Network


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

Hotspot


Dive into the research topics where Bibhash C. Paria is active.

Publication


Featured researches published by Bibhash C. Paria.


Endocrinology | 2014

Prostaglandins are essential for cervical ripening in LPS-Mediated preterm birth but not term or antiprogestin-driven preterm ripening

Brenda Timmons; Jeff Reese; Simona Socrate; Noah J. Ehinger; Bibhash C. Paria; Ginger L. Milne; Meredith L. Akins; Richard J. Auchus; Donald D. McIntire; Michael House; Mala Mahendroo

Globally, an estimated 13 million preterm babies are born each year. These babies are at increased risk of infant mortality and life-long health complications. Interventions to prevent preterm birth (PTB) require an understanding of processes driving parturition. Prostaglandins (PGs) have diverse functions in parturition, including regulation of uterine contractility and tissue remodeling. Our studies on cervical remodeling in mice suggest that although local synthesis of PGs are not increased in term ripening, transcripts encoding PG-endoperoxide synthase 2 (Ptgs2) are induced in lipopolysaccharide (LPS)-mediated premature ripening. This study provides evidence for two distinct pathways of cervical ripening: one dependent on PGs derived from paracrine or endocrine sources and the other independent of PG actions. Cervical PG levels are increased in LPS-treated mice, a model of infection-mediated PTB, consistent with increases in PG synthesizing enzymes and reduction in PG-metabolizing enzymes. Administration of SC-236, a PTGS2 inhibitor, along with LPS attenuated cervical softening, consistent with the essential role of PGs in LPS-induced ripening. In contrast, during term and preterm ripening mediated by the antiprogestin, mifepristone, cervical PG levels, and expression of PG synthetic and catabolic enzymes did not change in a manner that supports a role for PGs. These findings in mice, supported by correlative studies in women, suggest PGs do not regulate all aspects of the parturition process. Additionally, it suggests a need to refocus current strategies toward developing therapies for the prevention of PTB that target early, pathway-specific processes rather than focusing on common late end point mediators of PTB.


Pediatric Research | 2012

Isoprostanes as physiological mediators of transition to newborn life: novel mechanisms regulating patency of the term and preterm ductus arteriosus

Jian Xiong Chen; Patrick W. O'Mara; Stanley D. Poole; Naoko Brown; Noah J. Ehinger; James C. Slaughter; Bibhash C. Paria; Judy L. Aschner; Jeff Reese

Background:Increased oxygen tension at birth regulates physiologic events that are essential to postnatal survival, but the accompanying oxidative stress may also generate isoprostanes. We hypothesized that isoprostanes regulate ductus arteriosus (DA) function during postnatal vascular transition.Methods:Isoprostanes were measured by gas chromatography–mass spectrometry. DA tone was assessed by pressure myography. Gene expression was measured by quantitative PCR.Results:Oxygen exposure was associated with increased 8-iso-prostaglandin (PG)F2α in newborn mouse lungs. Both 8-iso-PGE2 and 8-iso-PGF2α induced concentration-dependent constriction of the isolated term DA, which was reversed by the thromboxane A2 (TxA2) receptor antagonist SQ29548. SQ29548 pretreatment unmasked an isoprostane-induced DA dilation mediated by the EP4 PG receptor. Exposure of the preterm DA to 8-iso-PGE2 caused unexpected DA relaxation that was reversed by EP4 antagonism. In contrast, exposure to 8-iso-PGF2α caused preterm DA constriction via TxA2 receptor activation. Further investigation revealed the predominance of the TxA2 receptor at term, whereas the EP4 receptor was expressed and functionally active from mid-gestation onward.Conclusion:This study identifies a novel physiological role for isoprostanes during postnatal vascular transition and provide evidence that oxidative stress may act on membrane lipids to produce vasoactive mediators that stimulate physiological DA closure at birth or induce pathological patency of the preterm DA.


Physiological Genomics | 2014

Transcriptional profiling reveals ductus arteriosus-specific genes that regulate vascular tone

Elaine L. Shelton; Gerren Ector; Cristi L. Galindo; Christopher W. Hooper; Naoko Brown; Irene Wilkerson; Elise R. Pfaltzgraff; Bibhash C. Paria; Robert B. Cotton; Jason Z. Stoller; Jeff Reese

Failure of the ductus arteriosus (DA) to close at birth can lead to serious complications. Conversely, certain profound congenital cardiac malformations require the DA to be patent until corrective surgery can be performed. In each instance, clinicians have a very limited repertoire of therapeutic options at their disposal - indomethacin or ibuprofen to close a patent DA (PDA) and prostaglandin E1 to maintain patency of the DA. Neither treatment is specific to the DA and both may have deleterious off-target effects. Therefore, more therapeutic options specifically targeted to the DA should be considered. We hypothesized the DA possesses a unique genetic signature that would set it apart from other vessels. A microarray was used to compare the genetic profiles of the murine DA and ascending aorta (AO). Over 4,000 genes were differentially expressed between these vessels including a subset of ion channel-related genes. Specifically, the alpha and beta subunits of large-conductance calcium-activated potassium (BKCa) channels are enriched in the DA. Gain- and loss-of-function studies showed inhibition of BKCa channels caused the DA to constrict, while activation caused DA relaxation even in the presence of O2. This study identifies subsets of genes that are enriched in the DA that may be used to develop DA-specific drugs. Ion channels that regulate DA tone, including BKCa channels, are promising targets. Specifically, BKCa channel agonists like NS1619 maintain DA patency even in the presence of O2 and may be clinically useful.


PLOS ONE | 2015

High-Throughput Screening of Myometrial Calcium-Mobilization to Identify Modulators of Uterine Contractility.

Jennifer L. Herington; Daniel R. Swale; Naoko Brown; Elaine L. Shelton; Hyehun Choi; Charles H. Williams; Charles C. Hong; Bibhash C. Paria; Jerod S. Denton; Jeff Reese

The uterine myometrium (UT-myo) is a therapeutic target for preterm labor, labor induction, and postpartum hemorrhage. Stimulation of intracellular Ca2+-release in UT-myo cells by oxytocin is a final pathway controlling myometrial contractions. The goal of this study was to develop a dual-addition assay for high-throughput screening of small molecular compounds, which could regulate Ca2+-mobilization in UT-myo cells, and hence, myometrial contractions. Primary murine UT-myo cells in 384-well plates were loaded with a Ca2+-sensitive fluorescent probe, and then screened for inducers of Ca2+-mobilization and inhibitors of oxytocin-induced Ca2+-mobilization. The assay exhibited robust screening statistics (Z´ = 0.73), DMSO-tolerance, and was validated for high-throughput screening against 2,727 small molecules from the Spectrum, NIH Clinical I and II collections of well-annotated compounds. The screen revealed a hit-rate of 1.80% for agonist and 1.39% for antagonist compounds. Concentration-dependent responses of hit-compounds demonstrated an EC50 less than 10μM for 21 hit-antagonist compounds, compared to only 7 hit-agonist compounds. Subsequent studies focused on hit-antagonist compounds. Based on the percent inhibition and functional annotation analyses, we selected 4 confirmed hit-antagonist compounds (benzbromarone, dipyridamole, fenoterol hydrobromide and nisoldipine) for further analysis. Using an ex vivo isometric contractility assay, each compound significantly inhibited uterine contractility, at different potencies (IC50). Overall, these results demonstrate for the first time that high-throughput small-molecules screening of myometrial Ca2+-mobilization is an ideal primary approach for discovering modulators of uterine contractility.


American Journal of Physiology-heart and Circulatory Physiology | 2014

Aminoglycoside-mediated relaxation of the ductus arteriosus in sepsis-associated PDA

Megan Vucovich; Robert B. Cotton; Elaine L. Shelton; Jeremy A. Goettel; Noah J. Ehinger; Stanley D. Poole; Naoko Brown; James L. Wynn; Bibhash C. Paria; James C. Slaughter; Reese H. Clark; Mario A. Rojas; Jeff Reese

Sepsis is strongly associated with patency of the ductus arteriosus (PDA) in critically ill newborns. Inflammation and the aminoglycoside antibiotics used to treat neonatal sepsis cause smooth muscle relaxation, but their contribution to PDA is unknown. We examined whether: 1) lipopolysaccharide (LPS) or inflammatory cytokines cause relaxation of the ex vivo mouse DA; 2) the aminoglycosides gentamicin, tobramycin, or amikacin causes DA relaxation; and 3) newborn infants treated with aminoglycosides have an increased risk of symptomatic PDA (sPDA). Changes in fetal mouse DA tone were measured by pressure myography in response to LPS, TNF-α, IFN-γ, macrophage-inflammatory protein 2, IL-15, IL-13, CXC chemokine ligand 12, or three aminoglycosides. A clinical database of inborn patients of all gestations was analyzed for association between sPDA and aminoglycoside treatment. Contrary to expectation, neither LPS nor any of the inflammatory mediators caused DA relaxation. However, each of the aminoglycosides caused concentration-dependent vasodilation in term and preterm mouse DAs. Pretreatment with indomethacin and N-(G)-nitro-L-arginine methyl ester did not prevent gentamicin-induced DA relaxation. Gentamicin-exposed DAs developed less oxygen-induced constriction than unexposed DAs. Among 488,349 infants who met the study criteria, 40,472 (8.3%) had sPDA. Confounder-adjusted odds of sPDA were higher in gentamicin-exposed infants, <25 wk and >32 wk. Together, these findings suggest that factors other than inflammation contribute to PDA. Aminoglycoside-induced vasorelaxation and inhibition of oxygen-induced DA constriction support the paradox that antibiotic treatment of sepsis may contribute to DA relaxation. This association was also found in newborn infants, suggesting that antibiotic selection may be an important consideration in efforts to reduce sepsis-associated PDA.


Journal of Molecular and Cellular Cardiology | 2013

Cimetidine-associated patent ductus arteriosus is mediated via a cytochrome P450 mechanism independent of H2 receptor antagonism

Robert B. Cotton; Lisa Shah; Stanley D. Poole; Noah J. Ehinger; Naoko Brown; Elaine L. Shelton; James C. Slaughter; H. Scott Baldwin; Bibhash C. Paria; Jeff Reese

Persistent patency of the ductus arteriosus (PDA) is a common problem in preterm infants. The antacid cimetidine is a potent antagonist of the H2 histamine receptor but it also inhibits certain cytochrome P450 enzymes (CYPs), which may affect DA patency. We examined whether cimetidine contributes to PDA and is mediated by CYP inhibition rather than H2 blockade. Analysis of a clinical trial to prevent lung injury in premature infants revealed a significant association between cimetidine treatment and PDA. Cimetidine and ranitidine, both CYP inhibitors as well as H2 blockers, caused relaxation of the term and preterm mouse DA. CYP enzymes that are inhibited by cimetidine were expressed in DA subendothelial smooth muscle. The selective CYP3A inhibitor ketoconazole induced greater DA relaxation than cimetidine, whereas famotidine and other H2 antagonists with less CYP inhibitory effects caused less dilation. Histamine receptors were developmentally regulated and localized in DA smooth muscle. However, cimetidine caused DA relaxation in histamine-deficient mice, consistent with CYP inhibition, not H2 antagonism, as the mechanism for PDA. Oxygen-induced DA constriction was inhibited by both cimetidine and famotidine. These studies show that antacids and other compounds with CYP inhibitory properties pose a significant and previously unrecognized risk for PDA in critically ill newborn infants.


Proceedings of the 2011 Biomedical Sciences and Engineering Conference: Image Informatics and Analytics in Biomedicine | 2011

Detecting changes during pregnancy with Raman spectroscopy

Elizabeth Vargis; C. Nathan Webb; Bibhash C. Paria; Kelly Bennett; Jeff Reese; Ayman Al-Hendy; Anita Mahadevan-Jansen

Preterm labor is the second leading cause of neonatal mortality and leads to a myriad of complications like delayed development and cerebral palsy. Currently, there is no way to accurately predict preterm labor, making its prevention and treatment virtually impossible. While there are some at-risk patients, over half of all preterm births do not fall into any high-risk category. This study seeks to predict and prevent preterm labor by using Raman spectroscopy to detect changes in the cervix during pregnancy. Since Raman spectroscopy has been used to detect cancers in vivo in organs like the cervix and skin, it follows that spectra will change over the course of pregnancy. Previous studies have shown that fluorescence decreased during pregnancy and increased during post-partum exams to pre-pregnancy levels. We believe significant changes will occur in the Raman spectra obtained during the course of pregnancy. In this study, Raman spectra from the cervix of pregnant mice will be acquired. Specific changes that occur due to cervical softening or changes in hormonal levels will be observed to understand the changes that occur before and after labor in the cervix.


