused on discovery or prediction, rather than on impacts of intervention or policy. CONCLUSIONS The most notable finding was that grants focused on cancer versus other diseases reflected a small percentage of the portfolio, highlighting the need to encourage more cancer systems epidemiology research. Opportunities include encouraging more multiscale research and continuing the support for broad examination of domains in these studies. Finally, the nascent discipline of systems epidemiology could benefit from the creation of standard terminology and definitions to guide future progress.
Collapse
Affiliation(s)
- Marissa M. Shams-White
- Risk Factor Assessment Branch, Epidemiology and Genomics Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Rolando Barajas
- Genomics Epidemiology Branch, Epidemiology and Genomics Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Roxanne E. Jensen
- Outcomes Research Branch, Healthcare Delivery Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Melissa Rotunno
- Genomics Epidemiology Branch, Epidemiology and Genomics Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Hannah Dueck
- Tumor Biology and Microenvironment Branch, Division of Cancer Biology, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Elizabeth M. Ginexi
- Office of Behavioral and Social Sciences Research, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Scott D. Rogers
- Epidemiology and Genomics Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Elizabeth M. Gillanders
- Genomics Epidemiology Branch, Epidemiology and Genomics Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Leah E. Mechanic
- Genomics Epidemiology Branch, Epidemiology and Genomics Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, Maryland, United States of America
| |
Collapse
|
9
|
Olsen KS, Holden M, Thalabard JC, Rasmussen Busund LT, Lund E, Holden L. Global blood gene expression profiles following a breast cancer diagnosis-Clinical follow-up in the NOWAC post-genome cohort. PLoS One 2021; 16:e0246650. [PMID: 33684121 PMCID: PMC7939296 DOI: 10.1371/journal.pone.0246650] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 01/23/2021] [Indexed: 11/19/2022] Open
Abstract
Objective This explorative study aimed to assess if there are any time-dependent blood gene expression changes during the first one to eight years after breast cancer diagnosis, which can be linked to the clinical outcome of the disease. Material and methods A random distribution of follow-up time from breast cancer diagnosis till blood sampling was obtained by a nested, matched case-control design in the Norwegian Women and Cancer Post-genome Cohort. From 2002–5, women were invited to donate blood samples, regardless of any cancer diagnosis. At end of the study period in 2015, any cancer diagnoses in the 50 000 participants were obtained via linkage to the Norwegian Cancer Registry. For each breast cancer patient (n = 415), an age- and storage time-matched control was drawn. The design gave a uniform, random length of follow-up time, independent of cancer stage. Differences in blood gene expression between breast cancer cases and controls were identified using the Bioconductor R-package limma, using a moving window in time, to handle the varying time elapsed from diagnosis to blood sample. Results The number of differentially expressed genes between cases and controls were close to 2,000 in the first year after diagnosis, but fell sharply the second year. During the next years, a transient second increase was observed, but only in women with metastatic disease who later died, both compared to invasive cases that survived (p<0,001) and to metastatic cases that survived (p = 0.024). Among the differentially expressed genes there was an overrepresentation of heme metabolism and T cell-related processes. Conclusion This explorative analysis identified changing trajectories in the years after diagnosis, depending on clinical stage. Hypothetically, this could represent the escape of the metastatic cancer from the immune system.
Collapse
Affiliation(s)
| | | | | | - Lill-Tove Rasmussen Busund
- UiT The Arctic University of Norway, Tromsø, Norway
- The University Hospital of North Norway, Tromsø, Norway
| | - Eiliv Lund
- UiT The Arctic University of Norway, Tromsø, Norway
- The Cancer Registry of Norway, Oslo, Norway
| | | |
Collapse
|
10
|
Ma H, Shen H. From human genome epidemiology to systems epidemiology: current progress and future perspective. J Biomed Res 2020; 34:323-327. [PMID: 32648851 PMCID: PMC7540239 DOI: 10.7555/jbr.34.20200027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
The recent progress in human genome epidemiology (HuGE) is already having a profound impact on the practice of medicine and public health. First, the success of genome-wide association studies has greatly expanded the direction and content of epidemiological researches, including revealing new genetic mechanisms of complex diseases, identifying new targets for therapeutic interventions, and improving application in early screening of high-risk populations. At the same time, large-scale genomic studies make it possible to efficiently explore the gene-environment interactions, which will help better understand the biological pathways of complex diseases and identify individuals who may be more susceptible to diseases. Additionally, the emergence of systems epidemiology aims to integrate multi-omics together with epidemiological data to create a systems network that can comprehensively characterize the diverse range of factors contributing to disease development. These progress will help to apply HuGE findings into practice to improve the health of individuals and populations.
Collapse
Affiliation(s)
- Hongxia Ma
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Hongbing Shen
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| |
Collapse
|
11
|
A Call for Systems Epidemiology to Tackle the Complexity of Schistosomiasis, Its Control, and Its Elimination. Trop Med Infect Dis 2019; 4:tropicalmed4010021. [PMID: 30699922 PMCID: PMC6473336 DOI: 10.3390/tropicalmed4010021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 01/22/2019] [Accepted: 01/24/2019] [Indexed: 12/20/2022] Open
Abstract
Ever since the first known written report of schistosomiasis in the mid-19th century, researchers have aimed to increase knowledge of the parasites, their hosts, and the mechanisms contributing to infection and disease. This knowledge generation has been paramount for the development of improved intervention strategies. Yet, despite a broad knowledge base of direct risk factors for schistosomiasis, there remains a paucity of information related to more complex, interconnected, and often hidden drivers of transmission that hamper intervention successes and sustainability. Such complex, multidirectional, non-linear, and synergistic interdependencies are best understood by looking at the integrated system as a whole. A research approach able to address this complexity and find previously neglected causal mechanisms for transmission, which include a wide variety of influencing factors, is needed. Systems epidemiology, as a holistic research approach, can integrate knowledge from classical epidemiology, with that of biology, ecology, social sciences, and other disciplines, and link this with informal, tacit knowledge from experts and affected populations. It can help to uncover wider-reaching but difficult-to-identify processes that directly or indirectly influence exposure, infection, transmission, and disease development, as well as how these interrelate and impact one another. Drawing on systems epidemiology to address persisting disease hotspots, failed intervention programmes, and systematically neglected population groups in mass drug administration programmes and research studies, can help overcome barriers in the progress towards schistosomiasis elimination. Generating a comprehensive view of the schistosomiasis system as a whole should thus be a priority research agenda towards the strategic goal of morbidity control and transmission elimination.
Collapse
|
12
|
Lund E, Rasmussen Busund LT, Thalabard JC. Rethinking the carcinogenesis of breast cancer: The theory of breast cancer as a child deficiency disease or a pseudo semi-allograft. Med Hypotheses 2018; 120:76-80. [PMID: 30220347 DOI: 10.1016/j.mehy.2018.08.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 08/12/2018] [Accepted: 08/22/2018] [Indexed: 11/19/2022]
Abstract
The theory of breast cancer as a child deficiency disease is an inversion of the current paradigm, which considers full-term pregnancies to be a protective factor and uses nulliparous women as the reference group. Instead, the theory of breast cancer as a child deficiency disease says that women with the highest parity (about 20, which is the limit of human fertility) are those with the lowest risk and should be used as the reference group in risk estimations. This theory is explained biologically by converting parity from the simple value of number of children into an understanding of the long-lasting biological and immunological effects of pregnancy. These effects can be reflected, as measured by functional genomics, in gene expression of the immune cells in the blood. Each pregnancy represents a unique fetus or semi-allograft, which provokes the creation and deposit of memory cell clones in the mother. Gene expression levels have been found to change linearly with number of full-term pregnancies in healthy women, but not in breast cancer patients. High hormone levels are necessary for a successful pregnancy, as they modulate the immune response from adaptive to innate in order to protect the fetus (considered as a semi-allograft) from rejection. At the end of the pregnancy, hormone levels drop, and the immune system recognizes the semi-allograft, but not in time for rejection to occur before birth. High hormones levels are also classified as carcinogens illustrating that carcinogenesis in the breast could be viewed as a war or balance between later exposures to hormonal carcinogens and the protection of the immune system. We propose that breast tumors are pseudo semi-allografts made up of transformed breast tissue cells. Assuming that the sensitivity to the exposure to increased levels of endogenous or exogenous hormones in women with breast cancer mimic those that occur in pregnancy, these breast tumor cells are protected against the body's immune reaction, just as the fetus is during pregnancy. However, with more pregnancies, the potential to eradicate the pseudo semi-allograft might increase due to enhanced immune surveillance. The theory of breast cancer as a child deficiency disease proposes that the protective effect of pregnancy on breast cancer incidence via the immune system is independent of other risk factors.
Collapse
Affiliation(s)
- Eiliv Lund
- UiT, The Arctic University of Norway, Tromsø, Norway; The Cancer Registry of Norway, Oslo, Norway.
| | - Lill-Tove Rasmussen Busund
- UiT, The Arctic University of Norway, Tromsø, Norway; The University Hospital of North Norway, Tromsø, Norway
| | | |
Collapse
|
13
|
Xia S, Zhou XN, Liu J. Systems thinking in combating infectious diseases. Infect Dis Poverty 2017; 6:144. [PMID: 28893320 PMCID: PMC5594605 DOI: 10.1186/s40249-017-0339-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 07/26/2017] [Indexed: 12/18/2022] Open
Abstract
The transmission of infectious diseases is a dynamic process determined by multiple factors originating from disease pathogens and/or parasites, vector species, and human populations. These factors interact with each other and demonstrate the intrinsic mechanisms of the disease transmission temporally, spatially, and socially. In this article, we provide a comprehensive perspective, named as systems thinking, for investigating disease dynamics and associated impact factors, by means of emphasizing the entirety of a system’s components and the complexity of their interrelated behaviors. We further develop the general steps for performing systems approach to tackling infectious diseases in the real-world settings, so as to expand our abilities to understand, predict, and mitigate infectious diseases.
Collapse
Affiliation(s)
- Shang Xia
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, 200025, People's Republic of China.,Key Laboratory of Parasite and Vector Biology, National Health and Family Planning Commission, Shanghai, 200025, People's Republic of China.,WHO Collaborating Centre for Tropical Diseases, Shanghai, 200025, People's Republic of China.,CDC-NIPD & HKBU-CSD Joint Research Laboratory for Intelligent Disease Surveillance and Control, Shanghai, 200025, People's Republic of China
| | - Xiao-Nong Zhou
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, 200025, People's Republic of China.,Key Laboratory of Parasite and Vector Biology, National Health and Family Planning Commission, Shanghai, 200025, People's Republic of China.,WHO Collaborating Centre for Tropical Diseases, Shanghai, 200025, People's Republic of China.,CDC-NIPD & HKBU-CSD Joint Research Laboratory for Intelligent Disease Surveillance and Control, Shanghai, 200025, People's Republic of China
| | - Jiming Liu
- Department of Computer Science, Hong Kong Baptist University, Kowloon Tong, Hong Kong. .,CDC-NIPD & HKBU-CSD Joint Research Laboratory for Intelligent Disease Surveillance and Control, Shanghai, 200025, People's Republic of China.
| |
Collapse
|
14
|
Visceral adipose tissue but not subcutaneous adipose tissue is associated with urine and serum metabolites. PLoS One 2017; 12:e0175133. [PMID: 28403191 PMCID: PMC5389790 DOI: 10.1371/journal.pone.0175133] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 03/21/2017] [Indexed: 12/27/2022] Open
Abstract
Obesity is a complex multifactorial phenotype that influences several metabolic pathways. Yet, few studies have examined the relations of different body fat compartments to urinary and serum metabolites. Anthropometric phenotypes (visceral adipose tissue (VAT), subcutaneous adipose tissue (SAT), the ratio between VAT and SAT (VSR), body mass index (BMI), waist circumference (WC)) and urinary and serum metabolite concentrations measured by nuclear magnetic resonance spectroscopy were measured in a population-based sample of 228 healthy adults. Multivariable linear and logistic regression models, corrected for multiple testing using the false discovery rate, were used to associate anthropometric phenotypes with metabolites. We adjusted for potential confounding variables: age, sex, smoking, physical activity, menopausal status, estimated glomerular filtration rate (eGFR), urinary glucose, and fasting status. In a fully adjusted logistic regression model dichotomized for the absence or presence of quantifiable metabolite amounts, VAT, BMI and WC were inversely related to urinary choline (ß = -0.18, p = 2.73*10−3), glycolic acid (ß = -0.20, 0.02), and guanidinoacetic acid (ß = -0.12, p = 0.04), and positively related to ethanolamine (ß = 0.18, p = 0.02) and dimethylamine (ß = 0.32, p = 0.02). BMI and WC were additionally inversely related to urinary glutamine and lactic acid. Moreover, WC was inversely associated with the detection of serine. VAT, but none of the other anthropometric parameters, was related to serum essential amino acids, such as valine, isoleucine, and phenylalanine among men. Compared to other adiposity measures, VAT demonstrated the strongest and most significant relations to urinary and serum metabolites. The distinct relations of VAT, SAT, VSR, BMI, and WC to metabolites emphasize the importance of accurately differentiating between body fat compartments when evaluating the potential role of metabolic regulation in the development of obesity-related diseases, such as insulin resistance, type 2 diabetes, and cardiovascular disease.
Collapse
|
15
|
A powerful weighted statistic for detecting group differences of directed biological networks. Sci Rep 2016; 6:34159. [PMID: 27686331 PMCID: PMC5054825 DOI: 10.1038/srep34159] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 09/08/2016] [Indexed: 12/15/2022] Open
Abstract
Complex disease is largely determined by a number of biomolecules interwoven into networks, rather than a single biomolecule. Different physiological conditions such as cases and controls may manifest as different networks. Statistical comparison between biological networks can provide not only new insight into the disease mechanism but statistical guidance for drug development. However, the methods developed in previous studies are inadequate to capture the changes in both the nodes and edges, and often ignore the network structure. In this study, we present a powerful weighted statistical test for group differences of directed biological networks, which is independent of the network attributes and can capture the changes in both the nodes and edges, as well as simultaneously accounting for the network structure through putting more weights on the difference of nodes locating on relatively more important position. Simulation studies illustrate that this method had better performance than previous ones under various sample sizes and network structures. One application to GWAS of leprosy successfully identifies the specific gene interaction network contributing to leprosy. Another real data analysis significantly identifies a new biological network, which is related to acute myeloid leukemia. One potential network responsible for lung cancer has also been significantly detected. The source R code is available on our website.
Collapse
|
16
|
Yuan Z, Ji J, Zhang T, Liu Y, Zhang X, Chen W, Xue F. A novel chi-square statistic for detecting group differences between pathways in systems epidemiology. Stat Med 2016; 35:5512-5524. [PMID: 27605026 DOI: 10.1002/sim.7094] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 08/01/2016] [Accepted: 08/16/2016] [Indexed: 12/15/2022]
Abstract
Traditional epidemiology often pays more attention to the identification of a single factor rather than to the pathway that is related to a disease, and therefore, it is difficult to explore the disease mechanism. Systems epidemiology aims to integrate putative lifestyle exposures and biomarkers extracted from multiple omics platforms to offer new insights into the pathway mechanisms that underlie disease at the human population level. One key but inadequately addressed question is how to develop powerful statistics to identify whether one candidate pathway is associated with a disease. Bearing in mind that a pathway difference can result from not only changes in the nodes but also changes in the edges, we propose a novel statistic for detecting group differences between pathways, which in principle, captures the nodes changes and edge changes, as well as simultaneously accounting for the pathway structure simultaneously. The proposed test has been proven to follow the chi-square distribution, and various simulations have shown it has better performance than other existing methods. Integrating genome-wide DNA methylation data, we analyzed one real data set from the Bogalusa cohort study and significantly identified a potential pathway, Smoking → SOCS3 → PIK3R1, which was strongly associated with abdominal obesity. The proposed test was powerful and efficient at identifying pathway differences between two groups, and it can be extended to other disciplines that involve statistical comparisons between pathways. The source code in R is available on our website. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Zhongshang Yuan
- Department of Biostatistics, School of Public Health, Shandong University, Jinan, 250012, Shandong, China
| | - Jiadong Ji
- Department of Biostatistics, School of Public Health, Shandong University, Jinan, 250012, Shandong, China
| | - Tao Zhang
- Department of Biostatistics, School of Public Health, Shandong University, Jinan, 250012, Shandong, China.,Department of Epidemiology, Tulane University Health Sciences Center, Tulane University, New Orleans, LA, U.S.A
| | - Yi Liu
- Department of Biostatistics, School of Public Health, Shandong University, Jinan, 250012, Shandong, China
| | - Xiaoshuai Zhang
- Department of Biostatistics, School of Public Health, Shandong University, Jinan, 250012, Shandong, China
| | - Wei Chen
- Department of Epidemiology, Tulane University Health Sciences Center, Tulane University, New Orleans, LA, U.S.A
| | - Fuzhong Xue
- Department of Biostatistics, School of Public Health, Shandong University, Jinan, 250012, Shandong, China
| |
Collapse
|
17
|
Lund E, Holden L, Bøvelstad H, Plancade S, Mode N, Günther CC, Nuel G, Thalabard JC, Holden M. A new statistical method for curve group analysis of longitudinal gene expression data illustrated for breast cancer in the NOWAC postgenome cohort as a proof of principle. BMC Med Res Methodol 2016; 16:28. [PMID: 26944545 PMCID: PMC4779232 DOI: 10.1186/s12874-016-0129-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 02/25/2016] [Indexed: 01/28/2023] Open
Abstract
Background The understanding of changes in temporal processes related to human carcinogenesis is limited. One approach for prospective functional genomic studies is to compile trajectories of differential expression of genes, based on measurements from many case-control pairs. We propose a new statistical method that does not assume any parametric shape for the gene trajectories. Methods The trajectory of a gene is defined as the curve representing the changes in gene expression levels in the blood as a function of time to cancer diagnosis. In a nested case–control design it consists of differences in gene expression levels between cases and controls. Genes can be grouped into curve groups, each curve group corresponding to genes with a similar development over time. The proposed new statistical approach is based on a set of hypothesis testing that can determine whether or not there is development in gene expression levels over time, and whether this development varies among different strata. Curve group analysis may reveal significant differences in gene expression levels over time among the different strata considered. This new method was applied as a “proof of concept” to breast cancer in the Norwegian Women and Cancer (NOWAC) postgenome cohort, using blood samples collected prospectively that were specifically preserved for transcriptomic analyses (PAX tube). Cohort members diagnosed with invasive breast cancer through 2009 were identified through linkage to the Cancer Registry of Norway, and for each case a random control from the postgenome cohort was also selected, matched by birth year and time of blood sampling, to create a case-control pair. After exclusions, 441 case-control pairs were available for analyses, in which we considered strata of lymph node status at time of diagnosis and time of diagnosis with respect to breast cancer screening visits. Results The development of gene expression levels in the NOWAC postgenome cohort varied in the last years before breast cancer diagnosis, and this development differed by lymph node status and participation in the Norwegian Breast Cancer Screening Program. The differences among the investigated strata appeared larger in the year before breast cancer diagnosis compared to earlier years. Conclusions This approach shows good properties in term of statistical power and type 1 error under minimal assumptions. When applied to a real data set it was able to discriminate between groups of genes with non-linear similar patterns before diagnosis.
Collapse
Affiliation(s)
- Eiliv Lund
- Department of Community Medicine, Pb. 5060, UiT The Arctic University of Norway, 9037, Tromsø, Norway.
| | | | - Hege Bøvelstad
- Department of Community Medicine, Pb. 5060, UiT The Arctic University of Norway, 9037, Tromsø, Norway.
| | - Sandra Plancade
- INRA, UR1404 Unité Mathématiques et Informatique Appliquées du Génome à l'Environnement, Jouy-en-Josas, France.
| | - Nicolle Mode
- Department of Community Medicine, Pb. 5060, UiT The Arctic University of Norway, 9037, Tromsø, Norway.
| | | | - Gregory Nuel
- CNRS, INSMI Stochastics and Biology Group (PSB) LPMA, UPMC, Sorbonne University, Paris, France.
| | | | | |
Collapse
|
18
|
Ji J, Yuan Z, Zhang X, Xue F. A powerful score-based statistical test for group difference in weighted biological networks. BMC Bioinformatics 2016; 17:86. [PMID: 26867929 PMCID: PMC4751708 DOI: 10.1186/s12859-016-0916-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 01/29/2016] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Complex disease is largely determined by a number of biomolecules interwoven into networks, rather than a single biomolecule. A key but inadequately addressed issue is how to test possible differences of the networks between two groups. Group-level comparison of network properties may shed light on underlying disease mechanisms and benefit the design of drug targets for complex diseases. We therefore proposed a powerful score-based statistic to detect group difference in weighted networks, which simultaneously capture the vertex changes and edge changes. RESULTS Simulation studies indicated that the proposed network difference measure (NetDifM) was stable and outperformed other methods existed, under various sample sizes and network topology structure. One application to real data about GWAS of leprosy successfully identified the specific gene interaction network contributing to leprosy. For additional gene expression data of ovarian cancer, two candidate subnetworks, PI3K-AKT and Notch signaling pathways, were considered and identified respectively. CONCLUSIONS The proposed method, accounting for the vertex changes and edge changes simultaneously, is valid and powerful to capture the group difference of biological networks.
Collapse
Affiliation(s)
- Jiadong Ji
- Department of Biostatistics, School of Public Health, Shandong University, PO Box 100, Jinan, 250012, Shandong, China.
| | - Zhongshang Yuan
- Department of Biostatistics, School of Public Health, Shandong University, PO Box 100, Jinan, 250012, Shandong, China.
| | - Xiaoshuai Zhang
- Department of Biostatistics, School of Public Health, Shandong University, PO Box 100, Jinan, 250012, Shandong, China.
| | - Fuzhong Xue
- Department of Biostatistics, School of Public Health, Shandong University, PO Box 100, Jinan, 250012, Shandong, China.
| |
Collapse
|
19
|
Mejía-Aranguré JM. Molecular epidemiology of acute leukemia in children: causal model, interaction of three factors-susceptibility, environmental exposure and vulnerability period. BOLETIN MEDICO DEL HOSPITAL INFANTIL DE MEXICO 2016; 73:55-63. [PMID: 29421234 DOI: 10.1016/j.bmhimx.2015.12.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 12/03/2015] [Indexed: 01/07/2023] Open
Abstract
Acute leukemias have a huge morphological, cytogenetic and molecular heterogeneity and genetic polymorphisms associated with susceptibility. Every leukemia presents causal factors associated with the development of the disease. Particularly, when three factors are present, they result in the development of acute leukemia. These phenomena are susceptibility, environmental exposure and a period that, for this model, has been called the period of vulnerability. This framework shows how the concepts of molecular epidemiology have established a reference from which it is more feasible to identify the environmental factors associated with the development of leukemia in children. Subsequently, the arguments show that only susceptible children are likely to develop leukemia once exposed to an environmental factor. For additional exposure, if the child is not susceptible to leukemia, the disease does not develop. In addition, this exposure should occur during a time window when hematopoietic cells and their environment are more vulnerable to such interaction, causing the development of leukemia. This model seeks to predict the time when the leukemia develops and attempts to give a context in which the causality of childhood leukemia should be studied. This information can influence and reduce the risk of a child developing leukemia.
