Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Stephen D. Hursting is active.

Publication


Featured researches published by Stephen D. Hursting.


Cell | 2006

Genomic instability and aging-like phenotype in the absence of mammalian SIRT6

Raul Mostoslavsky; Katrin F. Chua; David B. Lombard; Wendy W. Pang; Miriam R. Fischer; Lionel Gellon; Pingfang Liu; Gustavo Mostoslavsky; Sonia Franco; Michael M. Murphy; Kevin D. Mills; Parin Patel; Joyce T. Hsu; Andrew L. Hong; Ethan Ford; Hwei Ling Cheng; Caitlin Kennedy; Nomeli P. Nunez; Roderick T. Bronson; David Frendewey; Wojtek Auerbach; David M. Valenzuela; Margaret Karow; Michael O. Hottiger; Stephen D. Hursting; J. Carl Barrett; Leonard Guarente; Richard C. Mulligan; Bruce Demple; George D. Yancopoulos

The Sir2 histone deacetylase functions as a chromatin silencer to regulate recombination, genomic stability, and aging in budding yeast. Seven mammalian Sir2 homologs have been identified (SIRT1-SIRT7), and it has been speculated that some may have similar functions to Sir2. Here, we demonstrate that SIRT6 is a nuclear, chromatin-associated protein that promotes resistance to DNA damage and suppresses genomic instability in mouse cells, in association with a role in base excision repair (BER). SIRT6-deficient mice are small and at 2-3 weeks of age develop abnormalities that include profound lymphopenia, loss of subcutaneous fat, lordokyphosis, and severe metabolic defects, eventually dying at about 4 weeks. We conclude that one function of SIRT6 is to promote normal DNA repair, and that SIRT6 loss leads to abnormalities in mice that overlap with aging-associated degenerative processes.


Nutrition and Cancer | 1999

Phytoestrogen intake and prostate cancer: a case-control study using a new database.

Sara S. Strom; Yuko Yamamura; Cherie M. Duphorne; Margaret R. Spitz; Richard J. Babaian; Patricia C. Pillow; Stephen D. Hursting

In the last several years, attention has been focused on comparing the Western diet, which is rich in fat, protein, and refined carbohydrates, with the Asian diet, which is rich in phytoestrogens, as a possible explanation for the contrasting rates of clinically relevant prostate cancer. Phytoestrogens, plant-derived nutrients, include several isoflavones, flavonoids, lignans, phytosterols, and coumestans, some of which have been postulated as having anticarcinogenic properties. Using a new database, we examined the role of phytoestrogen intake and prostate cancer risk in 83 Caucasian cases and 107 controls. Controls reported consuming higher amounts of foods containing genistein, daidzein, and coumestrol and lower amounts of foods containing campesterol and stigmasterol. Multivariate analysis, after adjustment for age, family history of prostate cancer, alcohol consumption, and total calorie intake, showed an inverse association between coumestrol (p = 0.03) and daidzein (p = 0.07) and prostate cancer risk. Genistein, the most studied phytoestrogen, showed a slight protective effect (p = 0.26). However, a positive association was found between campesterol (p = 0.08) and stigmasterol (p = 0.03) and risk of prostate cancer. These results are suggestive of a possible relationship between phytoestrogen intake and prostate cancer risk. Larger comprehensive studies are needed to further refine the role of phytoestrogen intake in prostate cancer risk.


Annals of the New York Academy of Sciences | 2003

Environmental Exposure, DNA Methylation, and Gene Regulation

Shuanfang Li; Stephen D. Hursting; Barbara J. Davis; John A. McLachlan; J. Carl Barrett

Abstract: DNA methylation is an epigenetic mechanism that regulates chromosomal stability and gene expression. Abnormal DNA methylation patterns have been observed in many types of human tumors, including those of the breast, prostate, colon, thyroid, stomach, uterus, and cervix. We and others have shown that exposure to a wide variety of xenobiotics during critical periods of mammalian development can persistently alter the pattern of DNA methylation, resulting in potentially adverse biological effects such as aberrant gene expression. Thus, this epigenetic mechanism may underlie the observed increased risk in adulthood of several chronic diseases, including cancer, in response to xenobiotic exposures early in life. We present here the lessons learned from studies on the effects of perinatal diethylstilbesterol (DES) exposure on the methylation pattern of the promoters of several estrogen‐responsive genes associated with the development of reproductive organs. Perinatal DES exposure, which induces epithelial tumors of the uterus in mice and is associated with several reproductive tract abnormalities and increased vaginal and cervical cancer risk in women, provides a clear example of how estrogenic xenobiotic exposure during a critical period of development can abnormally demethylate DNA sequences during organ development and possibly increase cancer risk later in life. In addition, nutritional factors and stress may also alter DNA methylation during early life and modulate the risk of cancer and other chronic diseases in adulthood. We suggest that DNA methylation status may be influenced by environmental exposures in early life, leading to increased risk of cancer in adulthood.


Journal of Clinical Oncology | 2010

Energy Balance, Host-Related Factors, and Cancer Progression

Stephen D. Hursting; Nathan A. Berger

Obesity is associated with an increased risk and worsened prognosis for many types of cancer, but the mechanisms underlying the obesity-cancer progression link are poorly understood. Several energy balance-related host factors are known to influence tumor progression and/or treatment responsiveness after cancer develops, and these have been implicated as key contributors to the complex effects of obesity on cancer outcome. These host factors include leptin, adiponectin, steroid hormones, reactive oxygen species associated with inflammation, insulin, insulin-like growth factor-1, and sirtuins. Each of these host factors is considered in this article in the context of energy balance and cancer progression. In addition, future research directions in this field are discussed, including the importance of study designs addressing energy balance across the life course, the development and application of highly relevant animal models, potential roles of cancer stem cells in the response to energy balance modulation, and emerging pharmacologic approaches that target energy balance-related pathways.


Carcinogenesis | 2010

Calories and carcinogenesis: lessons learned from 30 years of calorie restriction research

Stephen D. Hursting; Sarah M. Smith; Laura M. Lashinger; Alison E. Harvey; Susan N. Perkins

Calorie restriction (CR) is arguably the most potent, broadly acting dietary regimen for suppressing the carcinogenesis process, and many of the key studies in this field have been published in Carcinogenesis. Translation of the knowledge gained from CR research in animal models to cancer prevention strategies in humans is urgently needed given the worldwide obesity epidemic and the established link between obesity and increased risk of many cancers. This review synthesizes the evidence on key biological mechanisms underlying many of the beneficial effects of CR, with particular emphasis on the impact of CR on growth factor signaling pathways and inflammatory processes and on the emerging development of pharmacological mimetics of CR. These approaches will facilitate the translation of CR research into effective strategies for cancer prevention in humans.


Annals of the New York Academy of Sciences | 2011

The growing challenge of obesity and cancer: an inflammatory issue

Alison E. Harvey; Laura M. Lashinger; Stephen D. Hursting

The prevalence of obesity, an established risk factor for many cancers, has risen steadily for the past several decades in the United States and in many parts of the world. This review synthesizes the evidence on key biological mechanisms underlying the obesity–cancer link, with particular emphasis on the impact of energy balance modulation, such as diet‐induced obesity and calorie restriction, on growth factor signaling pathways and inflammatory processes. Particular attention is placed on the proinflammatory environment associated with the obese state, specifically highlighting the involvement of obesity‐associated hormones/growth factors in crosstalk between macrophages, adipocytes, and epithelial cells in many cancers. Understanding the contribution of obesity to growth factor signaling and chronic inflammation provides mechanistic targets for disrupting the obesity–cancer link.


The Prostate | 2001

Leptin and prostate cancer

Shine Chang; Stephen D. Hursting; John H. Contois; Sara S. Strom; Yuko Yamamura; Richard J. Babaian; Patricia Troncoso; Peter T. Scardino; Thomas M. Wheeler; Christopher I. Amos; Margaret R. Spitz

Higher prostate cancer mortality rates among US immigrants from countries with lower rates suggest environmental influences on prostate carcinogenesis (e.g., diet, body composition).


Nutrition and Cancer | 1999

Development of a database for assessing dietary phytoestrogen intake

Patricia C. Pillow; Cherie M. Duphorne; Shine Chang; John H. Contois; Sara S. Strom; Margaret R. Spitz; Stephen D. Hursting

For the past two decades, epidemiologists have observed lower risks of lung, breast, prostate, colon, and other cancers in populations that frequently consume fruits and vegetables. Numerous phytoestrogens have been shown to be anticarcinogenic under experimental conditions and may account for at least part of the cancer-prevention effects of fruit and vegetable consumption. These plant constituents include isoflavonoids, coumestans, lignans, phytosterols, and flavonoids. DietSys, the nutrient analysis program associated with the National Cancer Institute Health Habits and History Questionnaire (HHHQ), and other nationally available nutrient analysis databases do not fully assess these constituents. Therefore, we modified DietSys to include these components in foods on the basis of published values. In addition, as part of an epidemiological study of prostate cancer, we modified the food-frequency component of the HHHQ to include the main foods contributing to phytoestrogen intake. Although there are limitations to the consistency and quality of many of the values because they were gathered from a variety of sources, our approach should provide a useful first tool for assessing the epidemiological association between phytoestrogen consumption and cancer risk. Furthermore, this work has already facilitated the identification of the major dietary contributors with phytoestrogen activity and prioritized future laboratory analyses of specific foods toward the development of a more complete and accurate database.


Cancer Prevention Research | 2008

Dietary Energy Balance Modulates Signaling through the Akt/Mammalian Target of Rapamycin Pathways in Multiple Epithelial Tissues

Tricia Moore; Linda Beltrán; Steve Carbajal; Sara S. Strom; Jeanine Traag; Stephen D. Hursting; John DiGiovanni

Abstract The prevalence of obesity, an established risk factor for several types of cancer, has increased steadily over the past several decades in the United States. New targets and strategies for offsetting the effect of obesity on cancer risk are urgently needed. In the present study, we examined the effect of dietary energy balance manipulation on steady-state signaling in multiple epithelial tissues, with a focus on the Akt and mammalian target of rapamycin (mTOR) pathways. For these experiments, male FVB/N and C57BL/6 and female ICR mice were maintained on a control (10 kcal% fat) diet, a diet-induced obesity (DIO; 60 kcal% fat) regimen, or a 30% calorie restriction (CR) regimen for 15 to 17 weeks. Relative to the control group, the DIO regimen increased, whereas CR decreased, circulating insulin-like growth factor-I (IGF-I) as has previously been reported. Western blot analyses showed that the DIO regimen enhanced, whereas CR inhibited, activation of Akt and mTOR, regardless of epithelial tissue or genetic background. In contrast, activation of AMP-activated protein kinase was modulated by dietary energy balance manipulation in the liver but not in the epidermis or dorsolateral prostate. Western blot analyses of epidermal extracts taken from ICR mice also revealed reduced activation of both the IGF-I receptor and epidermal growth factor receptor in CR mice, compared with control mice or mice maintained on the DIO regimen. Taken together, these novel findings suggest that dietary energy balance modulates signaling through cell-surface receptors (i.e., IGF-I receptor and epidermal growth factor receptor), affecting activation of multiple downstream pathways including Akt and mTOR, thus providing important dietary and pharmacologic targets for disrupting the obesity-cancer link.


Sports Medicine | 2008

Physical activity and cancer prevention : pathways and targets for intervention.

Connie J. Rogers; Lisa H. Colbert; John W. Greiner; Susan N. Perkins; Stephen D. Hursting

The prevalence of obesity, an established epidemiological risk factor for many cancers, has risen steadily for the past several decades in the US and many other countries. Particularly alarming are the increasing rates of obesity among children, portending continuing increases in the rates of obesity and obesity-related cancers for many years to come. Modulation of energy balance, via increased physical activity, has been shown in numerous comprehensive epidemiological reviews to reduce cancer risk. Unfortunately, the effects and mechanistic targets of physical activity interventions on the carcinogenesis process have not been thoroughly characterized.Studies to date suggest that exercise can exert its cancer-preventive effects at many stages during the process of carcinogenesis, including both tumour initiation and progression. As discussed in this review, exercise may be altering tumour initiation events by modifying carcinogen activation, specifically by enhancing the cytochrome P450 system and by enhancing selective enzymes in the carcinogen detoxification pathway, including, but not limited to, glutathione-S-transferases. Furthermore, exercise may reduce oxidative damage by increasing a variety of anti-oxidant enzymes, enhancing DNA repair systems and improving intracellular protein repair systems.In addition to altering processes related to tumour initiation, exercise may also exert a cancer-preventive effect by dampening the processes involved in the promotion and progression stages of carcinogenesis, including scavenging reactive oxygen species (ROS); altering cell proliferation, apoptosis and differentiation; decreasing inflammation; enhancing immune function; and suppressing angiogenesis. A paucity of data exists as to whether exercise may be working as an anti-promotion strategy via altering ROS in initiated or preneoplastic models; therefore, no conclusions can be made about this possible mechanism. The studies directly examining cell proliferation and apoptosis have shown that exercise can enhance both processes, which is difficult to interpret in the context of carcinogenesis. Studies examining the relationship between exercise and chronic inflammation suggest that exercise may reduce pro-inflammatory mediators and reduce the state of low-grade, chronic inflammation. Additionally, exercise has been shown to enhance components of the innate immune response (i.e. macrophage and natural killer cell function). Finally, only a limited number of studies have explored the relationship between exercise and angiogenesis; therefore, no conclusions can be made currently about the role of exercise in the angiogenesis process as it relates to tumour progression.In summary, exercise can alter biological processes that contribute to both antiinitiation and anti-progression events in the carcinogenesis process. However, more sophisticated, detailed studies are needed to examine each of the potential mechanisms contributing to an exercise-induced decrease in carcinogenesis in order to determine the minimum dose, duration and frequency of exercise needed to yield significant cancer-preventive effects, and whether exercise can be used prescriptively to reverse the obesity-induced physiological changes that increase cancer risk.

Collaboration


Dive into the Stephen D. Hursting's collaboration.

Top Co-Authors

Avatar

Susan N. Perkins

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Laura W. Bowers

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Laura M. Lashinger

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Emily L. Rossi

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jackie A. Lavigne

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

David Berrigan

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Nomeli P. Nunez

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Sarah M. Dunlap

University of Texas at Austin

View shared research outputs
Researchain Logo
Decentralizing Knowledge