Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 417
Filter
Add more filters

Complementary Medicines
Country/Region as subject
Publication year range
1.
Theriogenology ; 219: 157-166, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38432143

ABSTRACT

To understand better the role that kisspeptin plays in regulating seasonal and estrous cycle changes in the mare, this study investigated the number, location and interactions between GnRH, kisspeptin and RFRP-3 neurons in the equine hypothalamus. Hypothalami were collected from mares during the non-breeding season, vernal transition and various stages of the breeding season. Fluorescent immunohistochemistry was used to label the neuropeptides of interest. GnRH cells were observed primarily in the arcuate nucleus (ARC), while very few labeled cells were identified in the pre-optic area (POA). Kisspeptin cells were identified primarily in the ARC, with a small number of cells observed dorsal to the ARC, surrounding the third ventricle (3V). The mean number of kisspeptin cells varied between animals and typically showed no pattern associated with season or stage of estrous cycle, but a seasonal difference was identified in the ARC population. Small numbers of RFRP-3 cells were observed in the ARC, ventromedial hypothalamus (VMH) and dorsomedial hypothalamus (DMH). The mean number of RFRP-3 cells appeared higher in pre-ovulatory animals compared to all other stages. The percentage of GnRH cell bodies with kisspeptin appositions did not change with season or stage of estrous cycle. The percentage of kisspeptin cells receiving inputs from RFRP-3 fibers did not vary with season or stage of estrous cycle. These interactions suggest the possibility of the presence of an ultra-short loop feedback system between these three peptides. The changes in RFRP-3 neurons suggest the possibility of a role in the regulation of reproduction in the horse, but it is unlikely to be as a gonadotropin inhibitory factor.


Subject(s)
Gonadotropin-Releasing Hormone , Neuropeptides , Horses , Animals , Female , Gonadotropin-Releasing Hormone/metabolism , Kisspeptins/metabolism , Seasons , Neuropeptides/physiology , Hypothalamus/metabolism , Estrous Cycle/physiology , Neurons
2.
Endocrinology ; 163(2)2022 02 01.
Article in English | MEDLINE | ID: mdl-34967900

ABSTRACT

For billions of years before electric light was invented, life on Earth evolved under the pattern of light during the day and darkness during the night. Through evolution, nearly all organisms internalized the temporal rhythm of Earth's 24-hour rotation and evolved self-sustaining biological clocks with a ~24-hour rhythm. These internal rhythms are called circadian rhythms, and the molecular constituents that generate them are called molecular circadian clocks. Alignment of molecular clocks with the environmental light-dark rhythms optimizes physiology and behavior. This phenomenon is particularly true for reproductive function, in which seasonal breeders use day length information to time yearly changes in fertility. However, it is becoming increasingly clear that light-induced disruption of circadian rhythms can negatively impact fertility in nonseasonal breeders as well. In particular, the luteinizing hormone surge promoting ovulation is sensitive to circadian disruption. In this review, we will summarize our current understanding of the neuronal networks that underlie circadian rhythms and the luteinizing hormone surge.


Subject(s)
Circadian Rhythm/physiology , Luteinizing Hormone/metabolism , Nerve Net/physiology , Neurons/physiology , Animals , Circadian Clocks , Circadian Rhythm/genetics , Circadian Rhythm Signaling Peptides and Proteins , Female , Gonadotropin-Releasing Hormone , Hypothalamus , Kisspeptins , Light , Male , Neuropeptides/physiology , Rodentia , Suprachiasmatic Nucleus/physiology , Transcription Factors/physiology
3.
Peptides ; 146: 170665, 2021 12.
Article in English | MEDLINE | ID: mdl-34600038

ABSTRACT

Pyrokinins (PKs) are pleiotropic neuropeptides with significant roles in invertebrate physiology. Although functions of PKs are known in insects, there is a lack of knowledge of PK-encoding genes and PKs functions in ticks. Herein the first tick cDNAs of the capability (capa) gene were cloned from the southern cattle tick, Rhipicephalus microplus (Acari: Ixodidae), and the blacklegged tick, Ixodes scapularis. Each cDNA encoded one periviscerokinin and five different pyrokinins. Two PKs were identical in sequence in the two species. The three PKs unique to R. microplus (Rhimi-CAPA-PK1, -PK2, and -PK5) were tested on the recombinant R. microplus pyrokinin receptor using a calcium bioluminescence assay. The Rhimi-CAPA-PKs acted as agonists with EC50s ranging from 101-188 nM. Twenty PK analogs designed for enhanced bioavailability and biostability were tested on the receptor. Five of these were designed based on the sequences of the three unique Rhimi-CAPA-PKs. Eight PK analogs were also agonists; four of them were full agonists that exhibited comparable efficacy to the native Rhimi-CAPA-PKs, with EC50 ranging from 401 nM-1.9 µM. The structure-activity relationships (SAR) of all analogs were analyzed. Our results suggested that a positively charged, basic lysine at the variable position X of the PK active core (FXPRLamide) conferred enhanced affinity to the analogs in their interaction with the tick receptor. These analogs are promising tools to elucidate the pyrokinin function in ticks in vivo as these analogs are expected to have prolonged hemolymph residence time in comparison to the native peptides.


Subject(s)
Arthropod Proteins/genetics , DNA, Complementary/genetics , Ixodes/physiology , Neuropeptides/physiology , Rhipicephalus/physiology , Amino Acid Sequence , Animals , Arthropod Proteins/metabolism , Base Sequence , Cloning, Molecular , Neuropeptides/chemistry , Neuropeptides/metabolism , RNA, Messenger/genetics , Receptors, G-Protein-Coupled/agonists , Structure-Activity Relationship
4.
J Neurosci ; 41(33): 7148-7159, 2021 08 18.
Article in English | MEDLINE | ID: mdl-34210784

ABSTRACT

Following stroke, the survival of neurons and their ability to reestablish connections is critical to functional recovery. This is strongly influenced by the balance between neuronal excitation and inhibition. In the acute phase of experimental stroke, lethal hyperexcitability can be attenuated by positive allosteric modulation of GABAA receptors (GABAARs). Conversely, in the late phase, negative allosteric modulation of GABAAR can correct the suboptimal excitability and improves both sensory and motor recovery. Here, we hypothesized that octadecaneuropeptide (ODN), an endogenous allosteric modulator of the GABAAR synthesized by astrocytes, influences the outcome of ischemic brain tissue and subsequent functional recovery. We show that ODN boosts the excitability of cortical neurons, which makes it deleterious in the acute phase of stroke. However, if delivered after day 3, ODN is safe and improves motor recovery over the following month in two different paradigms of experimental stroke in mice. Furthermore, we bring evidence that, during the subacute period after stroke, the repairing cortex can be treated with ODN by means of a single hydrogel deposit into the stroke cavity.SIGNIFICANCE STATEMENT Stroke remains a devastating clinical challenge because there is no efficient therapy to either minimize neuronal death with neuroprotective drugs or to enhance spontaneous recovery with neurorepair drugs. Around the brain damage, the peri-infarct cortex can be viewed as a reservoir of plasticity. However, the potential of wiring new circuits in these areas is restrained by a chronic excess of GABAergic inhibition. Here we show that an astrocyte-derived peptide, can be used as a delayed treatment, to safely correct cortical excitability and facilitate sensorimotor recovery after stroke.


Subject(s)
Diazepam Binding Inhibitor/therapeutic use , GABA-A Receptor Agonists/therapeutic use , Neurons/drug effects , Neuropeptides/therapeutic use , Peptide Fragments/therapeutic use , Receptors, GABA-A/drug effects , Stroke/drug therapy , Adult , Animals , Astrocytes/metabolism , Cortical Spreading Depression/physiology , Diazepam Binding Inhibitor/deficiency , Diazepam Binding Inhibitor/physiology , Drug Implants , Evoked Potentials, Somatosensory , Female , GABA-A Receptor Agonists/pharmacology , Humans , Hydrogels , Infarction, Middle Cerebral Artery/drug therapy , Intracranial Thrombosis/drug therapy , Intracranial Thrombosis/etiology , Light , Mice , Mice, Inbred C57BL , N-Methylaspartate/toxicity , Neurons/physiology , Neuropeptides/deficiency , Neuropeptides/physiology , Patch-Clamp Techniques , Peptide Fragments/deficiency , Peptide Fragments/physiology , Rats , Rose Bengal/radiation effects , Rose Bengal/toxicity , Single-Blind Method , Stroke/etiology
5.
Rev Neurol ; 71(12): 460-466, 2020 Dec 16.
Article in Spanish | MEDLINE | ID: mdl-33319349

ABSTRACT

INTRODUCTION: The orexinergic system is one of the chemical mediators that modulate the gut-brain axis, given the involvement of hypothalamic orexin A (OXA) in gastrointestinal motility and secretion, and the presence of OXA in enteroendocrine cells of the intestinal mucosa and in primary afferent neurons of the mesenteric plexus, permitting its participation in gut-brain signaling. AIM: The source of OXA and the signal(s) triggering its peripheral release are not fully understood, and it is not known whether it acts on orexigenic receptors in peripheral tissues to meet physiological or pathological demands. The aim of this review is to address these questions in the light of new data indicating that OXA may have functions in the gut-brain axis that go beyond its participation in energy homeostasis. DEVELOPMENT: OXA in the enteric system protects against systemic and central inflammation, and hypothalamic OXA orchestrates numerous peripheral effects to suppress the systemic inflammatory response. For this reason, OXA may act as an immunomodulator in chronic inflammations or autoimmune diseases. OXA is also involved in the stress response, regulating physiological responses to emotional or stressful stimuli. CONCLUSIONS: OXA exerts anti-inflammatory and gastroprotective effects on the intestinal mucosa; however, it may increase the response to external and/or internal stress in individuals with chronic inflammation, exacerbating the gastrointestinal inflammation. Hence, pharmacologic interventions in the orexinergic system have been proposed to treat diseases in which intestinal hypersensitivity is combined with appetite loss, sleep disturbance, stress, and anxiety.


TITLE: Orexina A como mediadora en el diálogo intestino-cerebro.Introducción. Entre los mediadores químicos que modulan el eje intestino-cerebro debe incluirse el sistema orexinérgico, ya que la orexina A (OXA) hipotalámica interviene en la motilidad y en la secreción gastrointestinal. También está presente en las células enteroendocrinas de la mucosa intestinal y en las neuronas aferentes primarias del plexo mientérico, y puede intervenir en la señalización intestino-cerebro. Objetivo. No se conoce con exactitud la fuente ni la señal que originan la liberación de OXA periférica, ni tampoco si actúa en los receptores orexinérgicos de los tejidos periféricos ante demandas fisiológicas o patológicas. Esta revisión intenta analizar estas cuestiones a la luz de nuevos datos que indican que la OXA en el eje intestino-cerebro puede tener funciones más allá de su participación en la homeostasis energética. Desarrollo. La OXA en el sistema entérico protege de la inflamación sistémica y central, y en el hipotálamo orquesta numerosos efectos periféricos para suprimir la respuesta inflamatoria sistémica. Por ello, podría actuar como sustancia inmunomoduladora en inflamaciones crónicas o en enfermedades autoinmunitarias. La OXA también se relaciona con la respuesta de estrés, regulando las respuestas fisiológicas a estímulos emocionales o estresantes. Conclusiones. Aunque la OXA tiene efectos antiinflamatorios y gastroprotectores de la mucosa intestinal, en procesos de inflamación crónica podría incrementar la respuesta a estímulos estresantes, tanto externos como internos, y exacerbar la inflamación gastrointestinal. Por ello, se han propuesto intervenciones farmacológicas sobre el sistema orexinérgico como tratamiento para enfermedades en las que la hipersensibilidad intestinal coexiste con pérdida de apetito, alteraciones del sueño, estrés y ansiedad.


Subject(s)
Gastrointestinal Tract/immunology , Gastrointestinal Tract/physiology , Orexins/immunology , Orexins/physiology , Signal Transduction/physiology , Animals , Humans , Hypothalamus/metabolism , Hypothalamus/physiology , Mice , Neuroimmunomodulation/physiology , Neurons/physiology , Neuropeptides/immunology , Neuropeptides/metabolism , Neuropeptides/physiology , Neurotransmitter Agents/immunology , Neurotransmitter Agents/metabolism , Neurotransmitter Agents/physiology , Orexin Receptors/physiology , Orexins/metabolism , Psychological Distress
6.
Front Immunol ; 11: 2119, 2020.
Article in English | MEDLINE | ID: mdl-33072073

ABSTRACT

Neurotoxicity is a common side effect of chemotherapeutics that often leads to the development of chemotherapy-induced peripheral neuropathy (CIPN). The peptide Prokineticin 2 (PK2) has a key role in experimental models of CIPN and can be considered an insult-inducible endangering mediator. Since primary afferent sensory neurons are highly sensitive to anticancer drugs, giving rise to dysesthesias, the aim of our study was to evaluate the alterations induced by vincristine (VCR) and bortezomib (BTZ) exposure in sensory neuron cultures and the possible preventive effect of blocking PK2 signaling. Both VCR and BTZ induced a concentration-dependent reduction of total neurite length that was prevented by the PK receptor antagonist PC1. Antagonizing the PK system also reduced the upregulation of PK2, PK-R1, TLR4, IL-6, and IL-10 expression induced by chemotherapeutic drugs. In conclusion, inhibition of PK signaling with PC1 prevented the neurotoxic effects of chemotherapeutics, suggesting a promising strategy for neuroprotective therapies against the sensory neuron damage induced by exposure to these drugs.


Subject(s)
Antineoplastic Agents/toxicity , Bortezomib/toxicity , Gastrointestinal Hormones/antagonists & inhibitors , Nerve Tissue Proteins/antagonists & inhibitors , Neuropeptides/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Neurotoxicity Syndromes/prevention & control , Sensory Receptor Cells/drug effects , Triazines/pharmacology , Vincristine/toxicity , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Down-Regulation , Drug Evaluation, Preclinical , Gastrointestinal Hormones/physiology , Gene Expression Regulation/drug effects , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/physiology , Neurites/drug effects , Neurites/ultrastructure , Neuroimmunomodulation/drug effects , Neuropeptides/physiology , Neuroprotective Agents/therapeutic use , RNA, Messenger/biosynthesis , Sensory Receptor Cells/physiology , Sensory Receptor Cells/ultrastructure , Triazines/therapeutic use
7.
Sci Rep ; 10(1): 16947, 2020 10 12.
Article in English | MEDLINE | ID: mdl-33046791

ABSTRACT

Protein 4.1N, a member of the protein 4.1 family, is highly expressed in the brain. But its function remains to be fully defined. Using 4.1N-/- mice, we explored the function of 4.1N in vivo. We show that 4.1N-/- mice were born at a significantly reduced Mendelian ratio and exhibited high mortality between 3 to 5 weeks of age. Live 4.1N-/- mice were smaller than 4.1N+/+ mice. Notably, while there were no significant differences in organ/body weight ratio for most of the organs, the testis/body and ovary/body ratio were dramatically decreased in 4.1N-/- mice, demonstrating selective effects of 4.1N deficiency on the development of the reproductive systems. Histopathology of the reproductive organs showed atrophy of both testis and ovary. Specifically, in the testis there is a lack of spermatogenesis, lack of leydig cells and lack of mature sperm. Similarly, in the ovary there is a lack of follicular development and lack of corpora lutea formation, as well as lack of secretory changes in the endometrium. Examination of pituitary glands revealed that the secretory granules were significantly decreased in pituitary glands of 4.1N-/- compared to 4.1N+/+. Moreover, while GnRH was expressed in both neuronal cell body and axons in the hypothalamus of 4.1N+/+ mice, it was only expressed in the cell body but not the axons of 4.1N-/- mice. Our findings uncover a novel role for 4.1N in the axis of hypothalamus-pituitary gland-reproductive system.


Subject(s)
Cytoskeletal Proteins/deficiency , Cytoskeletal Proteins/physiology , Genitalia/metabolism , Genitalia/pathology , Membrane Proteins/deficiency , Membrane Proteins/physiology , Neuropeptides/deficiency , Neuropeptides/physiology , Neurosecretory Systems/metabolism , Neurosecretory Systems/pathology , Animals , Cytoskeletal Proteins/genetics , Female , Gonadotropin-Releasing Hormone/genetics , Gonadotropin-Releasing Hormone/metabolism , Humans , Hypothalamus/metabolism , Hypothalamus/pathology , Male , Membrane Proteins/genetics , Mice, Knockout , Neuropeptides/genetics , Organ Size , Ovary/pathology , Pituitary Gland/metabolism , Pituitary Gland/pathology , Spermatogenesis/genetics , Testis/pathology
8.
J Neuroendocrinol ; 31(12): e12807, 2019 12.
Article in English | MEDLINE | ID: mdl-31679160

ABSTRACT

The interaction of animals with conspecifics, termed social behaviour, has a major impact on the survival of many vertebrate species. Neuropeptide hormones modulate the underlying physiology that governs social interactions, and many findings concerning the neuroendocrine mechanisms of social behaviours have been extrapolated from animal models to humans. Neurones expressing neuropeptides show similar distribution patterns within the hypothalamic nucleus, even when evolutionarily distant species are compared. During evolution, hypothalamic neuropeptides and releasing hormones have retained not only their structures, but also their biological functions, including their effects on behaviour. Here, we review the current understanding of the mechanisms of social behaviours in several classes of animals, such as worms, insects and fish, as well as laboratory, wild and domesticated mammals.


Subject(s)
Hypothalamus/physiology , Neuropeptides/physiology , Social Behavior , Animals
9.
Ann Anat ; 225: 28-32, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31195095

ABSTRACT

Anterior cruciate ligament (ACL) tears is a devastating injury and one of the most common knee injuries experienced by athletes in the United States. Although patients reach maximal subjective improvement by one-year following ACL reconstruction, many patients often experience moderate to severe post-operative pain. Opioids, intra-articular injections, and regional anesthesia have been previously implemented to mediate post-operative pain. However, chronic opioid usage has become an epidemic in the United States. Alternative analgesic modalities, such as nerve blocks, have been implemented in clinical practice to provide adequate pain relief and minimize opioid usage. Periarticular injections targeted towards local neurological structures performed concomitantly with nerve blocks provides superior pain relief and satisfaction than isolated nerve blocks. Therefore, it is imperative for physicians to understand local neurological anatomy around the knee joint in order to provide adequate analgesia while minimizing opioid consumption. This purpose of this investigation is to summarize (1) neurogenic origins of pain generators and mediators in sites affected by ACL reconstruction and autograft harvest sites and (2) analgesia utilized in ACL reconstruction.


Subject(s)
Anterior Cruciate Ligament Reconstruction , Anterior Cruciate Ligament/anatomy & histology , Knee Joint/blood supply , Knee Joint/innervation , Pain, Postoperative/etiology , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/adverse effects , Anesthesia, Local , Anesthetics, Local/administration & dosage , Anterior Cruciate Ligament/innervation , Anterior Cruciate Ligament/surgery , Autografts , Ion Channels/metabolism , Mechanoreceptors/physiology , Nerve Block , Neuropeptides/metabolism , Neuropeptides/physiology , Nociception/physiology , Nociceptors/physiology
10.
Med Hypotheses ; 126: 66-68, 2019 May.
Article in English | MEDLINE | ID: mdl-31010502

ABSTRACT

Narcolepsy with cataplexy is a lifelong sleep disorder associated with orexin/hypocretin deficiency in the central nervous system. In addition to a genetic predisposition, a variety of environmental factors, such as influenza viruses, have been implicated in the pathogenesis of the disease. In this article, a hypothesis is proposed that environmental agents access the olfactory bulb and trigger neuroinflammation, which in turn induces neurodegeneration of orexinergic neurons in the lateral hypothalamus and other neuronal subpopulations regulating the sleep-wake cycle, which triggers the development of narcolepsy.


Subject(s)
Narcolepsy/physiopathology , Olfactory Bulb/physiopathology , Animals , Cataplexy , Cytokines/metabolism , Humans , Hypothalamus/metabolism , Hypothalamus/physiopathology , Inflammation , Intracellular Signaling Peptides and Proteins , Mice , Models, Anatomic , Neurons/physiology , Neuropeptides/physiology , Olfactory Bulb/metabolism , Orexins/metabolism , Sleep , Wakefulness
11.
Semin Reprod Med ; 37(4): 155-165, 2019 07.
Article in English | MEDLINE | ID: mdl-31972860

ABSTRACT

Puberty is a fundamental developmental event in the lifespan of any individual, when sexual and somatic maturation is completed, and reproductive capacity is achieved. While the tempo of puberty is under strong genetic determination, it is also modulated by a wide array of internal and environmental cues, including, prominently, nutritional and metabolic signals. In the last decade, our understanding of the neurohormonal basis of normal puberty and its perturbations has enlarged considerably. This is illustrated by the elucidation of the essential roles of kisspeptins, encoded by the Kiss1 gene, in the hypothalamic circuits controlling puberty. Moreover, other neuropeptide pathways, convergent with kisspeptin signaling, have been pointed out as important coregulators of pubertal timing. These include the cotransmitters of Kiss1 neurons in the arcuate nucleus (ARC), neurokinin B, and dynorphin, as well as melanocortins, produced by ARC neurons expressing proopiomelanocortin, which are endowed with key roles also in the control of metabolic homeostasis. This neuropeptide setup seemingly participates, in a coordinated manner, in transmitting the regulatory actions of metabolic cues on pubertal maturation. In this function, cellular metabolic sensors, such as the AMP-activated protein kinase, and the fuel-sensing deacetylase, SIRT1, have also been shown recently to contribute to the metabolic regulation of puberty. Altogether, elucidation of the physiological roles of these signals and regulatory circuits will help uncover the intimacies of the brain control of puberty, and its alterations in conditions of metabolic stress, ranging from subnutrition to obesity.


Subject(s)
Neuropeptides/physiology , Puberty/physiology , Animals , Humans , Hypothalamus/metabolism , Hypothalamus/physiology , Kisspeptins/metabolism , Neurokinin B/metabolism , Neurons/metabolism , Neurons/physiology , Neuropeptides/pharmacology , Puberty/drug effects , Reproduction/physiology , Sexual Maturation/drug effects , Sexual Maturation/physiology , Signal Transduction/drug effects , Signal Transduction/physiology
12.
Ann Endocrinol (Paris) ; 80(1): 38-46, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30243474

ABSTRACT

The present review focused on the most important effects of leptin on the hypothalamus and on how leptin regulates neuropeptides associated with food intake and GnRH secretion. This review of the literature suggests that a reduction in leptin serum concentrations results from lower body energy reserves or poor energy availability, leading to hypothalamic secretion of neuropeptides such as NPY/AgRP and QRFP to stimulate food intake. Under these negative metabolic conditions, GnRH secretion is reduced, impairing reproductive functions. In contrast, when metabolic status is inversed by an increase in food availability, energy reserves or both, leptin serum concentrations increase to an action threshold reversing the pattern of secretion: i.e., reducing NPY/AgRP and QRFP and increasing POMC and Kisspeptin, and thereby reducing food intake and stimulating GnRH secretion to promote reproductive function.


Subject(s)
Eating/physiology , Gonadotropin-Releasing Hormone/metabolism , Leptin/physiology , Neuropeptides/physiology , Agouti-Related Protein/metabolism , Animals , Energy Metabolism/physiology , Homeostasis/physiology , Humans , Hypothalamus/physiology , Kisspeptins/metabolism , Leptin/blood , Neuropeptide Y/metabolism , Pro-Opiomelanocortin/metabolism , Reproduction/physiology
13.
Gen Comp Endocrinol ; 260: 164-170, 2018 05 01.
Article in English | MEDLINE | ID: mdl-28951261

ABSTRACT

Probing previously unknown neuropeptides and/or peptide hormones is essential for our understanding of the regulation of energy homeostasis in the brain. We recently performed a cDNA subtractive screening of the chicken hypothalamus, which contained one of the feeding and energy metabolic centers. We found a gene encoding a novel protein of 182 amino acid residues, including one putative small secretory protein of 80 amino acid residues. The C-terminal amino acids of the small protein were Gly-Leu-NH2, and as a result, the small protein was termed neurosecretory protein GL (NPGL). Subcutaneous and intracerebroventricular infusions of NPGL increased body mass gain in chicks, suggesting a central role for this protein in regulating growth and energy homeostasis. A database search revealed that the Npgl gene is conserved in vertebrates, including mice and rats. This review summarizes the advances in the characterization, localization, and biological action of NPGL, in birds and rodents.


Subject(s)
Chickens , Energy Metabolism/genetics , Feeding Behavior/physiology , Mice , Nerve Tissue Proteins/physiology , Animals , Chickens/genetics , Chickens/metabolism , Homeostasis , Hypothalamus/metabolism , Mice/genetics , Mice/metabolism , Neuropeptides/physiology , Rats/genetics , Rats/metabolism , Vertebrates
14.
Clin Chem ; 64(1): 173-182, 2018 01.
Article in English | MEDLINE | ID: mdl-29097517

ABSTRACT

BACKGROUND: The global rise in the prevalence of obesity and associated comorbidities such as type 2 diabetes, cardiovascular disease, and cancer represents a major public health concern. CONTENT: Studies in rodents with the use of global and targeted gene disruption, and mapping of neurocircuitry by using optogenetics and designer receptors exclusively activated by designer drugs (DREADDs) have greatly advanced our understanding of the neural control of body weight. In conjunction with analytical chemistry techniques involving classical immunoassays and mass spectrometry, many neuropeptides that are key to energy homeostasis have been identified. The actions of neuropeptides are diverse, from paracrine modulation of local neurotransmission to hormonal control of distant target organs. SUMMARY: Multiple hormones, such as the adipocyte-derived leptin, insulin, and gut hormones, and nutrients signal peripheral energy state to the central nervous system. Neurons in distinct areas of the hypothalamus and brainstem integrate and translate this information by both direct inhibitory/excitatory projections and anorexigenic or orexigenic neuropeptides into actions on food intake and energy expenditure. The importance of these neuropeptides in human energy balance is most powerfully illustrated by genetic forms of obesity that involve neuropeptides such as melanocortin-4-receptor (MC4R) deficiency. Drugs that mimic the actions of neuropeptides are being tested for the treatment of obesity. Successful therapeutic strategies in obesity will require in-depth knowledge of the neuronal circuits they are working in, the downstream targets, and potential compensatory mechanisms.


Subject(s)
Metabolic Diseases/physiopathology , Neuropeptides/physiology , Obesity/physiopathology , Animals , Appetite/physiology , Energy Metabolism , Feeding Behavior , Humans , Hypothalamus/metabolism , Neurons/metabolism , Receptor, Melanocortin, Type 4/metabolism , Signal Transduction
15.
Physiol Behav ; 190: 43-60, 2018 06 01.
Article in English | MEDLINE | ID: mdl-28624479

ABSTRACT

We tested the hypothesis that the effects of food restriction on behavioral motivation are mediated by one or both of the RFamide peptides, RFamide-related peptide-3 (RFRP-3) and kisspeptin (Kp) in female Syrian hamsters (Mesocricetus auratus). Female hamsters fed ad libitum and given a choice between food and adult male hamsters are highly motivated to visit males instead of food on all four days of the estrous cycle, but after 8days of mild food restriction (75% of ad libitum intake) they shift their preference toward food every day of the estrous cycle until the day of estrus, when they shift their preference back toward the males. In support of a role for RFRP-3 in these behavioral changes, the preference for food and the activation of RFRP-3-immunoreactive (Ir) cells in the dorsomedial hypothalamus (DMH) showed the same estrous cycle pattern in food-restricted females, but no association was observed between behavior and the activation of Kp cells in the hypothalamic arcuate nucleus or preoptic area. Next, we tested the hypothesis that food-restriction-induced activation of RFRP-3-Ir cells is modulated by high levels of ovarian steroids at the time of estrus. In support of this idea, on nonestrous days, mild food restriction increased activation of RFRP-3-Ir cells, but failed to do so on the day of estrus even though this level of food restriction did not significantly decrease circulating concentrations of estradiol or progesterone. Furthermore, in ovariectomized females, food-restriction-induced increases in activation of RFRP-3-Ir cells were blocked by systemic treatment with progesterone alone, estradiol plus progesterone, but not estradiol alone. Central infusion with RFRP-3 in ad libitum-fed females significantly decreased sexual motivation and produced significant increases in 90-minute food hoarding, in support of the hypothesis that elevated central levels of RFRP-3 are sufficient to create the shift in behavioral motivation in females fed ad libitum. Together, these results are consistent with the hypothesis that high levels of ingestive motivation are promoted during the nonfertile phase of the estrous cycle by elevated activation of RFRP-3-Ir cells, and RFRP-3-Ir cellular activation is modulated by ovarian steroids around the time of estrus, thereby diverting attention away from food and increasing sexual motivation.


Subject(s)
Estrous Cycle/physiology , Food Deprivation/physiology , Kisspeptins/physiology , Motivation/physiology , Neuropeptides/physiology , Animals , Caloric Restriction , Cricetinae , Estradiol/blood , Estradiol/pharmacology , Female , Hypothalamus/metabolism , Male , Mesocricetus , Microinjections , Neuropeptides/metabolism , Neuropeptides/pharmacology , Ovariectomy , Progesterone/blood , Progesterone/pharmacology
16.
Neuropeptides ; 65: 21-27, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28433253

ABSTRACT

The central role of neuropeptide S (NPS), identified as the endogenous ligand for GPR154, now named neuropeptide S receptor (NPSR), has not yet been fully clarified. We examined the central role of NPS for body temperature, energy expenditure, locomotor activity and adrenal hormone secretion in rats. Intracerebroventricular (icv) injection of NPS increased body temperature in a dose-dependent manner. Energy consumption and locomotor activity were also significantly increased by icv injection of NPS. In addition, icv injection of NPS increased the peripheral blood concentration of adrenalin and corticosterone. Pretreatment with the ß1- and ß2-adrenergic receptor blocker timolol inhibited the NPS-induced increase of body temperature. The expression of both NPS mRNA in the brainstem and NPSR mRNA in the hypothalamus showed a nocturnal rhythm with a peak occurring during the first half of the dark period. To examine whether the endogenous NPS is involved in regulation of body temperature, NPSR antagonist SHA68 was administered one hour after darkness. SHA68 attenuated the nocturnal rise of body temperature. These results suggest that NPS contributes to the regulation of the sympathetic nervous system.


Subject(s)
Motor Activity , Neuropeptides/physiology , Sympathetic Nervous System/physiology , Thermogenesis , Adrenergic beta-Antagonists/administration & dosage , Animals , Brain Stem/metabolism , Circadian Rhythm , Corticosterone/blood , Energy Metabolism , Epinephrine/blood , Hypothalamus/metabolism , Male , Neuropeptides/administration & dosage , Oxazolidinones/administration & dosage , Pyrazines/administration & dosage , RNA, Messenger/metabolism , Rats, Wistar , Receptors, Neuropeptide/antagonists & inhibitors
17.
J Biol Regul Homeost Agents ; 30(3): 883-888, 2016.
Article in English | MEDLINE | ID: mdl-27655516

ABSTRACT

The 77 amino prepropeptide apelin has been isolated from bovine stomach tissue and several smaller fragments, including apelin-13, showed high affinity for the orphan APJ receptor. The distribution of apelinergic fibers and receptors in the hypothalamus may suggest a role of apelin-13 on energy balance regulation, albeit the studies reporting the acute effects of apelin on feeding control are inconsistent. Considering the possible involvement of apelinergic system on hypothalamic appetite controlling network, in the present study we evaluated in the rat the effects of intrahypothalamic apelin-13 injection on food intake and the involvement of orexigenic and anorexigenic hypothalamic peptides and neurotransmitters. Eighteen rats (6 for each group of treatment) were injected into the ARC with either vehicle or apelin-13 (1-2 µg/rat). Food intake and hypothalamic peptide and neurotransmitter levels were evaluated 2 and 24 h after injection. Compared to vehicle, apelin-13 administration increased food intake both 2 and 24 h following treatment. This effect could be related to inhibited cocaine- and amphetamine-regulated transcript (CART) gene expression and serotonin (5-hydroxytryptamine, 5-HT) synthesis and release, and increased orexin A gene expression in the hypothalamus.


Subject(s)
Appetite/drug effects , Arcuate Nucleus of Hypothalamus/drug effects , Feeding Behavior/drug effects , Intercellular Signaling Peptides and Proteins/therapeutic use , Animals , Appetite/physiology , Arcuate Nucleus of Hypothalamus/physiology , Electric Stimulation , Feeding Behavior/physiology , Gene Expression Regulation/drug effects , Hypothalamus/metabolism , Hypothalamus/ultrastructure , Injections , Intercellular Signaling Peptides and Proteins/administration & dosage , Male , Motor Activity/drug effects , Neuropeptides/genetics , Neuropeptides/physiology , Neurotransmitter Agents/genetics , Neurotransmitter Agents/physiology , Rats , Rats, Sprague-Dawley , Serotonin/physiology , Synaptosomes/metabolism
18.
J Neuroendocrinol ; 28(9)2016 09.
Article in English | MEDLINE | ID: mdl-27369805

ABSTRACT

RFamide-related peptides (RFRPs) have been heavily implicated in the control of reproductive function subsequent to their discovery more than 16 years ago. However, recent studies using genetic and pharmacological tools have challenged their importance in regulating the hypothalamic-pituitary-gonadal axis. It is generally accepted that RFRPs act as part of a wider RFamide system, which involves two receptors, called the neuropeptide FF receptors (NPFFR1 and R2), and includes the closely-related neuropeptide NPFF. NPFF has been studied ever since the 1980s and many of the functions of NPFF are also shared by RFRPs. The current review questions whether these functions of NPFF are indeed specific to just NPFF alone and presents evidence from both neuroendocrine and pharmacological perspectives. Furthermore, recently emerging new functions of RFRPs are discussed with the overall goal of clarifying the functions of RFRPs beyond the hypothalamic-pituitary-gonadal axis.


Subject(s)
Brain/physiology , Gonadotrophs/physiology , Neuropeptides/physiology , Receptors, Neuropeptide/physiology , Animals , Anxiety , Gonads/innervation , Gonads/physiology , Humans , Hypothalamus/physiology , Neural Pathways/physiology , Pituitary Gland/physiology , Reproduction , Stress, Psychological
19.
Article in English | MEDLINE | ID: mdl-27387442

ABSTRACT

Maintaining adaptive control of behavior and physiology is the main strategy used by animals in responding to changes of food resources. To investigate the effects of random food deprivation (FD) and refeeding on energy metabolism and behavior in Apodemus chevrieri, we acclimated adult males to FD for 4weeks, then refed them ad libitum for 4weeks (FD-Re group). During the period of FD, animals were fed ad libitum for 4 randomly assigned days each week, and deprived of food the other 3days. A control group was fed ad libitum for 8weeks. At 4 and 8weeks we measured body mass, thermogenesis, serum leptin levels, body composition, gastrointestinal tract morphology, behavior and hypothalamic neuropeptide expression. At 4weeks, food intake, gastrointestinal mass, neuropeptide Y (NPY) and agouti-related protein (AgRP) mRNA expressions increased and thermogenesis, leptin levels, pro-opiomelanocortin (POMC) and cocaine- and amphetamine-regulated transcript (CART) expressions decreased in FD compared with controls. FD also showed more resting behavior and less activity than the controls on ad libitum day. There were no differences between FD-Re and controls at 8weeks, indicating significant plasticity. These results suggested that animals can compensate for unpredictable reduction in food availability by increasing food intake and reducing energy expended through thermogenesis and activity. Leptin levels, NPY, AgRP, POMC, and CART mRNA levels may also regulate energy metabolism. Significant plasticity in energy metabolism and behavior was shown by A. chevrieri over a short timescale, allowing them to adapt to food shortages in nutritionally unpredictable environments.


Subject(s)
Food Deprivation/physiology , Hypothalamus/physiology , Murinae/physiology , Neuropeptides/physiology , Agouti-Related Protein/genetics , Agouti-Related Protein/physiology , Animals , Basal Metabolism/genetics , Basal Metabolism/physiology , Behavior, Animal/physiology , Body Composition , Body Weight , Eating/genetics , Eating/physiology , Energy Metabolism , Leptin/blood , Male , Murinae/genetics , Murinae/psychology , Nerve Tissue Proteins/genetics , Neuropeptide Y/genetics , Neuropeptide Y/physiology , Neuropeptides/genetics , Pro-Opiomelanocortin/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Thermogenesis/genetics , Thermogenesis/physiology
20.
Med Hypotheses ; 93: 30-3, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27372853

ABSTRACT

Over the past decades, life-styles changing have led to exacerbated food and caloric intake and a reduction in energy expenditure. Obesity, main outcome of these changes, increases the risk for developing type 2 diabetes, cardiovascular disease and metabolic syndrome, the leading cause of death in adult and middle age population. Body weight and energy homeostasis are maintained via complex interactions between orexigenic and anorexigenic neuropeptides that take place predominantly in the hypothalamus. Overeating may disrupt the mechanisms of feeding control, by decreasing the expression of proopiomelanocortin (POMC) and α-melanocyte stimulating hormone (α-MSH) and increasing orexigenic neuropeptide Y (NPY) and agouti-related peptide (AgRP), which leads to a disturbance in appetite control and energy balance. Studies have shown that regular physical exercise might decrease body-weight, food intake and improve the metabolic profile, however until the currently there is no consensus about its effects on the expression of orexigenic/anorexigenic neuropeptides expression. Therefore, we propose that the type and length of physical exercise affect POMC/αMSH and NPY/AgRP systems differently and plays an important role in feeding behavior. Moreover, based on the present reports, we hypothesize that increased POMC/αMSH overcome NPY/AgRP expression decreasing food intake in long term physical exercise and that results in amelioration of several conditions related to overweight and obesity.


Subject(s)
Appetite Regulation , Exercise , Hypothalamus/physiology , Neuropeptides/physiology , Agouti-Related Protein/physiology , Animals , Body Weight , Eating , Energy Metabolism , Feeding Behavior , Humans , Models, Theoretical , Neuropeptide Y/physiology , Obesity , Overweight , Pro-Opiomelanocortin/physiology , alpha-MSH/physiology
SELECTION OF CITATIONS
SEARCH DETAIL