Network


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

Hotspot


Dive into the research topics where Prakash Jagtap is active.

Publication


Featured researches published by Prakash Jagtap.


Nature Reviews Drug Discovery | 2005

Poly(ADP-ribose) polymerase and the therapeutic effects of its inhibitors

Prakash Jagtap; Csaba Szabó

Poly(ADP-ribose) polymerases (PARPs) are involved in the regulation of many cellular functions. Three consequences of the activation of PARP1, which is the main isoform of the PARP family, are particularly important for drug development: first, its role in DNA repair; second, its capacity to deplete cellular energetic pools, which culminates in cell dysfunction and necrosis; and third, its capacity to promote the transcription of pro-inflammatory genes. Consequently, pharmacological inhibitors of PARP have the potential to enhance the cytotoxicity of certain DNA-damaging anticancer drugs, reduce parenchymal cell necrosis (for example, in stroke or myocardial infarction) and downregulate multiple simultaneous pathways of inflammation and tissue injury (for example, in circulatory shock, colitis or diabetic complications). The first ultrapotent novel PARP inhibitors have now entered human clinical trials. This article presents an overview of the principal pathophysiological pathways and mechanisms that are governed by PARP, followed by the main structures and therapeutic actions of various classes of novel PARP inhibitors.


Nature Medicine | 2001

Diabetic endothelial dysfunction: the role of poly(ADP-ribose) polymerase activation

Francisco Garcia Soriano; László Virág; Prakash Jagtap; Éva Szabó; Jon G. Mabley; Lucas Liaudet; Anita Marton; Dale G. Hoyt; Kanneganti Murthy; Andrew L. Salzman; Garry J. Southan; Csaba Szabó

Diabetic patients frequently suffer from retinopathy, nephropathy, neuropathy and accelerated atherosclerosis. The loss of endothelial function precedes these vascular alterations. Here we report that activation of poly(ADP-ribose) polymerase (PARP) is an important factor in the pathogenesis of endothelial dysfunction in diabetes. Destruction of islet cells with streptozotocin in mice induced hyperglycemia, intravascular oxidant production, DNA strand breakage, PARP activation and a selective loss of endothelium-dependent vasodilation. Treatment with a novel potent PARP inhibitor, starting after the time of islet destruction, maintained normal vascular responsiveness, despite the persistence of severe hyperglycemia. Endothelial cells incubated in high glucose exhibited production of reactive nitrogen and oxygen species, consequent single-strand DNA breakage, PARP activation and associated metabolic and functional impairment. Basal and high-glucose-induced nuclear factor-κB activation were suppressed in the PARP-deficient cells. Our results indicate that PARP may be a novel drug target for the therapy of diabetic endothelial dysfunction.


Critical Care Medicine | 2002

Novel phenanthridinone inhibitors of poly(adenosine 5'-diphosphate-ribose) synthetase: Potent cytoprotective and antishock agents

Prakash Jagtap; Francisco Garcia Soriano; László Virág; Lucas Liaudet; Jon G. Mabley; Éva Szabó; György Haskó; Anita Marton; Clara Batista Lorigados; Ferenc Gallyas; Balazs Sumegi; Dale G. Hoyt; Erkan Baloglu; John VanDuzer; Andrew L. Salzman; Garry J. Southan; Csaba Szabó

ObjectiveTo synthesize novel inhibitors of the nuclear enzyme poly(adenosine 5′-diphosphate [ADP]-ribose) synthetase (PARS), also known as poly(ADP-ribose) polymerase (PARP), and to test them in in vitro models of oxidant-induced cytotoxicity and in endotoxin and splanchnic occlusion-reperfusion-induced shock. DesignRandomized, prospective laboratory study. SettingResearch laboratory. SubjectsMurine macrophages, thymocytes, and endothelial cells; Balb/c mice and Wistar rats. InterventionsMacrophages and endothelial cells were treated with peroxynitrite and bleomycin to induce PARS activation, and thymocytes were treated with peroxynitrite to induce cell necrosis. Novel PARS inhibitors were synthesized and used to reduce PARS activation and to reverse cytotoxicity. Balb/c mice were subjected to splanchnic occlusion and reperfusion and were pretreated with various doses (1–10 mg/kg intraperitoneally) of PJ34, a selected, potent, water-soluble PARS inhibitor. The passage of fluorescein isothiocyanate-conjugated dextran (4 kDa) was analyzed in everted gut ileal sacs incubated ex vivo as an index of gut permeability. Wistar rats were subjected to Escherichia coli bacterial lipopolysaccharide (40 mg/kg intraperitoneally). PJ34 was also used at 10 mg/kg intraperitoneally, 1 hr before lipopolysaccharide or at 25 mg/kg intraperitoneally 1 hr after lipopolysaccharide treatment. Serum concentrations of indicators or multiple organ injury, concentrations of various proinflammatory mediators, and tissue concentrations of myeloperoxidase and malondialdehyde were measured. In addition, survival rates and vascular contractile and relaxant responses were recorded. Measurements and Main ResultsAppropriate modifications of the phenanthridinone core structure yielded significant increases in the potency of the compounds, both as PARS inhibitors and as cytoprotective agents. The compound N-(6-oxo-5,6-dihydro-phenanthridin-2-yl) -N,N-dimethylacetamide (designated as PJ34) was one of the potent PARS inhibitors of the series, and it dose-dependently protected against thymocyte necrosis, with a half-maximal restoration of cell viability of 35 nM and complete protection at 200 nM. PARS activation also was visualized by immunohistochemistry and was dose-dependently suppressed by PJ34. The effect of PJ34 was dose-dependently reversed by excess nicotinamide adenine dinucleotide (oxidized). The PARS inhibitors dose-dependently suppressed proinflammatory cytokine and chemokine production and restored viability in immunostimulated macrophages. PJ34 was selected for the subsequent in vivo studies. PJ34 significantly protected against splanchnic reperfusion-induced intestinal hyperpermeability in the mouse. PJ34 reduced peak plasma concentrations of tumor necrosis factor-&agr;, interleukin-1&bgr;, and nitrite/nitrate in the plasma of lipopolysaccharide-treated rats. PJ34 ameliorated the lipopolysaccharide-induced increases in indexes of liver and kidney failure and concentrations of myeloperoxidase and malondialdehyde in the lung and gut. Lipopolysaccharide elicited vascular dysfunction, which was normalized by PJ34. Lipopolysaccharide-induced mortality was reduced by PJ34 (both pre- and posttreatment). ConclusionsThe novel series of phenanthridinone PARS inhibitors have potent cytoprotective effects in vitro and significant protective effects in shock and reperfusion injury in rodent models in vivo.


Inflammation Research | 2001

Anti-inflammatory effects of a novel, potent inhibitor of poly (ADP-ribose) polymerase

Jon G. Mabley; Prakash Jagtap; Mauro Perretti; Stephen J. Getting; Andrew L. Salzman; László Virág; Éva Szabó; Francisco Garcia Soriano; Lucas Liaudet; Ge Abdelkarim; György Haskó; Anita Marton; G. J. Southan; Csaba Szabó

Abstract.Objective and design: Oxygen- and nitrogen-derived free radicals and oxidants play an important role in the pathogenesis of various forms of inflammation. Recent work emphasizes the importance of oxidant-induced DNA strand breakage and activation of the nuclear enzyme poly(ADP-ribose) polymerase (PARP) in the pathogenesis of various inflammatory diseases. We have recently demonstrated the efficacy of PJ34, a novel, potent phenanthridinone derivative PARP inhibitor, in rodent models of diabetic vascular dysfunction and stroke. Here we tested the efficacy of PARP inhibition in various models of local inflammation in rodents.¶Materials and methods: PJ34 (at doses of 0.03-30 mg/kg) was tested in rats and mice subjected to standard models of inflammation, with relevant parameters of inflammation measured using standard methods.¶Results: PJ34 treatment (s.c, i.p. and i.v.) dose-dependently suppressed neutrophil infiltration and nitric oxide (but not KC and IL-1β) production in peritonitis. In a model of systemic endotoxemia, PJ34 pretreatment significantly reduced plasma levels of TNF-α, IL-1β and nitrite/nitrate (breakdown products of nitric oxide) production. PJ34 treatment (oral gavage) induced a significant suppression of the inflammatory response in dextran sulfate colitis, multiple low dose streptozotocin diabetes and cyclophosphamide-accelerated autoimmune diabetes in the non-obese diabetic mice, and reduced the degree of mononuclear cell infiltration into the iris in an endotoxin-induced uveitis model. Delaying the start of PJ34 administration in the colitis model conferred significant protective effects, while in the arthritis model the post-treatment paradigm lacked protective effects.¶Conclusions: PJ34 provides significant, dose-dependent, anti-inflammatory effects in a variety of local inflammation models. Some of its actions are maintained in the post-treatment regimen and/or after discontinuation of treatment. We conclude that PARP inhibition offers a powerful means for reducing the severity of various forms of local inflammatory responses.


The Annals of Thoracic Surgery | 2002

Myocardial protection by PJ34, a novel potent poly (ADP-ribose) synthetase inhibitor.

Renato Faro; Yoshiya Toyoda; James D. McCully; Prakash Jagtap; Éva Szabó; László Virág; Cesario Bianchi; Sidney Levitsky; Csaba Szabó; Frank W. Sellke

BACKGROUND The activation of poly (ADP-ribose) synthetase plays an important role in the pathogenesis leading to myocardial ischemia-reperfusion injury. The aim of this study was to determine if a novel potent inhibitor of poly (ADP-ribose) synthetase, PJ34, provides myocardial protection. METHODS Pigs were subjected to 60 minutes of regional ischemia followed by 180 minutes of reperfusion. Ten mg/kg of PJ34 (PJ34; n = 6) was administrated intravenously (treated group) from 15 to 5 minutes before reperfusion followed by 3 mg/kg/hour of PJ34 from 5 minutes before reperfusion to the end of 180 minutes reperfusion. Control pigs (n = 7) received vehicle only. Arterial and left ventricular pressure and coronary flow were monitored. RESULTS The PJ34 showed significant reduction on infarct size (37.5%+/-4.5% and 50.5%+/-4.8% of the area at risk) for PJ34 and control pigs groups, respectively, (p < 0.05). Significant reduction in postsystolic shortening, as well as improvement on segment shortening, and positive first derivative of pressure over time (+dP/dt) maximum were also observed in PJ34 versus control pigs (p < 0.05). CONCLUSIONS Our results suggest that PJ34 provides cardioprotection by decreasing myocardial infarct size and enhancing postischemic regional and global functional recovery.


Progress in the chemistry of organic natural products. Progrès dans la chimie des substances organiques naturelles | 2002

The Chemistry of Taxol and Related Taxoids

David G. I. Kingston; Prakash Jagtap; Haiqing Yuan; L. Samala

The anticancer compound taxol (1.1.1) was originally isolated in the late 1960’s from the bark of the Western Yew, Taxus brevifolia, and its structure was published in 1971 in a paper that has been cited well over 1000 times since its publication (1). Taxol was originally isolated on the basis of its cytotoxicity and antileukemic activity, but it was not actively developed as a drug for several years because of its scarcity (the initial yield from yew bark was 0.02%) and because of its lack of water-solubility. Fortunately additional testing at the National Cancer Institute revealed its activity against various human solid tumor xenografts in nude mice, and a program to develop taxol as a drug was initiated in 1977. A key discovery from the Horwitz laboratory in 1979, in a paper that has also been cited over 1000 times since its publication, showed that taxol had a unique mechanism of action, binding to polymerized tubulin and stabilizing microtubules to dissociation (2). These twin discoveries of taxol’s activity and its mechanism of action provided the impetus for continued clinical trials in the face of some initial disappointments, and the first positive clinical results against ovarian cancer were announced in 1989 (3). Following these results taxol was also found to be effective against breast cancer (4) and was licensed to Bristol-Myers Squibb; its sales are estimated at


Bioorganic & Medicinal Chemistry | 2001

Synthesis and bioactivity of 2,4-diacyl analogues of paclitaxel.

Mahendra D. Chordia; Haiqing Yuan; Prakash Jagtap; John F. Kadow; Byron H. Long; Craig R. Fairchild; Kathy A. Johnston; David G. I. Kingston

1.6 billion in 2000 (5).


Tetrahedron Letters | 1999

A facile N-debenzoylation of paclitaxel: Conversion of paclitaxel to docetaxel

Prakash Jagtap; David G. I. Kingston

The 2,4-diacyl paclitaxel analogues 8a-8r were prepared from paclitaxel by acylation of 4-deacetyl-2-debenzoylpaclitaxel 1,2-carbonate (3) followed either by hydrolysis of the carbonate and acylation or by direct treatment of the carbonate with an aryllithium. Some of the resulting derivatives showed significantly improved tubulin assembly activity and cytotoxicity as compared with paclitaxel; in some cases this improvement was especially significant for paclitaxel-resistant cell lines.


Tetrahedron Letters | 1992

Convenient Synthesis of Dimethyl-1-Aryl-4-Hydroxy-N-Methylcarbazole-2,3-Dicarboxylates via Michael Initiated Ring Closure Methodology

Raghao S. Mali; Prakash Jagtap

Abstract An efficient and regioselective method for the N-debenzoylation of paclitaxel has been developed, and has been applied to the conversion of paclitaxel to 10-acetyldocetaxel and to docetaxel.


Archive | 2001

Local and systemic inflammation: role of poly (ADP-ribose) synthetase activation by reactive nitrogen species

Jon G. Mabley; Lucas Liaudet; Francisco Garcia Soriano; László Virág; Prakash Jagtap; Anita Marton; Clara Batista Lorigados; Ferenc Gallyas; Éva Szabó; Galaleldin E. Abdelkarim; György Haskó; Garry J. Southan; Andrew L. Salzman; Csaba Szabó

Abstract A convenient method has been described for the synthesis of dimethyl-1-aryl-4-hydroxy-N-methylcarbazole-2,3-dicarboxylates ( 3a–d ) from ethyl N-methyl-2-benzylindole-3-carboxylates ( 2a–d ).

Collaboration


Dive into the Prakash Jagtap's collaboration.

Top Co-Authors

Avatar

Andrew L. Salzman

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Csaba Szabo

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Csaba Szabó

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alexei Degterev

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Junying Yuan

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Gregory D. Cuny

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

William Williams

Thomas Jefferson University

View shared research outputs
Researchain Logo
Decentralizing Knowledge