Network


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

Hotspot


Dive into the research topics where Erica A. Dale is active.

Publication


Featured researches published by Erica A. Dale.


Journal of Applied Physiology | 2012

Severe acute intermittent hypoxia elicits phrenic long-term facilitation by a novel adenosine-dependent mechanism

Nicole L. Nichols; Erica A. Dale; Gordon S. Mitchell

Acute intermittent hypoxia [AIH; 3, 5-min episodes; 35-45 mmHg arterial PO(2) (Pa(O(2)))] elicits serotonin-dependent phrenic long-term facilitation (pLTF), a form of phrenic motor facilitation (pMF) initiated by G(q) protein-coupled metabotropic 5-HT(2) receptors. An alternate pathway to pMF is induced by G(s) protein-coupled metabotropic receptors, including adenosine A(2A) receptors. AIH-induced pLTF is dominated by the serotonin-dependent pathway and is actually restrained via inhibition from the adenosine-dependent pathway. Here, we hypothesized that severe AIH shifts pLTF from a serotonin-dependent to an adenosine-dependent form of pMF. pLTF induced by severe (25-30 mmHg Pa(O(2))) and moderate (45-55 mmHg Pa(O(2))) AIH were compared in anesthetized rats, with and without intrathecal (C4) spinal A(2A) (MSX-3, 130 ng/kg, 12 μl) or 5-HT receptor antagonist (methysergide, 300 μg/kg, 15 μl) injections. During severe, but not moderate AIH, progressive augmentation of the phrenic response during hypoxic episodes was observed. Severe AIH (78% ± 8% 90 min post-AIH, n = 6) elicited greater pLTF vs. moderate AIH (41% ± 12%, n = 8; P < 0.05). MSX-3 (28% ± 6%; n = 6; P < 0.05) attenuated pLTF following severe AIH, but enhanced pLTF following moderate AIH (86% ± 26%; n = 8; P < 0.05). Methysergide abolished pLTF after moderate AIH (12% ± 5%; n = 6; P = 0.035), but had no effect after severe AIH (66 ± 13%; n = 5; P > 0.05). Thus severe AIH shifts pLTF from a serotonin-dependent to an adenosine-dependent mechanism; the adenosinergic pathway inhibits the serotonergic pathway following moderate AIH. Here we demonstrate a novel adenosine-dependent pathway to pLTF following severe AIH. Shifts in the mechanisms of respiratory plasticity provide the ventilatory control system greater flexibility as challenges that differ in severity are confronted.


American Journal of Respiratory and Critical Care Medicine | 2013

Intermittent Hypoxia and Stem Cell Implants Preserve Breathing Capacity in a Rodent Model of Amyotrophic Lateral Sclerosis

Nicole L. Nichols; Genevieve Gowing; Irawan Satriotomo; Lisa J. Nashold; Erica A. Dale; Masatoshi Suzuki; Pablo Avalos; Patrick Mulcrone; Jacalyn McHugh; Clive N. Svendsen; Gordon S. Mitchell

RATIONALE Amyotrophic lateral sclerosis (ALS) is a devastating motor neuron disease causing paralysis and death from respiratory failure. Strategies to preserve and/or restore respiratory function are critical for successful treatment. Although breathing capacity is maintained until late in disease progression in rodent models of familial ALS (SOD1(G93A) rats and mice), reduced numbers of phrenic motor neurons and decreased phrenic nerve activity are observed. Decreased phrenic motor output suggests imminent respiratory failure. OBJECTIVES To preserve or restore phrenic nerve activity in SOD1(G93A) rats at disease end stage. METHODS SOD1(G93A) rats were injected with human neural progenitor cells (hNPCs) bracketing the phrenic motor nucleus before disease onset, or exposed to acute intermittent hypoxia (AIH) at disease end stage. MEASUREMENTS AND MAIN RESULTS The capacity to generate phrenic motor output in anesthetized rats at disease end stage was: (1) transiently restored by a single presentation of AIH; and (2) preserved ipsilateral to hNPC transplants made before disease onset. hNPC transplants improved ipsilateral phrenic motor neuron survival. CONCLUSIONS AIH-induced respiratory plasticity and stem cell therapy have complementary translational potential to treat breathing deficits in patients with ALS.


The Journal of Neuroscience | 2012

Cervical Spinal Erythropoietin Induces Phrenic Motor Facilitation via Extracellular Signal-Regulated Protein Kinase and Akt Signaling

Erica A. Dale; Irawan Satriotomo; Gordon S. Mitchell

Erythropoietin (EPO) is typically known for its role in erythropoiesis but is also a potent neurotrophic/neuroprotective factor for spinal motor neurons. Another trophic factor regulated by hypoxia-inducible factor-1, vascular endothelial growth factor (VEGF), signals via ERK and Akt activation to elicit long-lasting phrenic motor facilitation (pMF). Because EPO also signals via ERK and Akt activation, we tested the hypothesis that EPO elicits similar pMF. Using retrograde labeling and immunohistochemical techniques, we demonstrate in adult, male, Sprague Dawley rats that EPO and its receptor, EPO-R, are expressed in identified phrenic motor neurons. Intrathecal EPO at C4 elicits long-lasting pMF; integrated phrenic nerve burst amplitude increased >90 min after injection (63 ± 12% baseline 90 min after injection; p < 0.001). EPO increased phosphorylation (and presumed activation) of ERK (1.6-fold vs controls; p < 0.05) in phrenic motor neurons; EPO also increased pAkt (1.6-fold vs controls; p < 0.05). EPO-induced pMF was abolished by the MEK/ERK inhibitor U0126 [1,4-diamino-2,3-dicyano-1,4-bis(o-aminophenylmercapto)butadiene] and the phosphatidylinositol 3-kinase/Akt inhibitor LY294002 [2-(4-morpholinyl)-8-phenyl-1(4H)-benzopyran-4-one], demonstrating that ERK MAP kinases and Akt are both required for EPO-induced pMF. Pretreatment with U0126 and LY294002 decreased both pERK and pAkt in phrenic motor neurons (p < 0.05), indicating a complex interaction between these kinases. We conclude that EPO elicits spinal plasticity in respiratory motor control. Because EPO expression is hypoxia sensitive, it may play a role in respiratory plasticity in conditions of prolonged or recurrent low oxygen.


The Journal of Neuroscience | 2015

Phrenic Long-Term Facilitation Requires PKCθ Activity within Phrenic Motor Neurons

Michael J. Devinney; Daryl P. Fields; Adrianne G. Huxtable; Timothy J. Peterson; Erica A. Dale; Gordon S. Mitchell

Acute intermittent hypoxia (AIH) induces a form of spinal motor plasticity known as phrenic long-term facilitation (pLTF); pLTF is a prolonged increase in phrenic motor output after AIH has ended. In anesthetized rats, we demonstrate that pLTF requires activity of the novel PKC isoform, PKCθ, and that the relevant PKCθ is within phrenic motor neurons. Whereas spinal PKCθ inhibitors block pLTF, inhibitors targeting other PKC isoforms do not. PKCθ is highly expressed in phrenic motor neurons, and PKCθ knockdown with intrapleural siRNAs abolishes pLTF. Intrapleural siRNAs targeting PKCζ, an atypical PKC isoform expressed in phrenic motor neurons that underlies a distinct form of phrenic motor plasticity, does not affect pLTF. Thus, PKCθ plays a critical role in spinal AIH-induced respiratory motor plasticity, and the relevant PKCθ is localized within phrenic motor neurons. Intrapleural siRNA delivery has considerable potential as a therapeutic tool to selectively manipulate plasticity in vital respiratory motor neurons.


Journal of Applied Physiology | 2014

Adrenergic α1 receptor activation is sufficient, but not necessary for phrenic long-term facilitation

Adrianne G. Huxtable; P.M. MacFarlane; S. Vinit; Nicole L. Nichols; Erica A. Dale; Gordon S. Mitchell

Acute intermittent hypoxia (AIH; three 5-min hypoxic episodes) causes a form of phrenic motor facilitation (pMF) known as phrenic long-term facilitation (pLTF); pLTF is initiated by spinal activation of Gq protein-coupled 5-HT2 receptors. Because α1 adrenergic receptors are expressed in the phrenic motor nucleus and are also Gq protein-coupled, we hypothesized that α1 receptors are sufficient, but not necessary for AIH-induced pLTF. In anesthetized, paralyzed, and ventilated rats, episodic spinal application of the α1 receptor agonist phenylephrine (PE) elicited dose-dependent pMF (10 and 100 μM, P < 0.05; but not 1 μM). PE-induced pMF was blocked by the α1 receptor antagonist prazosin (1 mM; -20 ± 20% at 60 min, -5 ± 21% at 90 min; n = 6). Although α1 receptor activation is sufficient to induce pMF, it was not necessary for AIH-induced pLTF because intrathecal prazosin (1 mM) did not alter AIH-induced pLTF (56 ± 9% at 60 min, 78 ± 12% at 90 min; n = 9). Intravenous (iv) prazosin (150 μg/kg) appeared to reduce pLTF (21 ± 9% at 60 min, 26 ± 8% at 90 min), but this effect was not significant. Hypoglossal long-term facilitation was unaffected by intrathecal prazosin, but was blocked by iv prazosin (-4 ± 14% at 60 min, -13 ± 18% at 90 min), suggesting different LTF mechanisms in different motor neuron pools. In conclusion, Gq protein-coupled α1 adrenergic receptors evoke pMF, but they are not necessary for AIH-induced pLTF.


Respiratory Physiology & Neurobiology | 2013

Spinal vascular endothelial growth factor (VEGF) and erythropoietin (EPO) induced phrenic motor facilitation after repetitive acute intermittent hypoxia.

Erica A. Dale; Gordon S. Mitchell

Vascular endothelial growth factor (VEGF) and erythropoietin (EPO) exert neurotrophic and neuroprotective effects in the CNS. We recently demonstrated that VEGF, EPO and their receptors (VEGF-R2, EPO-R) are expressed in phrenic motor neurons, and that cervical spinal VEGF-R2 and EPO-R activation elicit long-lasting phrenic motor facilitation (pMF). Since VEGF, VEGF-R, EPO, and EPO-R are hypoxia-regulated genes, and repetitive exposure to acute intermittent hypoxia (rAIH) up-regulates these molecules in phrenic motor neurons, we tested the hypothesis that 4 weeks of rAIH (10 episodes per day, 3 days per week) enhances VEGF- or EPO-induced pMF. We confirm that cervical spinal VEGF and EPO injections elicit pMF. However, neither VEGF- nor EPO-induced pMF was affected by rAIH pre-conditioning (4 wks). Although our data confirm that spinal VEGF and EPO may play an important role in respiratory plasticity, we provide no evidence that rAIH amplifies their impact. Further experiments with more robust protocols are warranted.


Experimental Neurology | 2017

Phrenic motor neuron TrkB expression is necessary for acute intermittent hypoxia-induced phrenic long-term facilitation

Erica A. Dale; Daryl P. Fields; Michael J. Devinney; Gordon S. Mitchell

ABSTRACT Phrenic long‐term facilitation (pLTF) is a form of hypoxia‐induced spinal respiratory motor plasticity that requires new synthesis of brain derived neurotrophic factor (BDNF) and activation of its high‐affinity receptor, tropomyosin receptor kinase B (TrkB). Since the cellular location of relevant TrkB receptors is not known, we utilized intrapleural siRNA injections to selectively knock down TrkB receptor protein within phrenic motor neurons. TrkB receptors within phrenic motor neurons are necessary for BDNF‐dependent acute intermittent hypoxia‐induced pLTF, demonstrating that phrenic motor neurons are a critical site of respiratory motor plasticity. HighlightsAcute intermittent hypoxia‐induced respiratory motor plasticity requires BDNF signaling.Phrenic motor neurons express the high affinity BDNF receptor TrkB.Intrapleural siRNA targeting TrkB receptors decreased TrkB receptor expression selectively on phrenic motor neurons.Intrapleural siRNA targeting TrkB receptors abolished BDNF dependent, hypoxia‐induced respiratory motor plasticity.Phrenic motor neurons are a critical site for spinal respiratory motor plasticity.


Neuroscience | 2016

Repetitive acute intermittent hypoxia increases growth/neurotrophic factor expression in non-respiratory motor neurons

Irawan Satriotomo; Nicole L. Nichols; Erica A. Dale; A.T. Emery; Jenny Marie Dahlberg; Gordon S. Mitchell

Repetitive acute intermittent hypoxia (rAIH) increases growth/trophic factor expression in respiratory motor neurons, thereby eliciting spinal respiratory motor plasticity and/or neuroprotection. Here we demonstrate that rAIH effects are not unique to respiratory motor neurons, but are also expressed in non-respiratory, spinal alpha motor neurons and upper motor neurons of the motor cortex. In specific, we used immunohistochemistry and immunofluorescence to assess growth/trophic factor protein expression in spinal sections from rats exposed to AIH three times per week for 10weeks (3×wAIH). 3×wAIH increased brain-derived neurotrophic factor (BDNF), its high-affinity receptor, tropomyosin receptor kinase B (TrkB), and phosphorylated TrkB (pTrkB) immunoreactivity in putative alpha motor neurons of spinal cervical 7 (C7) and lumbar 3 (L3) segments, as well as in upper motor neurons of the primary motor cortex (M1). 3×wAIH also increased immunoreactivity of vascular endothelial growth factor A (VEGFA), the high-affinity VEGFA receptor (VEGFR-2) and an important VEGF gene regulator, hypoxia-inducible factor-1α (HIF-1α). Thus, rAIH effects on growth/trophic factors are characteristic of non-respiratory as well as respiratory motor neurons. rAIH may be a useful tool in the treatment of disorders causing paralysis, such as spinal injury and motor neuron disease, as a pretreatment to enhance motor neuron survival during disease, or as preconditioning for cell-transplant therapies.


Archive | 2014

Therapeutic Potential of Intermittent Hypoxia: Lessons from Respiratory Motor Plasticity

Angela Navarrete-Opazo; Erica A. Dale; Gordon S. Mitchell

Intermittent hypoxia (IH) is a subject of considerable interest since it has both beneficial and adverse effects. Unfortunately, a lack of consistency in the use of the term “intermittent hypoxia” has led to considerable confusion in the field. In reviewing available literature, the physiological and pathological impact of IH appears to be highly associated with the effective IH “dose.” IH consisting of modest hypoxic episodes (≥9 % inspired O2) and lesser numbers of hypoxia/reoxygenation events per day (≤15 cycles/day) is generally associated with beneficial effects in multiple body systems. In contrast, severe hypoxic episodes (<9 % inspired oxygen) and more frequent hypoxic episodes per day (40–2,400 cycles/day) shift the balance towards morbidity. In accordance, the impact of IH on the neural system controlling breathing is critically dependent on variables including the pattern of hypoxia (intermittent versus sustained), the severity of hypoxia within episodes, and the overall duration of IH exposure (minutes to years). A low IH “dose” (few episodes, moderate hypoxia) elicits serotonin-dependent spinal, respiratory motor plasticity that may be harnessed as a therapeutic approach to improve respiratory function in clinical conditions that impair breathing, such as cervical spinal injury. With a similar protocol but more severe hypoxic episodes, a distinct adenosine-dependent mechanism of spinal respiratory motor plasticity is observed. The cumulative effectives of repeated, low-dose IH (metaplasticity) suggest that repetitive, acute IH may represent a simple, safe, and effective treatment to promote meaningful therapeutic benefit in a range of clinical conditions that compromise respiratory (and nonrespiratory) somatic motor function.


Experimental Neurology | 2012

Repetitive acute intermittent hypoxia increases expression of proteins associated with plasticity in the phrenic motor nucleus.

Irawan Satriotomo; Erica A. Dale; Jenny Marie Dahlberg; Gordon S. Mitchell

Collaboration


Dive into the Erica A. Dale's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Irawan Satriotomo

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Nicole L. Nichols

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Clive N. Svendsen

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Adrianne G. Huxtable

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Angela Navarrete-Opazo

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Daryl P. Fields

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Jenny Marie Dahlberg

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Lisa J. Nashold

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Michael J. Devinney

University of Wisconsin-Madison

View shared research outputs
Researchain Logo
Decentralizing Knowledge