Network


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

Hotspot


Dive into the research topics where Joanne R. Lupton is active.

Publication


Featured researches published by Joanne R. Lupton.


Prostaglandins Leukotrienes and Essential Fatty Acids | 2009

Dietary docosahexaenoic and eicosapentaenoic acid: Emerging mediators of inflammation

Robert S. Chapkin; Wooki Kim; Joanne R. Lupton; David N. McMurray

The inflammatory response is designed to help fight and clear infection, remove harmful chemicals, and repair damaged tissue and organ systems. Although this process, in general, is protective, the failure to resolve the inflammation and return the target tissue to homeostasis can result in disease, including the promotion of cancer. A plethora of published literature supports the contention that dietary n-3 polyunsaturated fatty acids (PUFA), and eicosapentaenoic (EPA, 20:5n-3) and docosahexaenoic acid (DHA, 22:6n-3) in particular, are important modulators of a hosts inflammatory/immune responses. The following review describes a mechanistic model that may explain, in part, the pleiotropic anti-inflammatory and immunosuppressive properties of EPA and DHA. In this review, we focus on salient studies that address three overarching mechanisms of n-3 PUFA action: (i) modulation of nuclear receptor activation, i.e., nuclear factor-kappaB (NF-kappaB) suppression; (ii) suppression of arachidonic acid-cyclooxygenase-derived eicosanoids, primarily prostaglandin E(2)-dependent signaling; and (iii) alteration of the plasma membrane micro-organization (lipid rafts), particularly as it relates to the function of Toll-like receptors (TLRs), and T-lymphocyte signaling molecule recruitment to the immunological synapse (IS). We propose that lipid rafts may be targets for the development of n-3 PUFA-containing dietary bioactive agents to down-modulate inflammatory and immune responses and for the treatment of autoimmune and chronic inflammatory diseases.


The FASEB Journal | 2004

n-3 PUFA alter caveolae lipid composition and resident protein localization in mouse colon

David W.L. Ma; Jeongmin Seo; Laurie A. Davidson; Evelyn S. Callaway; Yang-Yi Fan; Joanne R. Lupton; Robert S. Chapkin

Caveolae, by virtue of their unique lipid environment, serve as signaling platforms that regulate cellular events. Perturbations in caveolae lipid composition have been shown in vitro to displace proteins from lipid microdomains, thereby altering their functionality and subsequent downstream signaling. Because membrane remodeling may not be accurately represented by using pharmacological treatments and in vitro models, we investigated the in vivo ability of dietary n‐3 polyunsaturated fatty acids (PUFA) to alter caveolae lipid environment and the compartmentalization of resident proteins in mouse colonic mucosa. n‐3 PUFA were examined for their chemoprotective, membrane lipid‐modifying properties. Colonic caveolae in mice fed n‐6 or n‐3 PUFA enriched diets were characteristically enriched in cholesterol, sphingomyelin, and caveolin‐1. n‐3 PUFA feeding, compared with n‐6 PUFA, significantly altered colonic caveolae microenvironment by increasing phospholipid n‐3 fatty acyl content and reducing both cholesterol (by 46%) and caveolin‐1 (by 53%), without altering total cellular levels. Concomitantly, localization of caveolae‐resident signaling proteins H‐Ras and eNOS in colonic caveolae was decreased by n‐3 PUFA, by 45 and 56%, respectively. The distribution of non‐caveolae proteins K‐Ras and clathrin was unaffected. Moreover, EGF‐stimulated H‐Ras, but not K‐Ras activation was significantly suppressed following n‐3 PUFA feeding, in parallel with the selective alterations in their microlocalization. These findings reveal a novel modality by which n‐3 PUFA remodel membrane microdomains in vivo and thereby alter caveolae protein localization and functionality.


Cancer Research | 2004

Chemopreventive n-3 Polyunsaturated Fatty Acids Reprogram Genetic Signatures during Colon Cancer Initiation and Progression in the Rat

Laurie A. Davidson; Danh V. Nguyen; Regina Hokanson; Evelyn S. Callaway; Robert B. Isett; Nancy D. Turner; Edward R. Dougherty; Naisyin Wang; Joanne R. Lupton; Raymond J. Carroll; Robert S. Chapkin

The mechanisms by which n-3 polyunsaturated fatty acids (PUFAs) decrease colon tumor formation have not been fully elucidated. Examination of genes up- or down-regulated at various stages of tumor development via the monitoring of gene expression relationships will help to determine the biological processes ultimately responsible for the protective effects of n-3 PUFA. Therefore, using a 3 × 2 × 2 factorial design, we used Codelink DNA microarrays containing ∼9000 genes to help decipher the global changes in colonocyte gene expression profiles in carcinogen-injected Sprague Dawley rats. Animals were assigned to three dietary treatments differing only in the type of fat (corn oil/n-6 PUFA, fish oil/n-3 PUFA, or olive oil/n-9 monounsaturated fatty acid), two treatments (injection with the carcinogen azoxymethane or with saline), and two time points (12 hours and 10 weeks after first injection). Only the consumption of n-3 PUFA exerted a protective effect at the initiation (DNA adduct formation) and promotional (aberrant crypt foci) stages. Importantly, microarray analysis of colonocyte gene expression profiles discerned fundamental differences among animals treated with n-3 PUFA at both the 12 hours and 10-week time points. Thus, in addition to demonstrating that dietary fat composition alters the molecular portrait of gene expression profiles in the colonic epithelium at both the initiation and promotional stages of tumor development, these findings indicate that the chemopreventive effect of fish oil is due to the direct action of n-3 PUFA and not to a reduction in the content of n-6 PUFA.


Journal of Nutrition | 2007

Immunomodulatory Effects of (n-3) Fatty Acids: Putative Link to Inflammation and Colon Cancer

Robert S. Chapkin; Laurie A. Davidson; Lan Ly; Brad R. Weeks; Joanne R. Lupton; David N. McMurray

Chronic inflammation and colorectal cancer are closely linked. Although the overall mechanisms of inflammation-associated gastrointestinal carcinogenesis are complex, it is clear that antiinflammatory therapy is efficacious against neoplastic progression and malignant conversion. From a dietary perspective, fish oil containing (n-3) polyunsaturated fatty acids (PUFAs) has antiinflammatory properties, but for years the mechanism has remained obscure. Of relevance to the immune system in the intestine, we showed that (n-3) PUFA feeding alters the balance between CD4+ T-helper (Th1 and Th2) subsets by directly suppressing Th1 cell development (i.e., clonal expansion). This is noteworthy because Th1 cells mediate inflammatory diseases and resistance to intracellular pathogens or allergic hypersensitivity, and Th2 cells mediate resistance to extracellular pathogens. Therefore, any changes induced by (n-3) PUFAs in T-cell subset balance and function are important because the outcome is expected to suppress the development of autoimmune diseases and possibly the occurrence of colon cancer. Precisely how the immunomodulatory effects of (n-3) PUFAs influence inflammation-associated colonic tumor development is the subject of an ongoing investigation.


Carcinogenesis | 2009

n-3 Polyunsaturated fatty acids modulate carcinogen-directed non-coding microRNA signatures in rat colon

Laurie A. Davidson; Naisyin Wang; Manasvi S. Shah; Joanne R. Lupton; Ivan Ivanov; Robert S. Chapkin

We have hypothesized that dietary modulation of intestinal non-coding RNA [microRNA (miRNA)] expression may contribute to the chemoprotective effects of nutritional bioactives (fish oil and pectin). To fully understand the effects of these agents on the expression of miRNAs, Sprague-Dawley rats were fed diets containing corn oil or fish oil with pectin or cellulose and injected with azoxymethane (AOM, a colon-specific carcinogen) or saline (control). Real-time polymerase chain reaction using miRNA-specific primers and Taq Man probes was carried out to quantify effects on miRNA expression in colonic mucosa. From 368 mature miRNAs assayed, at an early stage of cancer progression (10 week post AOM injection), let-7d, miR-15b, miR-107, miR-191 and miR-324-5p were significantly (P < 0.05) affected by diet x carcinogen interactions. Overall, fish oil fed animals exhibited the smallest number of differentially expressed miRNAs (AOM versus saline treatment). With respect to the tumor stage (34 week post AOM injection), 46 miRNAs were dysregulated in adenocarcinomas compared with normal mucosa from saline-injected animals. Of the 27 miRNAs expressed at higher (P < 0.05) levels in tumors, miR-34a, 132, 223 and 224 were overexpressed at >10-fold. In contrast, the expression levels of miR-192, 194, 215 and 375 were dramatically reduced (< or = 0.32-fold) in adenocarcinomas. These results demonstrate for the first time the utility of the rat AOM model and the novel role of fish oil in protecting the colon from carcinogen-induced miRNA dysregulation.


Cancer Research | 2008

Reduced Colitis-Associated Colon Cancer in Fat-1 (n-3 Fatty Acid Desaturase) Transgenic Mice

Qian Jia; Joanne R. Lupton; Roger Smith; Brad R. Weeks; Evelyn S. Callaway; Laurie A. Davidson; Wooki Kim; Yang Yi Fan; Peiying Yang; Robert A. Newman; Jing X. Kang; David N. McMurray; Robert S. Chapkin

Bioactive food components containing n-3 polyunsaturated fatty acids (PUFA) modulate multiple determinants that link inflammation to cancer initiation and progression. Therefore, in this study, fat-1 transgenic mice, which convert endogenous n-6 PUFA to n-3 PUFA in multiple tissues, were injected with azoxymethane followed by three cycles of dextran sodium sulfate (DSS) to induce colitis-associated cancer. Fat-1 mice exhibited a reduced number of colonic adenocarcinomas per mouse (1.05 +/- 0.29 versus 2.12 +/- 0.51, P = 0.033), elevated apoptosis (P = 0.03), and a decrease in n-6 PUFA-derived eicosanoids, compared with wild-type (wt) mice. To determine whether the chemoprotective effects of n-3 PUFA could be attributed to its pleiotropic anti-inflammatory properties, colonic inflammation and injury scores were evaluated 5 days after DSS exposure followed by either a 3-day or 2-week recovery period. There was no effect of n-3 PUFA at 3 days. However, following a 2-week recovery period, colonic inflammation and ulceration scores returned to pretreatment levels compared with 3-day recovery only in fat-1 mice. For the purpose of examining the specific reactivity of lymphoid elements in the intestine, CD3(+) T cells, CD4(+) T helper cells, and macrophages from colonic lamina propria were quantified. Comparison of 3-day versus 2-week recovery time points revealed that fat-1 mice exhibited decreased (P < 0.05) CD3(+), CD4(+) T helper, and macrophage cell numbers per colon as compared with wt mice. These results suggest that the antitumorigenic effect of n-3 PUFA may be mediated, in part, via its anti-inflammatory properties.


Cancer Letters | 1994

Butyrate as a differentiating agent: pharmacokinetics, analogues and current status.

Harold L. Newmark; Joanne R. Lupton; Charles W. Young

The field of butyrate-induced differentiation of neoplastic and non-neoplastic cells is reviewed and possible clinical correlations considered with regard to butyrate, butyrate prodrugs and butyrate analogues. These topics are discussed from the point of view of the concentrations required in vitro for biologic effect, and likely pharmacokinetic behavior in vivo.


Current Opinion in Gastroenterology | 2007

Colon cancer, fatty acids and anti-inflammatory compounds.

Robert S. Chapkin; David N. McMurray; Joanne R. Lupton

Purpose of review To outline recent findings on the efficacy of n-3 polyunsaturated fatty acids in the prevention/treatment of inflammatory bowel disease and colorectal cancer. Recent findings Compelling data indicate a functional link between chronic inflammation and colon cancer. With respect to environmental risk factors, there is growing evidence that long-chain n-3 polyunsaturated fatty acids found in fish oil suppress inflammatory bowel diseases and colon cancer risk in humans. Unfortunately, the molecular basis of the effect of n-3 polyunsaturated fatty acids on inflammation/colitis-associated colon cancer risk is still largely obscure. In this review, we focus on recent studies which address three emerging mechanisms of n-3 polyunsaturated fatty acids action: (1) metabolic interconversion into bioactive eicosanoids, (2) modulation of nuclear receptor activation, and (3) alteration of membrane phospholipid composition and functionality of lipid microdomains. Summary The consumption of dietary fish oil may prove to be an effective adjuvant therapy in colon cancer. Therefore, it is both appropriate and timely to determine precisely how n-3 polyunsaturated fatty acids modulate cell signaling networks, and reduce the risk of developing colon cancer and inflammatory disorders of the intestine.


Chemistry and Physics of Lipids | 2008

Mechanisms by which docosahexaenoic acid and related fatty acids reduce colon cancer risk and inflammatory disorders of the intestine

Robert S. Chapkin; Jeongmin Seo; David N. McMurray; Joanne R. Lupton

A growing body of epidemiological, clinical, and experimental evidence has underscored both the pharmacological potential and the nutritional value of dietary fish oil enriched in very long chain n-3 PUFAs such as docosahexaenoic acid (DHA, 22:6, n-3) and eicosapentaenoic acid (EPA, 20:5, n-3). The broad health benefits of very long chain n-3 PUFAs and the pleiotropic effects of dietary fish oil and DHA have been proposed to involve alterations in membrane structure and function, eicosanoid metabolism, gene expression and the formation of lipid peroxidation products, although a comprehensive understanding of the mechanisms of action has yet to be elucidated. In this review, we present data demonstrating that DHA selectively modulates the subcellular localization of lipidated signaling proteins depending on their transport pathway, which may be universally applied to other lipidated protein trafficking. An interesting possibility raised by the current observations is that lipidated proteins may exhibit different subcellular distribution profiles in various tissues, which contain a distinct membrane lipid composition. In addition, the current findings clearly indicate that subcellular localization of proteins with a certain trafficking pathway can be subjected to selective regulation by dietary manipulation. This form of regulated plasma membrane targeting of a select subset of upstream signaling proteins may provide cells with the flexibility to coordinate the arrangement of signaling translators on the cell surface. Ultimately, this may allow organ systems such as the colon to optimally decode, respond, and adapt to the vagaries of an ever-changing extracellular environment.


Experimental Biology and Medicine | 2010

Apigenin and naringenin suppress colon carcinogenesis through the aberrant crypt stage in azoxymethane-treated rats

Tety Leonardi; Jairam Vanamala; Stella S. Taddeo; Laurie A. Davidson; Mary E. Murphy; Bhimanagouda S. Patil; Naisyin Wang; Raymond J. Carroll; Robert S. Chapkin; Joanne R. Lupton; Nancy D. Turner

Epidemiological evidence suggests that a diet abundant in fruits and vegetables may protect against colon cancer. Bioactive compounds, including flavonoids and limonoids, have been shown to possess antiproliferative and antitumorigenic effects in various cancer models. This experiment investigated the effects of four citrus flavonoids and one limonoid mixture at the promotion stage of chemically induced colon cancer in rats. Male Sprague–Dawley rats (n = 10 rats/group) were randomly allocated to one of six diets formulated to contain 0.1% apigenin, 0.02% naringenin, 0.1% hesperidin, 0.01% nobiletin, 0.035% limonin glucoside/obacunone glucoside mixture or a control diet (0% flavonoid/limonoid). Rats received experimental diets for 10 weeks and were injected with azoxymethane (15 mg/kg) at weeks 3 and 4. Excised colons were evaluated for aberrant crypt foci (ACF) formation, colonocyte proliferation (proliferating cell nuclear antigen assay), apoptosis (terminal deoxynucleotidyl transferase dUTP nick end labeling assay) and expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) (immunoblotting). When compared with the control diet, apigenin lowered the number of high multiplicity ACF (HMACF >4 aberrant crypts/focus) by 57% (P < 0.05), while naringenin lowered both the number of HMACF by 51% (P < 0.05) and the proliferative index by 32% (P < 0.05). Both apigenin and naringenin increased apoptosis of luminal surface colonocytes (78% and 97%, respectively; P < 0.05) when compared with the control diet. Hesperidin, nobiletin and the limonin glucoside/obacunone glucoside mixture did not affect these variables. The colonic mucosal protein levels of iNOS or COX-2 were not different among the six diet groups. The ability of dietary apigenin and naringenin to reduce HMACF, lower proliferation (naringenin only) and increase apoptosis may contribute toward colon cancer prevention. However, these effects were not due to mitigation of iNOS and COX-2 protein levels at the ACF stage of colon cancer.

Collaboration


Dive into the Joanne R. Lupton's collaboration.

Researchain Logo
Decentralizing Knowledge