Network


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

Hotspot


Dive into the research topics where Aixia Ren is active.

Publication


Featured researches published by Aixia Ren.


Journal of Biological Chemistry | 2013

Multi-drug Resistance Protein 4 (MRP4)-mediated Regulation of Fibroblast Cell Migration Reflects a Dichotomous Role of Intracellular Cyclic Nucleotides

Chandrima Sinha; Aixia Ren; Kavisha Arora; Chang Suk Moon; Sunitha Yarlagadda; Weiqiang Zhang; Satish Cheepala; John D. Schuetz; Anjaparavanda P. Naren

Background: MRP4 is an endogenous transporter of cyclic nucleotides that can regulate cell migration. The role of MRP4 in fibroblast migration is unknown. Results: MRP4-deficient fibroblasts migrate faster and have a moderately higher level of intracellular cyclic nucleotides. Conclusion: Inhibition of MRP4 increases fibroblast migration via alteration of intracellular cyclic nucleotide levels. Significance: Inhibition of MRP4 facilitates wound repair. It has long been known that cyclic nucleotides and cyclic nucleotide-dependent signaling molecules control cell migration. However, the concept that it is not just the absence or presence of cyclic nucleotides, but a highly coordinated balance between these molecules that regulates cell migration, is new and revolutionary. In this study, we used multidrug resistance protein 4 (MRP4)-expressing cell lines and MRP4 knock-out mice as model systems and wound healing assays as the experimental system to explore this unique and emerging concept. MRP4, a member of a large family of ATP binding cassette transporter proteins, localizes to the plasma membrane and functions as a nucleotide efflux transporter and thus plays a role in the regulation of intracellular cyclic nucleotide levels. Here, we demonstrate that mouse embryonic fibroblasts (MEFs) isolated from Mrp4−/− mice have higher intracellular cyclic nucleotide levels and migrate faster compared with MEFs from Mrp4+/+ mice. Using FRET-based cAMP and cGMP sensors, we show that inhibition of MRP4 with MK571 increases both cAMP and cGMP levels, which results in increased migration. In contrast to these moderate increases in cAMP and cGMP levels seen in the absence of MRP4, a robust increase in cAMP levels was observed following treatment with forskolin and isobutylmethylxanthine, which decreases fibroblast migration. In response to externally added cell-permeant cyclic nucleotides (cpt-cAMP and cpt-cGMP), MEF migration appears to be biphasic. Altogether, our studies provide the first experimental evidence supporting the novel concept that balance between cyclic nucleotides is critical for cell migration.


American Journal of Physiology-lung Cellular and Molecular Physiology | 2011

Regulatory domain phosphorylation to distinguish the mechanistic basis underlying acute CFTR modulators.

Louise C. Pyle; Annette Ehrhardt; Lisa High Mitchell; Lijuan Fan; Aixia Ren; Anjaparavanda P. Naren; Yao Li; John P. Clancy; Graeme B. Bolger; Eric J. Sorscher; Steven M. Rowe

Modulator compounds intended to overcome disease-causing mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) show significant promise in clinical testing for cystic fibrosis. However, the mechanism(s) of action underlying these compounds are not fully understood. Activation of CFTR ion transport requires PKA-regulated phosphorylation of the regulatory domain (R-D) and dimerization of the nucleotide binding domains. Using a newly developed assay, we evaluated nine compounds including both CFTR potentatiators and activators discovered via various high-throughput screening strategies to acutely augment CFTR activity. We found considerable differences in the effects on R-D phosphorylation. Some (including UC(CF)-152) stimulated robust phosphorylation, and others had little effect (e.g., VRT-532 and VX-770). We then compared CFTR activation by UC(CF)-152 and VRT-532 in Ussing chamber studies using two epithelial models, CFBE41o(-) and Fischer rat thyroid cells, expressing various CFTR forms. UC(CF)-152 activated wild-type-, G551D-, and rescued F508del-CFTR currents but did not potentiate cAMP-mediated CFTR activation. In contrast, VRT-532 moderately activated CFTR short-circuit current and strongly potentiated forskolin-mediated current. Combined with the result that UC(CF)-152, but not VRT-532 or VX-770, acts by increasing CFTR R-D phosphorylation, these findings indicate that potentiation of endogenous cAMP-mediated activation of mutant CFTR is not due to a pathway involving augmented R-D phosphorylation. This study presents an assay useful to distinguish preclinical compounds by a crucial mechanism underlying CFTR activation, delineates two types of compound able to acutely augment CFTR activity (e.g., activators and potentiators), and demonstrates that a number of different mechanisms can be successfully employed to activate mutant CFTR.


Pflügers Archiv: European Journal of Physiology | 2013

Compartmentalization of cyclic nucleotide signaling: a question of when, where, and why?

Kavisha Arora; Chandrima Sinha; Weiqiang Zhang; Aixia Ren; Chang Suk Moon; Sunitha Yarlagadda; Anjaparavanda P. Naren

Preciseness of cellular behavior depends upon how an extracellular cue mobilizes a correct orchestra of cellular messengers and effector proteins spatially and temporally. This concept, termed compartmentalization of cellular signaling, is now known to form the molecular basis of many aspects of cellular behavior in health and disease. The cyclic nucleotides cyclic adenosine monophosphate and cyclic guanosine monophosphate are ubiquitous cellular messengers that can be compartmentalized in three ways: first, by their physical containment; second, by formation of multiple protein signaling complexes; and third, by their selective depletion. Compartmentalized cyclic nucleotide signaling is a very prevalent response among all cell types. In order to understand how it becomes relevant to cellular behavior, it is important to know how it is executed in cells to regulate physiological responses and, also, how its execution or dysregulation can lead to a pathophysiological condition, which forms the scope of the presented review.


Biochemical Journal | 2011

Functional regulation of cystic fibrosis transmembrane conductance regulator-containing macromolecular complexes: a small-molecule inhibitor approach

Weiqiang Zhang; Himabindu Penmatsa; Aixia Ren; Chandanamali Punchihewa; Andrew Lemoff; Bing Yan; Naoaki Fujii; Anjaparavanda P. Naren

CFTR (cystic fibrosis transmembrane conductance regulator) has been shown to form multiple protein macromolecular complexes with its interacting partners at discrete subcellular microdomains to modulate trafficking, transport and signalling in cells. Targeting protein-protein interactions within these macromolecular complexes would affect the expression or function of the CFTR channel. We specifically targeted the PDZ domain-based LPA2 (type 2 lysophosphatidic acid receptor)-NHERF2 (Na+/H+ exchanger regulatory factor-2) interaction within the CFTR-NHERF2-LPA2-containing macromolecular complexes in airway epithelia and tested its regulatory role on CFTR channel function. We identified a cell-permeable small-molecule compound that preferentially inhibits the LPA2-NHERF2 interaction. We show that this compound can disrupt the LPA2-NHERF2 interaction in cells and thus compromises the integrity of macromolecular complexes. Functionally, it elevates cAMP levels in proximity to CFTR and upregulates its channel activity. The results of the present study demonstrate that CFTR Cl- channel function can be finely tuned by modulating PDZ domain-based protein-protein interactions within the CFTR-containing macromolecular complexes. The present study might help to identify novel therapeutic targets to treat diseases associated with dysfunctional CFTR Cl- channels.


Journal of Biological Chemistry | 2015

Compartmentalized Accumulation of cAMP near Complexes of Multidrug Resistance Protein 4 (MRP4) and Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) Contributes to Drug-induced Diarrhea

Changsuk Moon; Weiqiang Zhang; Aixia Ren; Kavisha Arora; Chandrima Sinha; Sunitha Yarlagadda; Koryse Woodrooffe; John D. Schuetz; Koteswara Rao Valasani; Hugo R. de Jonge; Shiva Kumar Shanmukhappa; Mohamed T. Shata; Randal K. Buddington; Kaushik Parthasarathi; Anjaparavanda P. Naren

Background: Diarrhea is an adverse side effect associated with many therapeutics. Results: Irinotecan induced hyperactive cystic fibrosis transmembrane conductance regulator (CFTR) function by inhibiting multidrug resistance protein 4 (MRP4) and formation of MRP4-CFTR macromolecular complexes. Conclusion: MRP4-CFTR-containing macromolecular complexes play an important role in drug-induced diarrhea. Significance: These studies help define molecular mechanisms of drug-induced diarrhea. Diarrhea is one of the most common adverse side effects observed in ∼7% of individuals consuming Food and Drug Administration (FDA)-approved drugs. The mechanism of how these drugs alter fluid secretion in the gut and induce diarrhea is not clearly understood. Several drugs are either substrates or inhibitors of multidrug resistance protein 4 (MRP4), such as the anti-colon cancer drug irinotecan and an anti-retroviral used to treat HIV infection, 3′-azido-3′-deoxythymidine (AZT). These drugs activate cystic fibrosis transmembrane conductance regulator (CFTR)-mediated fluid secretion by inhibiting MRP4-mediated cAMP efflux. Binding of drugs to MRP4 augments the formation of MRP4-CFTR-containing macromolecular complexes that is mediated via scaffolding protein PDZK1. Importantly, HIV patients on AZT treatment demonstrate augmented MRP4-CFTR complex formation in the colon, which defines a novel paradigm of drug-induced diarrhea.


Biochemistry | 2014

Stabilizing rescued surface-localized δf508 CFTR by potentiation of its interaction with Na(+)/H(+) exchanger regulatory factor 1.

Kavisha Arora; Changsuk Moon; Weiqiang Zhang; Sunitha Yarlagadda; Himabindu Penmatsa; Aixia Ren; Chandrima Sinha; Anjaparavanda P. Naren

Cystic fibrosis (CF) is a recessive genetic disease caused by mutations in CFTR, a plasma-membrane-localized anion channel. The most common mutation in CFTR, deletion of phenylalanine at residue 508 (ΔF508), causes misfolding of CFTR resulting in little or no protein at the plasma membrane. The CFTR corrector VX-809 shows promise for treating CF patients homozygous for ΔF508. Here, we demonstrate the significance of protein–protein interactions in enhancing the stability of the ΔF508 CFTR mutant channel protein at the plasma membrane. We determined that VX-809 prolongs the stability of ΔF508 CFTR at the plasma membrane. Using competition-based assays, we demonstrated that ΔF508 CFTR interacts poorly with Na+/H+ exchanger regulatory factor 1 (NHERF1) compared to wild-type CFTR, and VX-809 significantly increased this binding affinity. We conclude that stabilized CFTR–NHERF1 interaction is a determinant of the functional efficiency of rescued ΔF508 CFTR. Our results demonstrate the importance of macromolecular-complex formation in stabilizing rescued mutant CFTR at the plasma membrane and suggest this to be foundational for the development of a new generation of effective CFTR-corrector-based therapeutics.


Journal of Biological Chemistry | 2013

MAST205 Competes with Cystic Fibrosis Transmembrane Conductance Regulator (CFTR)-associated Ligand for Binding to CFTR to Regulate CFTR-mediated Fluid Transport

Aixia Ren; Weiqiang Zhang; Sunitha Yarlagadda; Chandrima Sinha; Kavisha Arora; Chang Suk Moon; Anjaparavanda P. Naren

Background: CFTR is an important cAMP-regulated chloride channel. Results: MAST205 and CAL compete for binding to CFTR to regulate the expression level and function of CFTR. Conclusion: MAST205 is a regulator for CFTR. Significance: Targeting the MAST205-CFTR complex has potential clinical implications for treating CFTR-related diseases such as cystic fibrosis and secretory diarrheas. The PDZ (postsynaptic density-95/discs large/zona occludens-1) domain-based interactions play important roles in regulating the expression and function of the cystic fibrosis transmembrane conductance regulator (CFTR). Several PDZ domain-containing proteins (PDZ proteins for short) have been identified as directly or indirectly interacting with the C terminus of CFTR. To better understand the regulation of CFTR processing, we conducted a genetic screen and identified MAST205 (a microtubule-associated serine/threonine kinase with a molecular mass of 205 kDa) as a new CFTR regulator. We found that overexpression of MAST205 increased the expression of CFTR and augmented CFTR-mediated fluid transport in a dose-dependent manner. Conversely, knockdown of MAST205 inhibited CFTR function. The PDZ motif of CFTR is required for the regulatory role of MAST205 in CFTR expression and function. We further demonstrated that MAST205 and the CFTR-associated ligand competed for binding to CFTR, which facilitated the processing of CFTR and consequently up-regulated the expression and function of CFTR at the plasma membrane. More importantly, we found that MAST205 could facilitate the processing of F508del-CFTR mutant and augment its quantity and channel function at the plasma membrane. Taken together, our data suggest that MAST205 plays an important role in regulating CFTR expression and function. Our findings have important clinical implications for treating CFTR-associated diseases such as cystic fibrosis and secretory diarrheas.


Journal of Biological Chemistry | 2014

Asymmetrical Macromolecular Complex Formation of Lysophosphatidic Acid Receptor 2 (LPA2) Mediates Gradient Sensing in Fibroblasts

Aixia Ren; Changsuk Moon; Weiqiang Zhang; Chandrima Sinha; Sunitha Yarlagadda; Kavisha Arora; Xusheng Wang; Junming Yue; Kaushik Parthasarathi; Rick Heil-Chapdelaine; Gabor Tigyi; Anjaparavanda P. Naren

Background: Chemotaxis is a fundamental process in many physiological and pathological events. Results: An LPA gradient induces a spatiotemporally restricted decrease in the mobility of LPA2 indicative of its cytoplasmic anchorage to NHERF2, the cytoskeleton and PLCβ, which causes a gradient of localized Ca2+ puffs. Conclusion: Asymmetrical macromolecular complex formation by LPA2 mediates gradient sensing. Significance: Our finding provides a new mechanistic basis to help understand chemotactic gradient sensing. Chemotactic migration of fibroblasts toward growth factors relies on their capacity to sense minute extracellular gradients and respond to spatially confined receptor-mediated signals. Currently, mechanisms underlying the gradient sensing of fibroblasts remain poorly understood. Using single-particle tracking methodology, we determined that a lysophosphatidic acid (LPA) gradient induces a spatiotemporally restricted decrease in the mobility of LPA receptor 2 (LPA2) on chemotactic fibroblasts. The onset of decreased LPA2 mobility correlates to the spatial recruitment and coupling to LPA2-interacting proteins that anchor the complex to the cytoskeleton. These localized PDZ motif-mediated macromolecular complexes of LPA2 trigger a Ca2+ puff gradient that governs gradient sensing and directional migration in response to LPA. Disruption of the PDZ motif-mediated assembly of the macromolecular complex of LPA2 disorganizes the gradient of Ca2+ puffs, disrupts gradient sensing, and reduces the directional migration of fibroblasts toward LPA. Our findings illustrate that the asymmetric macromolecular complex formation of chemoattractant receptors mediates gradient sensing and provides a new mechanistic basis for models to describe gradient sensing of fibroblasts.


Cellular Signalling | 2015

PKA and actin play critical roles as downstream effectors in MRP4-mediated regulation of fibroblast migration.

Chandrima Sinha; Aixia Ren; Kavisha Arora; Chang Suk Moon; Sunitha Yarlagadda; Koryse Woodrooffe; Songbai Lin; John D. Schuetz; Assem G. Ziady; Anjaparavanda P. Naren

Multidrug resistance protein 4 (MRP4), a member of the ATP binding cassette transporter family, functions as a plasma membrane exporter of cyclic nucleotides. Recently, we demonstrated that fibroblasts lacking the Mrp4 gene migrate faster and contain higher cyclic-nucleotide levels. Here, we show that cAMP accumulation and protein kinase A (PKA) activity are higher and polarized in Mrp4(-/-) fibroblasts, versus Mrp4(+/+) cells. MRP4-containing macromolecular complexes isolated from these fibroblasts contained several proteins, including actin, which play important roles in cell migration. We found that actin interacts with MRP4, predominantly at the plasma membrane, and an intact actin cytoskeleton is required to restrict MRP4 to specific microdomains of the plasma membrane. Our data further indicated that the enhanced accumulation of cAMP in Mrp4(-/-) fibroblasts facilitates cortical actin polymerization in a PKA-dependent manner at the leading edge, which in turn increases the overall rate of cell migration to accelerate the process of wound healing. Disruption of actin polymerization or inhibition of PKA activity abolished the effect of MRP4 on cell migration. Together, our findings suggest a novel cAMP-dependent mechanism for MRP4-mediated regulation of fibroblast migration whereby PKA and actin play critical roles as downstream effectors.


Gastroenterology | 2012

Mo2042 Identification of a Novel CFTR Interactome in Contributing Diarrhea in Inflammatory Bowel Disease

Kavisha Arora; Sunitha Yarlagadda; Aixia Ren; Weiqiang Zhang; Chandrima Sinha; Anjaparavanda P. Naren

Background: Inflammatory bowel disease (IBD) is a group of disorders in which the gut suffers from inflammation. Among the few predisposing genetic factors identified in IBD, a cGMP/cAMP-regulated chloride (Cl-) channel CFTR has emerged as one. Our lab and others have reported involvement of CFTR in formation of multiple protein complexes that can regulate the channel function in healthy and diseased states. In our current study, we aim to elucidate a molecular mechanism in the form of a signaling protein complex of CFTR that contributes to disease progression in IBD. Method: (a) Protein-protein interactions were studied using biochemical methods (protein pairwise binding assay,macromolecular complex assembly assay, and co-immunoprecipitation) and In Vitro methods for studying protein interaction in live cell (N-FRETc) in HEK293 cells. (b) Immunohistochemistry was done on mouse colon tissue for studying protein expression and distribution. Real time PCR was performed to monitor relative expression levels of several transcripts under inflammatory conditions. (c) CFTR dependent chloride secretion was monitored using iodide efflux, chloride efflux and short circuit measurements in colonic epithelial cell line HT29CL19A. (d) Dextran sodium sulfate (DSS) induced colitis mouse model was used in Nherf2+/+ and Nherf2-/C57BL/6 male mice 7-8 weeks of age. Results: (a) Upregulation of iNOS increased CFTR channel function by at least two folds in In Vivo and In Vitro models of inflammation. The phenomenon involves cGMP dependent signaling mechanism. (b) CFTR interacts with iNOS mediated by NHERF2 in a PDZ domain-dependent manner. (c) Diarrhea is attenuated in Nherf2-/compared to Nherf2+/+ mice in DSS-induced colitis model which simulates IBD. Conclusion: (a) A macromolecular complex of CFTR, NHERF2 and iNOS is integrative to CFTR dependent diarrhea in IBD. This compartmentalization of proteins and consequently of the signaling effectors enables a rapid and fine-tuned regulation of CFTR function. (b) Such signaling modules that are part of pathological profile can be approached as targets for improvement of IBD conditions.

Collaboration


Dive into the Aixia Ren's collaboration.

Top Co-Authors

Avatar

Anjaparavanda P. Naren

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Weiqiang Zhang

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Kavisha Arora

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Sunitha Yarlagadda

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Chandrima Sinha

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Chang Suk Moon

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Changsuk Moon

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Himabindu Penmatsa

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

John D. Schuetz

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Kaushik Parthasarathi

University of Tennessee Health Science Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge