Network


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

Hotspot


Dive into the research topics where Jiney Jose is active.

Publication


Featured researches published by Jiney Jose.


Angewandte Chemie | 2014

Differential sensing of MAP kinases using SOX-peptides.

Diana Zamora-Olivares; Tamer S. Kaoud; Jiney Jose; Andrew D. Ellington; Kevin N. Dalby; Eric V. Anslyn

Five SOX peptides are used to classify the MAPK groups and isoforms thereof using chemometrics. The score plots show excellent classification and accuracy, while support vector machine analysis leads to the quantification of ERK and an ERK inhibitor concentration in kinase mixtures. Examination of the loading plots reveals cross-reactivity among the peptides, and some unexpected surprises.


Anesthesia & Analgesia | 2015

Development of Rapidly Metabolized and Ultra-Short-Acting Ketamine Analogs.

Martyn Harvey; Jamie Sleigh; Logan J. Voss; Jiney Jose; Swarna A. Gamage; Frederik B. Pruijn; Sarath Liyanage; William A. Denny

BACKGROUND: Ketamine is a well-established, rapidly acting dissociative anesthetic. Clinical use is limited by prolonged psychotomimetic phenomena on emergence, often requiring the coadministration of additional hypnotic drugs. We hypothesized that the development of ketamine ester analogs with ultrashort offset times might markedly reduce the dysphoric emergence phenomena and, hence, increase the utility of a ketamine-like hypnotic and analgesic. Here, we describe the results of studies that seek to define the pharmacology of 5 esters of ((1-(2-chlorophenyl)-2-oxocyclohexyl)amino)pentanoate hydrochloride, the first ketamine analogs designed to be susceptible to ultrarapid metabolism. METHODS: Five norketamine ester analogs (R1–R5) were compared by ability to produce loss of righting and nociceptive blunting in rats. Toxicity testing was performed for 2 analogs (R1, R5) with 50% lethal dose (LD50) estimation in rats. In vitro metabolic stability was tested in rabbit plasma and whole blood by high-performance liquid chromatography. Behavioral and hemodynamic effects were observed in rabbits. We estimated the pharmacokinetics of these analogs in rabbits. RESULTS: All 5 norketamine esters produced rapid loss of righting reflex and diminished pedal withdrawal with ultrarapid offset in the models studied (return of righting reflex 87 seconds [interquartile range (IQR) 78–131] R1 vs 996 seconds [IQR 840–1304] ketamine in rats; P < 0.01). The LD50 was comparable to that of ketamine (LD50 R1 50.2 mg/kg [95% confidence interval, 30–63]). For all analogs, hydrolysis to sole carboxylic acid derivatives was most rapid in vivo (clearance 1.61 L/kg/min R1 [IQR 0.40–2.42]), followed by whole blood and then plasma. Analog R5 demonstrated relatively greater nociceptive blunting than hypnotic effect (P < 0.001; pedal withdrawal score comparison with R1). CONCLUSIONS: The 5 norketamine ester analogs retain the hypnotic characteristics of the parent compound, yet display rapid offset due to ultrarapid metabolism.


ACS Chemical Biology | 2012

Manipulating JNK Signaling with (−)-Zuonin A

Tamer S. Kaoud; Heekwang Park; Shreya Mitra; Chunli Yan; Chun Chia Tseng; Yue Shi; Jiney Jose; Juliana M. Taliaferro; Kiyoun Lee; Pengyu Ren; Jiyong Hong; Kevin N. Dalby

Recently, in a virtual screening strategy to identify new compounds targeting the D-recruitment site (DRS) of the c-Jun N-terminal kinases (JNKs), we identified the natural product (-)-zuonin A. Here we report the asymmetric synthesis of (-)-zuonin A and its enantiomer (+)-zuonin A. A kinetic analysis for the inhibition of c-Jun phosphorylation by (-)-zuonin A revealed a mechanism of partial competitive inhibition. Its binding is proposed to weaken the interaction of c-Jun to JNK by approximately 5-fold, without affecting the efficiency of phosphorylation within the complex. (-)-Zuonin A inhibits the ability of both MKK4 and MKK7 to phosphorylate and activate JNK. The binding site of (-)-zuonin A is predicted by docking and molecular dynamics simulation to be located in the DRS of JNK. (+)-Zuonin A also binds JNK but barely impedes the binding of c-Jun. (-)-Zuonin A inhibits the activation of JNK, as well as the phosphorylation of c-Jun in anisomycin-treated HEK293 cells, with the inhibition of JNK activation being more pronounced. (-)-Zuonin A also inhibits events associated with constitutive JNK2 activity, including c-Jun phosphorylation, basal Akt activation, and MDA-MB-231 cell migration. Mutations in the predicted binding site for (-)-zuonin A can render it significantly more or less sensitive to inhibition than wild type JNK2, allowing for the design of potential chemical genetic experiments. These studies suggest that the biological activity reported for other lignans, such as saucerneol F and zuonin B, may be the result of their ability to impede protein-protein interactions within MAPK cascades.


Bioorganic & Medicinal Chemistry | 2013

Structure-activity relationships for ketamine esters as short-acting anaesthetics.

Jiney Jose; Swarna A. Gamage; Martyn Harvey; Logan J. Voss; Jamie Sleigh; William A. Denny

A series of aliphatic esters of the non-opioid anaesthetic/analgesic ketamine were prepared and their properties as shorter-acting analogues of ketamine itself were explored in an infused rat model, measuring the time after infusion to recover from both the anaesthetic (righting reflex) and analgesic (response to stimulus) effects. The potency of the esters as sedatives was not significantly related to chain length, but Me, Et and i-Pr esters were the more dose potent (up to twofold less than ketamine), whereas n-Pr esters were less potent (from 2- to 6-fold less than ketamine). For the Me, Et and i-Pr esters recovery from anaesthesia was 10-15-fold faster than from ketamine itself, and for the n-Pr esters it was 20-25-fold faster than from ketamine. A new dimethylamino ketamine derivative (homoketamine) had ketamine-like sedative effects but was slightly less potent than, but ester analogues of homoketamine had very weak sedative effects.


ChemBioChem | 2014

Reversible Covalent Inhibition of eEF-2K by Carbonitriles

Ashwini K. Devkota; Ramakrishna Edupuganti; Chunli Yan; Yue Shi; Jiney Jose; Qiantao Wang; Tamer S. Kaoud; Eun Jeong Cho; Pengyu Ren; Kevin N. Dalby

eEF‐2K is a potential target for treating cancer. However, potent specific inhibitors for this enzyme are lacking. Previously, we identified 2,6‐diamino‐4‐(2‐fluorophenyl)‐4H‐thiopyran‐3,5‐dicarbonitrile (DFTD) as an inhibitor of eEF‐2K. Here we describe its mechanism of action against eEF‐2K, on the basis of kinetic, mutational, and docking studies, and use chemoinformatic approaches to identify a similar class of carbonitrile‐containing compounds that exhibit the same mechanism of action. We show that DFTD behaves as a reversible covalent inhibitor of eEF‐2K with a two‐step mechanism of inhibition: a fast initial binding step, followed by a slower reversible inactivation step. Molecular docking suggests that a nitrile group of DFTD binds within 4.5 Å of the active site Cys146 to form a reversible thioimidate adduct. Because Cys146 is not conserved amongst other related kinases, targeting this residue holds promise for the development of selective covalent inhibitors of eEF‐2K.


Pharmacology | 2015

Determination of the Hypnotic Potency in Rats of the Novel Ketamine Ester Analogue SN 35210

Martyn Harvey; James Wallace Sleigh; Logan J. Voss; Frederik B. Pruijn; Jiney Jose; Swarma Gamage; William A. Denny

Background: Ketamine is a rapidly acting dissociative anaesthetic drug with additional sympathomimetic, analgesic, and antidepressant properties. Despite these advantages, clinical use is curtailed by prolonged psychomimetic effects apparent over the entire dose spectrum. In this study, we report on the hypnotic potency of SN 35210, the first ketamine ester-analogue designed for rapid offset via esterase-mediated hydrolysis. Methods: Thirty-three adult Sprague Dawley rats received intravenous racemic ketamine (n = 14), racemic SN 35210 (n = 19), S-enantiomer SN 35210 (n = 17), or R-enantiomer SN 35210 (n = 15), in crossover design. The ability to induce loss of righting reflex (LORR) at a given dose, the duration of righting reflex loss, and the time to return of normal behaviours were recorded. The ED50 for LORR was determined for all agents. Results: The ED50 for righting reflex loss was racemic ketamine 9.6 (95% CI 8.5-10.9) mg/kg, racemic SN 35210 10.4 (95% CI 9.5-11.5) mg/kg, S-enantiomer SN 35210 10.6 (95% CI 9.1-11.8), and R-enantiomer SN 35210 10.3 (95% CI 9.1-11.4) mg/kg. The duration of righting reflex loss and time to return to normal behaviours were approximately 5 times greater for racemic ketamine than all 3 SN 35210 ester analogues. Conclusions: Racemic, and R and S-enantiomer SN 35210, produced LORR in rats at similar doses to the parent compound ketamine. The duration of righting reflex loss, and duration of behavioural aberration, was significantly reduced for all SN 35210 analogues.


Bioorganic & Medicinal Chemistry Letters | 2017

Benzenesulphonamide inhibitors of the cytolytic protein perforin

Julie A. Spicer; Christian Miller; Patrick D. O'Connor; Jiney Jose; Kristiina M. Huttunen; Jagdish K. Jaiswal; William A. Denny; Hedieh Akhlaghi; Kylie A. Browne; Joseph A. Trapani

Graphical abstract


European Journal of Medicinal Chemistry | 2017

Substituted arylsulphonamides as inhibitors of perforin-mediated lysis

Julie A. Spicer; Christian Miller; Patrick D. O'Connor; Jiney Jose; Kristiina M. Huttunen; Jagdish K. Jaiswal; William A. Denny; Hedieh Akhlaghi; Kylie A. Browne; Joseph A. Trapani

The structure-activity relationships for a series of arylsulphonamide-based inhibitors of the pore-forming protein perforin have been explored. Perforin is a key component of the human immune response, however inappropriate activity has also been implicated in certain auto-immune and therapy-induced conditions such as allograft rejection and graft versus host disease. Since perforin is expressed exclusively by cells of the immune system, inhibition of this protein would be a highly selective strategy for the immunosuppressive treatment of these disorders. Compounds from this series were demonstrated to be potent inhibitors of the lytic action of both isolated recombinant perforin and perforin secreted by natural killer cells in vitro. Several potent and soluble examples were assessed for in vivo pharmacokinetic properties and found to be suitable for progression to an in vivo model of transplant rejection.


Bioorganic & Medicinal Chemistry Letters | 2018

A mitochondria-selective near-infrared-emitting fluorescent dye for cellular imaging studies

Peter J. Choi; Katsuya Noguchi; Munetaka Ishiyama; William A. Denny; Jiney Jose

This communication details the synthesis, evaluation of photophysical properties, and cellular imaging studies of cyanine chromophore based fluorescent dye 1 as a selective imaging agent for mitochondria.


ACS Medicinal Chemistry Letters | 2017

Serotonin Analogues as Inhibitors of Breast Cancer Cell Growth

Jiney Jose; Clint D. J. Tavares; Nancy D. Ebelt; Alessia Lodi; Ramakrishna Edupuganti; Xuemei Xie; Ashwini K. Devkota; Tamer S. Kaoud; Carla L. Van Den Berg; Eric V. Anslyn; Stefano Tiziani; Chandra Bartholomeusz; Kevin N. Dalby

Serotonin (5-hydroxytryptamine, 5-HT) is a critical local regulator of epithelial homeostasis in the breast and exerts its actions through a number of receptors. Dysregulation of serotonin signaling is reported to contribute to breast cancer pathophysiology by enhancing cell proliferation and promoting resistance to apoptosis. Preliminary analyses indicated that the potent 5-HT1B/1D serotonin receptor agonist 5-nonyloxytryptamine (5-NT), a triptan-like molecule, induced cell death in breast cancer cell lines. Thus, we synthesized a series of novel alkyloxytryptamine analogues, several of which decreased the viability of various human cancer cell lines. Proteomic and metabolomic analyses showed that compounds 6 and 10 induced apoptosis and interfered with signaling pathways that regulate protein translation and survival, such as the Akt/mTOR pathway, in triple-negative breast cancer cells.

Collaboration


Dive into the Jiney Jose's collaboration.

Top Co-Authors

Avatar

Kevin N. Dalby

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Tamer S. Kaoud

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ashwini K. Devkota

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chunli Yan

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Eric V. Anslyn

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Pengyu Ren

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Jihyun Park

University of Texas at Austin

View shared research outputs
Researchain Logo
Decentralizing Knowledge