Network


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

Hotspot


Dive into the research topics where David A. Lovejoy is active.

Publication


Featured researches published by David A. Lovejoy.


Peptides | 2010

Behavioral effects of neuropeptides in rodent models of depression and anxiety

Susan Rotzinger; David A. Lovejoy; Laura A. Tan

In recent years, studies have advocated neuropeptide systems as modulators for the behavioral states found in mood disorders such as depression and anxiety disorders. Neuropeptides have been tested in traditional animal models and screening procedures that have been validated by known antidepressants and anxiolytics. However, it has become clear that although these tests are very useful, neuropeptides have distinct behavioral effects and dose-dependent characteristics, and therefore, use of these tests with neuropeptides must be done with an understanding of their unique characteristics. This review will focus on the behavioral actions of neuropeptides and their synthetic analogs, particularly in studies utilizing various preclinical tests of depression and anxiety. Specifically, the following neuropeptide systems will be reviewed: corticotropin-releasing factor (CRF), urocortin (Ucn), teneurin C-terminal associated peptide (TCAP), neuropeptide Y (NPY), arginine vasopressin (AVP), oxytocin, the Tyr-MIF-1 family, cholecystokinin (CCK), galanin, and substance P. These neuropeptide systems each have a unique role in the regulation of stress-like behavior, and therefore provide intriguing therapeutic targets for mood disorder treatment.


The Journal of Comparative Neurology | 2003

Urocortin in the central nervous system of a primate (Cebus apella): sequencing, immunohistochemical, and hybridization histochemical characterization.

Luciana Auxiliadora de Paula Vasconcelos; Cynthia J. Donaldson; Luciane V. Sita; Cláudio Aparecido Casatti; Claudimara F.P. Lotfi; Liqun Wang; M.Z. Alia Cadinouche; Lúcio Frigo; Carol F. Elias; David A. Lovejoy; Jackson C. Bittencourt

The urocortin (UCN)‐like immunoreactivity and UCN mRNA distribution in various regions of the nonprimate mammalian brain have been reported. However, the Edinger‐Westphal nucleus (EW) appears to be the only brain site where UCN expression is conserved across species. Although UCN peptides are present throughout vertebrate phylogeny, the functional roles of both UCN and EW remain poorly understood. Therefore, a study focused on UCN system organization in the primate brain is warranted. By using immunohistochemistry (single and double labeling) and in situ hybridization, we have characterized the organization of UCN‐expressing cells and fibers in the central nervous system and pituitary of the capuchin monkey (Cebus apella). In addition, the sequence of the prepro‐UCN was determined to establish the level of structural conservation relative to the human sequence. To understand the relationship of acetylcholine cells in the EW, a colocalization study comparing choline acetyltransferase (ChAT) and UCN was also performed. The cloned monkey prepro‐UCN is 95% identical to the human preprohormone across the matched sequences. By using an antiserum raised against rat UCN and a probe generated from human cDNA, we found that the EW is the dominant site for UCN expression, although UCN mRNA is also expressed in spinal cord lamina IX. Labeled axons and terminals were distributed diffusely throughout many brain regions and along the length of the spinal cord. Of particular interest were UCN‐immunoreactive inputs to the medial preoptic area, the paraventricular nucleus of the hypothalamus, the oral part of the spinal trigeminal nucleus, the flocculus of the cerebellum, and the spinal cord laminae VII and X. We found no UCN hybridization signal in the pituitary. In addition, we observed no colocalization between ChAT and UCN in EW neurons. Our results support the hypothesis that the UCN system might participate in the control of autonomic, endocrine, and sensorimotor functions in primates. J. Comp. Neurol. 463:157–175, 2003.


General and Comparative Endocrinology | 2011

Stress and reproduction: Controversies and challenges

Dhan Chand; David A. Lovejoy

Inhibition of reproductive function by the activation of the stress-response has been observed since times of antiquity, however delineating a molecular mechanism by which this occurs in vertebrates continues to present a major challenge. Because recent genome sequencing programs have identified the presence of numerous paralogous peptides and receptors, our understanding of the complexity of the interaction between the reproductive and stress axes has expanded. At the neuroendocrine level, numerous studies have focused on the interaction between the corticotropin-releasing factor (CRF) and gonadotropin-releasing hormone (GnRH) systems in vertebrates. Moreover, most of these studies have been performed using rodent models and may not be completely relevant for non-mammalian vertebrates. A further problem lies in the variation of the functional expression of paralogous genes in the different taxa. In particular, the urocortin 2 and GnRH-II systems have been lost in some lineages, where its function has been taken over by urocortin 3 and GnRH-I, respectively. Establishing an integrated model that incorporates all paralogous systems for both the stress and reproductive system remains to be achieved.


General and Comparative Endocrinology | 2010

Characterization of a corticotropin-releasing factor (CRF)/diuretic hormone-like peptide from tunicates: Insight into the origins of the vertebrate CRF family

David A. Lovejoy; Dalia Barsyte-Lovejoy

The corticotropin-releasing factor (CRF) superfamily of peptides includes the four paralogous vertebrate peptide lineages of CRF, urotensin-1/urocortin/sauvagine, urocortin 2 and urocortin 3, as well as the diuretic hormones (DH) and peptides of the arthropods. However, there are considerable sequence differences between the group of vertebrate peptides and those of the arthropods, notably insects. Because of the likely incidence of the formation of paralogous forms within the insects and the great variation in primary structures among these peptides, establishing the structure of the ancestral vertebrate version has not been possible. We screened the genomes of the tunicates, Ciona intestinalis and Ciona savignii, in silico, using the various conserved motifs of both the vertebrate CRF paralogues and the insect diuretic hormone sequences to identify the structure of the Ciona CRF/DH-like peptide genes. A single peptide gene was found in both genomes that possessed motifs reflective of both groups of peptides. These structures suggest a single CRF-like peptide was inherited by vertebrates and possibly chordates. Moreover, the conserved structure of the CRF peptide may have become constrained once it became associated with the regulation of the hypothalamus-pituitary-adrenal/interrenal axis.


Neuroscience | 2007

Regulation of neurite growth in immortalized mouse hypothalamic neurons and rat hippocampal primary cultures by teneurin C-terminal-associated peptide-1.

A. Al Chawaf; K. St. Amant; Denise D. Belsham; David A. Lovejoy

Teneurins are a highly conserved family of four type II transmembrane proteins that are expressed in the CNS. The protein possesses several functional domains including a unique bioactive 40-41 amino acid sequence at the extracellular terminus. Synthetic versions of this teneurin C-terminal-associated peptide (TCAP) can modulate cyclic AMP accumulation, cell proliferation and teneurin mRNA levels in vitro. Furthermore, i.c.v. injections of TCAP-1 into rat brain induce major changes in acoustic startle response behavior 3 weeks after administration, suggesting that the peptide may act to alter interneuron communication via changes in neurite and axon outgrowth. Synthetic mouse/rat TCAP-1 was used to treat cultured immortalized mouse hypothalamic cells, to determine if TCAP-1 could directly regulate neurite and axon growth. TCAP-1-treated cells showed a significant increase in the length of neurites accompanied by a marked increase in beta-tubulin transcription and translation as determined by real-time PCR and Western blot analysis, respectively. Changes in alpha-actinin-4 transcription and beta-actin protein expression were also noted. Immunofluorescence confocal microscopy using beta-tubulin antiserum showed enhanced resolution of beta-tubulin cytoskeletal elements throughout the cell. In order to determine if the effects of TCAP-1 could be reproduced in primary neuronal cultures, primary cultures of E18 rat hippocampal cells were treated with 100 nM TCAP-1. The TCAP-1-treated hippocampal cultures showed a significant increase in both the number of cells, dendritic branching and the presence of large and fasciculated beta-tubulin immunoreactive axons. These data suggest that TCAP acts, in part, as a functional region of the teneurins to regulate neurite and axonal growth of neurons.


Vitamins and Hormones Series | 2005

Gonadotropin‐Releasing Hormone: Gene Evolution, Expression, and Regulation

Denise D. Belsham; David A. Lovejoy

The gonadotropin-releasing hormone (GnRH) gene is a superb example of the diverse regulation that is required to maintain the function of an evolutionarily conserved and fundamental gene. Because reproductive capacity is critical to the survival of the species, physiological homeostasis dictates optimal conditions for reproductive success, and any perturbation from this balance may affect GnRH expression. These disturbances may include alterations in signals dictated by stress, nutritional imbalance, body weight, and neurological problems; therefore, changes in other neuroendocrine systems may directly influence the hypothalamic-pituitary-gonadal axis through direct regulation of GnRH. Thus, to maintain optimal reproductive capacity, the regulation of the GnRH gene is tightly constrained by a number of diverse signaling pathways and neuromodulators. In this review, we summarize what is currently known of GnRH gene structure, the location and function of the two isoforms of the GnRH gene, some of the many hormones and neuromodulators found to affect GnRH expression, and the molecular mechanisms responsible for the regulation of the GnRH gene. We also discuss the latest models used to study the transcriptional regulation of the GnRH gene, from cell models to evolving in vivo technologies. Although we have come a long way in the last two decades toward uncovering the intricacies behind the control of the GnRH neuron, there remain vast distances to cover before direct therapeutic manipulation of the GnRH gene to control reproductive competence is possible.


General and Comparative Endocrinology | 2009

Structural evolution of urotensin-I: Reflections of life before corticotropin releasing factor

David A. Lovejoy

Peptides have a long evolutionary history that predates the appearance of metazoans. The corticotropin releasing factor (CRF) family of peptides is among the most ancient peptide lineages. The identification and characterization of urotensin-I and related orthologues led the way for the elucidation of the entire CRF peptide family. A comparative analysis of the CRF paralogue sequences suggest that CRF is the most derived of these peptides and has lost many of its ancestral characteristics after it became associated with the hypothalamic-pituitary-adrenal/interrenal (HPA/I axis). In vertebrates, the urotensin-I group of orthologues, which includes sauvagine and urocortin, possess a number of shared characteristics that may be indicative of the ancestral peptide. Given the early origin of the CRF family peptides, it is likely that other peptide lineages are distantly related to the CRF family. In silico or cDNA library screening using probes based on urotensin-I/urocortin characteristics have been used to identify novel CRF family and related sequences that provide clues the evolutionary origin of the CRF family.


Frontiers in Neuroscience | 2013

Appetite-suppressing effects and interactions of centrally administered corticotropin-releasing factor, urotensin I and serotonin in rainbow trout (Oncorhynchus mykiss)

Van A. Ortega; David A. Lovejoy; Nicholas J. Bernier

Corticotropin-releasing factor (CRF), urotensin I (UI) and serotonin (5-HT) are generally recognized as key regulators of the anorexigenic stress response in vertebrates, yet the proximal effects and potential interactions of these central messengers on food intake in salmonids are not known. Moreover, no study to date in fishes has compared the appetite-suppressing effects of CRF and UI using species-specific peptides. Therefore, the objectives of this study were to (1) assess the individual effects of synthesized rainbow trout CRF (rtCRF), rtUI as well as 5-HT on food intake in rainbow trout, and (2) determine whether the CRF and serotonergic systems interact in the regulation of food intake in this species. Intracerebroventricular (icv) injections of rtCRF and rtUI both suppressed food intake in a dose-related manner but rtUI [ED50 = 17.4 ng/g body weight (BW)] was significantly more potent than rtCRF (ED50 = 105.9 ng/g BW). Co-injection of either rtCRF or rtUI with the CRF receptor antagonist α-hCRF(9–41) blocked the reduction in food intake induced by CRF-related peptides. Icv injections of 5-HT also inhibited feeding in a dose-related manner (ED50 = 14.7 ng/g BW) and these effects were blocked by the serotonergic receptor antagonist methysergide. While the anorexigenic effects of 5-HT were reversed by α-hCRF(9–41) co-injection, the appetite-suppressing effects of either rtCRF or rtUI were not affected by methysergide co-injection. These results identify CRF, UI and 5-HT as anorexigenic agents in rainbow trout, and suggest that 5-HT-induced anorexia may be at least partially mediated by CRF- and/or UI-secreting neurons.


Peptides | 2007

Corticotropin-releasing factor (CRF)-induced behaviors are modulated by intravenous administration of teneurin C-terminal associated peptide-1 (TCAP-1)

Arij Al Chawaf; Karen Xu; Laura A. Tan; Franco J. Vaccarino; David A. Lovejoy; Susan Rotzinger

The teneurin C-terminal associated peptides (TCAP) are a recently discovered family of bioactive peptides that can attenuate aspects of the behavioral stress responses of rats. Because TCAP has some structural similarity to the corticotropin-releasing factor (CRF) family of peptides, and modulates elements of the stress response, TCAP may act to modulate CRF actions in vivo. This hypothesis was tested by investigating anxiety-related behaviors in male rats following repeated intravenous (IV) TCAP-1 administration with either an acute intracerebroventricular (ICV) or IV CRF challenge. TCAP-1 alone did not affect behavioral responses significantly, however did significantly affect CRF-regulated behaviors depending on CRFs mode of injection. In both the elevated plus-maze and the open field tests, TCAP-1 had an anxiolytic effect on ICV CRF responses as indicated by decreased stretched-attend postures in the elevated plus maze (p<0.05), and increased center time and center entries in the open field (p<0.05). However, prior TCAP-1 treatment has an anxiogenic effect on the IV CRF-induced behaviors (decreased center entries and total distance in the open field (p<0.05)). TCAP-1s actions are not mediated through acute changes in glucocorticoid levels and may occur via a central action in the brain. A fluorescently (FITC)-labeled TCAP-1 analog was IV-administered to investigate whether IV TCAP-1 has the potential to regulate central mechanisms by crossing the blood-brain barrier. FITC-TCAP-1 was detected in blood vessels and fibers in the brain indicating that uptake into the brain is a possible route for its interaction with CRF and its receptors. Thus, TCAP may modulate CRF-associated behaviors by a direct action in the CNS.


Brain Research | 2007

Teneurin carboxy (C)-terminal associated peptide-1 inhibits alkalosis-associated necrotic neuronal death by stimulating superoxide dismutase and catalase activity in immortalized mouse hypothalamic cells.

Gina Trubiani; Arij Al Chawaf; Denise D. Belsham; Dalia Barsyte-Lovejoy; David A. Lovejoy

The teneurins and the teneurin C-terminal-associated peptides (TCAP) are implicated in the regulation of neuron growth and differentiation. However, current observations suggest that TCAP-1 may also have a neuroprotective action during times of pH-induced cellular stress in the brain such as during hypoxia-ischemia and brain alkalosis. To test this hypothesis, we cultured a TCAP-1-responsive mouse hypothalamic cell line, N38, using media buffered at pHs 6.8, 7.4, 8.0 and 8.4 subsequently treated with 100 nM TCAP-1. TCAP-1 significantly inhibited the decline in cell proliferation at pHs 8.0 and 8.4 as determined by direct cell viability assays and decreased the incidence of cells showing necrotic morphology. In addition, TCAP-1 decreased the number of cells undergoing necrosis by 4- to 5-fold as measured by uptake of ethidium homodimer III. Moreover, TCAP-1 significantly decreased the incidence of superoxide radicals and increased superoxide dismutase 1 (SOD1) expression. These results were accompanied by an increase in the SOD copper chaperone expression and increased catalase activity and expression. The results indicate that TCAP may play a neuroprotective role during periods of pH stress by upregulating oxygen radical scavenging systems. Thus, the TCAP-teneurin system may be part of a mechanism to protect neurons during trauma, such as hypoxia and ischemia.

Collaboration


Dive into the David A. Lovejoy's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Susan Rotzinger

University Health Network

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Karen Xu

University of Toronto

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge