Network


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

Hotspot


Dive into the research topics where Yang-Yi Fan is active.

Publication


Featured researches published by Yang-Yi Fan.


Journal of Immunology | 2004

Dietary docosahexaenoic acid suppresses T cell protein kinase Cθ lipid raft recruitment and IL-2 production

Yang-Yi Fan; Lan H. Ly; Rola Barhoumi; David N. McMurray; Robert S. Chapkin

To date, the proximal molecular targets through which dietary n-3 polyunsaturated fatty acids (PUFA) suppress the inflammatory process have not been elucidated. Because cholesterol and sphingolipid-enriched rafts have been proposed as platforms for compartmentalizing dynamically regulated signaling assemblies at the plasma membrane, we determined the in vivo effects of fish oil and highly purified docosahexaenoic acid (DHA; 22:6n-3) on T cell microdomain lipid composition and the membrane subdomain distribution of signal-transducing molecules (protein kinase C (PKC)θ, linker for activation of T cells, and Fas/CD95), before and after stimulation. Mice were fed diets containing 5 g/100 g corn oil (control), 4 g/100 g fish oil (contains a mixture of n-3 PUFA) plus 1 g/100 g corn oil, or 4 g/100 g corn oil plus 1 g/100 g DHA ethyl ester for 14 days. Dietary n-3 PUFA were incorporated into splenic T cell lipid raft and soluble membrane phospholipids, resulting in a 30% reduction in raft sphingomyelin content. In addition, polyclonal activation-induced colocalization of PKCθ with lipid rafts was reduced by n-3 PUFA feeding. With respect to PKCθ effector pathway signaling, both AP-1 and NF-κB activation, IL-2 secretion, and lymphoproliferation were inhibited by fish oil feeding. Similar results were obtained when purified DHA was fed. These data demonstrate for the first time that dietary DHA alters T cell membrane microdomain composition and suppresses the PKCθ signaling axis.


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.


Journal of Immunology | 2008

n-3 Polyunsaturated Fatty Acids Suppress the Localization and Activation of Signaling Proteins at the Immunological Synapse in Murine CD4+ T Cells by Affecting Lipid Raft Formation

Wooki Kim; Yang-Yi Fan; Rola Barhoumi; Roger Smith; David N. McMurray; Robert S. Chapkin

The molecular properties of immunosuppressive n-3 polyunsaturated fatty acids (PUFA) have not been fully elucidated. Using CD4+ T cells from wild-type control and fat-1 transgenic mice (enriched in n-3 PUFA), we show that membrane raft accumulation assessed by Laurdan (6-dodecanoyl-2-dimethyl aminonaphthalene) labeling was enhanced in fat-1 cells following immunological synapse (IS) formation by CD3-specific Ab expressing hybridoma cells. However, the localization of protein kinase Cθ, phospholipase Cγ-1, and F-actin into the IS was suppressed. In addition, both the phosphorylation status of phospholipase Cγ-1 at the IS and cell proliferation as assessed by CFSE labeling and [3H]thymidine incorporation were suppressed in fat-1 cells. These data imply that lipid rafts may be targets for the development of dietary agents for the treatment of autoimmune and chronic inflammatory diseases.


Lipids | 2002

Dietary n-3 PUFA alter colonocyte mitochondrial membrane composition and function

Robert S. Chapkin; Mee Young Hong; Yang-Yi Fan; Laurie A. Davidson; Lisa M. Sanders; Cara E. Henderson; Rola Barhoumi; Robert C. Burghardt; Nancy D. Turner; Joanne R. Lupton

There is experimental evidence that dietary fish oil, which contains the n−3 fatty acid family, i.e., EPA and DHA, protects against colon tumor development, in part by increasing apoptosis. Since mitochondria can act as central executioners of apoptosis, we hypothesized that EPA and DHA incorporation into colonocyte mitochondrial membranes, owing to their high degree of unsaturation, would enhance susceptibility to damage by reactive oxygen species (ROS) generated via oxidative phosphorylation. This, in turn, would compromise mitochondrial function, thereby initiating apoptosis. To test this hypothesis, colonic crypts were isolated from rats fed either fish oil, purified n−3 fatty acid ethyl esters, or corn oil (control). Dietary lipid source had no effect on colonic mitochondrial phospholipid class mole percentages, although incorporation of EPA and DHA was associated with a reduction in n−6 fatty acids known to enhance colon tumor development, i.e., linoleic acid (LNA) and its metabolic product, arachidonic acid (ARA). Select compositional changes in major phospholipid pools were correlated to alterations in mitochondrial function as assessed by confocal microscopy. The mol% sum of LNA plus ARA in cardiolipin was inversely correlated with ROS (P=0.024). Ethanolamine glycerophospholipid ARA (P=0.046) and choline glycerophospholipid INA (P=0.033) levels were positively correlated to mitochondrial membrane potential. In contrast, ethanolamine glycerophospholipid EPA (P=0.042) and DHA (P=0.024), levels were negatively correlated to mitochondrial membrane potential. Additionally, EPA and DHA levels in choline glycerophospholipids (P=0.026) were positively correlated with caspase 3 activity. These data provide evidence in vivo indicating that dietary FPA and DHA induce compositional changes in colonic mitochondrial membrane phospholipids that facilitate appotosis.


Biochimica et Biophysica Acta | 1993

Purified dietary n − 3 polyunsaturated fatty acids alter diacylglycerol mass and molecular species composition in concanavalin A-stimulated murine splenocytes

Kara Hosack Fowler; David N. McMurray; Yang-Yi Fan; Harold M. Aukema; Robert S. Chapkin

A low-dose, short-term dietary supplementation with highly purified (n-3) fatty acid ethyl esters was studied in mice to determine the effect on splenic cell membrane diacylglycerol mass and composition. Mice were fed diets containing either 3% safflower oil (SAF) ethyl esters, 2% SAF plus 1% eicosapentaenoic acid ethyl ester (EPA), or 2% SAF plus 1% docosahexaenoic acid ethyl ester (DHA). Following a 10-day feeding period, pathogen-free mice were sacrificed and splenic cells isolated and stimulated with concanavalin A (Con A) at 10 micrograms/ml. After 0 min (basal), 5 min, and 180 min, 1,2-diacyl, 1-O-alkyl-2-acyl, and 1-O-alkenyl-2-acyl-sn-glycerol subclasses were isolated and quantitated by HPLC. Diacylglycerol (DAG) was found to be the major diradylglycerol (DG) component in murine splenocytes. DHA-fed mice had significantly (P < 0.05) higher levels of DAG at all stimulation time points relative to EPA and SAF animals. Significant effects (P < 0.05) of diet, time, and a diet x time interaction (P < 0.05) were noted for various DAG molecular species. In general, a significantly higher (n-3) polyunsaturated fatty acid (PUFA) content in the EPA and DHA groups, and a significantly higher (n-6) PUFA content in the SAF group was noted. 18:0-22:5(n-3), 18:1-22:5(n-3) and 16:1-20:5(n-3) species were present only in EPA and DHA-DAG, confirming the incorporation of (n-3) fatty acids into splenocyte DAG. The data indicate that the molecular species composition of murine splenocyte DAG is significantly modulated by low-dose, short-term EPA and DHA feeding. In addition, substitution of SAF with DHA results in an increase in DAG mass. These alterations could potentially influence signal transduction pathways regulating lymphocyte function.


British Journal of Nutrition | 2011

Dietary fish oil and curcumin combine to modulate colonic cytokinetics and gene expression in dextran sodium sulphate-treated mice

Qian Jia; Ivan Ivanov; Zlatomir Z. Zlatev; Robert C. Alaniz; Brad R. Weeks; Evelyn S. Callaway; Jennifer S. Goldsby; Laurie A. Davidson; Yang-Yi Fan; Lan Zhou; Joanne R. Lupton; David N. McMurray; Robert S. Chapkin

Both fish oil (FO) and curcumin have potential as anti-tumour and anti-inflammatory agents. To further explore their combined effects on dextran sodium sulphate (DSS)-induced colitis, C57BL/6 mice were randomised to four diets (2 × 2 design) differing in fatty acid content with or without curcumin supplementation (FO, FO+2 % curcumin, maize oil (control, MO) or MO+2 % curcumin). Mice were exposed to one or two cycles of DSS in the drinking-water to induce either acute or chronic intestinal inflammation, respectively. FO-fed mice exposed to the single-cycle DSS treatment exhibited the highest mortality (40 %, seventeen of forty-three) compared with MO with the lowest mortality (3 %, one of twenty-nine) (P = 0·0008). Addition of curcumin to MO increased (P = 0·003) mortality to 37 % compared with the control. Consistent with animal survival data, following the one- or two-cycle DSS treatment, both dietary FO and curcumin promoted mucosal injury/ulceration compared with MO. In contrast, compared with other diets, combined FO and curcumin feeding enhanced the resolution of chronic inflammation and suppressed (P < 0·05) a key inflammatory mediator, NF-κB, in the colon mucosa. Mucosal microarray analysis revealed that dietary FO, curcumin and FO plus curcumin combination differentially modulated the expression of genes induced by DSS treatment. These results suggest that dietary lipids and curcumin interact to regulate mucosal homeostasis and the resolution of chronic inflammation in the colon.


Journal of Nutrition | 2009

Dietary Curcumin and Limonin Suppress CD4 T-Cell Proliferation and Interleukin-2 Production in Mice

Wooki Kim; Yang-Yi Fan; Roger Smith; Bhimanagouda S. Patil; Guddadarangavvanahally K. Jayaprakasha; David N. McMurray; Robert S. Chapkin

Phytochemicals may reduce chronic inflammation and cancer risk in part by modulating T-cell nuclear factor-kappaB (NF-kappaB) activation. Therefore, we examined the effects of curcumin (Cur) and limonin (Lim) feeding on NF-kappaB-dependent CD4(+) T-cell proliferation. DO11.10 transgenic mice (n = 5-7) were fed diets containing 1% Cur or 0.02% Lim combined with either (n-6) PUFA [5% corn oil (CO)] or (n-3) PUFA [4% fish oil+1% corn oil (FO)] for 2 wk, followed by splenic CD4(+) T-cell isolation and stimulation with ovalbumin peptide 323-339 (OVA) and antigen-presenting cells from mice fed a conventional nonpurified rodent diet. Both Cur and Lim diets suppressed (P < 0.05) NF-kappaB p65 nuclear translocation in activated CD4(+) T-cells. In contrast, activator protein-1 (c-Jun) and nuclear factor of activated T-cells c1 were not affected compared with the CO control diet (no Cur or Lim). CD4(+) T-cell proliferation in response to either mitogenic anti-CD3/28 monoclonal antibodies (mAb) or antigenic stimulation by OVA was also suppressed (P < 0.05) by Cur as assessed by carboxyfluorescein succinimidyl ester staining. In contrast, interleukin-2 production was not directly associated with NF-kappaB status. Interestingly, dietary combination with FO enhanced the suppressive effects (P < 0.05) of Cur or Lim with respect to CD4(+) T-cell proliferation in response to anti-CD3/28 mAb. These results suggest that combination chemotherapy (FO+Cur or Lim) may favorably modulate CD4(+) T-cell-mediated inflammation.


American Journal of Physiology-cell Physiology | 1999

Antagonism of CD95 signaling blocks butyrate induction of apoptosis in young adult mouse colonic cells

Yang-Yi Fan; Jianhu Zhang; Rola Barhoumi; Robert C. Burghardt; Nancy D. Turner; Joanne R. Lupton; Robert S. Chapkin

There is great interest in utilizing butyrate as a chemopreventive agent for colon tumorigenesis because of its ability to promote apoptosis in colon tumor cell lines. Because CD95 (APO-1/Fas) transduces signals resulting in apoptosis, we tested the hypothesis that butyrate-dependent colonocyte apoptosis is mediated by this death receptor. Butyrate (1 mM) exposure for 24 h upregulated expression of Fas and its ligand in young adult mouse colon (YAMC) cells. To delineate the proapoptotic effect of butyrate and to avoid the confounding effects of detachment from the extracellular matrix, adherent cell apoptosis was monitored as loss of plasma membrane asymmetry and dissipation of mitochondrial membrane potential (ΔΨmt) by laser cytometry. Soluble Fas receptor protein (Fas:Fc chimera) and caspase inhibitors (z-VAD-fmk and z-IETD-fmk) blocked butyrate induction of apoptosis. Treatment with Fas agonistic antibody (clone Jo-2) significantly induced cell death, indicating that Fas in colonocytes is functional. In addition, butyrate promoted apoptosis by inducing loss of ΔΨmt and phospholipid asymmetry of the plasma membrane after 12 and 24 h of exposure, respectively, before cell detachment. Therefore, Fas receptor-dependent signal transduction is involved in butyrate induction of apoptosis in colonocytes.There is great interest in utilizing butyrate as a chemopreventive agent for colon tumorigenesis because of its ability to promote apoptosis in colon tumor cell lines. Because CD95 (APO-1/Fas) transduces signals resulting in apoptosis, we tested the hypothesis that butyrate-dependent colonocyte apoptosis is mediated by this death receptor. Butyrate (1 mM) exposure for 24 h upregulated expression of Fas and its ligand in young adult mouse colon (YAMC) cells. To delineate the proapoptotic effect of butyrate and to avoid the confounding effects of detachment from the extracellular matrix, adherent cell apoptosis was monitored as loss of plasma membrane asymmetry and dissipation of mitochondrial membrane potential (DeltaPsi(mt)) by laser cytometry. Soluble Fas receptor protein (Fas:Fc chimera) and caspase inhibitors (z-VAD-fmk and z-IETD-fmk) blocked butyrate induction of apoptosis. Treatment with Fas agonistic antibody (clone Jo-2) significantly induced cell death, indicating that Fas in colonocytes is functional. In addition, butyrate promoted apoptosis by inducing loss of DeltaPsi(mt) and phospholipid asymmetry of the plasma membrane after 12 and 24 h of exposure, respectively, before cell detachment. Therefore, Fas receptor-dependent signal transduction is involved in butyrate induction of apoptosis in colonocytes.


Biochemical Journal | 2012

n-3 polyunsaturated fatty acids suppress phosphatidylinositol 4,5-bisphosphate-dependent actin remodelling during CD4+ T-cell activation.

Tim Y. Hou; Jennifer M. Monk; Yang-Yi Fan; Rola Barhoumi; Yong Q. Chen; Gonzalo M. Rivera; David N. McMurray; Robert S. Chapkin

n-3 PUFA (polyunsaturated fatty acids), i.e. DHA (docosahexaenoic acid), found in fish oil, exhibit anti-inflammatory properties; however, the molecular mechanisms remain unclear. Since PtdIns(4,5)P2 resides in raft domains and DHA can alter the size of rafts, we hypothesized that PtdIns(4,5)P2 and downstream actin remodelling are perturbed by the incorporation of n-3 PUFA into membranes, resulting in suppressed T-cell activation. CD4+ T-cells isolated from Fat-1 transgenic mice (membranes enriched in n-3 PUFA) exhibited a 50% decrease in PtdIns(4,5)P2. Upon activation by plate-bound anti-CD3/anti-CD28 or PMA/ionomycin, Fat-1 CD4+ T-cells failed to metabolize PtdIns(4,5)P2. Furthermore, actin remodelling failed to initiate in Fat-1 CD4+ T-cells upon stimulation; however, the defect was reversed by incubation with exogenous PtdIns(4,5)P2. When Fat-1 CD4+ T-cells were stimulated with anti-CD3/anti-CD28-coated beads, WASP (Wiskott-Aldrich syndrome protein) failed to translocate to the immunological synapse. The suppressive phenotype, consisting of defects in PtdIns(4,5)P2 metabolism and actin remodelling, were recapitulated in CD4+ T-cells isolated from mice fed on a 4% DHA triacylglycerol-enriched diet. Collectively, these data demonstrate that n-3 PUFA, such as DHA, alter PtdIns(4,5)P2 in CD4+ T-cells, thereby suppressing the recruitment of WASP to the immunological synapse, and impairing actin remodelling in CD4+ T-cells.


PLOS ONE | 2010

Incorporation of a Dietary Omega 3 Fatty Acid Impairs Murine Macrophage Responses to Mycobacterium tuberculosis

Diana L. Bonilla; Lan H. Ly; Yang-Yi Fan; Robert S. Chapkin; David N. McMurray

Background Beside their health benefits, dietary omega 3 polyunsaturated fatty acids (n-3 PUFA) might impair host resistance to Mycobacterium tuberculosis (Mtb) by creating an immunosuppressive environment. We hypothesized that incorporation of n-3 PUFA suppresses activation of macrophage antimycobacterial responses and favors bacterial growth, in part, by modulating the IFNγ-mediated signaling pathway. Methodology/Principal Findings Murine macrophage-like J774A.1 cells were incubated with bovine serum albumin (BSA)-conjugated docosahexaenoic acid (DHA; 22:6n-3) or BSA alone, activated with recombinant IFNγ, and infected with a virulent strain (H37Rv) of M. tuberculosis. The fatty acid composition of macrophage membranes was modified significantly by DHA treatment. DHA-treated macrophages were less effective in controlling intracellular mycobacteria and showed impaired oxidative metabolism and reduced phagolysosome maturation. Incorporation of DHA resulted in defective macrophage activation, as characterized by reduced production of pro-inflammatory cytokines (TNFα, IL-6 and MCP-1), and lower expression of co-stimulatory molecules (CD40 and CD86). DHA treatment impaired STAT1 phosphorylation and colocalization of the IFNγ receptor with lipid rafts, without affecting surface expression of IFNγ receptor. Conclusions/Significance We conclude that DHA reduces the ability of J774A.1 cells to control M. tuberculosis in response to activation by IFNγ, by modulation of IFNγ receptor signaling and function, suggesting that n-3 PUFA-enriched diets may have a detrimental effect on host immunity to tuberculosis.

Collaboration


Dive into the Yang-Yi Fan's collaboration.

Researchain Logo
Decentralizing Knowledge