Network


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

Hotspot


Dive into the research topics where Joseph W. Lynch is active.

Publication


Featured researches published by Joseph W. Lynch.


The Journal of Membrane Biology | 1991

Liquid junction potentials and small cell effects in patch-clamp analysis

Peter H. Barry; Joseph W. Lynch

Since the early 1980s, the patch-clamp technique (Hamill et al., 1981) has been of particular value in investigating the properties of ion channels in cells. When used in either the intact or excised configurations, the properties of individual ionic channels can be directly measured. In addition, the whole-cell configuration can be used to investigate the total response of the full complement of channels in a cell. The whole-cell configuration is of particular value in exploring the properties of very small cells which are not readily accessible to conventional microelectrode techniques. in all of the above measurements, there are two potential sources of error, in every situation there may be significant errors due to uncompensated junction potentials, which m a y a p p e a r to be eliminated by the normal zeroing procedure whereby residual potentials between pipette and bath solutions are offset prior to patch formation. In addition, in the intact and whole-cell patch configurations, the effect of the cells being small can introduce radical errors in the measurement of single-channel and whole-cell properties. The aim of this review is firstly to outline the contribution of such junction potentials and the errors resulting from measurements on small cells and secondly to indicate how adequate junction potential corrections can be applied and the true values of underlying membrane parameters determined for such cells. Where necessary, appropriate equations have been presented. Much of the material is a review of published work. However, the review also seeks to extend the implications of that work and, in particular, it also includes (in an appendix) a timedependent solution of the current relaxation following a channel closure.


Pharmacology & Therapeutics | 1997

The glycine receptor

Sundran Rajendra; Joseph W. Lynch; Peter R. Schofield

The inhibitory glycine receptor (GlyR) is a member of the ligand-gated ion channel receptor superfamily. The GlyR comprises a pentameric complex that forms a chloride-selective transmembrane channel, which is predominantly expressed in the spinal cord and brain stem. We review the pharmacological and physiological properties of the GlyR and relate this information to more recent insights that have been obtained through the cloning and recombinant expression of the GlyR subunits. We also discuss insights into our understanding of GlyR structure and function that have been obtained by the genetic characterisation of various heritable disorders of glycinergic neurotransmission.


Neuropharmacology | 2009

Native glycine receptor subtypes and their physiological roles

Joseph W. Lynch

The glycine receptor chloride channel (GlyR), a member of the pentameric Cys-loop ion channel receptor family, mediates inhibitory neurotransmission in the spinal cord, brainstem and retina. They are also found presynaptically, where they modulate neurotransmitter release. Functional GlyRs are formed from a total of five subunits (alpha1-alpha4, beta). Although alpha subunits efficiently form homomeric GlyRs in recombinant expression systems, homomeric alpha1, alpha3 and alpha4 GlyRs are weakly expressed in adult neurons. In contrast, alpha2 homomeric GlyRs are abundantly expressed in embryonic neurons, although their numbers decline sharply by adulthood. Numerous lines of biochemical, biophysical, pharmacological and genetic evidence suggest the majority of glycinergic neurotransmission in adults is mediated by heteromeric alpha1beta GlyRs. Immunocytochemical co-localisation experiments suggest the presence of alpha2beta, alpha3beta and alpha4beta GlyRs at synapses in the adult mouse retina. Immunocytochemical and electrophysiological evidence also implicates alpha3beta GlyRs as important mediators of glycinergic inhibitory neurotransmission in nociceptive sensory neuronal circuits in peripheral laminae of the spinal cord dorsal horn. It is yet to be determined why multiple GlyR synaptic subtypes are differentially distributed in these and possibly other locations. The development of pharmacological agents that can discriminate strongly between different beta subunit-containing GlyR isoforms will help to address this issue, and thereby provide important insights into a variety of central nervous system functions including retinal signal processing and spinal pain mechanisms. Finally, agents that selectively potentiate different GlyR isoforms may be useful as therapeutic lead compounds for peripheral inflammatory pain and movement disorders such as spasticity.


The EMBO Journal | 1997

Identification of intracellular and extracellular domains mediating signal transduction in the inhibitory glycine receptor chloride channel.

Joseph W. Lynch; Sundran Rajendra; Kerrie D. Pierce; Cheryl A. Handford; Peter H. Barry; Peter R. Schofield

Fast synaptic neurotransmission is mediated by transmitter‐activated conformational changes in ligand‐gated ion channel receptors, culminating in opening of the integral ion channel pore. Human hereditary hyperekplexia, or startle disease, is caused by mutations in both the intracellular or extracellular loops flanking the pore‐lining M2 domain of the glycine receptor α1 subunit. These flanking domains are designated the M1‐M2 loop and the M2‐M3 loop respectively. We show that four startle disease mutations and six additional alanine substitution mutations distributed throughout both loops result in uncoupling of the ligand binding sites from the channel activation gate. We therefore conclude that the M1‐M2 and M2‐M3 loops act in parallel to activate the channel. Their locations strongly suggest that they act as hinges governing allosteric control of the M2 domain. As the members of the ligand‐gated ion channel superfamily share a common structure, this signal transduction model may apply to all members of this superfamily.


Neuron | 1995

Mutation of an arginine residue in the human glycine receptor transforms β-alanine and taurine from agonists into competitive antagonists

Sundran Rajendra; Joseph W. Lynch; Kerrie D. Pierce; Chris French; Peter H. Barry; Peter R. Schofield

Agonist binding to the inhibitory glycine receptor (GlyR) initiates the opening of a chloride-selective channel that modulates the neuronal membrane potential. Point mutations of the GlyR, substituting Arg-271 with either Leu or Gln, have been shown to underlie the inherited neurological disorder startle disease (hyperekplexia). We show that these substitutions result in the redistribution of GlyR single-channel conductances to lower conductance levels. Additionally, the binding of the glycinergic agonists beta-alanine and taurine to mutated GlyRs does not initiate a chloride current, but instead competitively antagonizes currents activated by glycine. These findings are consistent with mutations of Arg-271 resulting in the uncoupling of the agonist binding process from the channel activation mechanism of the receptor.


Biophysical Journal | 1989

Action potentials initiated by single channels opening in a small neuron (rat olfactory receptor)

Joseph W. Lynch; Peter H. Barry

Rat olfactory receptor neurons were enzymatically dissociated and studied with the cell-attached configuration of the patch-clamp technique. Biphasic current waveforms induced across the membrane patch by intracellular action potentials were observed in approximately 5% of cells studied. In one cell in particular, current injected by the opening of a single channel initiated an action potential in the remainder of the cell each time the channel opened. A conventional type of electrical model of the cell and patch allowed the accurate modeling of cell excitability. The same model was used to explain the shape of the action potential current waveforms induced across the patch. The analysis indicated that the whole cell resistance (Ro) was approximately 40 G omega and the membrane capacitance (Co) was close to the standard value of 1 microF.cm-2. In addition, the threshold potential change necessary to initiate an action potential (Vth) was approximately 13 mV and a minimum current injection of 1 pA was required to depolarize the cell to spike threshold. When the smaller size of mammalian receptors are taken into account, membrane electrical properties were found to be consistent with those of salamander cells investigated by others using whole-cell recording. The analysis also revealed possible errors in the determination of single-channel conductances and reversal potentials by cell-attached recording from small cells.


The Journal of Neuroscience | 2010

Pathophysiological Mechanisms of Dominant and Recessive GLRA1 Mutations in Hyperekplexia

Seo-Kyung Chung; Jean-François Vanbellinghen; Jonathan G. L. Mullins; Angela Robinson; Janina Hantke; C. L. Hammond; Daniel F. Gilbert; Michael Freilinger; Monique M. Ryan; Michael C. Kruer; Amira Masri; Candan Gürses; Colin D. Ferrie; Kirsten Harvey; Rita Shiang; John Christodoulou; Frederick Andermann; Eva Andermann; Rhys Huw Thomas; Robert J. Harvey; Joseph W. Lynch; Mark I. Rees

Hyperekplexia is a rare, but potentially fatal, neuromotor disorder characterized by exaggerated startle reflexes and hypertonia in response to sudden, unexpected auditory or tactile stimuli. This disorder is primarily caused by inherited mutations in the genes encoding the glycine receptor (GlyR) α1 subunit (GLRA1) and the presynaptic glycine transporter GlyT2 (SLC6A5). In this study, systematic DNA sequencing of GLRA1 in 88 new unrelated human hyperekplexia patients revealed 19 sequence variants in 30 index cases, of which 21 cases were inherited in recessive or compound heterozygote modes. This indicates that recessive hyperekplexia is far more prevalent than previous estimates. From the 19 GLRA1 sequence variants, we have investigated the functional effects of 11 novel and 2 recurrent mutations. The expression levels and functional properties of these hyperekplexia mutants were analyzed using a high-content imaging system and patch-clamp electrophysiology. When expressed in HEK293 cells, either as homomeric α1 or heteromeric α1β GlyRs, subcellular localization defects were the major mechanism underlying recessive mutations. However, mutants without trafficking defects typically showed alterations in the glycine sensitivity suggestive of disrupted receptor function. This study also reports the first hyperekplexia mutation associated with a GlyR leak conductance, suggesting tonic channel opening as a new mechanism in neuronal ligand-gated ion channels.


Current Pharmaceutical Design | 2007

Molecular Pharmacology of the Glycine Receptor Chloride Channel

Timothy I. Webb; Joseph W. Lynch

The glycine receptor (GlyR) Cl(-) channel belongs to the cysteine-loop family of ligand-gated ion channel receptors. It is best known for mediating inhibitory neurotransmission in motor and sensory reflex circuits of the spinal cord, although glycinergic synapses are also present in the brain stem, cerebellum and retina. Extrasynaptic GlyRs are widely distributed throughout the central nervous system and they are also found in sperm and macrophages. A total of 5 GlyR subunits (alpha1-4 and beta) have been identified. Embryonic receptors comprise alpha2 homomers whereas adult receptors comprise predominantly alpha beta heteromers in a 2:3 stoichiometry. Notably, the alpha3 subunit is present in synaptic GlyRs that mediate inhibitory neurotransmission onto spinal nociceptive neurons. These receptors are specifically inhibited by inflammatory mediators, implying a role for alpha3-containing GlyRs in inflammatory pain sensitisation. Because molecules that increase GlyR current may have clinical potential as muscle relaxant and peripheral analgesic drugs, this review focuses on the molecular pharmacology of GlyR potentiating substances. Of all GlyR potentiating substances identified to date, we conclude that 5HT(3)R antagonists such as tropisetron offer the most promise as therapeutic lead compounds. However, one problem is that that virtually all known GlyR potentiating compounds, including tropisetron analogues, lack specificity for the GlyR. Another is that almost nothing is known about the pharmacological properties of alpha3-containing GlyRs, which is the subtype of choice for targeting by novel antinociceptive agents. These issues need to be addressed before GlyR-specific therapeutics can be developed.


Molecular Brain Research | 1996

The human glycine receptor β subunit : primary structure, functional characterisation and chromosomal localisation of the human and murine genes

Cheryl A. Handford; Joseph W. Lynch; Elizabeth Baker; Graham C. Webb; Judith H. Ford; Grant R. Sutherland; Peter R. Schofield

The inhibitory glycine receptor (GlyR) is a pentameric receptor comprised of alpha and beta subunits, of which the beta subunit has not been characterised in humans. A 2106 bp cDNA, isolated from a human hippocampal cDNA library, contained an open reading frame of 497 amino acids which encodes the beta subunit of the human GlyR. The mature human GlyR beta polypeptide displays 99% amino acid identity with the rat GlyR beta subunit and 48% identity with the human GlyR alpha 1 subunit. Neither [3H]strychnine binding nor glycine-gated currents were detected when the human GlyR beta subunit cDNA was expressed in the human embryonic kidney 293 cell line. However, co-expression of the beta subunit cDNA with the alpha 1 subunit cDNA resulted in expression of functional GlyRs which showed a 4-fold reduction in the EC50 values when compared to alpha 1 homomeric GlyRs. Glycine-gated currents of alpha 1/beta GlyRs were 17-fold less sensitive than homomeric alpha 1 GlyRs to the antagonists picrotoxin, picrotoxinin and picrotin, providing clear evidence that heteromeric alpha 1/beta GlyRs were expressed. The beta subunit appears to play a structural rather than ligand binding role in GlyR function. Fluorescence in situ hybridisation was used to localise the gene encoding the human GlyR beta subunit (GLRB) to chromosome 4q32, a position syntenic with mouse chromosome 3. In situ hybridisation using the human GlyR beta subunit cDNA showed that the murine GlyR beta subunit gene (Glrb) maps to the spastic (spa) locus on mouse chromosome 3 at bands E3-F1. This is consistent with the recent finding that a mutation in the murine GlyR beta subunit causes the spa phenotype. It also raises the possibility that mutations in the human beta subunit gene may cause inherited disorders of the startle response.


The Journal of Neuroscience | 2008

A Cation-π Interaction in the Binding Site of the Glycine Receptor Is Mediated by a Phenylalanine Residue

Stephan A. Pless; Kat S. Millen; Ariele P. Hanek; Joseph W. Lynch; Henry A. Lester; Sarah C. R. Lummis; Dennis A. Dougherty

Cys-loop receptor binding sites characteristically contain many aromatic amino acids. In nicotinic ACh and 5-HT3 receptors, a Trp residue forms a cation-π interaction with the agonist, whereas in GABAA receptors, a Tyr performs this role. The glycine receptor binding site, however, contains predominantly Phe residues. Homology models suggest that two of these Phe side chains, Phe159 and Phe207, and possibly a third, Phe63, are positioned such that they could contribute to a cation-π interaction with the primary amine of glycine. Here, we test this hypothesis by incorporation of a series of fluorinated Phe derivatives using unnatural amino acid mutagenesis. The data reveal a clear correlation between the glycine EC50 value and the cation-π binding ability of the fluorinated Phe derivatives at position 159, but not at positions 207 or 63, indicating a single cation-π interaction between glycine and Phe159. The data thus provide an anchor point for locating glycine in its binding site, and demonstrate for the first time a cation-π interaction between Phe and a neurotransmitter.

Collaboration


Dive into the Joseph W. Lynch's collaboration.

Top Co-Authors

Avatar

Peter H. Barry

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Peter R. Schofield

Neuroscience Research Australia

View shared research outputs
Top Co-Authors

Avatar

Sahil Talwar

University of Queensland

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Qiang Shan

University of Queensland

View shared research outputs
Top Co-Authors

Avatar

Timothy Lynagh

University of Copenhagen

View shared research outputs
Top Co-Authors

Avatar

Daniel F. Gilbert

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar

Pankaj Sah

University of Queensland

View shared research outputs
Researchain Logo
Decentralizing Knowledge