Network


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

Hotspot


Dive into the research topics where Abhay Kulkarni is active.

Publication


Featured researches published by Abhay Kulkarni.


PLOS ONE | 2014

Transient receptor potential ankyrin 1 receptor activation in vitro and in vivo by pro-tussive agents: GRC 17536 as a promising anti-tussive therapeutic.

Indranil Mukhopadhyay; Abhay Kulkarni; Sarika Aranake; Pallavi Karnik; Mahesh Shivarama Shetty; Sandeep Thorat; Indraneel Ghosh; Dinesh Pradeep Wale; Vikram Mansingh Bhosale; Neelima Khairatkar-Joshi

Cough is a protective reflex action that helps clear the respiratory tract which is continuously exposed to airborne environmental irritants. However, chronic cough presents itself as a disease in its own right and despite its global occurrence; the molecular mechanisms responsible for cough are not completely understood. Transient receptor potential ankyrin1 (TRPA1) is robustly expressed in the neuronal as well as non-neuronal cells of the respiratory tract and is a sensor of a wide range of environmental irritants. It is fast getting acceptance as a key biological sensor of a variety of pro-tussive agents often implicated in miscellaneous chronic cough conditions. In the present study, we demonstrate in vitro direct functional activation of TRPA1 receptor by citric acid which is routinely used to evoke cough in preclinical and clinical studies. We also show for the first time that a potent and selective TRPA1 antagonist GRC 17536 inhibits citric acid induced cellular Ca+2 influx in TRPA1 expressing cells and the citric acid induced cough response in guinea pigs. Hence our data provides a mechanistic link between TRPA1 receptor activation in vitro and cough response induced in vivo by citric acid. Furthermore, we also show evidence for TRPA1 activation in vitro by the TLR4, TLR7 and TLR8 ligands which are implicated in bacterial/respiratory virus pathogenesis often resulting in chronic cough. In conclusion, this study highlights the potential utility of TRPA1 antagonist such as GRC 17536 in the treatment of miscellaneous chronic cough conditions arising due to diverse causes but commonly driven via TRPA1.


Pharmaceuticals | 2016

Blocking TRPA1 in Respiratory Disorders: Does It Hold a Promise?

Indranil Mukhopadhyay; Abhay Kulkarni; Neelima Khairatkar-Joshi

Transient Receptor Potential Ankyrin 1 (TRPA1) ion channel is expressed abundantly on the C fibers that innervate almost entire respiratory tract starting from oral cavity and oropharynx, conducting airways in the trachea, bronchi, terminal bronchioles, respiratory bronchioles and upto alveolar ducts and alveoli. Functional presence of TRPA1 on non-neuronal cells got recognized recently. TRPA1 plays a well-recognized role of “chemosensor”, detecting presence of exogenous irritants and endogenous pro-inflammatory mediators that are implicated in airway inflammation and sensory symptoms like chronic cough, asthma, chronic obstructive pulmonary disease (COPD), allergic rhinitis and cystic fibrosis. TRPA1 can remain activated chronically due to elevated levels and continued presence of such endogenous ligands and pro-inflammatory mediators. Several selective TRPA1 antagonists have been tested in animal models of respiratory disease and their performance is very promising. Although there is no TRPA1 antagonist in advanced clinical trials or approved on market yet to treat respiratory diseases, however, limited but promising evidences available so far indicate likelihood that targeting TRPA1 may present a new therapy in treatment of respiratory diseases in near future. This review will focus on in vitro, animal and human evidences that strengthen the proposed role of TRPA1 in modulation of specific airway sensory responses and also on preclinical and clinical progress of selected TRPA1 antagonists.


Bioorganic & Medicinal Chemistry Letters | 2017

Discovery of furan and dihydrofuran-fused tricyclic benzo[d]imidazole derivatives as potent and orally efficacious microsomal prostaglandin E synthase-1 (mPGES-1) inhibitors: Part-1

Nagarajan Muthukaman; Macchindra Tambe; Sanjay Deshmukh; Dnyandeo Pisal; Shital Tondlekar; Mahamadhanif S. Shaikh; Neelam Sarode; Vidya Ganapati Kattige; Monali Pisat; Pooja Sawant; Srinivasa Honnegowda; Vikas Karande; Abhay Kulkarni; Dayanidhi Behera; Satyawan Jadhav; Ramchandra R. Sangana; Girish S. Gudi; Neelima Khairatkar-Joshi; Laxmikant Atmaram Gharat

This letter describes the synthesis and biological evaluation of furan and dihydrofuran-fused tricyclic benzo[d]imidazole derivatives as novel mPGES-1 inhibitors, capable of inhibiting an increased PGE2 production in the disease state. Structure-activity optimization afforded many potent mPGES-1 inhibitors having <50 nM potencies in the A549 cellular assay and adequate metabolic stability in liver microsomes. Lead compounds 8l and 8m demonstrated reasonable in vitro pharmacology and pharmacokinetic properties over other compounds. In particular, 8m revealed satisfactory oral pharmacokinetics and bioavailability in multiple species like rat, guinea pig, dog and cynomolgus monkey. In addition, the representative compound 8m showed in vivo efficacy by inhibiting LPS-induced thermal hyperalgesia with an ED50 of 14.3 mg/kg in guinea pig.


Bioorganic & Medicinal Chemistry Letters | 2017

Tricyclic 4,4-dimethyl-3,4-dihydrochromeno[3,4-d]imidazole derivatives as microsomal prostaglandin E2 synthase-1 (mPGES-1) inhibitors: SAR and in vivo efficacy in hyperalgesia pain model

Nagarajan Muthukaman; Macchindra Tambe; Mahamadhanif S. Shaikh; Dnyandeo Pisal; Sanjay Deshmukh; Shital Tondlekar; Neelam Sarode; Lakshminarayana Narayana; Jitendra Maganbhai Gajera; Vidya Ganapati Kattige; Srinivasa Honnegowda; Vikas Karande; Abhay Kulkarni; Dayanidhi Behera; Satyawan Jadhav; Girish S. Gudi; Neelima Khairatkar-Joshi; Laxmikant Atmaram Gharat

A series of substituted tricyclic 4,4-dimethyl-3,4-dihydrochromeno[3,4-d]imidazole derivatives have been synthesized and their mPGES-1 biological activity has been disclosed in detail. Structure-activity relationship (SAR) optimization provided inhibitors with excellent mPGES-1 potency and low to moderate PGE2 release A549 cell potency. Among the mPGES-1 inhibitors studied, 7, 9 and 11l provided excellent selectivity over COX-2 (>200-fold) and >70-fold selectivity for COX-1 except 11l, which exhibited dual mPGES-1/COX-1 activity. Furthermore, the above tested mPGES-1 inhibitors demonstrated good metabolic stability in liver microsomes, high plasma protein binding (PPB) and no significant inhibition observed in clinically relevant CYP isoforms. Besides, selected mPGES-1 tool compounds 9 and 11l provided good in vivo pharmacokinetic profile and oral bioavailability (%F=33 and 85). Additionally, the representative mPGES-1 tool compounds 9 and 11l revealed moderate in vivo efficacy in the LPS-induced thermal hyperalgesia guinea pig pain model.


Bioorganic & Medicinal Chemistry Letters | 2018

Alleviating CYP and hERG liabilities by structure optimization of dihydrofuran-fused tricyclic benzo[d]imidazole series – Potent, selective and orally efficacious microsomal prostaglandin E synthase-1 (mPGES-1) inhibitors: Part-2

Nagarajan Muthukaman; Sanjay Deshmukh; Macchindra Tambe; Dnyandeo Pisal; Shital Tondlekar; Mahamadhanif S. Shaikh; Neelam Sarode; Vidya Ganapati Kattige; Pooja Sawant; Monali Pisat; Vikas Karande; Srinivasa Honnegowda; Abhay Kulkarni; Dayanidhi Behera; Satyawan Jadhav; Ramchandra R. Sangana; Girish S. Gudi; Neelima Khairatkar-Joshi; Laxmikant Atmaram Gharat

In an effort to identify CYP and hERG clean mPGES-1 inhibitors from the dihydrofuran-fused tricyclic benzo[d]imidazole series lead 7, an extensive structure-activity relationship (SAR) studies were performed. Optimization of A, D and E-rings in 7 afforded many potent compounds with human whole blood potency in the range of 160-950 nM. Selected inhibitors 21d, 21j, 21m, 21n, 21p and 22b provided selectivity against COX-enzymes and mPGES-1 isoforms (mPGES-2 and cPGES) along with sufficient selectivity against prostanoid synthases. Most of the tested analogs demonstrated required metabolic stability in liver microsomes, low hERG and CYP liability. Oral pharmacokinetics and bioavailability of lead compounds 21j, 21m and 21p are discussed in multiple species like rat, guinea pig, dog, and cynomolgus monkey. Besides, these compounds revealed low to moderate activity against human pregnane X receptor (hPXR). The selected lead 21j further demonstrated in vivo efficacy in acute hyperalgesia (ED50: 39.6 mg/kg) and MIA-induced osteoarthritic pain models (ED50: 106 mg/kg).


Bioorganic & Medicinal Chemistry Letters | 2018

Discovery of 5-(2-chloro-4′-(1H-imidazol-1-yl)-[1,1′-biphenyl]-4-yl)-1H-tetrazole as potent and orally efficacious S-nitrosoglutathione reductase (GSNOR) inhibitors for the potential treatment of COPD

Nagarajan Muthukaman; Sanjay Deshmukh; Shital Tondlekar; Macchindra Tambe; Dnyandeo Pisal; Neelam Sarode; Siddharth Mhatre; Samitabh Chakraborti; Daisy Manish Shah; Vikram Mansingh Bhosale; Abhay Kulkarni; Mahamad Yunnus A. Mahat; Satyawan Jadhav; Girish S. Gudi; Neelima Khairatkar-Joshi; Laxmikant Atmaram Gharat

Endogenous nitrosothiols (SNOs) including S-nitrosoglutathione (GSNO) serve as reservoir for bioavailable nitric oxide (NO) and mediate NO-based signaling, inflammatory status and smooth muscle function in the lung. GSNOR inhibition increases pulmonary GSNO and induces bronchodilation while reducing inflammation in lung diseases. In this letter, design, synthesis and structure-activity relationships (SAR) of novel imidazole-biaryl-tetrazole based GSNOR inhibitors are described. Many potent inhibitors (30, 39, 41, 42, 44, 45 and 58) were identified with low nanomolar activity (IC50s: <15 nM) along with adequate metabolic stability. Lead compounds 30 and 58 exhibited good exposure and oral bioavailability in mouse pharmacokinetic (PK) study. Compound 30 was selected for further profiling and revealed comparable mouse and rat GSNOR potency, high selectivity against alcohol dehydrogenase (ADH) and carbonyl reductase (CBR1) family of enzymes, low efflux ratio and permeability in PAMPA, a high permeability in CALU-3 assay, significantly low hERG activity and minimal off-target activity. Further, an in vivo efficacy of compound 30 is disclosed in cigarette smoke (CS) induced mouse model for COPD.


European Respiratory Journal | 2015

LATE-BREAKING ABSTRACT: Novel inhaled ROR/c inhibitor for potential treatment of COPD

Srinivasa Honnegowda; Abraham Thomas; Sachin Sundarlal Chaudhari; Sanjib Das; Daisy Manish Shah; Malini Bajpai; Indrani Sarkar; Abhay Kulkarni; Girish S. Gudi; Satyawan Jadhav; Neelima Khairatkar-Joshi

Background: Antibody mediated IL-17 blockade is remarkably successful in autoimmune disease treatment. Recent studies implicate IL-17 in airway diseases. Increased IL-17 immuno-reactivity and Th17 cells in lung tissue and IL-17 level in bronchoalveolar lavage fluid (BALF) is reported in human COPD patients and tobacco-smoke-exposed rats. Anti-IL-17 Ab attenuates airway inflammation in these animals. In human COPD patients, lung IL-17 immuno-reactivity correlates with disease severity. Recent studies also implicate IL-17 in COPD exacerbations. Hence IL-17 blockade seems a very attractive approach in potentially addressing the anti-inflammatory treatment gap in COPD management. IL-17 production is modulated by RORγt / RORc receptor in mice / man and offers a novel mechanism for IL-17 blockade. Objective: We synthesized several novel, potent, selective RORc blockers and evaluated efficacy of the series lead called “Compound A” in animal model of COPD. Results: Compound A is highly selective and potent preclinical lead with IC 50 ofless than 40 nM in RORc binding and PBMC- IL-17 release assay. In vivo, it produced ∼ 75 % inhibition of IL-17 release induced by LPS and anti-CD3. Its efficacy in a COPD was evaluated in cigarette smoke mouse model under acute (7 days) and chronic (3 months) conditions. Compound A showed promising efficacy in suppressing lung cellular infiltration by 50-70% upon intranasal administration. In chronic model, the compound showed significant decrease of lung emphysema, epithelial IL-17 immuno-reactivity and RORgt/IL-17 positive lung cells. Conclusion: Our RORc antagonist is a novel, inhalable IL-17 blocker for potential treatment of COPD.


European Journal of Medicinal Chemistry | 2017

A2B adenosine receptor antagonists: Design, synthesis and biological evaluation of novel xanthine derivatives

Sujay Basu; Dinesh Barawkar; Vidya Ramdas; Yogesh Waman; Meena V. Patel; Anil Panmand; Sachin Thorat; Rajesh Bonagiri; Dilip Jadhav; Partha Mukhopadhyay; Vandna Prasad; B. Srinivasa Reddy; Arnab Goswami; Sandhya Chaturvedi; Suraj Menon; Azfar Quraishi; Indraneel Ghosh; Sushant Dusange; Shalini Paliwal; Abhay Kulkarni; Vikas Karande; Rhishikesh Thakre; Gaurav Bedse; Sreekanth R. Rouduri; Jayasagar Gundu; Venkata Palle; Anita Chugh; Kasim A. Mookhtiar


European Journal of Medicinal Chemistry | 2017

Design and synthesis of novel xanthine derivatives as potent and selective A2B adenosine receptor antagonists for the treatment of chronic inflammatory airway diseases

Sujay Basu; Dinesh Barawkar; Vidya Ramdas; Meena V. Patel; Yogesh Waman; Anil Panmand; Sachin Thorat; Minakshi Naykodi; Arnab Goswami; B. Srinivasa Reddy; Vandna Prasad; Sandhya Chaturvedi; Azfar Quraishi; Suraj Menon; Shalini Paliwal; Abhay Kulkarni; Vikas Karande; Indraneel Ghosh; Syed Mustafa; Siddhartha De; Vaibhav Jain; Ena Ray Banerjee; Sreekanth R. Rouduri; Venkata Palle; Anita Chugh; Kasim A. Mookhtiar


Bioorganic & Medicinal Chemistry Letters | 2016

Discovery of 2-((2-chloro-6-fluorophenyl)amino)-N-(3-fluoro-5-(trifluoromethyl)phenyl)-1-methyl-7,8-dihydro-1H-[1,4]dioxino[2',3':3,4]benzo[1,2-d]imidazole-5-carboxamide as potent, selective and efficacious microsomal prostaglandin E2 synthase-1 (mPGES-1) inhibitor.

Nagarajan Muthukaman; Sanjay Deshmukh; Neelam Sarode; Shital Tondlekar; Macchindra Tambe; Dnyandeo Pisal; Mahamadhanif S. Shaikh; Vidya Ganapati Kattige; Srinivasa Honnegowda; Vikas Karande; Abhay Kulkarni; Satyawan Jadhav; Mahamad Yunnus A. Mahat; Girish S. Gudi; Neelima Khairatkar-Joshi; Laxmikant Atmaram Gharat

Collaboration


Dive into the Abhay Kulkarni's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Arnab Goswami

Post Graduate Institute of Medical Education and Research

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge