Network


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

Hotspot


Dive into the research topics where Gaelle Spagnol is active.

Publication


Featured researches published by Gaelle Spagnol.


American Journal of Physiology-heart and Circulatory Physiology | 2012

Cardiomyocyte ATP release through pannexin 1 aids in early fibroblast activation

Elena Dolmatova; Gaelle Spagnol; Daniela Boassa; Jennifer R. Baum; Kimberly Keith; Cinzia Ambrosi; Maria I. Kontaridis; Paul L. Sorgen; Gina E. Sosinsky; Heather S. Duffy

Fibrosis following myocardial infarction is associated with increases in arrhythmias and sudden cardiac death. Initial steps in the development of fibrosis are not clear; however, it is likely that cardiac fibroblasts play an important role. In immune cells, ATP release from pannexin 1 (Panx1) channels acts as a paracrine signal initiating activation of innate immunity. ATP has been shown in noncardiac systems to initiate fibroblast activation. Therefore, we propose that ATP release through Panx1 channels and subsequent fibroblast activation in the heart drives the development of fibrosis in the heart following myocardial infarction. We identified for the first time that Panx1 is localized within sarcolemmal membranes of canine cardiac myocytes where it directly interacts with the postsynaptic density 95/Drosophila disk large/zonula occludens-1-containing scaffolding protein synapse-associated protein 97 via its carboxyl terminal domain (amino acids 300-357). Induced ischemia rapidly increased glycosylation of Panx1, resulting in increased trafficking to the plasma membrane as well as increased interaction with synapse-associated protein 97. Cellular stress enhanced ATP release from myocyte Panx1 channels, which, in turn, causes fibroblast transformation to the activated myofibroblast phenotype via activation of the MAPK and p53 pathways, both of which are involved in the development of cardiac fibrosis. ATP release through Panx1 channels in cardiac myocytes during ischemia may be an early paracrine event leading to profibrotic responses to ischemic cardiac injury.


Journal of Biological Chemistry | 2009

Characterization of the Structure and Intermolecular Interactions between the Connexin40 and Connexin43 Carboxyl-terminal and Cytoplasmic Loop Domains

Denis Bouvier; Gaelle Spagnol; Sylvie Chenavas; Fabien Kieken; Heidi Vitrac; Sarah Brownell; Admir Kellezi; Vincent Forge; Paul L. Sorgen

Gap junctions are intercellular channels that allow the passage of ions, small molecules, and second messengers that are essential for the coordination of cellular function. They are formed by two hemichannels, each constituted by the oligomerization of six connexins (Cx). Among the 21 different human Cx isoforms, studies have suggested that in the heart, Cx40 and Cx43 can oligomerize to form heteromeric hemichannels. The mechanism of heteromeric channel regulation has not been clearly defined. Tissue ischemia leads to intracellular acidification and closure of Cx43 and Cx40 homomeric channels. However, coexpression of Cx40 and Cx43 in Xenopus oocytes enhances the pH sensitivity of the channel. This phenomenon requires the carboxyl-terminal (CT) part of both connexins. In this study we used different biophysical methods to determine the structure of the Cx40CT and characterize the Cx40CT/Cx43CT interaction. Our results revealed that the Cx40CT is an intrinsically disordered protein similar to the Cx43CT and that the Cx40CT and Cx43CT can interact. Additionally, we have identified an interaction between the Cx40CT and the cytoplasmic loop of Cx40 as well as between the Cx40CT and the cytoplasmic loop of Cx43 (and vice versa). Our studies support the “particle-receptor” model for pH gating of Cx40 and Cx43 gap junction channels and suggest that interactions between cytoplasmic regulatory domains (both homo- and hetero-connexin) could be important for the regulation of heteromeric channels.


Journal of Biological Chemistry | 2013

Effects of Phosphorylation on the Structure and Backbone Dynamics of the Intrinsically Disordered Connexin43 C-terminal Domain

Rosslyn Grosely; Jennifer L. Kopanic; Sarah Nabors; Fabien Kieken; Gaelle Spagnol; Mona Al-Mugotir; Sydney Zach; Paul L. Sorgen