Collapse
Affiliation(s)
- Juan Manuel Mejía-Aranguré
- Unidad de Investigación en Epidemiología Clínica, Unidad Médica de Alta Especialidad, Hospital de Pediatría, Centro Médico Nacional Siglo XXI and Coordinación de Investigación en Salud, Instituto Mexicano del Seguro Social, Ciudad de México, México.
| |
Collapse
|
20
|
Lund E, Plancade S, Nuel G, Bøvelstad H, Thalabard JC. A processual model for functional analyses of carcinogenesis in the prospective cohort design. Med Hypotheses 2015; 85:494-7. [PMID: 26187155 PMCID: PMC4579405 DOI: 10.1016/j.mehy.2015.07.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 07/08/2015] [Indexed: 12/04/2022]
Abstract
Traditionally, the prospective design has been chosen for risk factor analyses of lifestyle and cancer using mainly estimation by survival analysis methods. With new technologies, epidemiologists can expand their prospective studies to include functional genomics given either as transcriptomics, mRNA and microRNA, or epigenetics in blood or other biological materials. The novel functional analyses should not be assessed using classical survival analyses since the main goal is not risk estimation, but the analysis of functional genomics as part of the dynamic carcinogenic process over time, i.e., a "processual" approach. In the risk factor model, time to event is analysed as a function of exposure variables known at start of follow-up (fixed covariates) or changing over the follow-up period (time-dependent covariates). In the processual model, transcriptomics or epigenetics is considered as functions of time and exposures. The success of this novel approach depends on the development of new statistical methods with the capacity of describing and analysing the time-dependent curves or trajectories for tens of thousands of genes simultaneously. This approach also focuses on multilevel or integrative analyses introducing novel statistical methods in epidemiology. The processual approach as part of systems epidemiology might represent in a near future an alternative to human in vitro studies using human biological material for understanding the mechanisms and pathways involved in carcinogenesis.
Collapse
Affiliation(s)
- Eiliv Lund
- Department of Community Medicine, Faculty of Health Sciences, University of Tromsø, 9037 Tromsø, Norway.
| | | | - Gregory Nuel
- CNRS, INSMI Stochastics and Biology Group (PSB) LPMA, UPMC, Sorbonne University, France.
| | - Hege Bøvelstad
- Department of Community Medicine, Faculty of Health Sciences, University of Tromsø, 9037 Tromsø, Norway.
| | - Jean-Christophe Thalabard
- Department of Applied Mathematics, MAP5, 45 rue des Saints-Peres, University Paris Descartes, 75006 Paris, France.
| |
Collapse
|
21
|
Abstract
PURPOSE OF REVIEW Coffee is one of the most widely consumed beverages in the world and has been associated with many health conditions. This review examines the limitations of the classic epidemiological approach to studies of coffee and health, and describes the progress in systems epidemiology of coffee and its correlated constituent, caffeine. Implications and applications of this growing body of knowledge are also discussed. RECENT FINDINGS Population-based metabolomic studies of coffee replicate coffee-metabolite correlations observed in clinical settings but have also identified novel metabolites of coffee response, such as specific sphingomyelin derivatives and acylcarnitines. Genome-wide analyses of self-reported coffee and caffeine intake and serum levels of caffeine support an overwhelming role for caffeine in modulating the coffee consumption behavior. Interindividual variation in the physiological exposure or response to any of the many chemicals present in coffee may alter the persistence and magnitude of their effects. It is thus imperative that future studies of coffee and health account for this variation. SUMMARY Systems epidemiological approaches promise to inform causality, parse the constituents of coffee responsible for health effects, and identify the subgroups most likely to benefit from increasing or decreasing coffee consumption.
Collapse
Affiliation(s)
- Marilyn C Cornelis
- aDepartment of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois bDepartment of Nutrition, Harvard School of Public Health cChanning Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
22
|
Ji J, Yuan Z, Zhang X, Li F, Xu J, Liu Y, Li H, Wang J, Xue F. Detection for pathway effect contributing to disease in systems epidemiology with a case-control design. BMJ Open 2015; 5:e006721. [PMID: 25596199 PMCID: PMC4298111 DOI: 10.1136/bmjopen-2014-006721] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
OBJECTIVES Identification of pathway effects responsible for specific diseases has been one of the essential tasks in systems epidemiology. Despite some advance in procedures for distinguishing specific pathway (or network) topology between different disease status, statistical inference at a population level remains unsolved and further development is still needed. To identify the specific pathways contributing to diseases, we attempt to develop powerful statistics which can capture the complex relationship among risk factors. SETTING AND PARTICIPANTS Acute myeloid leukaemia (AML) data obtained from 133 adults (98 patients and 35 controls; 47% female). RESULTS Simulation studies indicated that the proposed Pathway Effect Measures (PEM) were stable; bootstrap-based methods outperformed the others, with bias-corrected bootstrap CI method having the highest power. Application to real data of AML successfully identified the specific pathway (Treg→TGFβ→Th17) effect contributing to AML with p values less than 0.05 under various methods and the bias-corrected bootstrap CI (-0.214 to -0.020). It demonstrated that Th17-Treg correlation balance was impaired in patients with AML, suggesting that Th17-Treg imbalance potentially plays a role in the pathogenesis of AML. CONCLUSIONS The proposed bootstrap-based PEM are valid and powerful for detecting the specific pathway effect contributing to disease, thus potentially providing new insight into the underlying mechanisms and ways to study the disease effects of specific pathways more comprehensively.
Collapse
Affiliation(s)
- Jiadong Ji
- Department of Epidemiology and Biostatistics, School of Public Health, Shandong University, Jinan, China
| | - Zhongshang Yuan
- Department of Epidemiology and Biostatistics, School of Public Health, Shandong University, Jinan, China
| | - Xiaoshuai Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Shandong University, Jinan, China
| | - Fangyu Li
- Department of Neurology, Capital Medical University, Xuanwu Hospital, Beijing, China
| | - Jing Xu
- Department of Epidemiology and Biostatistics, School of Public Health, Shandong University, Jinan, China
| | - Ying Liu
- Department of Public Health and Clinical Medicine, Umea University, Umea, Sweden
| | - Hongkai Li
- Department of Epidemiology and Biostatistics, School of Public Health, Shandong University, Jinan, China
| | - Jia Wang
- School of Mathematics, Shandong University, Jinan, China
| | - Fuzhong Xue
- Department of Epidemiology and Biostatistics, School of Public Health, Shandong University, Jinan, China
| |
Collapse
|
23
|
Dammann O, Gray P, Gressens P, Wolkenhauer O, Leviton A. Systems Epidemiology: What's in a Name? Online J Public Health Inform 2014; 6:e198. [PMID: 25598870 PMCID: PMC4292535 DOI: 10.5210/ojphi.v6i3.5571] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Systems biology is an interdisciplinary effort to integrate molecular, cellular, tissue, organ, and organism levels of function into computational models that facilitate the identification of general principles. Systems medicine adds a disease focus. Systems epidemiology adds yet another level consisting of antecedents that might contribute to the disease process in populations. In etiologic and prevention research, systems-type thinking about multiple levels of causation will allow epidemiologists to identify contributors to disease at multiple levels as well as their interactions. In public health, systems epidemiology will contribute to the improvement of syndromic surveillance methods. We encourage the creation of computational simulation models that integrate information about disease etiology, pathogenetic data, and the expertise of investigators from different disciplines.
Collapse
Affiliation(s)
- O. Dammann
- Dept of Public Health and Community Medicine, Tufts
University School of Medicine, Boston, MA
- Perinatal Epidemiology Unit, Dept. of Gynecology and
Obstetrics, Hannover Medical School, Hannover, Germany
| | - P. Gray
- Dept of Public Health and Community Medicine, Tufts
University School of Medicine, Boston, MA
| | - P. Gressens
- Inserm, U676, Paris, France
- Department of Perinatal Imaging and Health,
Department of Division of Imaging Sciences and Biomedical Engineering,
King’s College London, King’s Health Partners, St. Thomas’
Hospital, London, United Kingdom
| | - O. Wolkenhauer
- Department of Systems Biology and Bioinformatics,
University of Rostock, Rostock, Germany
- Stellenbosch Institute for Advanced Study (STIAS),
Stellenbosch, South Africa
| | - A. Leviton
- Neuroepidemiology Unit, Children’s Hospital,
Boston, MA
| |
Collapse
|
24
|
Vehik K, Ajami NJ, Hadley D, Petrosino JF, Burkhardt BR. The changing landscape of type 1 diabetes: recent developments and future frontiers. Curr Diab Rep 2013; 13:642-50. [PMID: 23912764 PMCID: PMC3827778 DOI: 10.1007/s11892-013-0406-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Type 1 diabetes (T1D) research has made great strides over the past decade with advances in understanding the pathogenesis, natural history, candidate environmental exposures, exposure triggering time, disease prediction, and diagnosis. Major monitoring efforts have provided baseline historical measures, leading to better epidemiological studies incorporating longitudinal biosamples (ie, biobanks), which have allowed for new technologies ('omics') to further expose the etiological agents responsible for the initiation, progression, and eventual clinical onset of T1D. These new frontiers have brought forth high-dimensionality data, which have furthered the evidence of the heterogeneous nature of T1D pathogenesis and allowed for a more mechanistic approach in understanding the etiology of T1D. This review will expand on the most recent advances in the quest for T1D determinants, drawing upon novel research tools that epidemiology, genetics, microbiology, and immunology have provided, linking them to the major hypotheses associated with T1D etiology, and discussing the future frontiers.
Collapse
Affiliation(s)
- Kendra Vehik
- Pediatrics Epidemiology Center, Morsani College of Medicine, University of South Florida, 3650 Spectrum Blvd, Ste 100, Tampa, FL, 33612, USA,
| | | | | | | | | |
Collapse
|
25
|
Cornelis MC, Hu FB. Systems Epidemiology: A New Direction in Nutrition and Metabolic Disease Research. Curr Nutr Rep 2013; 2. [PMID: 24278790 DOI: 10.1007/s13668-013-0052-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Systems epidemiology applied to the field of nutrition has potential to provide new insight into underlying mechanisms and ways to study the health effects of specific foods more comprehensively. Human intervention and population-based studies have identified i) common genetic factors associated with several nutrition-related traits and ii) dietary factors altering the expression of genes and levels of proteins and metabolites related to inflammation, lipid metabolism and/or gut microbial metabolism, results of high relevance to metabolic disease. System-level tools applied type 2 diabetes and related conditions have revealed new pathways that are potentially modified by diet and thus offer additional opportunities for nutritional investigations. Moving forward, harnessing the resources of existing large prospective studies within which biological samples have been archived and diet and lifestyle have been measured repeatedly within individual will enable systems-level data to be integrated, the outcome of which will be improved personalized optimal nutrition for prevention and treatment of disease.
Collapse
Affiliation(s)
- Marilyn C Cornelis
- Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts, USA
| | | |
Collapse
|
26
|
Abstract
Testosterone is the major circulating androgen in men but exhibits an age-related decline in the ageing male. Late-onset hypogonadism or androgen deficiency syndrome (ADS) is a 'syndromic' disorder including both a persistent low testosterone serum concentration and major clinical symptoms, including erectile dysfunction, low libido, decreased muscle mass and strength, increased body fat, decreased vitality or depressed mood. Given its unspecific symptoms, treatment goals and monitoring parameters, this review will outline the various uncertainties concerning the diagnosis, therapy and monitoring of ADS to date. Literature was identified primarily through searches for specific investigators in the PubMed database. No date or language limits were applied in the literature search for the present review. The current state of research, showing that metabolomics is starting to have an impact not only on disease diagnosis and prognosis but also on drug treatment efficacy and safety monitoring, will be presented, and the application of metabolomics to improve the clinical management of ADS will be discussed. Finally, the scientific opportunities presented by metabolomics and other -omics as novel and promising tools for biomarker discovery and individualised testosterone replacement therapy in men will be explored.
Collapse
Affiliation(s)
- Robin Haring
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Ferdinand-Sauerbruch-Strasse, D-17475 Greifswald, Germany.
| |
Collapse
|
27
|
Hu FB. Metabolic profiling of diabetes: from black-box epidemiology to systems epidemiology. Clin Chem 2011; 57:1224-6. [PMID: 21690202 DOI: 10.1373/clinchem.2011.167056] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
28
|
Mulvenna J, Yonglitthipagon P, Sripa B, Brindley PJ, Loukas A, Bethony JM. Banking on the future: biobanking for "omics" approaches to biomarker discovery for Opisthorchis-induced cholangiocarcinoma in Thailand. Parasitol Int 2011; 61:173-7. [PMID: 21855650 DOI: 10.1016/j.parint.2011.06.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2011] [Revised: 06/03/2011] [Accepted: 06/12/2011] [Indexed: 01/03/2023]
Abstract
Cholangiocarcinoma (CCA)--bile duct cancer--is associated with late presentation, poses challenges for diagnosis, and has high mortality. These features t highlight the desperate need for biomarkers than can be measured early and in accessible body fluids such as plasma of people at risk for developing this lethal cancer. In this manuscript, we address previous limitations in the discovery stage of biomarker(s) for CCA and indicate how new generation of "omics" technologies could be used for biomarker discovery in Thailand. A key factor in the success of this biomarker program for CCA is the combination of cutting edge technology with strategic sample acquisition by a biorepositories.
Collapse
Affiliation(s)
- Jason Mulvenna
- Queensland Tropical Health Alliance, James Cook University, Cairns, QLD 4878, Australia
| | | | | | | | | | | |
Collapse
|
29
|
Perera FP. Cancer: the big questions to address in coming years. Cancer Epidemiol Biomarkers Prev 2011; 20:571-3. [PMID: 21454418 DOI: 10.1158/1055-9965.epi-11-0184] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Frederica P Perera
- Columbia University School of Public Health, Department of Environmental Health Sciences, 722 West 168th Street, New York, NY 10032, USA.
| |
Collapse
|
30
|
McHale CM, Zhang L, Hubbard AE, Smith MT. Toxicogenomic profiling of chemically exposed humans in risk assessment. Mutat Res 2010; 705:172-83. [PMID: 20382258 PMCID: PMC2928857 DOI: 10.1016/j.mrrev.2010.04.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2010] [Accepted: 04/01/2010] [Indexed: 12/13/2022]
Abstract
Gene-environment interactions contribute to complex disease development. The environmental contribution, in particular low-level and prevalent environmental exposures, may constitute much of the risk and contribute substantially to disease. Systematic risk evaluation of the majority of human chemical exposures, has not been conducted and is a goal of regulatory agencies in the U.S. and worldwide. With the recent recognition that toxicological approaches more predictive of effects in humans are required for risk assessment, in vitro human cell line data as well as animal data are being used to identify toxicity mechanisms that can be translated into biomarkers relevant to human exposure studies. In this review, we discuss how data from toxicogenomic studies of exposed human populations can inform risk assessment, by generating biomarkers of exposure, early effect, and/or susceptibility, elucidating mechanisms of action underlying exposure-related disease, and detecting response at low doses. Good experimental design incorporating precise, individual exposure measurements, phenotypic anchors (pre-disease or traditional toxicological markers), and a range of relevant exposure levels, is necessary. Further, toxicogenomic studies need to be designed with sufficient power to detect true effects of the exposure. As more studies are performed and incorporated into databases such as the Comparative Toxicogenomics Database (CTD) and Chemical Effects in Biological Systems (CEBS), data can be mined for classification of newly tested chemicals (hazard identification), and, for investigating the dose-response, and inter-relationship among genes, environment and disease in a systems biology approach (risk characterization).
Collapse
Affiliation(s)
- Cliona M. McHale
- School of Public Health, Division of Environmental Health Sciences, University of California, Berkeley, CA 94720
| | - Luoping Zhang
- School of Public Health, Division of Environmental Health Sciences, University of California, Berkeley, CA 94720
| | - Alan E. Hubbard
- School of Public Health, Division of Biostatistics, University of California, Berkeley, CA 94720
| | - Martyn T. Smith
- School of Public Health, Division of Environmental Health Sciences, University of California, Berkeley, CA 94720
| |
Collapse
|
31
|
Chiu WA, Euling SY, Scott CS, Subramaniam RP. Approaches to advancing quantitative human health risk assessment of environmental chemicals in the post-genomic era. Toxicol Appl Pharmacol 2010; 271:309-23. [PMID: 20353796 DOI: 10.1016/j.taap.2010.03.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2010] [Revised: 03/19/2010] [Accepted: 03/22/2010] [Indexed: 10/19/2022]
Abstract
The contribution of genomics and associated technologies to human health risk assessment for environmental chemicals has focused largely on elucidating mechanisms of toxicity, as discussed in other articles in this issue. However, there is interest in moving beyond hazard characterization to making more direct impacts on quantitative risk assessment (QRA)--i.e., the determination of toxicity values for setting exposure standards and cleanup values. We propose that the evolution of QRA of environmental chemicals in the post-genomic era will involve three, somewhat overlapping phases in which different types of approaches begin to mature. The initial focus (in Phase I) has been and continues to be on "augmentation" of weight of evidence--using genomic and related technologies qualitatively to increase the confidence in and scientific basis of the results of QRA. Efforts aimed towards "integration" of these data with traditional animal-based approaches, in particular quantitative predictors, or surrogates, for the in vivo toxicity data to which they have been anchored are just beginning to be explored now (in Phase II). In parallel, there is a recognized need for "expansion" of the use of established biomarkers of susceptibility or risk of human diseases and disorders for QRA, particularly for addressing the issues of cumulative assessment and population risk. Ultimately (in Phase III), substantial further advances could be realized by the development of novel molecular and pathway-based biomarkers and statistical and in silico models that build on anticipated progress in understanding the pathways of human diseases and disorders. Such efforts would facilitate a gradual "reorientation" of QRA towards approaches that more directly link environmental exposures to human outcomes.
Collapse
Affiliation(s)
- Weihsueh A Chiu
- National Center for Environmental Assessment, U.S. Environmental Protection Agency, Washington DC, 20460, USA.
| | | | | | | |
Collapse
|
32
|
Dumeaux V, Olsen KS, Nuel G, Paulssen RH, Børresen-Dale AL, Lund E. Deciphering normal blood gene expression variation--The NOWAC postgenome study. PLoS Genet 2010; 6:e1000873. [PMID: 20300640 PMCID: PMC2837385 DOI: 10.1371/journal.pgen.1000873] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2009] [Accepted: 02/05/2010] [Indexed: 12/16/2022] Open
Abstract
There is growing evidence that gene expression profiling of peripheral blood cells is a valuable tool for assessing gene signatures related to exposure, drug-response, or disease. However, the true promise of this approach can not be estimated until the scientific community has robust baseline data describing variation in gene expression patterns in normal individuals. Using a large representative sample set of postmenopausal women (N = 286) in the Norwegian Women and Cancer (NOWAC) postgenome study, we investigated variability of whole blood gene expression in the general population. In particular, we examined changes in blood gene expression caused by technical variability, normal inter-individual differences, and exposure variables at proportions and levels relevant to real-life situations. We observe that the overall changes in gene expression are subtle, implying the need for careful analytic approaches of the data. In particular, technical variability may not be ignored and subsequent adjustments must be considered in any analysis. Many new candidate genes were identified that are differentially expressed according to inter-individual (i.e. fasting, BMI) and exposure (i.e. smoking) factors, thus establishing that these effects are mirrored in blood. By focusing on the biological implications instead of directly comparing gene lists from several related studies in the literature, our analytic approach was able to identify significant similarities and effects consistent across these reports. This establishes the feasibility of blood gene expression profiling, if they are predicated upon careful experimental design and analysis in order to minimize confounding signals, artifacts of sample preparation and processing, and inter-individual differences. As a major defence and transport system, blood cells are capable of adjusting gene expression in response to various clinical, biochemical, and pathological conditions. Here, we expand our understanding about the nature and extent of variation in gene expression from blood among healthy individuals. Using a large representative sample of postmenopausal women (N = 286) in the Norwegian Women and Cancer (NOWAC) postgenome study, we investigated blood gene expression changes due to normal inter-individuality (age, body mass index, fasting status), and exposure variables (smoking, hormone therapy, and medication use) at proportions and levels found in real life situations. Host genes were found to vary by inter-individual (i.e. fasting, BMI) and exposure (i.e. smoking) factors, and these gene lists may be used as a basis for further hypothesis development. Our study also establishes the feasibility of blood gene expression profiling for disease prediction, diagnosis, or prognosis, but underscores the necessity of care in study design and analysis to account for inter-individual differences and confounding signals.
Collapse
Affiliation(s)
- Vanessa Dumeaux
- Institute of Community Medicine, University of Tromsø, Tromsø, Norway.
| | | | | | | | | | | |
Collapse
|
33
|
Spitz MR, Bondy ML. The evolving discipline of molecular epidemiology of cancer. Carcinogenesis 2009; 31:127-34. [PMID: 20022891 DOI: 10.1093/carcin/bgp246] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Classical epidemiologic studies have made seminal contributions to identifying the etiology of most common cancers. Molecular epidemiology was conceived of as an extension of traditional epidemiology to incorporate biomarkers with questionnaire data to further our understanding of the mechanisms of carcinogenesis. Early molecular epidemiologic studies employed functional assays. These studies were hampered by the need for sequential and/or prediagnostic samples, viable lymphocytes and the uncertainty of how well these functional data (derived from surrogate lymphocytic tissue) reflected events in the target tissue. The completion of the Human Genome Project and Hapmap Project, together with the unparalleled advances in high-throughput genotyping revolutionized the practice of molecular epidemiology. Early studies had been constrained by existing technology to use the hypothesis-driven candidate gene approach, with disappointing results. Pathway analysis addressed some of the concerns, although the study of interacting and overlapping gene networks remained a challenge. Whole-genome scanning approaches were designed as agnostic studies using a dense set of markers to capture much of the common genome variation to study germ-line genetic variation as risk factors for common complex diseases. It should be possible to exploit the wealth of these data for pharmacogenetic studies to realize the promise of personalized therapy. Going forward, the temptation for epidemiologists to be lured by high-tech 'omics' will be immense. Systems Epidemiology, the observational prototype of systems biology, is an extension of classical epidemiology to include powerful new platforms such as the transcriptome, proteome and metabolome. However, there will always be the need for impeccably designed and well-powered epidemiologic studies with rigorous quality control of data, specimen acquisition and statistical analysis.
Collapse
Affiliation(s)
- Margaret R Spitz
- Department of Epidemiology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA.
| | | |
Collapse
|
34
|
Pleil JD. Influence of systems biology response and environmental exposure level on between-subject variability in breath and blood biomarkers. Biomarkers 2009; 14:560-71. [DOI: 10.3109/13547500903186460] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
35
|
Vlaanderen J, Moore LE, Smith MT, Lan Q, Zhang L, Skibola CF, Rothman N, Vermeulen R. Application of OMICS technologies in occupational and environmental health research; current status and projections. Occup Environ Med 2009; 67:136-43. [PMID: 19933307 DOI: 10.1136/oem.2008.042788] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OMICS technologies are relatively new biomarker discovery tools that can be applied to study large sets of biological molecules. Their application in human observational studies (HOS) has become feasible in recent years due to a spectacular increase in the sensitivity, resolution and throughput of OMICS-based assays. Although, the number of OMICS techniques is ever expanding, the five most developed OMICS technologies are genotyping, transcriptomics, epigenomics, proteomics and metabolomics. These techniques have been applied in HOS to various extents. However, their application in occupational environmental health (OEH) research has been limited. Here, we will discuss the opportunities these new techniques provide for OEH research. In addition we will address difficulties and limitations to the interpretation of the data that is generated by OMICS technologies. To illustrate the current status of the application of OMICS in OEH research, we will provide examples of studies that used OMICS technologies to investigate human health effects of two well-known toxicants, benzene and arsenic.
Collapse
Affiliation(s)
- J Vlaanderen
- Institute for Risk Assessment Sciences, Division of Environmental Epidemiology, University Utrecht, Po Box 80178, 3508 TD, Utrecht, the Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|