Network


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

Hotspot


Dive into the research topics where David A. Taylor-Fishwick is active.

Publication


Featured researches published by David A. Taylor-Fishwick.


Progress in Lipid Research | 2011

Functional and pathological roles of the 12- and 15-lipoxygenases

Anca D. Dobrian; David C. Lieb; Banumathi K. Cole; David A. Taylor-Fishwick; Swarup K. Chakrabarti; Jerry L. Nadler

The 12/15-lipoxygenase enzymes react with fatty acids producing active lipid metabolites that are involved in a number of significant disease states. The latter include type 1 and type 2 diabetes (and associated complications), cardiovascular disease, hypertension, renal disease, and the neurological conditions Alzheimers disease and Parkinsons disease. A number of elegant studies over the last thirty years have contributed to unraveling the role that lipoxygenases play in chronic inflammation. The development of animal models with targeted gene deletions has led to a better understanding of the role that lipoxygenases play in various conditions. Selective inhibitors of the different lipoxygenase isoforms are an active area of investigation, and will be both an important research tool and a promising therapeutic target for treating a wide spectrum of human diseases.


Annals of Surgery | 2004

A pentadecapeptide fragment of islet neogenesis-associated protein increases beta-cell mass and reverses diabetes in C57BL/6J mice.

Lawrence Rosenberg; Mark Lipsett; Ji-Won Yoon; Marc Prentki; Rennian Wang; Hee-Sook Jun; Gary L. Pittenger; David A. Taylor-Fishwick; Aaron I. Vinik

Objective:The objective of this study was to demonstrate that islet neogenesis-associated protein (INGAP) peptide, a pentadecapeptide containing the biologically active portion of native INGAP, increases functional β-cell mass in normal animals and can be used therapeutically to reverse hyperglycemia in streptozotocin-induced diabetes. Summary Background Data:INGAP, a 175 amino acid pancreatic acinar cell protein, has been suggested to be implicated in β-cell mass expansion. Methods:In the first part of this study, normoglycemic hamsters were administered either 500 &mgr;g INGAP peptide (n = 30) or saline (n = 20) intraperitoneally daily and sacrificed after 10 or 30 days of treatment. Blood glucose and insulin levels were measured, and a histologic and morphometric analysis of the pancreas was performed to determine the effect of INGAP peptide on the endocrine pancreas. In the second part of the study, 6- to 8-week-old C57BL/6J mice (n = 8) were administered multiple low doses of the β-cell toxin streptozotocin (STZ) inducing insulitis and hyperglycemia. The mice were then injected with INGAP peptide (n = 4) or saline (n = 4) for 39 days and sacrificed at 48 days. Two additional groups of diabetic mice were administered either a peptide composed of a scrambled sequence of amino acids from INGAP peptide (n = 5) or exendin-4 (n = 5), an incretin that has been associated with amelioration of hyperglycemia. Results:Islet cell neogenesis was stimulated in INGAP-treated hamsters by 10 days. At 30 days, the foci of new endocrine cells had the appearance of mature islets. There was a 75% increase in islet number, with normal circulating levels of blood glucose and insulin. Administration of INGAP peptide to diabetic mice reversed the diabetic state in all animals, and this was associated with increased expression of PDX-1 in duct cells and islet cell neogenesis with a reduction of insulitis in the new islets. Diabetic mice treated with exendin-4 or a scrambled INGAP peptide did not revert from hyperglycemia. Conclusion:Because there is a deficiency of β-cell mass in both type-1 and type-2 diabetes, INGAP peptide stimulation of fully functional neoislet differentiation may provide a novel approach for diabetes therapy.


Molecular and Cellular Endocrinology | 2012

Integration of pro-inflammatory cytokines, 12-lipoxygenase and NOX-1 in pancreatic islet beta cell dysfunction.

Jessica R. Weaver; Theodore R. Holman; Yumi Imai; Ajit Jadhav; Victor Kenyon; David J. Maloney; Jerry L. Nadler; Ganesha Rai; Anton Simeonov; David A. Taylor-Fishwick

Elevated cellular reactive species, which can be produced by diabetic serum conditions such as elevated inflammatory cytokines, lipotoxicity or glucotoxicity contribute to islet beta cell dysfunction and cell death. Cellular pathways that result in beta cell oxidative stress are poorly resolved. In this study, stimulation of human donor islets, primary mouse islets or homogeneous beta cell lines with a cocktail of inflammatory cytokines (TNFα, IL-1β, and INFγ) significantly induced NADPH oxidase-1 (NOX-1) gene expression (p<0.05). This pro-inflammatory cytokine cocktail concomitantly induced loss of islet glucose stimulated insulin response (p<0.05), elevated expression of MCP-1 (p<0.01), increased cellular reactive oxygen species (ROS) and induced cell death. Inhibitors of NADPH oxidase, apocynin and diphenyleneiodonium, and a dual selective NOX1/4 inhibitor, blocked ROS generation (p<0.01) and induction of MCP-1 (p<0.05) by pro-inflammatory cytokines in beta cells. It has previously been reported that pro-inflammatory cytokine stimulation induces 12-lipoxygenase (12-LO) expression in human islets. 12-Hydroxyeicosatetraenoic acid (12-HETE), a product of 12-LO activity, stimulated NOX-1 expression in human islets (p<0.05). A novel selective inhibitor of 12-LO blocked induction of NOX-1, production of ROS and pro-caspase 3 cleavage by pro-inflammatory cytokines in INS-1 beta cells (p<0.01). Inhibition was not seen with a structurally related but inactive analog. Importantly, islets from human type 2 diabetic donors have an elevated expression of NOX-1 (p<0.05). This study describes an integrated pathway in beta cells that links beta cell dysfunction induced by pro-inflammatory cytokines with 12-lipoxygenase and NADPH oxidase (NOX-1) activation. Inhibitors of this pathway may provide a new therapeutic strategy to preserve beta cell mass in diabetes.


The Journal of Clinical Endocrinology and Metabolism | 2010

12-Lipoxygenase Products Reduce Insulin Secretion and β-Cell Viability in Human Islets

Kaiwen Ma; Craig S. Nunemaker; R. Wu; Swarup K. Chakrabarti; David A. Taylor-Fishwick; J. L. Nadler

CONTEXTnInflammation is increasingly recognized as an important contributing factor in diabetes mellitus. Lipoxygenases (LOs) produce active lipids that promote inflammatory damage by catalyzing the oxidation of linoleic and arachidonic acid, and LO is expressed in rodent and human islets. Little is known about the differential effect of the various hydroxyeicosatetraenoic acids (HETEs) that result from LO activity in human islets.nnnOBJECTIVEnWe compared the effects of 12-LO products on human islet viability and function.nnnDESIGNnHuman islets were treated with stable compounds derived from LOs: 12(S)-HETE, 15HETE, 12HPETE, and 12RHETE and then examined for insulin secretion and islet viability. The p38-MAPK (p38) and JNK stress-activated pathways were investigated as mechanisms of 12-LO-mediated islet inhibition in rodent and human islets.nnnRESULTSnInsulin secretion was consistently reduced by 12(S)-HETE and 12HPETE. 12(S)-HETE at 1 nm reduced viability activity by 32% measured by MTT assay and increased cell death by 50% at 100 nm in human islets. These effects were partially reversed with lisofylline, a small-molecule antiinflammatory compound that protects mitochondrial function. 12(S)-HETE increased phosphorylated p38-MAPK (pp38) protein activity in human islets. Injecting 12-LO siRNA into C57BL/6 mice reduced 12-LO and pp38-MAPK protein levels in mouse islets. The addition of proinflammatory cytokines increased pp38 levels in normal mouse islets but not in siRNA-treated islets.nnnCONCLUSIONSnThese data suggest that 12(S)-HETE reduces insulin secretion and increases cell death in human islets. The 12-LO pathway is present in human islets, and expression is up-regulated by inflammatory cytokines. Reduction of 12-LO activity could thus provide a new therapeutic approach to protect human beta-cells from inflammatory injury.


Journal of Medicinal Chemistry | 2011

Discovery of Potent and Selective Inhibitors of Human Platelet type 12-Lipoxygenase

Victor Kenyon; Ganesha Rai; Ajit Jadhav; Lena Schultz; Michelle Armstrong; J. Brian Jameson; Steven Perry; Netra Joshi; James M. Bougie; William Leister; David A. Taylor-Fishwick; Jerry L. Nadler; Michael Holinstat; Anton Simeonov; David J. Maloney; Theodore R. Holman

We report the discovery of novel small molecule inhibitors of platelet-type 12-human lipoxygenase, which display nanomolar activity against the purified enzyme, using a quantitative high-throughput screen (qHTS) on a library of 153607 compounds. These compounds also exhibit excellent specificity, >50-fold selectivity vs the paralogues, 5-human lipoxygenase, reticulocyte 15-human lipoxygenase type-1, and epithelial 15-human lipoxygenase type-2, and >100-fold selectivity vs ovine cyclooxygenase-1 and human cyclooxygenase-2. Kinetic experiments indicate this chemotype is a noncompetitive inhibitor that does not reduce the active site iron. Moreover, chiral HPLC separation of two of the racemic lead molecules revealed a strong preference for the (-)-enantiomers (IC(50) of 0.43 ± 0.04 and 0.38 ± 0.05 μM) compared to the (+)-enantiomers (IC(50) of >25 μM for both), indicating a fine degree of selectivity in the active site due to chiral geometry. In addition, these compounds demonstrate efficacy in cellular models, which underscores their relevance to disease modification.


Pancreas | 2007

Intramuscular injection of islet neogenesis-associated protein peptide stimulates pancreatic islet neogenesis in healthy dogs.

Gary L. Pittenger; David A. Taylor-Fishwick; Robert H. Johns; Niculina Burcus; Srivenkat Kosuri; Aaron I. Vinik

Objectives: Diabetes is a serious health problem. It has been proposed that islet neogenesis from pancreatic progenitor cells may restore insulin secretion in diabetic mammals. Islet neogenesis- associated protein (INGAP) stimulates islet neogenesis; therefore, we hypothesized that it would stimulate islet neogenesis in dogs. Methods: Forty nondiabetic beagle dogs were randomly divided into 4 groups. Group 1 received daily intramuscular injections of vehicle, whereas the other 3 groups received daily INGAP104-118 injections of 0.5, 1.5, or 10 mg/kg. After 30 days, pancreatic tissues were collected, and RNA and histological sections were analyzed. Results: In dogs treated with 1.5 mg/kg INGAP104-118, there was a significant (P < 0.001) increase in the percentage of insulin-positive cells (P < 0.001) and insulin gene expression. There was a trend to increased insulin-positive cells and gene expression with treatments of 0.5 and 10 mg/kg peptide. Protein gene product 9.5-positive cells were increased with treatment. Conclusions: These results indicate that INGAP stimulates cells in the pancreatic duct epithelium of healthy dogs (putative islet progenitor cells) to develop along a neuroendocrine pathway and form new islets in response to INGAP peptide. The INGAP might be an effective therapy for diabetes.


Diabetes, Obesity and Metabolism | 2013

Interaction between cytokines and inflammatory cells in islet dysfunction, insulin resistance and vascular disease

Yumi Imai; Anca D. Dobrian; J. R. Weaver; Matthew Butcher; Banumathi K. Cole; Elena Galkina; Margaret Morris; David A. Taylor-Fishwick; J. L. Nadler

Inflammation is an established pathogenic player in insulin resistance, islet demise and atherosclerosis. The complex interactions between cytokines, immune cells and affected tissues result in sustained inflammation in diabetes and atherosclerosis. 12‐ and 15‐lipoxygenase (LO), such as 12/15‐LO, produces a variety of metabolites through peroxidation of fatty acids and potentially contributes to the complex molecular crosstalk at the site of inflammation. 12‐ and 15‐LO pathways are frequently activated in tissues affected by diabetes and atherosclerosis including adipose tissue (AT), islets and the vasculature. Moreover, mice with whole body and tissue‐specific knockout of 12/15‐LO are protected against insulin resistance, hyperglycaemia and atherosclerosis supporting functional contribution of 12‐ and 15‐LO pathways in diabetes and atherosclerosis. Recently, it has emerged that there is a temporal regulation of the particular isoforms of 12‐ and 15‐LO in human AT and islets during the development of type 1 and type 2 diabetes and obesity. Analyses of tissues affected by diabetes and atherosclerosis also implied the roles of interleukin (IL)‐12 and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase‐1 (NOX‐1) in islets and IL‐17A in atherosclerosis. Future studies should aim to test the efficacy of inhibitions of these mediators for treatment of diabetes and atherosclerosis.


Biochimica et Biophysica Acta | 2003

Cloning genomic INGAP: a Reg-related family member with distinct transcriptional regulation sites.

David A. Taylor-Fishwick; Sharon Rittman; Hidayah Kendall; Lipika Roy; Wenjing Shi; Yong Cao; Gary L. Pittenger; Aaron I. Vinik

The protein product of hamster islet neogenesis-associated protein (INGAP) cDNA induces new pancreatic islet development. Manipulation of this process provides a new therapeutic strategy for the treatment of diabetes. As regulators of INGAP gene expression are unknown over 6 kb of hamster genomic INGAP has been cloned. Sequence analysis has identified a 3 kb 5-prime region with core promoter elements that is rich in transcription factor binding sites and six exons for the coding region. Analysis of promoter activity reveals stimulus-responsive DNA elements which have been identified though deletion analysis. Comparison of transcription factor binding sites in INGAP to the related gene RegIIIdelta exposes potential sites for differential gene regulation.


Pancreas | 2008

The Reg family member INGAP is a marker of endocrine patterning in the embryonic pancreas.

Natasha S. Hamblet; Wenjing Shi; Aaron I. Vinik; David A. Taylor-Fishwick

Objective: Adult islet neogenesis is believed to recapitulate elements of pancreatic endocrine development. Identifying factors that regulate islet neogenesis-associated protein (INGAP) gene activity could provide links to pancreas development. Methods: Predicted transcriptional regulators of INGAP were screened in an INGAP-promoter-reporter assay. Based upon their temporal expression, the occurrence of INGAP-positive cells during pancreas embryonic development were studied. Results: Pancreatic transcription factors, PDX-1, Ngn3, NeuroD, and Isl-1, activated the INGAP promoter, but PAX4, PAX6, and Nkx2.2 did not. The INGAP-positive cells were present in the developing pancreatic bud of the mouse embryo. Emerging clusters of unorganized endocrine cells were INGAP positive. These cells coexpressed insulin or somatostatin, but glucagon-expressing cells remained distinct. The INGAP-positive cells were also detected in the maturing neonatal endocrine cells organized into islets. In direct contrast to the embryo, glucagon localized with most INGAP-positive cells in the postnatal endocrine cells. The INGAP-positive cells juxtaposed pancreatic duct cells. A subset of 5-bromo-2&vprime;-deoxyuridine-positive/INGAP-positive cells was detected in the neonatal pancreas. Conclusions: These data implicate INGAP and/or Reg family proteins in endocrine cell patterning during embryonic development and suggest that INGAP immunoreactivity is a key marker associated with early endocrine cells.


Journal of Medicinal Chemistry | 2014

Synthesis and Structure–Activity Relationship Studies of 4-((2-Hydroxy-3-methoxybenzyl)amino)benzenesulfonamide Derivatives as Potent and Selective Inhibitors of 12-Lipoxygenase

Diane K. Luci; J. Brian Jameson; Adam Yasgar; Giovanni Diaz; Netra Joshi; Auric Kantz; Kate Markham; Steve Perry; Norine Kuhn; Jennifer Yeung; Edward H. Kerns; Lena Schultz; Michael Holinstat; Jerry L. Nadler; David A. Taylor-Fishwick; Ajit Jadhav; Anton Simeonov; Theodore R. Holman; David J. Maloney

Human lipoxygenases (LOXs) are a family of iron-containing enzymes which catalyze the oxidation of polyunsaturated fatty acids to provide the corresponding bioactive hydroxyeicosatetraenoic acid (HETE) metabolites. These eicosanoid signaling molecules are involved in a number of physiologic responses such as platelet aggregation, inflammation, and cell proliferation. Our group has taken a particular interest in platelet-type 12-(S)-LOX (12-LOX) because of its demonstrated role in skin diseases, diabetes, platelet hemostasis, thrombosis, and cancer. Herein, we report the identification and medicinal chemistry optimization of a 4-((2-hydroxy-3-methoxybenzyl)amino)benzenesulfonamide-based scaffold. Top compounds, exemplified by 35 and 36, display nM potency against 12-LOX, excellent selectivity over related lipoxygenases and cyclooxygenases, and possess favorable ADME properties. In addition, both compounds inhibit PAR-4 induced aggregation and calcium mobilization in human platelets and reduce 12-HETE in β-cells.

Collaboration


Dive into the David A. Taylor-Fishwick's collaboration.

Top Co-Authors

Avatar

Aaron I. Vinik

Eastern Virginia Medical School

View shared research outputs
Top Co-Authors

Avatar

Jerry L. Nadler

Eastern Virginia Medical School

View shared research outputs
Top Co-Authors

Avatar

Gary L. Pittenger

Eastern Virginia Medical School

View shared research outputs
Top Co-Authors

Avatar

Jessica R. Weaver

Eastern Virginia Medical School

View shared research outputs
Top Co-Authors

Avatar

Ajit Jadhav

University of California

View shared research outputs
Top Co-Authors

Avatar

Anton Simeonov

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

David J. Maloney

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yumi Imai

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Anca D. Dobrian

Eastern Virginia Medical School

View shared research outputs
Researchain Logo
Decentralizing Knowledge