Network


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

Hotspot


Dive into the research topics where Khatuna Gagnidze is active.

Publication


Featured researches published by Khatuna Gagnidze.


Journal of Biological Chemistry | 2003

Activation of the murine interleukin-12 p40 promoter by functional interactions between NFAT and ICSBP

Chen Zhu; Kavitha N. Rao; Huabao Xiong; Khatuna Gagnidze; Fengling Li; Curt M. Horvath; Scott E. Plevy

Interleukin (IL)-12 is a heterodimeric cytokine that is critical for the development of a T-helper-1 immune response and immunity against intracellular pathogens. The IL-12 p40 gene product, expressed specifically in macrophages and dendritic cells, heterodimerizes with p35 to form bioactive IL-12, and heterodimerizes with p19 to comprise the cytokine IL-23. Regulation of the murine IL-12 p40 promoter is complex. Multiple cis-acting elements have been characterized that are involved in activation by bacterial products. However, molecular mechanisms through which interferon (IFN)-γ and bacterial products synergistically activate IL-12 p40 gene expression are less clear. In this study, a composite NFAT/ICSBP binding site at –68 to –54 is identified that is functionally important for p40 promoter activation by lipopolysaccharide (LPS) and LPS plus IFN-γ. DNA binding of NFAT and ICSBP is demonstrated on the endogenous promoter by chromatin immunoprecipitation. NFAT is required for ICSBP binding to this region. Overexpression of NFAT and ICSBP synergistically activates the p40 promoter. A dominant negative NFAT molecule attenuates LPS- and IFN-γ-activated endogenous IL-12 p40 mRNA expression. A physical association between NFAT and ICSBP in the absence of DNA is detected by co-immunoprecipitation of endogenous proteins. Three NFAT domains are required for ICSBP interaction. Finally, in LPS- and IFN-γ-activated RAW-264.7 cells, the association between NFAT and ICSBP is abrogated by IL-10 priming.


The EMBO Journal | 2011

AT1R–CB1R heteromerization reveals a new mechanism for the pathogenic properties of angiotensin II

Raphael Rozenfeld; Achla Gupta; Khatuna Gagnidze; Maribel P. Lim; Ivone Gomes; Dinah Lee-Ramos; Natalia Nieto; Lakshmi A. Devi

The mechanism of G protein‐coupled receptor (GPCR) signal integration is controversial. While GPCR assembly into hetero‐oligomers facilitates signal integration of different receptor types, cross‐talk between Gαi‐ and Gαq‐coupled receptors is often thought to be oligomerization independent. In this study, we examined the mechanism of signal integration between the Gαi‐coupled type I cannabinoid receptor (CB1R) and the Gαq‐coupled AT1R. We find that these two receptors functionally interact, resulting in the potentiation of AT1R signalling and coupling of AT1R to multiple G proteins. Importantly, using several methods, that is, co‐immunoprecipitation and resonance energy transfer assays, as well as receptor‐ and heteromer‐selective antibodies, we show that AT1R and CB1R form receptor heteromers. We examined the physiological relevance of this interaction in hepatic stellate cells from ethanol‐administered rats in which CB1R is upregulated. We found a significant upregulation of AT1R–CB1R heteromers and enhancement of angiotensin II‐mediated signalling, as compared with cells from control animals. Moreover, blocking CB1R activity prevented angiotensin II‐mediated mitogenic signalling and profibrogenic gene expression. These results provide a molecular basis for the pivotal role of heteromer‐dependent signal integration in pathology.


Journal of Biological Chemistry | 2001

Characterization of an activation protein-1-binding site in the murine interleukin-12 p40 promoter. Demonstration of novel functional elements by a reductionist approach.

Chen Zhu; Khatuna Gagnidze; James H. M. Gemberling; Scott E. Plevy

Interleukin (IL)-12 is a heterodimeric cytokine produced by macrophages in response to intracellular pathogens and provides an obligatory signal for the differentiation of T-helper-1 cells. We previously reported an analysis of the IL-12 p40 promoter in RAW264.7 macrophages. Multiple control elements were involved in activation of transcription by bacterial products. A critical control element, located between −96 and −88, interacts with C/EBP family members. In this study, using a strategy to demonstrate functional activity in a minimal promoter context, three novel cis-acting elements are found to have an important role in IL-12 p40 promoter activation by lipopolysaccharide. One of these elements is characterized in detail. Mutations from −79 to −74 in the murine IL-12 p40 promoter significantly reduce lipopolysaccharide-induced promoter activity. Electrophoretic mobility shift assays demonstrate binding of AP-1 family members to this region. Spacing between the C/EBP and AP-1 site is important for promoter activation, suggesting cooperativity between these elements. c-Jun and a mutant c-Jun molecule activate the IL-12 p40 promoter and synergistically activate the promoter when co-expressed with C/EBPβ. Finally, this region of the promoter is demonstrated to be a target for mitogen-activated protein kinase and toll-like receptor signaling pathways.


Molecular Pharmacology | 2006

Simultaneous Activation of the δ Opioid Receptor (δOR)/Sensory Neuron-Specific Receptor-4 (SNSR-4) Hetero-Oligomer by the Mixed Bivalent Agonist Bovine Adrenal Medulla Peptide 22 Activates SNSR-4 but Inhibits δOR Signaling

Andreas Breit; Khatuna Gagnidze; Lakshmi A. Devi; Monique Lagacé; Michel Bouvier

Hetero-oligomerization among G protein-coupled receptors has been proposed to contribute to signal integration. Because sensory neuron-specific receptors (SNSRs) and the opioid receptors (OR) share a common ligand, the bovine adrenal medulla peptide (BAM) 22, and have opposite effects on pain modulation, we investigated the possible consequences of δOR/SNSR-4 hetero-oligomerization on the signaling properties of both receptor subtypes. Bioluminescence resonance energy transfer revealed that the human δOR has similar propensity to homo-oligomerize and to form hetero-oligomers with human SNSR-4 when coexpressed in human embryonic kidney 293 cells. The hetero-oligomerization leads to a receptor form displaying unique functional properties. Individual activation of either δOR or SNSR-4 in cells coexpressing the two receptors led to the modulation of their respective signaling pathways; inhibition of adenylyl cyclase and activation of phospholipase C, respectively. In contrast, the δOR/SNSR-4 bivalent agonist BAM22, which could activate each receptor expressed individually, fully activated the SNSR-4-dependent phospholipase C but did not promote δOR-mediated inhibition of adenylyl cyclase in δOR/SNSR-4-coexpressing cells. Likewise, concomitant activation of the δOR/SNSR-4 hetero-oligomer by selective δOR and SNSR-4 agonists promoted SNSR-4 but not δOR signaling, revealing an agonist-dependent dominant-negative effect of SNSR-4 on δOR signaling. Furthermore, the δOR selective antagonist naltrexone trans-inhibited the SNSR-4-promoted phospholipase C activation mediated by BAM22 but not by the SNSR-4-selective agonists, suggesting a bivalent binding mode of BAM22 to the δOR/SNSR-4 hetero-oligomer. The observation that BAM22 inhibited the Leu-enkephalin-promoted cAMP inhibition in rat dorsal root ganglia neurons supports the potential physiological implication of such regulatory mechanism.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Stress and the dynamic genome: Steroids, epigenetics, and the transposome.

Richard G. Hunter; Khatuna Gagnidze; Bruce S. McEwen; Donald W. Pfaff

Stress plays a substantial role in shaping behavior and brain function, often with lasting effects. How these lasting effects occur in the context of a fixed postmitotic neuronal genome has been an enduring question for the field. Synaptic plasticity and neurogenesis have provided some of the answers to this question, and more recently epigenetic mechanisms have come to the fore. The exploration of epigenetic mechanisms recently led us to discover that a single acute stress can regulate the expression of retrotransposons in the rat hippocampus via an epigenetic mechanism. We propose that this response may represent a genomic stress response aimed at maintaining genomic and transcriptional stability in vulnerable brain regions such as the hippocampus. This finding and those of other researchers have made clear that retrotransposons and the genomic plasticity they permit play a significant role in brain function during stress and disease. These observations also raise the possibility that the transposome might have adaptive functions at the level of both evolution and the individual organism.


Endocrine Reviews | 2014

Implications of prenatal steroid perturbations for neurodevelopment, behavior, and autism.

Andrea C. Gore; Katherine M. Martien; Khatuna Gagnidze; Donald W. Pfaff

The prenatal brain develops under the influence of an ever-changing hormonal milieu that includes endogenous fetal gonadal and adrenal hormones, placental and maternal hormones, and exogenous substances with hormonal activity that can cross the placental barrier. This review discusses the influences of endogenous fetal and maternal hormones on normal brain development and potential consequences of pathophysiological hormonal perturbations to the developing brain, with particular reference to autism. We also consider the effects of hormonal pharmaceuticals used for assisted reproduction, the maintenance of pregnancy, the prevention of congenital adrenal hypertrophy, and hormonal contraceptives continued into an unanticipated pregnancy, among others. These treatments, although in some instances life-saving, may have unintended consequences on the developing fetuses. Additional concern is raised by fetal exposures to endocrine-disrupting chemicals encountered universally by pregnant women from food/water containers, contaminated food, household chemicals, and other sources. What are the potential outcomes of prenatal steroid perturbations on neurodevelopmental and behavioral disorders, including autism-spectrum disorders? Our purposes here are 1) to summarize some consequences of steroid exposures during pregnancy for the development of brain and behavior in the offspring; 2) to summarize what is known about the relationships between exposures and behavior, including autism spectrum disorders; 3) to discuss the molecular underpinnings of such effects, especially molecular epigenetic mechanisms of prenatal steroid manipulations, a field that may explain effects of direct exposures, and even transgenerational effects; and 4) for all of these, to add cautionary notes about their interpretation in the name of scientific rigor.


Journal of Neuroendocrinology | 2013

Early histone modifications in the ventromedial hypothalamus and preoptic area following oestradiol administration

Khatuna Gagnidze; Zachary M. Weil; Larissa C. Faustino; Sara M. Schaafsma; Donald W. Pfaff

Expression of the primary female sex behaviour, lordosis, in laboratory animals depends on oestrogen‐induced expression of progesterone receptor (PgR) within a defined cell group in the ventrolateral portion of the ventromedial nucleus of the hypothalamus (VMH). The minimal latency from oestradiol administration to lordosis is 18 h. During that time, ligand‐bound oestrogen receptors (ER), members of a nuclear receptor superfamily, recruit transcriptional coregulators, which induce covalent modifications of histone proteins, thus leading to transcriptional activation or repression of target genes. The present study aimed to investigate the early molecular epigenetic events underlying oestrogen‐regulated transcriptional activation of the Pgr gene in the VMH of female mice. Oestradiol (E2) administration induced rapid and transient global histone modifications in the VMH of ovariectomised female mice. Histone H3 N‐terminus phosphorylation (H3S10phK14Ac), acetylation (H3Ac) and methylation (H3K4me3) exhibited distinct temporal patterns facilitative to the induction of transcription. A transcriptional repressive (H3K9me3) modification showed a different temporal pattern. Collectively, this should create a permissive environment for the transcriptional activity necessary for lordosis, within 3–6 h after E2 treatment. In the VMH, changes in the H3Ac and H3K4me3 levels of histone H3 were also detected at the promoter region of the Pgr gene within the same time window, although they were delayed in the preoptic area. Moreover, examination of histone modifications associated with the promoter of another ER‐target gene, oxytocin receptor (Oxtr), revealed gene‐ and brain‐region specific effects of E2 treatment. In the VMH of female mice, E2 treatment resulted in the recruitment of ERα to the oestrogen‐response‐elements‐containing putative enhancer site of Pgr gene, approximately 200 kb upstream of the transcription start site, although it failed to increase ERα association with the more proximal promoter region. Finally, E2 administration led to significant changes in the mRNA expression of several ER coregulators in a brain‐region dependent manner. Taken together, these data indicate that, in the hypothalamus and preoptic area of female mice, early responses to E2 treatment involve highly specific changes in chromatin structure, dependent on cell group, gene, histone modification studied, promoter/enhancer site and time following E2.


Proceedings of the National Academy of Sciences of the United States of America | 2013

GPR171 is a hypothalamic G protein-coupled receptor for BigLEN, a neuropeptide involved in feeding

Ivone Gomes; Dipendra K. Aryal; Jonathan H. Wardman; Achla Gupta; Khatuna Gagnidze; Ramona M. Rodriguiz; Sanjai Kumar; William C. Wetsel; John E. Pintar; Lloyd D. Fricker; Lakshmi A. Devi

Significance The mechanism by which vertebrate animals control their body weight is a complex process involving a variety of molecules that regulate feeding and metabolism. Some of these molecules are neuropeptides that bind to specific receptors in feeding centers of the brain. One of the most abundant peptides in brain, LENSSPQAPARRLLPP (named BigLEN), has been proposed to function as a neuropeptide involved in regulating body weight, but the receptor through which this peptide acts had not been identified. We screened candidate receptors and found one, G protein-coupled receptor 171 (GPR171), that is activated by BigLEN. Additional studies showed that the BigLEN–GPR171 system plays an important role in regulating feeding and metabolism in mice. Thus, GPR171 is a potential target for developing antiobesity drugs. Multiple peptide systems, including neuropeptide Y, leptin, ghrelin, and others, are involved with the control of food intake and body weight. The peptide LENSSPQAPARRLLPP (BigLEN) has been proposed to act through an unknown receptor to regulate body weight. In the present study, we used a combination of ligand-binding and receptor-activity assays to characterize a Gαi/o protein-coupled receptor activated by BigLEN in the mouse hypothalamus and Neuro2A cells. We then selected orphan G protein-coupled receptors expressed in the hypothalamus and Neuro2A cells and tested each for activation by BigLEN. G protein-coupled receptor 171 (GPR171) is activated by BigLEN, but not by the C terminally truncated peptide LittleLEN. The four C-terminal amino acids of BigLEN are sufficient to bind and activate GPR171. Overexpression of GPR171 leads to an increase, and knockdown leads to a decrease, in binding and signaling by BigLEN and the C-terminal peptide. In the hypothalamus GPR171 expression complements the expression of BigLEN, and its level and activity are elevated in mice lacking BigLEN. In mice, shRNA-mediated knockdown of hypothalamic GPR171 leads to a decrease in BigLEN signaling and results in changes in food intake and metabolism. The combination of GPR171 shRNA together with neutralization of BigLEN peptide by antibody absorption nearly eliminates acute feeding in food-deprived mice. Taken together, these results demonstrate that GPR171 is the BigLEN receptor and that the BigLEN–GPR171 system plays an important role in regulating responses associated with feeding and metabolism in mice.


Journal of Medicinal Chemistry | 2008

Homology modeling and site-directed mutagenesis to identify selective inhibitors of endothelin-converting enzyme-2.

Khatuna Gagnidze; Sachchidanand; Raphael Rozenfeld; Mihaly Mezei; Ming-Ming Zhou; Lakshmi A. Devi

Endothelin-converting enzyme-2 (ECE-2), a member of M13 family of zinc metallopeptidases, has previously been shown to process a number of neuropeptides including those derived from prodynorphin, proenkephalin, proSAAS, and amyloid precursor protein. ECE-2, unlike ECE-1, exhibits restricted neuroendocrine distribution and acidic pH optimum; it is consistent with a role in the regulation of neuropeptide levels in vivo. Here, we report the generation of a three-dimensional (3D) molecular model of ECE-2 using the crystal structure of neprilysin (EC 3.4.24.11) as a template. On the basis of the predictions made from the molecular model, we mutated and tested two residues, Trp 148 and Tyr 563, in the catalytic site. The mutation of Tyr 563 was found to significantly affect the catalytic activity and inhibitor binding. The molecular model was used to virtually screen a small molecule library of 13 000 compounds. Among the top-scoring compounds three were found to inhibit ECE-2 with high affinity and exhibited specificity for ECE-2 compared to neprilysin. Thus, the model provides a new useful tool to probe the active site of ECE-2 and design additional selective inhibitors of this enzyme.


Proceedings of the National Academy of Sciences of the United States of America | 2017

Sex-specific gene-environment interactions underlying ASD-like behaviors

Sara M. Schaafsma; Khatuna Gagnidze; Anny Reyes; Natalie Norstedt; Karl Månsson; Kerel Francis; Donald W. Pfaff

Significance Autism spectrum disorders (ASDs) comprise a heterogeneous set of neurodevelopmental disorders. Although hundreds of genes have now been identified to be associated with ASD, genetic factors cannot fully explain ASD’s incidence. The early environment is now known to be pivotal in ASD’s etiology too. In the face of this complexity, one aspect of ASD has stood out constantly as a causative biological factor: the sex difference. Approximately 80% of the children diagnosed are boys. This current set of experiments tests, in an animal model, the “three-hit theory of autism,” which states that interactions among (i) being male, (ii) suffering early (especially, prenatal/immunological) stress, and (iii) having certain genetic mutations will predispose to an ASD diagnosis. The male bias in the incidence of autism spectrum disorders (ASDs) is one of the most notable characteristics of this group of neurodevelopmental disorders. The etiology of this sex bias is far from known, but pivotal for understanding the etiology of ASDs in general. Here we investigate whether a “three-hit” (genetic load × environmental factor × sex) theory of autism may help explain the male predominance. We found that LPS-induced maternal immune activation caused male-specific deficits in certain social responses in the contactin-associated protein-like 2 (Cntnap2) mouse model for ASD. The three “hits” had cumulative effects on ultrasonic vocalizations at postnatal day 3. Hits synergistically affected social recognition in adulthood: only mice exposed to all three hits showed deficits in this aspect of social behavior. In brains of the same mice we found a significant three-way interaction on corticotropin-releasing hormone receptor-1 (Crhr1) gene expression, in the left hippocampus specifically, which co-occurred with epigenetic alterations in histone H3 N-terminal lysine 4 trimethylation (H3K4me3) over the Crhr1 promoter. Although it is highly likely that multiple (synergistic) interactions may be at work, change in the expression of genes in the hypothalamic–pituitary–adrenal/stress system (e.g., Crhr1) is one of them. The data provide proof-of-principle that genetic and environmental factors interact to cause sex-specific effects that may help explain the male bias in ASD incidence.

Collaboration


Dive into the Khatuna Gagnidze's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lakshmi A. Devi

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Zachary M. Weil

The Ohio State University Wexner Medical Center

View shared research outputs
Top Co-Authors

Avatar

Achla Gupta

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Chen Zhu

Icahn School of Medicine at Mount Sinai

View shared research outputs
Researchain Logo
Decentralizing Knowledge