Archive | 1999

Cannabinoid Ligand-Receptor Signaling During Early Pregnancy in the Mouse

Bibhash C. Paria; Sanjoy K. Das; Sudhansu K. Dey

The recent identification and cloning of brain-type and spleen-type cannabinoid receptors (CB-1R and CB2-R, respectively) provide evidence that many of the effect of cannabinoids are mediated via these receptors. Using multiple appoaches (RT-PCR, Scatchard analysis, autoradiographic binding, cAMP assay, Western blotting, and immunocytochemistry), we demonstrated that functional CB1-R receptors are present in the preimplantation embryo and uterus. The levels of CB1-R in the embryo are much higher than those in the brain. Furthermore, the mouse uterus has the anandamide synthesizing and hydrolyzing capacities that are differentially regulated during the peri-implantation period. The uterus contains the highest levels of anandamide yet discovered in a mammalian tissue. These results suggest that preimplantation mouse embryos are possible targets for cannabinoid ligand-receptor signaling. Indeed, activation of embryonic cannabinoid receptors by natural and synthetic cannabinoid ligands interferes with preimplantaion embryo development, and this effect is completely reversed by a specific CB1-R antagonist. These results suggest that cannabinoid effects on embryo develpment are mediated by CB1-R We also observed that Δ9-tetrahydrocannabinol [(-)THC] infused in the presence of cytochrome P450 inhibitors interfered with blastocyst implantation. This adverse effect was reversed by coinfusion of CB1-R antagonist. Collectively, these results demonstrated that cannabinoid effects on embryo development and implantation are mediated by embryonic and/or uterine CB1-R.


Archive | 1999

Molecular Signaling in Implantation

Sanjoy K. Das; Bibhash C. Paria; Sudhansu K. Dey

The synchronized development of the preimplantation embryo to the blastocyst stage is essential to the process of implantation (1, 2). The establishment of a differentiated uterus for supporting embryo development and implantation is primarily dependent on the coordinated effects of progesterone (P4) and estrogen (1, 2). In the rodent, the first conspicuous sign that the implantation process has been initiated is an increased endometrial vascular permeability at the sites of blastocysts. This can be visualized as discrete blue bands along the uterus after an intravenous injection of a blue dye (1, 2). This increased vascular permeability coincides with the initial attachment reaction between the blastocyst trophectoderm and uterine luminal epithelium (3). In the mouse, the attachment reaction occurs in the evening (2200–2400 h) on day 4 of pregnancy (4) and is preceded by uterine luminal closure which in turn results in an intimate apposition of the trophectoderm with the luminal epithelium (1, 3, 5). The attachment reaction is followed by localized stromal decidualization and luminal epithelial apoptosis at the sites of blastocyst implantation (6).


American Journal of Reproductive Immunology | 2014

Interdisciplinary Collaborative Team for Blastocyst Implantation Research: inception and perspectives

Koji Yoshinaga; Mercy Prabhudas; Christopher J. Davies; Kenneth L. White; Kathleen M. Caron; Thaddeus G. Golos; Asgerally T. Fazleabas; Bibhash C. Paria; Gil Mor; Soumen Paul; Xiaoqin Ye; Sudhansu K. Dey; Thomas E. Spencer; Robert Michael Roberts

This study describes the currently active research team created by participation of investigators who are eager to work together with other investigators of different disciplines on implantation research. The name of this group is ‘Interdisciplinary Collaborative Team for Blastocyst Implantation Research’. From now on we will call the group the Team. It started with the publication of an NIH Program Announcement.

Collaboration


Dive into the Bibhash C. Paria's collaboration.

Top Co-Authors

Avatar

Jeff Reese

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Naoko Brown

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ayman Al-Hendy

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Noah J. Ehinger

Monroe Carell Jr. Children's Hospital at Vanderbilt

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge