Network


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

Hotspot


Dive into the research topics where Andelain Erickson is active.

Publication


Featured researches published by Andelain Erickson.


Scientific Reports | 2017

Multiple sodium channel isoforms mediate the pathological effects of Pacific ciguatoxin-1

Marco Inserra; Mathilde R. Israel; Ashlee Caldwell; Joel Castro; Jennifer R. Deuis; Andrea M. Harrington; Angelo Keramidas; Sonia Garcia-Caraballo; Jessica Maddern; Andelain Erickson; Luke Grundy; Grigori Y. Rychkov; Katharina Zimmermann; Richard J. Lewis; Stuart M. Brierley; Irina Vetter

Human intoxication with the seafood poison ciguatoxin, a dinoflagellate polyether that activates voltage-gated sodium channels (NaV), causes ciguatera, a disease characterised by gastrointestinal and neurological disturbances. We assessed the activity of the most potent congener, Pacific ciguatoxin-1 (P-CTX-1), on NaV1.1–1.9 using imaging and electrophysiological approaches. Although P-CTX-1 is essentially a non-selective NaV toxin and shifted the voltage-dependence of activation to more hyperpolarising potentials at all NaV subtypes, an increase in the inactivation time constant was observed only at NaV1.8, while the slope factor of the conductance-voltage curves was significantly increased for NaV1.7 and peak current was significantly increased for NaV1.6. Accordingly, P-CTX-1-induced visceral and cutaneous pain behaviours were significantly decreased after pharmacological inhibition of NaV1.8 and the tetrodotoxin-sensitive isoforms NaV1.7 and NaV1.6, respectively. The contribution of these isoforms to excitability of peripheral C- and A-fibre sensory neurons, confirmed using murine skin and visceral single-fibre recordings, reflects the expression pattern of NaV isoforms in peripheral sensory neurons and their contribution to membrane depolarisation, action potential initiation and propagation.


Journal of Immunology | 2017

G-CSF Receptor Blockade Ameliorates Arthritic Pain and Disease

Ming Chin Lee; James A. McCubbin; Anne D. Christensen; Daniel P. Poole; Pradeep Rajasekhar; TinaMarie Lieu; Nigel W. Bunnett; Sonia Garcia-Caraballo; Andelain Erickson; Stuart M. Brierley; Reem Saleh; Adrian Achuthan; Andrew J. Fleetwood; Robin L. Anderson; John A. Hamilton; Andrew D. Cook

G-CSF or CSF-3, originally defined as a regulator of granulocyte lineage development via its cell surface receptor (G-CSFR), can play a role in inflammation, and hence in many pathologies, due to its effects on mature lineage populations. Given this, and because pain is an extremely important arthritis symptom, the efficacy of an anti–G-CSFR mAb for arthritic pain and disease was compared with that of a neutrophil-depleting mAb, anti-Ly6G, in both adaptive and innate immune-mediated murine models. Pain and disease were ameliorated in Ag-induced arthritis, zymosan-induced arthritis, and methylated BSA/IL-1 arthritis by both prophylactic and therapeutic anti–G-CSFR mAb treatment, whereas only prophylactic anti-Ly6G mAb treatment was effective. Efficacy for pain and disease correlated with reduced joint neutrophil numbers and, importantly, benefits were noted without necessarily the concomitant reduction in circulating neutrophils. Anti–G-CSFR mAb also suppressed zymosan-induced inflammatory pain. A new G-CSF–driven (methylated BSA/G-CSF) arthritis model was established enabling us to demonstrate that pain was blocked by a cyclooxygenase-2 inhibitor, suggesting an indirect effect on neurons. Correspondingly, dorsal root ganglion neurons cultured in G-CSF failed to respond to G-CSF in vitro, and Csf3r gene expression could not be detected in dorsal root ganglion neurons by single-cell RT-PCR. These data suggest that G-CSFR/G-CSF targeting may be a safe therapeutic strategy for arthritis and other inflammatory conditions, particularly those in which pain is important, as well as for inflammatory pain per se.


The Journal of Physiology | 2018

Voltage‐gated sodium channels: (NaV)igating the field to determine their contribution to visceral nociception

Andelain Erickson; Annemie Deiteren; Andrea M. Harrington; Sonia Garcia-Caraballo; Joel Castro; Ashlee Caldwell; Luke Grundy; Stuart M. Brierley

Chronic visceral pain, altered motility and bladder dysfunction are common, yet poorly managed symptoms of functional and inflammatory disorders of the gastrointestinal and urinary tracts. Recently, numerous human channelopathies of the voltage‐gated sodium (NaV) channel family have been identified, which induce either painful neuropathies, an insensitivity to pain, or alterations in smooth muscle function. The identification of these disorders, in addition to the recent utilisation of genetically modified NaV mice and specific NaV channel modulators, has shed new light on how NaV channels contribute to the function of neuronal and non‐neuronal tissues within the gastrointestinal tract and bladder. Here we review the current pre‐clinical and clinical evidence to reveal how the nine NaV channel family members (NaV1.1–NaV1.9) contribute to abdominal visceral function in normal and disease states.


Toxins | 2017

Pain-Causing Venom Peptides: Insights into Sensory Neuron Pharmacology

Sina Jami; Andelain Erickson; Stuart M. Brierley; Irena Vetter

Venoms are produced by a wide variety of species including spiders, scorpions, reptiles, cnidarians, and fish for the purpose of harming or incapacitating predators or prey. While some venoms are of relatively simple composition, many contain hundreds to thousands of individual components with distinct pharmacological activity. Pain-inducing or “algesic” venom compounds have proven invaluable to our understanding of how physiological nociceptive neural networks operate. In this review, we present an overview of some of the diverse nociceptive pathways that can be modulated by specific venom components to evoke pain.


Pain | 2018

Tetrodotoxin-sensitive voltage-gated sodium channels regulate bladder afferent responses to distension

Luke Grundy; Andelain Erickson; Ashlee Caldwell; Sonia Garcia-Caraballo; Grigori Y. Rychkov; Andrea M. Harrington; Stuart M. Brierley

Abstract Interstitial cystitis/bladder pain syndrome (IC/BPS) is a prevalent, chronic bladder disorder that negatively impacts the quality of life for ∼5% of the western population. Hypersensitivity of mechanosensory afferents embedded within the bladder wall is considered a key component in mediating IC/BPS symptoms. Bladder infusion of voltage-gated sodium (Nav) channel blockers show clinical efficacy in treating IC/BPS symptoms; however, the current repertoire of Nav channels expressed by and contributing to bladder afferent function is unknown. We used single-cell reverse-transcription polymerase chain reaction of retrogradely traced bladder-innervating dorsal root ganglia (DRG) neurons to determine the expression profile of Nav channels, and patch-clamp recordings to characterise the contribution of tetrodotoxin-sensitive (TTX-S) and tetrodotoxin-resistant (TTX-R) Nav channels to total sodium current and neuronal excitability. We determined the TTX-S and TTX-R contribution to mechanosensitive bladder afferent responses ex vivo and spinal dorsal horn activation in vivo. Single-cell reverse-transcription polymerase chain reaction of bladder-innervating DRG neurons revealed significant heterogeneity in Nav channel coexpression patterns. However, TTX-S Nav channels contribute the vast majority of the total sodium current density and regulate the neuronal excitability of bladder DRG neurons. Furthermore, TTX-S Nav channels mediate almost all bladder afferent responses to distension. In vivo intrabladder infusion of TTX significantly reduces activation of dorsal horn neurons within the spinal cord to bladder distension. These data provide the first comprehensive analysis of Nav channel expression within sensory afferents innervating the bladder. They also demonstrate an essential role for TTX-S Nav channel regulation of bladder-innervating DRG neuroexcitability, bladder afferent responses to distension, and nociceptive signalling to the spinal cord.


JCI insight | 2018

NaV1.1 inhibition can reduce visceral hypersensitivity

Juan Salvatierra; Joel Castro; Andelain Erickson; Qian Li; João M. Bráz; John Gilchrist; Luke Grundy; Grigori Y. Rychkov; Annemie Deiteren; Rana Rais; Glenn F. King; Barbara S. Slusher; Allan I. Basbaum; Pankaj J. Pasricha; Stuart M. Brierley; Frank Bosmans

Functional bowel disorder patients can suffer from chronic abdominal pain, likely due to visceral hypersensitivity to mechanical stimuli. As there is only a limited understanding of the basis of chronic visceral hypersensitivity (CVH), drug-based management strategies are ill defined, vary considerably, and include NSAIDs, opioids, and even anticonvulsants. We previously reported that the 1.1 subtype of the voltage-gated sodium (NaV; NaV1.1) channel family regulates the excitability of sensory nerve fibers that transmit a mechanical pain message to the spinal cord. Herein, we investigated whether this channel subtype also underlies the abdominal pain that occurs with CVH. We demonstrate that NaV1.1 is functionally upregulated under CVH conditions and that inhibiting channel function reduces mechanical pain in 3 mechanistically distinct mouse models of chronic pain. In particular, we use a small molecule to show that selective NaV1.1 inhibition (a) decreases sodium currents in colon-innervating dorsal root ganglion neurons, (b) reduces colonic nociceptor mechanical responses, and (c) normalizes the enhanced visceromotor response to distension observed in 2 mouse models of irritable bowel syndrome. These results provide support for a relationship between NaV1.1 and chronic abdominal pain associated with functional bowel disorders.


The International Journal of Biochemistry & Cell Biology | 2018

Contribution of membrane receptor signalling to chronic visceral pain

Mahsa Sadeghi; Andelain Erickson; Joel Castro; Annemie Deiteren; Andrea M. Harrington; Luke Grundy; David J. Adams; Stuart M. Brierley

Irritable bowel syndrome and inflammatory bowel disease are major forms of chronic visceral pain, which affect over 15% of the global population. In order to identify new therapies, it is important to understand the underlying causes of chronic visceral pain. This review provides recent evidence demonstrating that inflammation or infection of the gastrointestinal tract triggers specific changes in the neuronal excitability of sensory pathways responsible for the transmission of nociceptive information from the periphery to the central nervous system. Specific changes in the expression and function of a variety of ion channels and receptors have been documented in inflammatory and chronic visceral pain conditions relevant to irritable bowel syndrome and inflammatory bowel disease. An increase in pro-nociceptive mechanisms enhances peripheral drive from the viscera and provides an underlying basis for enhanced nociceptive signalling during chronic visceral pain states. Recent evidence also highlights increases in anti-nociceptive mechanisms in models of chronic visceral pain, which present novel targets for pharmacological treatment of this condition.


Archive | 2018

Extrinsic Sensory Afferent Nerves Innervating the Gastrointestinal Tract in Health and Disease

Andrea M. Harrington; Joel Castro; Andelain Erickson; Luke Grundy; Stuart M. Brierley

Abstract Our gastrointestinal tract is innervated by specific classes of extrinsic sensory afferents, which follow either vagal or spinal pathways. Vagal afferents have cell bodies located within the nodose or jugular ganglia and project centrally to the brainstem. In contrast, spinal afferents have cell bodies in thoracic or sacral dorsal root ganglia and project centrally to the dorsal horn of the spinal cord. Together, these afferent pathways detect ongoing changes in the physical and chemical environment within the lumen and wall of our gastrointestinal tract. Consequently, these afferents convey the full spectrum of information that underlies the initiation of autonomic reflex control and all manner of visceral sensations, including hunger, fullness and urge. In pathological states these afferents also represent the underlying basis of ongoing symptoms such as bloating, discomfort and chronic visceral pain. This chapter will map out the anatomy and function of specific classes of sensory afferents innervating the gastrointestinal tract. Furthermore, it will highlight the key molecular mechanisms underlying the hypersensitivity of extrinsic sensory afferents innervating the gastrointestinal tract, which provides potential opportunities for the therapeutic treatment of gastrointestinal disorders.


The Journal of Physiology | 2018

Voltage-gated sodium channels: (NaV)igating the field to determine their contribution to visceral nociception: NaV channels and visceral pain

Andelain Erickson; Annemie Deiteren; Andrea M. Harrington; Sonia Garcia-Caraballo; Joel Castro; Ashlee Caldwell; Luke Grundy; Stuart M. Brierley


Venom to Drugs Conference 2014 | 2014

Discovery of voltage-gated sodium channel inhibitors from tarantula venoms

Fernanda C. Cardoso; Zoltan Dekan; Raveendra Anangi; K. J. Rosengren; Andelain Erickson; Irina Vetter; Volker Herzig; Paul F. Alewood; Glenn F. King; Richard J. Lewis

Collaboration


Dive into the Andelain Erickson's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joel Castro

University of Adelaide

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Glenn F. King

University of Queensland

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge