Network


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

Hotspot


Dive into the research topics where Evan M. Sloane is active.

Publication


Featured researches published by Evan M. Sloane.


Pain | 2005

Minocycline attenuates mechanical allodynia and proinflammatory cytokine expression in rat models of pain facilitation

Annemarie Ledeboer; Evan M. Sloane; Erin D. Milligan; Matthew G. Frank; John H. Mahony; Steven F. Maier; Linda R. Watkins

Activated glial cells (microglia and astroglia) in the spinal cord play a major role in mediating enhanced pain states by releasing proinflammatory cytokines and other substances thought to facilitate pain transmission. In the present study, we report that intrathecal administration of minocycline, a selective inhibitor of microglial cell activation, inhibits low threshold mechanical allodynia, as measured by the von Frey test, in two models of pain facilitation. In a rat model of neuropathic pain induced by sciatic nerve inflammation (sciatic inflammatory neuropathy, SIN), minocycline delayed the induction of allodynia in both acute and persistent paradigms. Moreover, minocycline was able to attenuate established SIN‐induced allodynia 1 day, but not 1 week later, suggesting a limited role of microglial activation in more perseverative pain states. Our data are consistent with a crucial role for microglial cells in initiating, rather than maintaining, enhanced pain responses. In a model of spinal immune activation by intrathecal HIV‐1 gp120, we show that the anti‐allodynic effects of minocycline are associated with decreased microglial activation, attenuated mRNA expression of interleukin‐1β (IL‐1β), tumor necrosis factor‐α (TNF‐α), IL‐1β‐converting enzyme, TNF‐α‐converting enzyme, IL‐1 receptor antagonist and IL‐10 in lumbar dorsal spinal cord, and reduced IL‐1β and TNF‐α levels in the CSF. In contrast, no significant effects of minocycline were observed on gp120‐induced IL‐6 and cyclooxygenase‐2 expression in spinal cord or CSF IL‐6 levels. Taken together these data highlight the importance of microglial activation in the development of exaggerated pain states.


European Journal of Neuroscience | 2005

Controlling pathological pain by adenovirally driven spinal production of the anti-inflammatory cytokine, interleukin-10.

Erin D. Milligan; Stephen J. Langer; Evan M. Sloane; Lin He; Julie Wieseler-Frank; Kevin A. O'Connor; David Martin; John Forsayeth; Steven F. Maier; Kirk W. Johnson; Raymond A. Chavez; Leslie A. Leinwand; Linda R. Watkins

Gene therapy for the control of pain has, to date, targeted neurons. However, recent evidence supports that spinal cord glia are critical to the creation and maintenance of pain facilitation through the release of proinflammatory cytokines. Because of the ability of interleukin‐10 (IL‐10) to suppress proinflammatory cytokines, we tested whether an adenoviral vector encoding human IL‐10 (AD‐h‐IL10) would block and reverse pain facilitation. Three pain models were examined, all of which are mediated by spinal pro‐inflammatory cytokines. Acute intrathecal administration of rat IL‐10 protein itself briefly reversed chronic constriction injury‐induced mechanical allodynia and thermal hyperalgesia. The transient reversal caused by IL‐10 protein paralleled the half‐life of human IL‐10 protein in the intrathecal space (t1/2 ∼ 2 h). IL‐10 gene therapy both prevented and reversed thermal hyperalgesia and mechanical allodynia, without affecting basal responses to thermal or mechanical stimuli. Extra‐territorial, as well as territorial, pain changes were reversed by this treatment. Intrathecal AD‐h‐IL10 injected over lumbosacral spinal cord led to elevated lumbosacral cerebrospinal fluid (CSF) levels of human IL‐10, with far less human IL‐10 observed in cervical CSF. In keeping with IL‐10s known anti‐inflammatory actions, AD‐h‐IL10 lowered CSF levels of IL‐1, relative to control AD. These studies support that this gene therapy approach provides an alternative to neuronally focused drug and gene therapies for clinical pain control.


Molecular Pain | 2005

Controlling neuropathic pain by adeno-associated virus driven production of the anti-inflammatory cytokine, interleukin-10

Erin D. Milligan; Evan M. Sloane; Stephen J. Langer; Pedro E. Cruz; Marucia Chacur; Leah Spataro; Julie Wieseler-Frank; Sayamwong E. Hammack; Steven F. Maier; Terence R. Flotte; John Forsayeth; Leslie A. Leinwand; Raymond A. Chavez; Linda R. Watkins

Despite many decades of drug development, effective therapies for neuropathic pain remain elusive. The recent recognition of spinal cord glia and glial pro-inflammatory cytokines as important contributors to neuropathic pain suggests an alternative therapeutic strategy; that is, targeting glial activation or its downstream consequences. While several glial-selective drugs have been successful in controlling neuropathic pain in animal models, none are optimal for human use. Thus the aim of the present studies was to explore a novel approach for controlling neuropathic pain. Here, an adeno-associated viral (serotype II; AAV2) vector was created that encodes the anti-inflammatory cytokine, interleukin-10 (IL-10). This anti-inflammatory cytokine is known to suppress the production of pro-inflammatory cytokines. Upon intrathecal administration, this novel AAV2-IL-10 vector was successful in transiently preventing and reversing neuropathic pain. Intrathecal administration of an AAV2 vector encoding beta-galactosidase revealed that AAV2 preferentially infects meningeal cells surrounding the CSF space. Taken together, these data provide initial support that intrathecal gene therapy to drive the production of IL-10 may prove to be an efficacious treatment for neuropathic pain.


Pain | 2006

Repeated intrathecal injections of plasmid DNA encoding interleukin-10 produce prolonged reversal of neuropathic pain

Erin D. Milligan; Evan M. Sloane; Stephen J. Langer; Travis S. Hughes; Brian M. Jekich; Matthew G. Frank; John H. Mahoney; Lindsay H. Levkoff; Steven F. Maier; Pedro E. Cruz; Terence R. Flotte; Kirk W. Johnson; Melissa M. Mahoney; Raymond A. Chavez; Leslie A. Leinwand; Linda R. Watkins

&NA; Neuropathic pain is a major clinical problem unresolved by available therapeutics. Spinal cord glia play a pivotal role in neuropathic pain, via the release of proinflammatory cytokines. Anti‐inflammatory cytokines, like interleukin‐10 (IL‐10), suppress proinflammatory cytokines. Thus, IL‐10 may provide a means for controlling glial amplification of pain. We recently documented that intrathecal IL‐10 protein resolves neuropathic pain, albeit briefly (˜2–3 h), given its short half‐life. Intrathecal gene therapy using viruses encoding IL‐10 can also resolve neuropathic pain, but for only ˜2 weeks. Here, we report a novel approach that dramatically increases the efficacy of intrathecal IL‐10 gene therapy. Repeated intrathecal delivery of plasmid DNA vectors encoding IL‐10 (pDNA‐IL‐10) abolished neuropathic pain for greater than 40 days. Naked pDNA‐IL‐10 reversed chronic constriction injury (CCI)‐induced allodynia both shortly after nerve injury as well as 2 months later. This supports that spinal proinflammatory cytokines are important in both the initiation and maintenance of neuropathic pain. Importantly, pDNA‐IL‐10 gene therapy reversed mechanical allodynia induced by CCI, returning rats to normal pain responsiveness, without additional analgesia. Together, these data suggest that intrathecal IL‐10 gene therapy may provide a novel approach for prolonged clinical pain control.


Pain | 2004

Peri-sciatic proinflammatory cytokines, reactive oxygen species, and complement induce mirror-image neuropathic pain in rats

Carin Twining; Evan M. Sloane; Erin D. Milligan; Marucia Chacur; David Martin; Stephen Poole; Henry Marsh; Steven F. Maier; Linda R. Watkins

&NA; In inflammatory neuropathy, immune activation near intact peripheral nerves induces mechanical allodynia. The identity of the peripheral immune product(s) that lead to these changes in pain behavior is unknown. The present series of studies utilized the sciatic inflammatory neuropathy (SIN) model to examine this question. Here, inflammatory neuropathy is created by injecting an immune activator (zymosan) around one sciatic nerve via an indwelling catheter. Our prior studies demonstrated that peri‐sciatic zymosan activated macrophages and neutrophils to release proinflammatory cytokines and reactive oxygen species (ROS). In addition, zymosan is a classical activator of the complement cascade. Thus the present series of experiments examined whether any of these inflammatory mediators are involved in the initial induction of SIN‐induced ipsilateral or bilateral allodynias. Peri‐sciatic injection of selective inhibitors/antagonists revealed that a number of immune products are early mediators of the resultant allodynias, including proinflammatory cytokines (tumor necrosis factor, interleukin‐1, and interleukin‐6), ROS, and complement. Thus these immune‐derived substances can markedly alter sensory nerve function at mid‐axon.


Journal of Neuroimmunology | 2008

Glia in pathological pain: a role for fractalkine.

Erin D. Milligan; Evan M. Sloane; Linda R. Watkins

Microglia and/or astrocytes play a significant role in the creation and maintenance of exaggerated pain states with inflammatory and/or neuropathic etiologies. The chemokine, fractalkine, has several functions, including the newly recognized role of mediating neuropathic pain conditions. Although constitutively expressed and released during inflammation, increased release of fractalkine binds to and activates microglia leading to pathological pain. We review the critical role of fractalkine in neuron-to-glial communication after peripheral nerve injury and inflammation and explore anti-inflammatory cytokines like interleukin-10 as a novel and effective approach for clinical pain control.


Neuroscience | 2010

Spinal upregulation of glutamate transporter GLT-1 by ceftriaxone: therapeutic efficacy in a range of experimental nervous system disorders

Khara M. Ramos; Makenzie T. Lewis; Kelly N. Morgan; Nicole Y. Crysdale; Juliet L. Kroll; Frederick R. Taylor; Jacqueline A. Harrison; Evan M. Sloane; Steven F. Maier; Linda R. Watkins

Glutamate neurotransmission is highly regulated, largely by glutamate transporters. In the spinal cord, the glutamate transporter GLT-1 is primarily responsible for glutamate clearance. Downregulation of GLT-1 can occur in activated astrocytes, and is associated with increased extracellular glutamate and neuroexcitation. Among other conditions, astrocyte activation occurs following repeated opioids and in models of chronic pain. If GLT-1 downregulation occurs in these states, GLT-1 could be a pharmacological target for improving opioid efficacy and controlling chronic pain. The present studies explored whether daily intrathecal treatment of rats with ceftriaxone, a beta-lactam antibiotic that upregulates GLT-1 expression, could prevent development of hyperalgesia and allodynia following repeated morphine, reverse pain arising from central or peripheral neuropathy, and reduce glial activation in these models. Ceftriaxone pre-treatment attenuated the development of hyperalgesia and allodynia in response to repeated morphine, and prevented associated astrocyte activation. In a model of multiple sclerosis (experimental autoimmune encephalomyelitis; EAE), ceftriaxone reversed tactile allodynia and halted the progression of motor weakness and paralysis. Similarly, ceftriaxone reversed tactile allodynia induced by chronic constriction nerve injury (CCI). EAE and CCI each significantly reduced the expression of membrane-bound, dimerized GLT-1 protein in lumbar spinal cord, an effect normalized by ceftriaxone. Lastly, ceftriaxone normalized CCI- and EAE-induced astrocyte activation in lumbar spinal cord. Together, these data indicate that increasing spinal GLT-1 expression attenuates opioid-induced paradoxical pain, alleviates neuropathic pain, and suppresses associated glial activation. GLT-1 therefore may be a therapeutic target that could improve available treatment options for patients with chronic pain.


Neuron Glia Biology | 2006

Intrathecal polymer-based interleukin-10 gene delivery for neuropathic pain

Erin D. Milligan; Ryan G. Soderquist; Stephanie M. Malone; John H. Mahoney; Travis S. Hughes; Stephen J. Langer; Evan M. Sloane; Steven F. Maier; Leslie A. Leinwand; Linda R. Watkins; Melissa J. Mahoney

Research on communication between glia and neurons has increased in the past decade. The onset of neuropathic pain, a major clinical problem that is not resolved by available therapeutics, involves activation of spinal cord glia through the release of proinflammatory cytokines in acute animal models of neuropathic pain. Here, we demonstrate for the first time that the spinal action of the proinflammatory cytokine, interleukin 1 (IL-1) is involved in maintaining persistent (2 months) allodynia induced by chronic-constriction injury (CCI). The anti-inflammatory cytokine IL-10 can suppress proinflammatory cytokines and spinal cord glial amplification of pain. Given that IL-1 is a key mediator of neuropathic pain, developing a clinically viable means of long-term delivery of IL-10 to the spinal cord is desirable. High doses of intrathecal IL-10-gene therapy using naked plasmid DNA (free pDNA-IL-10) is effective, but the dose required limits its potential clinical utility. Here we show that intrathecal gene therapy for neuropathic pain is improved sufficiently using two, distinct synthetic polymers, poly(lactic-co-glycolic) and polyethylenimine, that substantially lower doses of pDNA-IL-10 are effective. In conclusion, synthetic polymers used as i.t. gene-delivery systems are well-tolerated and improve the long-duration efficacy of pDNA-IL-10 gene therapy.


The Journal of Neuroscience | 2009

Enduring Reversal of Neuropathic Pain by a Single Intrathecal Injection of Adenosine 2A Receptor Agonists: A Novel Therapy for Neuropathic Pain

Lisa C. Loram; Jacqueline A. Harrison; Evan M. Sloane; Mark R. Hutchinson; Paige W. Sholar; Frederick R. Taylor; Debra Berkelhammer; Benjamen D. Coats; Stephen Poole; Erin D. Milligan; Steven F. Maier; Jayson Rieger; Linda R. Watkins

Previous studies of peripheral immune cells have documented that activation of adenosine 2A receptors (A2ARs) decrease proinflammatory cytokine release and increase release of the potent anti-inflammatory cytokine, interleukin-10 (IL-10). Given the growing literature supporting that glial proinflammatory cytokines importantly contribute to neuropathic pain and that IL-10 can suppress such pain, we evaluated the effects of intrathecally administered A2AR agonists on neuropathic pain using the chronic constriction injury (CCI) model. A single intrathecal injection of the A2AR agonists 4-(3-(6-amino-9-(5-cyclopropylcarbamoyl-3,4-dihydroxytetrahydrofuran-2-yl)-9H-purin-2-yl)prop-2-ynyl)piperidine-1-carboxylic acid methyl ester (ATL313) or 2-p-(2-carboxyethyl)phenethylamino-5′-N-ethylcarboxamido adenosine HCl (CGS21680), 10–14 d after CCI versus sham surgery, produced a long-duration reversal of mechanical allodynia and thermal hyperalgesia for at least 4 weeks. Neither drug altered the nociceptive responses of sham-operated controls. An A2AR antagonist [ZM241385 (4-(2-[7-amino-2-(2-furyl)(1,2,4)triazolo(2,3-a)(1,3,5)triazin-5-ylamino]ethyl)phenol)] coadministered intrathecally with ATL313 abolished the action of ATL313 in rats with neuropathy-induced allodynia but had no effect on allodynia in the absence of the A2AR agonist. ATL313 attenuated CCI-induced upregulation of spinal cord activation markers for microglia and astrocytes in the L4–L6 spinal cord segments both 1 and 4 weeks after a single intrathecal ATL313 administration. Neutralizing IL-10 antibodies administered intrathecally transiently abolished the effect of ATL313 on neuropathic pain. In addition, IL-10 mRNA was significantly elevated in the CSF cells collected from the lumbar region. Activation of A2ARs after intrathecal administration may be a novel, therapeutic approach for the treatment of neuropathic pain by increasing IL-10 in the immunocompetent cells of the CNS.


Brain Behavior and Immunity | 2009

Anti-inflammatory cytokine gene therapy decreases sensory and motor dysfunction in experimental Multiple Sclerosis: MOG-EAE behavioral and anatomical symptom treatment with cytokine gene therapy

Evan M. Sloane; Annemarie Ledeboer; W. Seibert; Benjamen D. Coats; M. van Strien; S.F. Maier; Kirk W. Johnson; Raymond A. Chavez; Linda R. Watkins; Leslie A. Leinwand; Erin D. Milligan; A.M. Van Dam

Multiple Sclerosis (MS) is an autoimmune inflammatory disease that presents clinically with a range of symptoms including motor, sensory, and cognitive dysfunction as well as demyelination and lesion formation in brain and spinal cord. A variety of animal models of MS have been developed that share many of the pathological hallmarks of MS including motor deficits (ascending paralysis), demyelination and axonal damage of central nervous system (CNS) tissue. In recent years, neuropathic pain has been recognized as a prevalent symptom of MS in a majority of patients. To date, there have been very few investigations into sensory disturbances in animal models of MS. The current work contains the first assessment of hind paw mechanical allodynia (von Frey test) over the course of a relapsing-remitting myelin oligodendrocyte glycoprotein induced experimental autoimmune encephalomyelitis (MOG-EAE) rat model of MS and establishes the utility of this model in examining autoimmune induced sensory dysfunction. We demonstrate periods of both decreased responsiveness to touch that precedes the onset of hind limb paralysis, and increased responsiveness (allodynia) that occurs during the period of motor deficit amelioration traditionally referred to as symptom remission. Furthermore, we tested the ability of our recently characterized anti-inflammatory IL-10 gene therapy to treat the autoimmune inflammation induced behavioral symptoms and tissue histopathological changes. This therapy is shown here to reverse inflammation induced paralysis, to reduce disease associated reduction in sensitivity to touch, to prevent the onset of allodynia, to reverse disease associated loss of body weight, and to suppress CNS glial activation associated with disease progression in this model.

Collaboration


Dive into the Evan M. Sloane's collaboration.

Top Co-Authors

Avatar

Linda R. Watkins

University of Colorado Boulder

View shared research outputs
Top Co-Authors

Avatar

Erin D. Milligan

University of Colorado Boulder

View shared research outputs
Top Co-Authors

Avatar

Steven F. Maier

University of Colorado Boulder

View shared research outputs
Top Co-Authors

Avatar

Leslie A. Leinwand

University of Colorado Boulder

View shared research outputs
Top Co-Authors

Avatar

John H. Mahoney

University of Colorado Boulder

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Stephen J. Langer

University of Colorado Boulder

View shared research outputs
Top Co-Authors

Avatar

Brian M. Jekich

University of Colorado Boulder

View shared research outputs
Top Co-Authors

Avatar

S.F. Maier

University of Colorado Boulder

View shared research outputs
Researchain Logo
Decentralizing Knowledge