Background: Phosphorylation of the connexin43 C-terminal (Cx43CT) domain regulates gap junction intercellular communication (GJIC). Results: Phosphorylation alters the α-helical propensity of the Cx43CT. Conclusion: Altering the conformational preference of the Cx43CT presents a novel mechanism for regulation of GJIC. Significance: Cx43CT residues susceptible to structural alterations are prime targets for chemical modulators of GJIC. Phosphorylation of the connexin43 C-terminal (Cx43CT) domain regulates gap junction intercellular communication. However, an understanding of the mechanisms by which phosphorylation exerts its effects is lacking. Here, we test the hypothesis that phosphorylation regulates Cx43 gap junction intercellular communication by mediating structural changes in the C-terminal domain. Circular dichroism and nuclear magnetic resonance were used to characterize the effects of phosphorylation on the secondary structure and backbone dynamics of soluble and membrane-tethered Cx43CT domains. Cx43CT phospho-mimetic isoforms, which have Asp substitutions at specific Ser/Tyr sites, revealed phosphorylation alters the α-helical content of the Cx43CT domain only when attached to the membrane. The changes in secondary structure are due to variations in the conformational preference and backbone flexibility of residues adjacent and distal to the site(s) of modification. In addition to the known direct effects of phosphorylation on molecular partner interactions, the data presented here suggest phosphorylation may also indirectly regulate binding affinity by altering the conformational preference of the Cx43CT domain.


PLOS ONE | 2016

Connexin43 Forms Supramolecular Complexes through Non-Overlapping Binding Sites for Drebrin, Tubulin, and ZO-1

Cinzia Ambrosi; Cynthia Ren; Gaelle Spagnol; Gabriel Cavin; Angela C. Cone; Elena E. Grintsevich; Gina E. Sosinsky; Paul L. Sorgen

Gap junctions are membrane specialization domains identified in most tissue types where cells abut each other. The connexin channels found in these membrane domains are conduits for direct cell-to-cell transfer of ions and molecules. Connexin43 (Cx43) is the most ubiquitous connexin, with critical roles in heart, skin, and brain. Several studies described the interaction between Cx43 and the cytoskeleton involving the actin binding proteins Zonula occludens (ZO-1) and drebrin, as well as with tubulin. However, a direct interaction has not been identified between drebrin and Cx43. In this study, co-IP and NMR experiments were used to demonstrate that the Cx43-CT directly interacts with the highly conserved N-terminus region of drebrin. Three Cx43-CT areas were found to be involved in drebrin binding, with residues 264–275 being critical for the interaction. Mimicking Src phosphorylation within this region (Y265) significantly disrupted the interaction between the Cx43-CT and drebrin. Immunofluorescence showed colocalization of Cx43, drebrin, and F-actin in astrocytes and Vero cells membrane, indicating that Cx43 forms a submembrane protein complex with cytoskeletal and scaffolding proteins. The co-IP data suggest that Cx43 indirectly interacts with F-actin through drebrin. Along with the known interaction of the Cx43-CT with ZO-1 and tubulin, the data presented here for drebrin indicate non-overlapping and separated binding sites for all three proteins for which simultaneous binding could be important in regulating cytoskeleton rearrangements, especially for neuronal migration during brain development.


Journal of Biological Chemistry | 2016

Structural Studies of the Nedd4 WW Domains and Their Selectivity for the Connexin43 (Cx43) Carboxyl Terminus.

Gaelle Spagnol; Fabien Kieken; Jennifer L. Kopanic; Hanjun Li; Sydney Zach; Kelly L. Stauch; Rosslyn Grosely; Paul L. Sorgen

Neuronal precursor cell-expressed developmentally down-regulated 4 (Nedd4) was the first ubiquitin protein ligase identified to interact with connexin43 (Cx43), and its suppressed expression results in accumulation of gap junction plaques at the plasma membrane. Nedd4-mediated ubiquitination of Cx43 is required to recruit Eps15 and target Cx43 to the endocytic pathway. Although the Cx43 residues that undergo ubiquitination are still unknown, in this study we address other unresolved questions pertaining to the molecular mechanisms mediating the direct interaction between Nedd4 (WW1–3 domains) and Cx43 (carboxyl terminus (CT)). All three WW domains display a similar three antiparallel β-strand structure and interact with the same Cx43CT 283PPXY286 sequence. Although Tyr286 is essential for the interaction, MAPK phosphorylation of the preceding serine residues (Ser(P)279 and Ser(P)282) increases the binding affinity by 2-fold for the WW domains (WW2 > WW3 ≫ WW1). The structure of the WW2·Cx43CT276–289(Ser(P)279, Ser(P)282) complex reveals that coordination of Ser(P)282 with the end of β-strand 3 enables Ser(P)279 to interact with the back face of β-strand 3 (Tyr286 is on the front face) and loop 2, forming a horseshoe-shaped arrangement. The close sequence identity of WW2 with WW1 and WW3 residues that interact with the Cx43CT PPXY motif and Ser(P)279/Ser(P)282 strongly suggests that the significantly lower binding affinity of WW1 is the result of a more rigid structure. This study presents the first structure illustrating how phosphorylation of the Cx43CT domain helps mediate the interaction with a molecular partner involved in gap junction regulation.


Journal of Cell Science | 2014

TC-PTP directly interacts with connexin43 to regulate gap junction intercellular communication

Hanjun Li; Gaelle Spagnol; Naava Naslavsky; Steve Caplan; Paul L. Sorgen

ABSTRACT Protein kinases have long been reported to regulate connexins; however, little is known about the involvement of phosphatases in the modulation of intercellular communication through gap junctions and the subsequent downstream effects on cellular processes. Here, we identify an interaction between the T-cell protein tyrosine phosphatase (TC-PTP, officially known as PTPN2) and the carboxyl terminus of connexin43 (Cx43, officially known as GJA1). Two cell lines, normal rat kidney (NRK) cells endogenously expressing Cx43 and an NRK-derived cell line expressing v-Src with temperature-sensitive activity, were used to demonstrate that EGF and v-Src stimulation, respectively, induced TC-PTP to colocalize with Cx43 at the plasma membrane. Cell biology experiments using phospho-specific antibodies and biophysical assays demonstrated that the interaction is direct and that TC-PTP dephosphorylates Cx43 residues Y247 and Y265, but does not affect v-Src. Transfection of TC-PTP also indirectly led to the dephosphorylation of Cx43 S368, by inactivating PKC&agr; and PKC&dgr;, with no effect on the phosphorylation of S279 and S282 (MAPK-dependent phosphorylation sites). Dephosphorylation maintained Cx43 gap junctions at the plaque and partially reversed the channel closure caused by v-Src-mediated phosphorylation of Cx43. Understanding dephosphorylation, along with the well-documented roles of Cx43 phosphorylation, might eventually lead to methods to modulate the regulation of gap junction channels, with potential benefits for human health.


Journal of Biological Chemistry | 2016

EHD3 Protein Is Required for Tubular Recycling Endosome Stabilization, and an Asparagine-Glutamic Acid Residue Pair within Its Eps15 Homology (EH) Domain Dictates Its Selective Binding to NPF Peptides

Kriti Bahl; Shuwei Xie; Gaelle Spagnol; Paul L. Sorgen; Naava Naslavsky; Steve Caplan

An elaborate network of dynamic lipid membranes, termed tubular recycling endosomes (TRE), coordinates the process of endocytic recycling in mammalian cells. The C-terminal Eps15 homology domain (EHD)-containing proteins have been implicated in the bending and fission of TRE, thus regulating endocytic recycling. EHD proteins have an EH domain that interacts with proteins containing an NPF motif. We found that NPF-containing EHD1 interaction partners such as molecules interacting with CasL-like1 (MICAL-L1) and Syndapin2 are essential for TRE biogenesis. Also crucial for TRE biogenesis is the generation of phosphatidic acid, an essential lipid component of TRE that serves as a docking point for MICAL-L1 and Syndapin2. EHD1 and EHD3 have 86% amino acid identity; they homo- and heterodimerize and partially co-localize to TRE. Despite their remarkable identity, they have distinct mechanistic functions. EHD1 induces membrane vesiculation, whereas EHD3 supports TRE biogenesis and/or stabilization by an unknown mechanism. While using phospholipase D inhibitors (which block the conversion of glycerophospholipids to phosphatidic acid) to deplete cellular TRE, we observed that, upon inhibitor washout, there was a rapid and dramatic regeneration of MICAL-L1-marked TRE. Using this “synchronized” TRE biogenesis system, we determined that EHD3 is involved in the stabilization of TRE rather than in their biogenesis. Moreover, we identify the residues Ala-519/Asp-520 of EHD1 and Asn-519/Glu-520 of EHD3 as defining the selectivity of these two paralogs for NPF-containing binding partners, and we present a model to explain the atomic mechanism and provide new insight for their differential roles in vesiculation and tubulation, respectively.


Journal of Biomolecular NMR | 2010

NMR structure note: UBA domain of CIP75

Fabien Kieken; Gaelle Spagnol; Vivian Su; Alan F. Lau; Paul L. Sorgen

Ubiquitination is a post-translational process in eukaryotes that covalently modifies substrate proteins with ubiquitin. Ubiquitin can act as a tag that signals the protein-transport machinery to shuttle the protein to the proteasome for degradation. Within the ubiquitin receptor family of proteins is a group that can directly connect ubiquitinated proteins to the proteasome for degradation. Of these receptors, the best-studied are the ubiquitin-like (UBL)ubiquitin-associated (UBA) proteins, Rad23A, PLIC2, and Ddi1. These proteins contain an UBL domain that interacts with the proteasome and the UBA domain that recognizes mono and polyubiquitinated proteins [see review (Su and Lau 2009)]. Recently, a potentially new member of this UBL-UBA domain-containing protein family, named connexin43interacting protein of approximately 75 kDa (CIP75), was identified in a yeast-two hybrid screen (mouse embryonic c-DNA library) to interact with the gap junction protein connexin43 (Cx43) (Li et al. 2008). CIP75 contains an UBL domain at its N-terminus and an UBA domain at its C-terminus; as well as a heat shock chaperonin-binding domain and a PEST sequence. PEST sequences have been shown to direct the ubiquitination and subsequent degradation of proteins undergoing rapid turnover (Roth et al. 1998). CIP75 has 596 amino acids and exhibits high sequence homology (75%) with the human A1Up protein (Davidson et al. 2000). The general domain organization of CIP75 also shows high similarity with the UBL-UBA domain-containing protein family members Ubiquilin-1, PLIC2, Rad23A, Rad23B, and the yeast protein Dsk2 (Li et al. 2008). CIP75 functions in a similar manner as these family members in that the UBL domain of CIP75 is essential for the interaction with the S2/RPN1 and S5a/ RPN10 subunits of the 19S subunit from the 26S proteasome complex. However, the UBA domain may associate with non-ubiquitinated Cx43, and this interaction appears to still regulate the turnover of Cx43 through the proteasomal pathway (Li et al. 2008). UBA domains typically consist of approximately 45 amino acids and are characterized by relatively poor sequence conservation (Hofmann and Bucher 1996). UBA domains adopt a common, compact fold comprising a bundle of three helices and the hydrophobic surface formed by the C-terminus of a-helix 1, loop 1, and a-helix 3, is the principal interface with ubiquitin [see review (Hurley et al. 2006)]. Studies using the two UBA domains from HHR23A established that there are functional differences (i.e. differential protein partners) between UBA domains (Withers-Ward et al. 2000), suggesting the hydrophobic surface may be a more general protein–protein interaction module. Support for this is provided from a survey of the ubiquitin interaction properties of 30 UBA domains, which Fabien Kieken and Gaelle Spagnol contributed equally to this work.


Channels | 2014

Structural order in Pannexin 1 cytoplasmic domains

Gaelle Spagnol; Paul L. Sorgen; David C. Spray

Pannexin 1 forms ion and metabolite permeable hexameric channels with abundant expression in the central nervous system and elsewhere. Although pannexin 1 does not form intercellular channels, a common channel topology and oligomerization state, as well as involvement of the intracellular carboxyl terminal (CT) domain in channel gating, is shared with connexins. In this study, we characterized the secondary structure of the mouse pannexin 1 cytoplasmic domains to complement structural studies of the transmembrane segments and compare with similar domains from connexins. A combination of structural prediction tools and circular dichroism revealed that, unlike connexins (predominately intrinsically disordered), cytosolic regions of pannexin 1 contain approximately 50% secondary structure, a majority being α-helical. Moreover, prediction of transmembrane domains uncovered a potential membrane interacting region (I360-G370) located upstream of the caspase cleavage site (D375-D378) within the pannexin 1 CT domain. The α-helical content of a peptide containing these domains (G357-S384) increased in the presence of detergent micelles providing evidence of membrane association. We also purified a pannexin 1 CT construct containing the caspase cleavage site (M374-C426), assigned the resonances by NMR, and confirmed cleavage by Caspase-3 in vitro. On the basis of these structural studies of the cytoplasmic domains of pannexin 1, we propose a mechanism for the opening of pannexin 1 channels upon apoptosis, involving structural changes within the CT domain.


International Journal of Molecular Sciences | 2018

Protein–Protein Interactions with Connexin 43: Regulation and Function

Paul L. Sorgen; Andrew J. Trease; Gaelle Spagnol; Mario Delmar; Morten Schak Nielsen

Connexins are integral membrane building blocks that form gap junctions, enabling direct cytoplasmic exchange of ions and low-molecular-mass metabolites between adjacent cells. In the heart, gap junctions mediate the propagation of cardiac action potentials and the maintenance of a regular beating rhythm. A number of connexin interacting proteins have been described and are known gap junction regulators either through direct effects (e.g., kinases) or the formation of larger multifunctional complexes (e.g., cytoskeleton scaffold proteins). Most connexin partners can be categorized as either proteins promoting coupling by stimulating forward trafficking and channel opening or inhibiting coupling by inducing channel closure, internalization, and degradation. While some interactions have only been implied through co-localization using immunohistochemistry, others have been confirmed by biophysical methods that allow detection of a direct interaction. Our understanding of these interactions is, by far, most well developed for connexin 43 (Cx43) and the scope of this review is to summarize our current knowledge of their functional and regulatory roles. The significance of these interactions is further exemplified by demonstrating their importance at the intercalated disc, a major hub for Cx43 regulation and Cx43 mediated effects.

Collaboration


Dive into the Gaelle Spagnol's collaboration.

Top Co-Authors

Avatar

Paul L. Sorgen

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Fabien Kieken

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Hanjun Li

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Andrew J. Trease

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jennifer L. Kopanic

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Kelly L. Stauch

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Rosslyn Grosely

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Steve Caplan

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Sydney Zach

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Admir Kellezi

University of Nebraska Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge