Network


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

Hotspot


Dive into the research topics where Jason M. Moreau is active.

Publication


Featured researches published by Jason M. Moreau.


Brain Research | 2012

Intermittent hypoxia and systemic leptin administration induces pSTAT3 and Fos/Fra-1 in the carotid body

Scott A. Messenger; Jason M. Moreau; John Ciriello

Glomus cells within the carotid body are known to respond to hypoxic stimuli. Recently, these cells have been shown to express the long form of the leptin receptor (Ob-Rb). However, whether these glomus cells expressing the Ob-Rb are activated by hypoxic stimuli is not known. Therefore, in this study we investigated whether intermittent hypoxia (IH) or changes in circulating levels of leptin induced phosphorylated signal transducer and activator of transcription 3 (pSTAT3), the immediate early gene c-fos protein, or fos-related antigen-1 protein (Fra-1) within carotid body glomus cells that expressed the Ob-Rb, and within neurons of the petrosal (PG) and nodose (NG) ganglia. Rats were subjected to IH (120 s normoxia, 80s hypoxia for 8h) or normoxia (8h), or intravenous injections of leptin (50 or 200 ng/0.1 mL) or the vehicle saline. Plasma leptin levels were measured in animals exposed to IH and normoxia. Exposure to 8h of IH increased plasma leptin levels greater than 2-fold compared to normoxic controls. Animals were then perfused with Zambonis fixative, and the region of the carotid bifurcation containing the carotid body and PG/NG complex was removed, paraffin embedded and sectioned at 6 μm for immunohistochemical processing. Carotid body glomus cells were identified by their expression of tyrosine hydroxylase immunoreactivity. These glomus cells also expressed the OB-Rb and were found to express pSTAT3-, fos-, and Fra-1-like immunoreactivity in response to both IH and systemic leptin injections. IH and leptin injections also increased fos and Fra-1 like expression in the PG, NG and jugular ganglion. Taken together, these data suggest IH alters circulating leptin which in turn activates directly carotid body glomus cells to exert a modulatory effect on the peripheral chemoreceptor reflex.


American Journal of Physiology-regulatory Integrative and Comparative Physiology | 2012

Leptin signaling in the nucleus of the solitary tract alters the cardiovascular responses to activation of the chemoreceptor reflex

John Ciriello; Jason M. Moreau

Circulating levels of leptin are elevated in individuals suffering from chronic intermittent hypoxia (CIH). Systemic and central administration of leptin elicits increases in sympathetic nervous activity (SNA), arterial pressure (AP), and heart rate (HR), and it attenuates the baroreceptor reflex, cardiovascular responses that are similar to those observed during CIH as a result of activation of chemoreceptors by the systemic hypoxia. Therefore, experiments were done in anesthetized Wistar rats to investigate the effects of leptin in nucleus of the solitary tract (NTS) on AP and HR responses, and renal SNA (RSNA) responses during activation of NTS neurons and the chemoreceptor reflex. Microinjection of leptin (5-100 ng; 20 nl) into caudal NTS pressor sites (l-glutamate; l-Glu; 0.25 M; 10 nl) elicited dose-related increases in AP, HR, and RSNA. Leptin microinjections (5 ng; 20 nl) into these sites potentiated the increase in AP and HR elicited by l-Glu. Additionally, bilateral injections of leptin (5 ng; 100 nl) into NTS potentiated the increase in AP and attenuated the bradycardia to systemic activation of the chemoreflex. In the Zucker obese rat, leptin injections into NTS neither elicited cardiovascular responses nor altered the cardiovascular responses to activation of the chemoreflex. Taken together, these data indicate that leptin exerts a modulatory effect on neuronal circuits within NTS that control cardiovascular responses elicited during the reflex activation of arterial chemoreceptors and suggest that increased AP and SNA observed in individuals with CIH may be due, in part, by leptins effects on the chemoreflex at the level of NTS.


Neuroscience | 2013

Systemic administration of leptin potentiates the response of neurons in the nucleus of the solitary tract to chemoreceptor activation in the rat

John Ciriello; Jason M. Moreau

Leptin microinjections into the nucleus of the solitary tract (NTS) have been shown to elicit sympathoexcitatory responses, and potentiate the cardiovascular responses to activation of the chemoreflex. In this study, experiments were done in Sprague-Dawley rats initially to provide a detailed mapping within the NTS complex of cells containing immunoreactivity to the long form of the leptin receptor (Ob-Rb). In a second series, this NTS region containing Ob-Rb immunoreactive cells was explored for single units antidromically activated by stimulation of pressor sites in the rostral ventrolateral medulla (RVLM). These antidromically identified neurons were then tested for their response to intra-carotid injections of leptin (50-100 ng/0.1 ml), and to activation of peripheral chemoreceptors following an injection of potassium cyanide (KCN) (80 μg/0.1 ml) into the carotid artery. Cells containing Ob-Rb-like immunoreactivity were found predominantly in the caudal NTS: within the medial, commissural and gelatinous (sub-postremal area) subnuclei of the NTS complex. Of 73 single units tested in these NTS regions, 48 were antidromically activated by stimulation of RVLM pressor sites and 25 of these single units responded with an increase in discharge rate after intra-carotid injections of leptin. In addition, 17 of these leptin responsive neurons were excited by the intra-carotid injections of KCN (80 μg/0.1 ml). Furthermore, the excitatory response of these single units to KCN was potentiated (59-83%) immediately following the leptin injection. These data indicate that leptin responsive neurons in NTS mediate chemoreceptor afferent information to pressor sites in the RVLM, and suggest that leptin may act as a facilitator on neuronal circuits within the NTS that potentiates the sympathoexcitatory responses elicited during the reflex activation of arterial chemoreceptors.


Neuroscience | 2013

Effects of acute intermittent hypoxia on energy balance and hypothalamic feeding pathways.

Jason M. Moreau; John Ciriello

This study was done to investigate the effects of acute intermittent hypoxia (IH) on metabolic factors associated with energy balance and body weight, and on hypothalamic satiety-inducing pathways. Adult male Sprague-Dawley rats were exposed to either 8h IH or normoxic control conditions. Food intake, locomotion and body weights were examined after IH. Additionally, plasma levels of leptin, adiponectin corticosterone, insulin and blood glucose were measured following exposure to IH. Furthermore, adipose tissue was removed and analyzed for leptin and adiponectin content. Finally, the hypothalamic arcuate nucleus (ARC) was assessed for alterations in protein signaling associated with satiety. IH reduced body weight, food intake and active cycle locomotion without altering adipose tissue mass. Leptin protein content was reduced while adiponectin content was elevated in adipose tissue after IH. Plasma concentration of leptin was significantly increased while adiponectin decreased after IH. No changes were found in plasma corticosterone, insulin and blood glucose. In ARC, phosphorylation of signal transducer and activator of transcription-3 and pro-opiomelanocortin (POMC) expression were elevated. In addition, POMC-expressing neurons were activated as determined by immediate early gene FRA-1/2 expression. Finally, ERK1/2 and its phosphorylation were reduced in response to IH. These data suggest that IH induces significant alterations to body energy balance through changes in the secretion of leptin which exert effects on satiety-inducing pathways within the hypothalamus.


Brain Research | 2013

Effect of chronic intermittent hypoxia on leptin and leptin receptor protein expression in the carotid body.

Scott A. Messenger; Jason M. Moreau; John Ciriello

This study was done to investigate whether chronic intermittent hypoxia (CIH) induced changes in leptin and leptin receptor protein levels, and known downstream mediators of leptin receptor signaling in the carotid body. Rats were subjected to CIH (120s normoxia, 80s hypoxia) or normoxia for 8h/day to either short term (7 days) or long term CIH exposure (95 days). After both 7 and 95 days of CIH, carotid body leptin protein expression was decreased, while protein levels of the long form leptin receptor (OB-Rb) were elevated. On the other hand, protein expression levels of the short form leptin receptor (OB-R100) were unchanged. Furthermore, phosphorylated signal transducer and activator of transcription 3 (pSTAT3) protein levels were found to be significantly decreased at only the 7 day period. On the other hand, suppressor of cytokine signaling 3 (SOCS3) protein levels were elevated at only the 7 day period, while phosphorylated extracellular-signal-regulated kinase 1/2 (pERK1/2) was elevated only at the 95 day period. In both the normoxia and the CIH groups, carotid body leptin was decreased at the 95 day period compared to 7 days. However, OB-Rb or Ob-R100 protein levels were not changed in the normoxic or CIH group at either time point. Furthermore, pSTAT3 protein levels were found to be significantly higher, while SOCS3 levels were significantly lower in the 95 day CIH group compared to the 7 day CIH group. Taken together, these data indicate that CIH induces changes in leptin and leptin downstream signaling proteins within the carotid bodies which may contribute to alterations in carotid chemoreceptor sensitivity.


Behavioural Brain Research | 2013

Nesfatin-1 induces Fos expression and elicits dipsogenic responses in subfornical organ

Jason M. Moreau; John Ciriello

Nesfatin-1 (Nes-1), an 82-amino acid protein cleaved from nucleobindin-2, has been suggested to play a role in ingestive behaviors. Intracerebroventricular (icv) injections of Nes-1 reduce water intake, although the sites of action for this effect are not known. Two series of experiments were done to identify potential sites of action of Nes-1 in drinking behavior. In the first series, icv injections of Nes-1 were made in urethane-anesthetized rats to investigate the distribution of neurons containing Fos-like immunoreactivity (Fos-ir) within the forebrain. Circumventricular organs, including subfornical organ (SFO), were found to contain neurons expressing Fos-ir. Additionally, several hypothalamic, thalamic and limbic nuclei also contained Fos-labeled neurons. As SFO is a pivotal central site in the regulation of water intake, a second series of experiments was done to investigate the role of direct injections of Nes-1 into SFO on water intake in conscious, freely moving rats. Nes-1 (2pmol) injections into SFO induced an increase in water intake compared to vehicle injections. However, when food was made available for ingestion after the Nes-1 injection, the dipsogenic effects of Nes-1 were attenuated. Additionally, the drinking response to Nes-1 was found to be more potent than that observed after injections of ANG II into SFO. Neither simultaneous injections ANG II nor the ANG II type-1 receptor blocker losartan affected the Nes-1 dipsogenic response. Taken together, these results suggest that Nes-1 is a potent dipsogenic agent in SFO, and that Nes-1 may act independently of the SFO angiotensinergic system to elicit the dipsogenic effect.


Brain Research | 2015

Chronic intermittent hypoxia induces changes in expression of synaptic proteins in the nucleus of the solitary tract

Jason M. Moreau; John Ciriello

Chronic intermittent hypoxia (CIH) has been shown to alter the response of neurons in the nucleus of the solitary tract (NTS) to activation of cardiovascular inputs. Although the mechanisms involved in these effects are not known, they may involve pre- and/or post-synaptic activity-dependent changes in the chemoreceptor afferent pathway at the level of NTS. To investigate this possibility, Sprague-Dawley rats were exposed to 7 or 95 days of CIH or normoxia. Arterial pressure (AP) and heart rates (HR) were measured at these time intervals in the conscious animal, and at each time point protein was also extracted from the caudal medial NTS and analyzed by western blot for the expression of brain-derived neurotrophic factor (BDNF), tropomyosin receptor kinase B (TrkB), synaptophysin and growth-associated protein-43 (GAP-43). AP was found not to be different between the CIH and normoxic animals at 7 days, although by 95 days of CIH exposure, AP was significantly elevated (124±6mmHg) compared to normoxic controls (107±4mmHg). After 7 days of CIH exposure, protein expression of BDNF and its receptor TrkB (isoforms gp95 and gp145) were found to be significantly elevated in NTS compared to normoxic controls. However, no changes were observed in synaptophysin, and GAP-43 protein expression. After 95 days of CIH exposure, BDNF, TrkB (gp95), synaptophysin, and GAP-43 protein expression were less abundant in NTS than in the normoxic controls. These data suggest that CIH may have induced neuroplasticity changes within chemoreceptor reflex pathways at the level of NTS that may be associated with the development of autonomic dysregulation often seen in patients with CIH associated with chronic sleep apnea.


American Journal of Physiology-regulatory Integrative and Comparative Physiology | 2012

Stanniocalcin-1 in the subfornical organ inhibits the dipsogenic response to angiotensin II

Jason M. Moreau; Waseem Iqbal; Jeffrey Turner; Graham F. Wagner; John Ciriello

Recently, receptors for the calcium-regulating glycoprotein hormone stanniocalcin-1 (STC-1) have been found within subfornical organ (SFO), a central structure involved in the regulation of electrolyte and body fluid homeostasis. However, whether SFO neurons produce STC-1 and how STC-1 may function in fluid homeostasis are not known. Two series of experiments were done in Sprague-Dawley rats to investigate whether STC-1 is expressed within SFO and whether it exerts an effect on water intake. In the first series, experiments were done to determine whether STC-1 was expressed within cells in SFO using immunohistochemistry, and whether protein and gene expression for STC-1 existed in SFO using Western blot and quantitative RT-PCR, respectively. Cells containing STC-1 immunoreactivity were found throughout the rostrocaudal extent of SFO. STC-1 protein expression within SFO was confirmed with Western blot, and SFO was also found to express STC-1 mRNA. In the second series, microinjections (200 nl) of STC-1, ANG II, a combination of the two or the vehicle were made into SFO in conscious, unrestrained rats. Water intake was measured at 0700 for a 1-h period after each injection in animals. Microinjections of STC-1 (17.6 or 176 nM) alone had no effect on water intake compared with controls. However, STC-1 not only attenuated the drinking responses to ANG II for about 30 min, but also decreased the total water intake over the 1-h period. These data suggest that STC-1 within the SFO may act in a paracrine/autocrine manner to modulate the neuronal responses to blood-borne ANG II. These findings also provide the first direct evidence of a physiological role for STC-1 in central regulation of body fluid homeostasis.


Neuroscience Letters | 2016

Effect of intermittent hypoxia on arcuate nucleus in the leptin-deficient rat.

John Ciriello; Jason M. Moreau; Aaron McCoy; Douglas L. Jones

Intermittent hypoxia (IH) is a major pathophysiological consequence of obstructive sleep apnea. Recently, it has been shown that IH results in changes in body energy balance, leptin secretion and concomitant alterations in arcuate nucleus (ARC). In this study, the role of leptin on these changes was investigated in leptin-deficient rats exposed to IH or normoxic control conditions. Body weights, consumatory and locomotor behaviours, and protein signaling in ARC were assessed immediately after IH exposure. Compared to normoxia, IH altered body weight, food intake, locomotor pattern, and the plasma concentration of leptin and angiotensin II in the wild-type rat. However, these changes were not observed in the leptin-deficient rat. Within ARC of wild-type animals, IH increased phosphorylated signal transducer and activator of transcription 3 and pro-opiomelanocortin protein expression, but not in the leptin-deficient rat. The long-form leptin receptor protein expression was not altered following IH in either rat strain. These data suggest that leptin is involved in mediating the alterations to body energy balance and ARC activity following IH.


Neuroscience | 2015

Effects of angiotensin II on leptin and downstream leptin signaling in the carotid body during acute intermittent hypoxia.

Jason M. Moreau; Scott A. Messenger; John Ciriello

Angiotensin II (ANG II) is known to promote leptin production and secretion. Although ANG II type 1 receptors (AT1Rs) and leptin are expressed within the carotid body, it is not known whether AT1R and leptin are co-expressed in the same glomus cells nor if these peptides are affected within the carotid body by intermittent hypoxia (IH). This study was done to investigate whether ANG II modulated leptin signaling in the carotid body during IH. Rats were treated with captopril (Capt) or the AT1R blocker losartan (Los) in the drinking water for 3days prior to being exposed to IH (8h) or normoxia (8h). IH induced increases in plasma ANG II and leptin compared to normoxic controls. Capt treatment abolished the plasma leptin changes to IH, whereas Los treatment had no effect on the IH induced increase in plasma leptin. Additionally, carotid body glomus cells containing both leptin and the long form of the leptin receptor (OB-Rb) were found to co-express AT1R protein, and IH increased the expression of only AT1R protein within the carotid body in both Capt- and non-Capt-treated animals. On the other hand, Los treatment did not modify AT1R protein expression to IH. Additionally, Capt and Los treatment eliminated the elevated carotid body leptin protein expression, and the changes in phosphorylated signal transducer and activator of transcription three protein, the short form of the leptin receptor (OB-R100), suppressor of cytokine signaling 3, and phosphorylated extracellular-signal-regulated kinase 1/2 protein expression induced by IH. However, Capt elevated the expression of OB-Rb protein, whereas Los abolished the changes in OB-Rb protein to IH. These findings, taken together with the previous observation that ANG II modifies carotid body chemosensitivity, suggest that the increased circulating levels of ANG II and leptin induced by IH act at the carotid body to alter leptin signaling within the carotid body which in turn may influence chemoreceptor function.

Collaboration


Dive into the Jason M. Moreau's collaboration.

Top Co-Authors

Avatar

John Ciriello

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Graham F. Wagner

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Douglas L. Jones

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Scott A. Messenger

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Jeffrey Turner

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Waseem Iqbal

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

T.H. Oiamo

University of Western Ontario

View shared research outputs
Researchain Logo
Decentralizing Knowledge