Network


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

Hotspot


Dive into the research topics where Kenneth B. Gagnon is active.

Publication


Featured researches published by Kenneth B. Gagnon.


Biochemical Journal | 2008

SPAK and OSR1: STE20 kinases involved in the regulation of ion homoeostasis and volume control in mammalian cells

Eric Delpire; Kenneth B. Gagnon

Since the discovery of an interaction between membrane transport proteins and the mammalian STE20 (sterile 20)-like kinases SPAK (STE20/SPS1-related proline/alanine-rich kinase) and OSR1 (oxidative stress-responsive kinase-1), a significant body of work has been performed probing the molecular physiology of these two kinases. To date, the function of SPAK and OSR1 is probably the best known of all mammalian kinases of the STE20 family. As they regulate by direct phosphorylation key ion transport mechanisms involved in fluid and ion homoeostasis, SPAK and OSR1 constitute key end-of-pathway effectors. Their significance in such fundamental functions as ion homoeostasis and cell volume control is evidenced by the evolutionary pressure that resulted in the duplication of the OSR1 gene in higher vertebrates. This review examines the distribution of these two kinases in the animal kingdom and tissue expression within a single organism. It also describes the main molecular features of these two kinases with emphasis on the interacting domain located at their extreme C-terminus. A large portion of the present review is devoted to the extensive biochemical and physiological studies that have resulted in our current understanding of SPAK/OSR1 function. Finally, as our understanding is a work in progress, we also identify unresolved questions and controversies that warrant further investigation.


Molecular and Cellular Biology | 2006

Characterization of SPAK and OSR1, Regulatory Kinases of the Na-K-2Cl Cotransporter

Kenneth B. Gagnon; Roger England; Eric Delpire

ABSTRACT Our recent studies demonstrate that SPAK (Ste20p-related Proline Alanine-rich Kinase), in combination with WNK4 [With No lysine (K) kinase], phosphorylates and stimulates the Na-K-2Cl cotransporter (NKCC1), whereas catalytically inactive SPAK (K104R) fails to activate the cotransporter. The catalytic domain of SPAK contains an activation loop between the well-conserved DFG and APE motifs. We speculated that four threonine residues (T231, T236, T243, and T247) in the activation loop might be sites of phosphorylation and kinase activation; therefore, we mutated each residue into an alanine. In this report, we demonstrate that coexpression of SPAK (T243A) or SPAK (T247A) with WNK4 not only prevented, but robustly inhibited, cotransporter activity in NKCC1-injected Xenopus laevis oocytes. These activation loop mutations produced an effect similar to that of the SPAK (K104R) mutant. In vitro phosphorylation experiments demonstrate that both intramolecular autophosphorylation of SPAK and phosphorylation of NKCC1 are significantly stronger in the presence of Mn2+ rather than Mg2+. We also show that SPAK activity is markedly inhibited by staurosporine and K252a, partially inhibited by N-ethylmaleimide and diamide, and unaffected by arsenite. OSR1, a kinase closely related to SPAK, exhibited similar kinase properties and similar functional activation of NKCC1 when coexpressed with WNK4.


Physiological Reviews | 2012

MOLECULAR PHYSIOLOGY OF SPAK AND OSR1: TWO STE20-RELATED PROTEIN KINASES REGULATING ION TRANSPORT

Kenneth B. Gagnon; Eric Delpire

SPAK (Ste20-related proline alanine rich kinase) and OSR1 (oxidative stress responsive kinase) are members of the germinal center kinase VI subfamily of the mammalian Ste20 (Sterile20)-related protein kinase family. Although there are 30 enzymes in this protein kinase family, their conservation across the fungi, plant, and animal kingdom confirms their evolutionary importance. Already, a large volume of work has accumulated on the tissue distribution, binding partners, signaling cascades, and physiological roles of mammalian SPAK and OSR1 in multiple organ systems. After reviewing this basic information, we will examine newer studies that demonstrate the pathophysiological consequences to SPAK and/or OSR1 disruption, discuss the development and analysis of genetically engineered mouse models, and address the possible role these serine/threonine kinases might have in cancer proliferation and migration.


Acta Physiologica | 2006

SPAK and OSR1, key kinases involved in the regulation of chloride transport.

Eric Delpire; Kenneth B. Gagnon

Reversible phosphorylation by protein kinases is probably one of the most important examples of post‐translational modification of ion transport proteins. Ste20‐related proline alanine‐rich kinase (SPAK) and oxidative stress response kinase (OSR1) are two serine/threonine kinases belonging to the germinal centre‐like kinase subfamily VI. Genetic analysis suggests that OSR1 evolved first, with SPAK arising following a gene duplication in vertebrate evolution. SPAK and OSR1 are two recently discovered kinases which have been linked to several key cellular processes, including cell differentiation, cell transformation and proliferation, cytoskeleton rearrangement, and most recently, regulation of ion transporters. Na–K–2Cl cotransporter activity is regulated by phosphorylation. Pharmacological evidence has identified several kinases and phosphatases which alter cotransporter function, however, no direct linkage between these enzymes and the cotransporter has been demonstrated. This article will review some of the physical and physiological properties of SPAK and OSR1, and present new evidence of a direct interaction between the Na–K–Cl cotransporter and the stress kinases.


American Journal of Physiology-cell Physiology | 2013

Physiology of SLC12 transporters: lessons from inherited human genetic mutations and genetically engineered mouse knockouts

Kenneth B. Gagnon; Eric Delpire

Among the over 300 members of the solute carrier (SLC) group of integral plasma membrane transport proteins are the nine electroneutral cation-chloride cotransporters belonging to the SLC12 gene family. Seven of these transporters have been functionally described as coupling the electrically silent movement of chloride with sodium and/or potassium. Although in silico analysis has identified two additional SLC12 family members, no physiological role has been ascribed to the proteins encoded by either the SLC12A8 or the SLC12A9 genes. Evolutionary conservation of this gene family from protists to humans confirms their importance. A wealth of physiological, immunohistochemical, and biochemical studies have revealed a great deal of information regarding the importance of this gene family to human health and disease. The sequencing of the human genome has provided investigators with the capability to link several human diseases with mutations in the genes encoding these plasma membrane proteins. The availability of bacterial artificial chromosomes, recombination engineering techniques, and the mouse genome sequence has simplified the creation of targeting constructs to manipulate the expression/function of these cation-chloride cotransporters in the mouse in an attempt to recapitulate some of these human pathologies. This review will summarize the three human disorders that have been linked to the mutation/dysfunction of the Na-Cl, Na-K-2Cl, and K-Cl cotransporters (i.e., Bartters, Gitlemans, and Andermanns syndromes), examine some additional pathologies arising from genetically modified mouse models of these cotransporters including deafness, blood pressure, hyperexcitability, and epithelial transport deficit phenotypes.


Cellular Physiology and Biochemistry | 2007

A single binding motif is required for SPAK activation of the Na-K-2Cl cotransporter.

Kenneth B. Gagnon; Roger England; Eric Delpire

Background: SPAK (Ste20p-related proline alanine-rich kinase) phosphorylates and activates NKCC1 (Na-K-2Cl cotransporter) in the presence of another serine/threonine kinase WNK4 (With No lysine (K)). However, whether or not the docking of SPAK to NKCC1 is a requirement for cotransporter activation has not been fully resolved. Methods: We mutated both SPAK binding motifs in the amino-terminal tail of NKCC1 and tested the interaction between SPAK and NKCC1 using a semi in vivo yeast two-hybrid assay, 32P-ATP in vitro phosphorylation assays, and 86Rb+ uptake (a K+ congener) assays in heterologously expressed Xenopus laevis oocytes. We also used site-directed mutagenesis to identify the principle phospho-regulatory threonine residues in the amino-terminal tail of NKCC1. Results: A single SPAK binding motif is necessary for isotonic NKCC1 activation. Mutation of the phenylalanine (F) residue within the motif abrogates binding and function. Phosphorylation of the cotransporter is markedly reduced in the absence of SPAK docking to NKCC1. Truncations of internal regions of the amino-terminus of NKCC1 do not disrupt protein structure enough to affect cotransporter function. Threonine residues (T206 and T211) are both identified as phospho-regulatory sites of NKCC1 function. Conclusion: We demonstrate that physical docking of SPAK to NKCC1 is necessary for cotransporter activity under both baseline and hyperosmotic conditions. We identify T206 and T211 as major phospho-acceptor sites involved in cotransporter function, with T206 common to two separate regulatory pathways: one involving SPAK, the other involving a still unknown kinase that is responsive to forskolin/PKA stimulation.


American Journal of Physiology-cell Physiology | 2010

On the substrate recognition and negative regulation of SPAK, a kinase modulating Na-K-2Cl cotransport activity

Kenneth B. Gagnon; Eric Delpire

Threonines targeted by Ste20-related proline-alanine-rich kinase (SPAK) for phosphorylation have been identified in Na+-K+-2Cl(-) cotransporter type 1 (NKCC1), NKCC2, and Na+-Cl(-) cotransporter (NCC). However, what constitutes the substrate recognition of the kinase is still unknown. Using site-directed mutagenesis and functional measurement of NKCC1 activity in Xenopus laevis oocytes, we determined that SPAK recognizes two threonine residues separated by four amino acids. Addition or removal of a single residue abrogated SPAK activation of NKCC1. Although both threonines are followed by hydrophobic residues, in vivo experiments have determined that SPAK activation of the cotransporter only requires a hydrophobic residue after the first threonine. Interestingly, downstream of the second threonine residue, we have identified a conserved aspartic acid residue which is critical for NKCC1 function. Mouse SPAK activity requires phosphorylation of two specific residues by WNK [with no lysine (K)] kinases: a threonine (T243) in the catalytic domain and a serine (S383) in the regulatory domain. We found that mutating the threonine residue into a glutamic acid (T243E) combined with mutation of the serine into an aspartic acid (S383D) rendered SPAK constitutively active. Surprisingly, alanine substitution of S383 or mutation of residues surrounding this residue also resulted in a constitutively active kinase. Interestingly, deletion of amino acids 356-398 identified another serine residue in the catalytic domain (S321) as another putative target of WNK phosphorylation. We found that WNK4 is capable of stimulating the deletion mutant when S321 is present, but not when S321 is mutated into an alanine.


Cellular Physiology and Biochemistry | 2001

K-Cl Cotransport: Immunohistochemical and Ion Flux Studies in Human Embryonic Kidney (HEK293) Cells Transfected with Full-Length and C-Terminal-Domain-Truncated KCC1 cDNAs

Peter K. Lauf; Jin Zhang; Kenneth B. Gagnon; Eric Delpire; Robert E.W. Fyffe; Norma C. Adragna

Coupled K and Cl movements are mediated by several isoforms of the K-Cl cotransporter (COT) encoded by the KCC genes. The ubiquitous KCC1 isoform, important for cell volume and ion homeostasis, has 12 transmembrane domains (Tmds), and cytoplasmic N- and C-terminal domains (Ntd and Ctd). This study investigates the cellular localization of KCC1 by confocal microscopy, activation of K-Cl COT by various non-osmotic and osmotic interventions with net unidirectional K and Rb fluxes at 37°C, and the effect of Ctd deletion on K-Cl COT regulation. Human embryonic kidney (HEK293) cells were transfected with full-length (fl) rabbit (rb)KCC1 and – CtdKCC1 cDNAs obtained after truncation at nucleotide 2011. Normal cells exposed to polyclonal anti-Ctd antibodies against Ctd epitopes within a 77 amino acid sequence (a.a.943-1020) revealed granular membrane and cytoplasmic immunostaining, presumably endogenous KCC1. Additional diffuse membrane and cytoplasmic immunofluorescence in flKCC1-transfected cells was absent in -CtdKCC1-transfected cells. Monoclonal antibodies against a c-myc epitope at the protein Ntd showed both membrane and cytosolic fluoresence. Basal and N-ethylmaleimide (NEM)-stimulated Rb influxes through K-Cl COT, calculated as Cl-dependent Rb fluxes, were 2-3-fold higher in flKCC1-transfected than in normal cells. NEM stimulation of K-Cl COT was highest in flKCC1-transfected cells, significantly lower in stably and abrogated in transiently –CtdKCC1-transfected cells. Furosemide, calyculin and genistein inhibited basal and NEM-stimulated K-Cl COT in normal and transfected cells. Staurosporine and hydroxylamine were ineffective stimulators. No effect of pH₀ changes (6.3-8.4) was observed in basal or NEM-stimulated K-Cl COT, in both normal and transfected cells. However, inhibition by NEM occurred at pH₀ 8.4. Furthermore, in a Cl-independent manner, NEM lowered cell K content by >30% and hypotonicity (210-70mOsM) stimulated furosemide-sensitive Rb influx and K loss. Thus, in cultured normal and KCC1-transfected cells, K-Cl COT shows significant differences from erythrocytes, and NEM and cell swelling open furosemide-sensitive and Cl-independent K/Rb channels. Failure of K-Cl COT in cells transfected with Ctd-truncated KCC1 to respond to NEM suggests a role of the Ctd for signal transduction.


Cellular Physiology and Biochemistry | 2007

Characterization of Glial Cell K-Cl Cotransport

Kenneth B. Gagnon; Norma C. Adragna; Robert E.W. Fyffe; Peter K. Lauf

Background: The molecular mechanism of K-Cl cotransport (KCC) consists of at least 4 isoforms, KCC 1, 2, 3, and 4 which, in multiple combinations, exist in most cells, including erythrocytes and neuronal cells. Methods: We utilized reverse-transcriptase-polymerase chain reaction (RT-PCR) and ion flux studies to characterize KCC activity in an immortalized in vitro cell model for fibrous astrocytes, the rat C6 glioblastoma cell. Isoform-specific sets of oligonucleotide primers were synthesized for NKCC1, KCC1, KCC2, KCC3, KCC4, and also for NKCC1 and actin. K-Cl cotransport activity was determined by measuring either the furosemide-sensitive, or the Cl<sup>-</sup>-dependent bumetanide-insensitive Rb<sup>+</sup> (a K<sup>+</sup> congener) influx in the presence of the Na/K pump inhibitor ouabain. Rb<sup>+</sup> influx was measured at a fixed external Cl concentrations, [Cl<sup>-</sup>]<sub>e</sub>, as a function of varying external Rb concentrations, [Rb<sup>+</sup>]<sub>e</sub>, and at a fixed [Rb<sup>+</sup>]<sub>e</sub> as a function of varying [Cl<sup>-</sup>]<sub>e</sub>, and with equimolar Cl replacement by anions of the chaotropic series. Results: RT-PCR of C6 glioblastoma (C6) cells identified mRNA for three KCC isoforms (1, 3, and 4). NKCC1 mRNA was also detected. The apparent K<sub>m</sub> for KCC-mediated Rb<sup>+</sup> influx was 15 mM [Rb<sup>+</sup>]<sub>e</sub>, and V<sub>max</sub> 12.5 nmol Rb<sup>+</sup> * mg protein<sup>-1</sup> * minute<sup>-1</sup>. The calculated apparent K<sub>m</sub> for external Cl<sup>-</sup> was 13 mM and V<sub>max</sub> 14.4 nmol Rb<sup>+</sup> * mg protein<sup>-1</sup> * minute<sup>-1</sup>. The anion selectivity sequence of the furosemide-sensitive Rb<sup>+</sup> influx was Cl<sup>-</sup>>>Br-=NO<sub>3</sub><sup>-</sup>>I<sup>-</sup>=SCN<sup>-</sup>>>Sfm<sup>-</sup> (sulfamate). Established activators of K-Cl cotransport, hyposmotic shock and N-ethylmaleimide (NEM) pretreatment, stimulated furosemide-sensitive Rb<sup>+</sup> influx. A ñ50% NEM-induced loss of intracellular K<sup>+</sup> was prevented by furosemide. Conclusion: We have identified by RT-PCR the presence of three distinct KCC isoforms (1, 3, and 4) in rat C6 glioblastoma cells, and functionally characterized the anion selectivity and kinetics of their collective sodium-independent cation-chloride cotransport activity.


Journal of Biological Chemistry | 2010

Multiple pathways for protein phosphatase 1 (PP1) regulation of Na-K-2Cl cotransporter (NKCC1) function: the N-terminal tail of the Na-K-2Cl cotransporter serves as a regulatory scaffold for Ste20-related proline/alanine-rich kinase (SPAK) AND PP1.

Kenneth B. Gagnon; Eric Delpire

The Na-K-2Cl cotransporter (NKCC1) participates in epithelial transport and in cell volume maintenance by mediating the movement of ions and water across plasma membranes. Functional studies have previously demonstrated that NKCC1 activity is stimulated by protein phosphatase 1 (PP1) inhibitors. In this study, we utilized both in vivo (heterologous cRNA expression in Xenopus laevis oocytes) and in vitro (32P-phosphorylation assays with glutathione S-transferase fusion proteins) experiments to determine whether PP1 exerts its inhibitory effect directly on the cotransporter, or indirectly by affecting the activating kinase. We found that PP1 reduced NKCC1 activity in oocytes under both isotonic and hypertonic conditions to the same level as in water-injected controls. Interestingly, mutation of key residues in the PP1 binding motif located in the N-terminal tail of NKCC1 significantly reduced the inhibitory effect of PP1. In vitro experiments performed with recombinant PP1, SPAK (Ste20-related proline/alanine-rich kinase, which activates NKCC1), and the N terminus of NKCC1 fused to glutathione S-transferase demonstrated that PP1 dephosphorylated both the kinase and the cotransporter in a time-dependent manner. More importantly, PP1 dephosphorylation of SPAK was significantly greater when protein-protein interaction between the kinase and the N-terminal tail of NKCC1 was present in the reaction, indicating the necessity of scaffolding the phosphatase and kinase in proximity to one another. Taken together, our data are consistent with PP1 inhibiting NKCC1 activity directly by dephosphorylating the cotransporter and indirectly by dephosphorylating SPAK.

Collaboration


Dive into the Kenneth B. Gagnon's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Roger England

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

José Ponce-Coria

National Autonomous University of Mexico

View shared research outputs
Top Co-Authors

Avatar

Norma Vázquez

National Autonomous University of Mexico

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge