Network


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

Hotspot


Dive into the research topics where Kevin A. Harvey is active.

Publication


Featured researches published by Kevin A. Harvey.


The FASEB Journal | 2000

Sphingosine 1-phosphate released from platelets during clotting accounts for the potent endothelial cell chemotactic activity of blood serum and provides a novel link between hemostasis and angiogenesis

Denis English; Zachary Welch; A. Thomas Kovala; Kevin A. Harvey; Olga V. Volpert; David N. Brindley; Joe G. N. Garcia

ABSTRACT Recent studies have identified factors responsible for angiogenesis within developing tumors, but mediators of vessel formation at sites of trauma, injury, and wound healing are not clearly established. Here we show that sphingosine 1‐phosphate (S1P) released by platelets during blood clotting is a potent, specific, and selective endothelial cell chemoattractant that accounts for most of the strong endothelial cell chemotactic activity of blood serum, an activity that is markedly diminished in plasma. Preincubation of endothelial cells with pertussis toxin inhibited this effect of S1P, demonstrating the involvement of a Gαi‐coupled receptor. After S1P‐induced migration, endothelial cells proliferated avidly and differentiated forming multicellular structures suggestive of early blood vessel formation. S1P was strikingly effective in enhancing the ability of fibroblast growth factor to induce angiogenesis in the avascular mouse cornea. Our results show that blood coagulation initiates endothelial cell angiogenic responses through the release of S1P, a potent endothelial cell chemoattractant that exerts its effects by activating a receptor‐dependent process.—English, D., Welch, Z., Kovala, A. T., Harvey, K., Volpert, O. V., Brindley, D. N., Garcia, J. G. N. Sphingosine 1‐phosphate released from platelets during clotting accounts for the potent endothelial cell chemotactic activity of blood serum and provides a novel link between hemostasis and angiogenesis. FASEB J. 14, 2255–2265 (2000)


International Journal of Cancer | 2005

Omega-3 polyunsaturated fatty acids attenuate breast cancer growth through activation of a neutral sphingomyelinase-mediated pathway

Min Wu; Kevin A. Harvey; Nargiz Ruzmetov; Zachary Welch; Laura Sech; Kim Jackson; William Stillwell; Gary P. Zaloga; Rafat A. Siddiqui

The effect of fish oils and their active omega‐3 fatty acid constituents, docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA), were investigated on breast cancer growth. In in vivo experiments, mice were fed diets that were rich in either omega‐3 (fish oil) or omega‐6 (corn oil) fatty acids. Three weeks after implantation of MDA‐MB‐231 breast cancer cells, the tumor volume and weight were significantly lower (p < 0.05) for mice fed the omega‐3 diets compared to those fed the omega‐6 diets. Dietary fish oil also caused a 40% (p < 0.05) increase in neutral sphingomyelinase (N‐SMYase) activity in the tumors. The tumor tissues from fish oil‐fed animals expressed elevated p21 (waf1/cip1) mRNA, whereas tumor tissues from corn oil‐fed animals exhibited undetectable levels of p21 expression. In in vitro experiments, at concentrations as low as 25 μM, DHA and EPA inhibited the growth of cultured MDA‐MB‐231 cells in a dose‐dependent manner by 20–25% (p < 0.05). N‐SMYase activity was also increased by 30–40% (p < 0.05) in the DHA‐ or EPA‐treated cells in which an increase in ceramide formation was observed. DHA and EPA were both observed to enhance membrane bleb formation and also to induce the expression of p21. Omega‐3 fatty acids‐induced bleb formation and p21 expression were inhibited by the N‐SMYase inhibitor GW4869, which also inhibited apoptosis by approximately 40% (p < 0.05). The results suggest that inhibition of breast cancer growth in nude mice by dietary fish oil and inhibition of breast cancer cell growth in culture by treatment with DHA and EPA is mediated by activation of N‐SMYase.


Biochimica et Biophysica Acta | 2001

Docosahexaenoic acid induces apoptosis in Jurkat cells by a protein phosphatase-mediated process.

Rafat A. Siddiqui; Laura J. Jenski; Kristiana Neff; Kevin A. Harvey; Richard J. Kovacs; William Stillwell

Docosahexaenoic acid (DHA) is an omega-3 fatty acid under intense investigation for its ability to modulate cancer cell growth and survival. This research was performed to study the cellular and molecular effects of DHA. Our experiments indicated that the treatment of Jurkat cells with DHA inhibited their survival, whereas similar concentrations (60 and 90 microM) of arachidonic acid and oleic acid had little effect. To explore the mechanism of inhibition, we used several measures of apoptosis to determine whether this process was involved in DHA-induced cell death in Jurkat cells. Caspase-3, an important cytosolic downstream regulator of apoptosis, is activated by death signals through proteolytic cleavage. Incubation of Jurkat cells with 60 and 90 microM DHA caused proteolysis of caspase-3 within 48 and 24 h, respectively. DHA treatment also caused the degradation of poly-ADP-ribose polymerase and DNA fragmentation as assayed by flow cytometric TUNEL (terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling) assay. These results indicate that DHA induces apoptosis in Jurkat leukemic cells. DHA-induced apoptosis was effectively inhibited by tautomycin and cypermethrin at concentrations that affect protein phosphatase 1 (PP1) and protein phosphatase 2B (PP2B) activities, respectively, implying a role for these phosphatases in the apoptotic pathway. Okadaic acid, an inhibitor of protein phosphatase 2A, had no effect on DHA-induced apoptosis. These results suggest that one mechanism through which DHA may control cancer cell growth is through apoptosis involving PP1/PP2B protein phosphatase activities.


Breast Cancer Research | 2005

Anticancer properties of propofol-docosahexaenoate and propofol-eicosapentaenoate on breast cancer cells

Rafat A. Siddiqui; Mustapha Zerouga; Min Wu; Alicia Castillo; Kevin A. Harvey; Gary P. Zaloga; William Stillwell

IntroductionEpidemiological evidence strongly links fish oil, which is rich in docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA), with low incidences of several types of cancer. The inhibitory effects of omega-3 polyunsaturated fatty acids on cancer development and progression are supported by studies with cultured cells and animal models. Propofol (2,6-diisopropylphenol) is the most extensively used general anesthetic–sedative agent employed today and is nontoxic to humans at high levels (50 μg/ml). Clinically relevant concentrations of propofol (3 to 8 μg/ml; 20 to 50 μM) have also been reported to have anticancer activities. The present study describes the synthesis, purification, characterization and evaluation of two novel anticancer conjugates, propofol-docosahexaenoate (propofol-DHA) and propofol-eicosapentaenoate (propofol-EPA).MethodsThe conjugates linking an omega-3 fatty acid, either DHA or EPA, with propofol were synthesized and tested for their effects on migration, adhesion and apoptosis on MDA-MB-231 breast cancer cells.ResultsAt low concentrations (25 μM), DHA, EPA or propofol alone or in combination had minimal effect on cell adhesion to vitronectin, cell migration against serum and the induction of apoptosis (only 5 to 15% of the cells became apoptotic). In contrast, the propofol-DHA or propofol-EPA conjugates significantly inhibited cell adhesion (15 to 30%) and migration (about 50%) and induced apoptosis (about 40%) in breast cancer cells.ConclusionThese results suggest that the novel propofol-DHA and propofol-EPA conjugates reported here may be useful for the treatment of breast cancer.


Chemistry and Physics of Lipids | 2008

Anticancer properties of oxidation products of docosahexaenoic acid.

Rafat A. Siddiqui; Kevin A. Harvey; William Stillwell

Docosahexaenoic acid (DHA) is the longest, most unsaturated, and hence, most oxidizable fatty acid commonly found in nature. The mechanisms behind DHAs many biological functions remain a subject of much debate. Here we review one important, but often unstudied, aspect of DHA function, namely, the potential role of its many oxidation products. We divide this review into camps, enzymatic and non-enzymatic oxidations, and report their effects primarily on induction of apoptosis in cancer cells. We conclude that the study of the effects of lipid peroxidation products on biochemical function will be a difficult but highly rewarding area for future studies.


Nutrition in Clinical Practice | 2007

Modulation of Lipid Rafts by Ω-3 Fatty Acids in Inflammation and Cancer: Implications for Use of Lipids During Nutrition Support

Rafat A. Siddiqui; Kevin A. Harvey; Gary P. Zaloga; William Stillwell

Current understanding of biologic membrane structure and function is largely based on the concept of lipid rafts. Lipid rafts are composed primarily of tightly packed, liquid-ordered sphingolipids/cholesterol/saturated phospholipids that float in a sea of more unsaturated and loosely packed, liquid-disordered lipids. Lipid rafts have important clinical implications because many important membrane-signaling proteins are located within the raft regions of the membrane, and alterations in raft structure can alter activity of these signaling proteins. Because rafts are lipid-based, their composition, structure, and function are susceptible to manipulation by dietary components such as ω-3 polyunsaturated fatty acids and by cholesterol depletion. We review how alteration of raft lipids affects the raft/nonraft localization and hence the function of several proteins involved in cell signaling. We focus our discussion of raft-signaling proteins on inflammation and cancer.


Clinical Nutrition | 2010

Long-chain saturated fatty acids induce pro-inflammatory responses and impact endothelial cell growth

Kevin A. Harvey; Candace L. Walker; Thomas M. Pavlina; Zhidong Xu; Gary P. Zaloga; Rafat A. Siddiqui

BACKGROUND & AIMS Saturated fatty acids (SFAs), significant components of enteral and parenteral formulations, have been linked to cardiovascular complications. However, the effect of SFAs upon vascular inflammation is less clear. Endothelial cells (EC) play an important role in the acute inflammatory responses. We, therefore, evaluated the acute effects of different chain-length SFAs upon EC functions. METHODS Endothelial cells were cultured with various SFAs. Growth and cytotoxicity were determined by WST-1 assay. Apoptosis and pro-inflammatory adhesion molecule (ICAM-1) expression was assayed using flow cytometry. Activation of NF-kappaB was analyzed using western blot analysis. RESULTS Long-chain SFAs (C14:0-C20:0) inhibited EC growth in a chain-length dependent manner. Medium-chain SFAs (C6:0-C12:0) did not significantly affect EC growth. In contrast, the short-chain SFA (C4:0) stimulated cellular growth. Stearic acid induced significantly more EC apoptosis and necrosis than palmitic acid or myristic acids. Stearic acid (>10muM) treatment also significantly increased ICAM-1 expression. Stearic acids pro-inflammatory response was confirmed by phosphorylation of IkappaB-alpha and NF-kappaB in a dose dependent manner. CONCLUSIONS Long-chain SFAs can induce pro-inflammatory responses and significantly impact growth and viability of EC. Our data suggest that the presence of long-chain SFAs in parenteral formulations may have harmful effects on the vascular system.


Nutrition and Cancer | 2005

Green tea polyphenols modulate secretion of urokinase plasminogen activator (uPA) and inhibit invasive behavior of breast cancer cells.

Veronika Slivova; Gary P. Zaloga; Stephen J. DeMichele; Pradip Mukerji; Yung-Sheng Huang; Rafat A. Siddiqui; Kevin A. Harvey; Tatiana Valachovicova; Daniel Sliva

Many epidemiological studies have suggested that consumption of green tea may decrease the risk of cancer. The chemopreventive effect of green tea polyphenols (GTP) has been demonstrated through the inhibition of cell proliferation and angiogenesis in cell culture and animal models of breast cancer. Metastasis of breast cancer is the major reason for the high mortality of breast cancer patients and is directly linked to the invasive behavior of breast cancer cells. Cancer metastasis consists of several interdependent processes including cancer cell adhesion, cancer cell migration, and invasion of cancer cells. In this study, we evaluated the effect of GTP on human breast cancer cells, and we show that in addition to inhibiting cell growth, GTP also suppressed the invasive behavior of MDA-MB-231 cells. These anti-invasive effects of GTP were the result of the inhibition of constitutively active transcription factors AP-1 and NF-κB, which further suppressed secretion of urokinase plasminogen activator (uPA) from breast cancer cells. Based on these results, it can be hypothesized that GTP treatment resulted in the inhibition of formation of signaling complexes responsible for cell adhesion and migration (uPA, uPA receptor, vitronectin, integrin receptor) and cell invasion (uPA, uPA receptor). Our results indicate that GTP may contribute to the anticancer effects of green tea by inhibiting the invasive behavior of cancer cells.


British Journal of Nutrition | 2008

Trans-fatty acids induce pro-inflammatory responses and endothelial cell dysfunction.

Kevin A. Harvey; Tyler Arnold; Tamkeen Rasool; Caryl J. Antalis; Steven J. Miller; Rafat A. Siddiqui

Epidemiological data indicate that there is a strong association between intake of trans-18 : 2 fatty acids (TFA) and sudden cardiac death. There is little known about the mechanisms by which TFA exert harmful effects on the cardiovascular system. The present in vitro study is the first to demonstrate the effects of membrane-incorporated C18 : 2 TFA on human aortic endothelial cell (HAEC) function. Trans-18 : 2 fatty acids were incorporated to a greater extent (2-fold) in the phospholipid fraction of endothelial cells than that of cis-18 : 2; furthermore, these fatty acids were enriched to a similar extent in the TAG fraction. Flow cytometric analysis indicated that TFA treatment of HAEC significantly increased the expression of endothelial adhesion molecules, including intercellular adhesion molecule-1 (CD54) and vitronectin receptor (CD51/CD61). Incorporation of TFA into membranes increased HAEC adhesion to fibronectin- or vitronectin-coated plates by 1.5- to 2-fold, respectively. Neutrophil and monocyte adhesion to HAEC monolayers was nearly proportional to adhesion molecule expression. TFA treatment also induced the release of monocyte chemoattractant protein-1 by nearly 3-fold in non-stimulated HAEC. Furthermore, we examined the role of TFA on in vitro angiogenic assays. Chemotactic migration of TFA-treated HAEC toward sphingosine-1-phosphate (SPP) was significantly increased compared with controls. Conversely, capillary morphogenesis of TFA-treated HAEC was significantly inhibited in response to SPP, suggesting that TFA incorporation suppresses endothelial cell differentiation. In conclusion, these in vitro studies demonstrated that TFA play a role in the induction of pro-inflammatory responses and endothelial cell dysfunction.


Journal of Lipid Research | 2010

Oleic acid inhibits stearic acid-induced inhibition of cell growth and pro-inflammatory responses in human aortic endothelial cells

Kevin A. Harvey; Candace L. Walker; Zhidong Xu; Phillip Whitley; Thomas M. Pavlina; Mary Hise; Gary P. Zaloga; Rafat A. Siddiqui

Saturated fatty acids (SFAs), significant components of both enteral/parenteral nutritional formulations (including diet), are linked to cardiovascular disease complications, such as atherosclerosis. We investigated whether oleic acid (C18:1n-9) reduces the growth inhibitory and pro-inflammatory effects of the stearic acid (C18:0) in human aortic endothelial cells (HAEC). Stearic acid induced growth inhibition at concentrations less than 50 μM, whereas higher concentrations invoked cytotoxicity. Stearic acid-induced growth inhibition and cytotoxic effects were eradicated upon cosupplementation with oleic acid (25 μM). Oleic acid (as low as 5 μM) also inhibited the stearic acid-induced increase in intercellular adhesion molecule-1 (ICAM-1) expression. Stearic acid-induced phosphorylation of nuclear factor-kappa B (NF-κB), a transcriptional regulator of ICAM-1, was also reduced by oleic acid. HAECs supplemented with either stearic or oleic acid resulted in cellular incorporation of C18:0 and C18:1n-9, respectively. Stearic acid primarily incorporated into phospholipids without increasing the total fatty acid content in HAECs. In contrast, oleic acid, with or without stearic acid, incorporated into both phospholipids and triglycerides, with a significant increase in total fatty acid amounts in triglycerides. Our data suggest that oleic acid has the ability to reduce the inflammatory effects of long-chain SFAs in HAECs through reducing cellular stearic acid incorporation and NF-κB activation.

Collaboration


Dive into the Kevin A. Harvey's collaboration.

Top Co-Authors

Avatar

Rafat A. Siddiqui

Indiana University – Purdue University Indianapolis

View shared research outputs
Top Co-Authors

Avatar

Zhidong Xu

Indiana University Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Daniel Sliva

Indiana University Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

James M. Thompson

Houston Methodist Hospital

View shared research outputs
Top Co-Authors

Avatar

Jan Jansen

Houston Methodist Hospital

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge