Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Integr Neurosci ; 23(3): 60, 2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-38538224

RESUMO

BACKGROUND: The medial prefrontal cortex (mPFC) is synaptically coupled to locus ceruleus (LC) located in the pontine tegmentum. The LC supplies norepinephrine (NE) to most of the central nervous system (CNS) via an elaborate efferent network. NE release in the cortex and various limbic structures regulates arousal, memory processes, adaptive behavior and cognitive control. METHODS: The study investigated the role of the mPFC-LC circuit in the cognitive behavior of mice. The mPFC efferents were inhibited optogenetically at the level of dorso-rostral pons by virally delivered ArchT opsin. The mice were implanted bilaterally with optic fibers transmitting yellow light and tested for anxiety-like behavior on Elevated O-maze (EOM), for long-term memory with Novel Object Recognition test (NOR), for problem-solving ability with Puzzle test and for learning with Cued Fear Conditioning (FC). In addition, we used anterograde transsynaptic viral tracing to map a possible anatomical circuit allowing the mPFC to modulate the activity of LC neurons, which supply NE to the main limbic structures with a functional role in cognitive behavior. RESULTS: The application of yellow light did not affect the anxiety-like behavior of the mice but impaired their ability to recognize a novel object and solve a problem. Optogenetic inhibition of mPFC to LC, in either acquisition or recall phase of FC similarly decreased freezing. The viral tracing identified the following tripartite circuits: mPFC-LC-dentate gyrus of the hippocampus (DG), mPFC-LC-amygdala (Amy), and mPFC-LC-mPFC. CONCLUSIONS: Our results reveal essential long-range regulatory circuits from the mPFC to LC and from LC to the limbic system that serves to optimize cognitive performance.


Assuntos
Locus Cerúleo , Optogenética , Camundongos , Animais , Locus Cerúleo/fisiologia , Neurônios/fisiologia , Córtex Pré-Frontal/fisiologia , Cognição
2.
Neuroscience ; 503: 17-27, 2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-36100034

RESUMO

We studied the effects of inflammatory pain on working memory and correlated the pain effects with changes in dendritic spine density and glutamate signaling in the medial prefrontal cortex (mPFC) of male and female mice. Injection of Complete Freund's Adjuvant (CFA) into the hind paw modeled inflammatory pain. The CFA equally decreased the mechanical thresholds in both sexes. The density of dendritic spines, as a marker for neuronal input, increased on the dendrites of both, pyramidal cells and interneurons in males but only on the dendrites of interneurons in CFA injected females. Next, we injected virus with glutamate sensor (pAAV5.hSyn.iGluSnFr) into the mPFC and used fiber photometry to record glutamate signaling during Y-maze spontaneous alternations test, which is a test for working memory in rodents. The detected fluorescent signal was higher during correct alternations when compared to incorrect alternations in both sexes. The CFA injection did not change the pattern of glutamate fluorescence during the test but the female mice made fewer incorrect alternations than their male counterparts. Furthermore, while the CFA injection decreased the expression of the glutamate transporter VGlut1 on the soma of mPFC neurons in both sexes, the decrease was sex dependent. We concluded that inflammatory pain, which increases sensory input into the mPFC neurons, may impair working memory by altering the glutamate signaling. The glutamate deficit that develops as a result of the pain is more pronounced in male mice in comparison to female mice.


Assuntos
Memória de Curto Prazo , Dor , Camundongos , Masculino , Feminino , Animais , Adjuvante de Freund/toxicidade , Dor/metabolismo , Transtornos da Memória/etiologia , Sistema X-AG de Transporte de Aminoácidos , Ácido Glutâmico , Inflamação/induzido quimicamente , Inflamação/metabolismo
3.
FASEB J ; 35(7): e21747, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34151467

RESUMO

We tested the hypothesis that the cognitive impairment associated with inflammatory pain may result from dysregulation of the top-down control of locus ceruleus's (LC) activity by the medial prefrontal cortex (mPFC). Injection of complete Freund's adjuvant (CFA) served as a model for inflammatory pain. The CFA injection decreased the thermal thresholds in both sexes but only the male mice showed increased anxiety-like behavior and diminished cognition, while the females were not affected. Increased calcium fluorescence, a marker for neuronal activity, was detected by photometry in the mPFC of males but not in females with CFA. Next, while chemogenetic inhibition of the projections from the mPFC to the LC improved the object recognition memory of males with pain, the inhibition of the mPFC to LC pathway in female mice produced anxiolysis and spatial memory deficits. The behavior results prompted us to compare the reciprocal innervation of mPFC and LC between the sexes. We used an anterograde transsynaptic tagging technique, which relies on postsynaptic cre transfer, to assess the innervation of LC by mPFC efferents. The males showed a higher rate of postsynaptic cre transfer into LC neurons from mPFC efferents than the females. And vice versa, a retrograde tracing experiment demonstrated that LC to mPFC projection neurons were more numerous in females when compared to males. In conclusion, we provide evidence that subtle differences in the reciprocal neuronal circuit between the LC and mPFC may contribute to sex differences associated with the adverse cognitive effects of inflammatory pain.


Assuntos
Inflamação/fisiopatologia , Locus Cerúleo/fisiopatologia , Dor/fisiopatologia , Córtex Pré-Frontal/fisiopatologia , Animais , Feminino , Masculino , Transtornos da Memória/fisiopatologia , Camundongos , Neurônios/fisiologia , Caracteres Sexuais , Memória Espacial/fisiologia
4.
Pain ; 161(8): 1861-1871, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32701845

RESUMO

We investigated the contribution of nucleus locus ceruleus (LC) to the development of pain-associated affective behavior. Mice of both sexes were subjected to sciatic nerve cuffing, a model of peripheral nerve injury, and monitored for 45 days. Although the thermal and mechanical thresholds were equally decreased in both males and females, only the male mice developed anxiodepressive-like behavior, which was complemented by suppressed hippocampal neurogenesis. Furthermore, the LC activity was lower in males when compared with females subjected to sciatic cuffing. Next, we used a chemogenetic approach to modulate the activity of LC projections to the dentate gyrus of the hippocampus in females without cuffs and in males with sciatic cuffs. Sustained inhibition of the LC projections to the dentate gyrus for 15 days induced anxiodepressive-like behavior and reduced the hippocampal neurogenesis in females. Activation of the LC projections to the dentate gyrus for 15 days prevented the development of anxiodepressive-like behavior and increased the hippocampal neurogenesis in males with cuffs. In sum, we demonstrated that the LC projections to the hippocampus link the sensory to the affective component of neuropathic injury and that the female mice are able to dissociate the nociception from affect by maintaining robust LC activity. The work provides evidence that sex differences in LC response to pain determine the sex differences in the development of pain phenotype.


Assuntos
Nervo Isquiático , Caracteres Sexuais , Animais , Feminino , Hipocampo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurogênese
5.
Neurobiol Stress ; 10: 100163, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31193505

RESUMO

Ensembles of principal neurons in the basolateral amygdala (BLA) generate the initial engrams for fear memories, while projections from the BLA to the medial prefrontal cortex (mPFC) are essential for the encoding, transfer and storage of remote fear memories. We tested the effects of chronic pain on remote fear memories in mice. Male mice underwent classic fear conditioning by pairing a single tone (conditional stimulus, CS) with a single electric foot shock (unconditional stimulus, US). Sciatic nerve constriction was used to induce neuropathic pain at various time points before or after the fear conditioning. The mice with sciatic nerve cuffs implanted 48 h after the fear conditioning showed an increased freezing response to CS when compared to mice without cuffs or when compared to mice in which the nerve cuffing was performed 48 h before the fear conditioning. The enhancing effect of pain on consolidated fear memory was further tested and mice in which the nerve cuffing was performed 14 days after the fear conditioning also showed an increased fear response when tested 56 days later. We used immunostaining to detect morphological changes in the BLA that could suggest a mechanism for the observed increase in fear response. We found an increased number of calbindin/parvalbumin positive neurons in the BLA and increased perisomatic density of GAD65 on projection neurons that connect BLA to mPFC in mice with nerve cuffs. Despite the strong increase of c-Fos expression in BLA and mPFC that was induced by fear recall, neither the BLA to mPFC nor the mPFC to BLA projection neurons were activated in mice with nerve cuffs. Furthermore, non-injured mice had an increased fear response when BLA to mPFC projections were inhibited by a chemogenetic method. In conclusion, this study provides evidence that persistent pain has a significant impact on consolidated fear memories. Very likely the underlying mechanism for this phenomenon is increased inhibitory input onto the BLA to mPFC projection neurons, possibly from neurons with induced parvalbumin expression. Conceivably, the increased fear response to consolidated fear memory is a harbinger for the later development of anxiety and depression symptoms associated with chronic pain.

6.
Neurobiol Dis ; 124: 364-372, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30572023

RESUMO

Enk neurons in CeA modulate the activity of the amygdala projection neurons and it is very likely that changes of Enk signaling cause the heightened anxiety that accompanies chronic pain. We use chemogenetics and transgenic mice to investigate the effects of acute and continuous activation of the amygdala Enk neurons on persistent pain and anxiodepressive-like behavior in mice. Enk-cre mice were injected bilaterally into the CeA with cre-activated AAV-DREADD/Gq/mCherry, while neuropathic pain was induced by sciatic nerve constriction. A single injection of DREADD's ligand CNO decreased the anxiety-like behavior in both, uninjured mice and in mice with neuropathic pain and produced robust analgesia that lasted for 24 h. Furthermore, the activation of Enk neurons by the DREADD ligand led to increased c-Fos expression in PKC-δ interneurons of the CeA and in non-serotonergic neurons in the ventrolateral periaqueductal gray (vlPAG), a brain structure that is an essential part of the descending pain inhibitory system. Next, we added CNO to the drinking water of the experimental mice for 14 days in order to assess the effects of continuous activation of CeA Enk interneurons on anxiodepressive-like behavior, which is affected by chronic pain. The prolonged activation of the CeA Enk interneurons reduced neohypophagia in the novelty suppressed feeding test and increased ΔFosB (a marker for sustained neuronal activation) in the vlPAG of mice with chronic pain. All together, the results of our experiments point to an important role of the CeA Enk neurons in the control of both nociception and emotion. Activation of Enk neurons resulted in sustained analgesia accompanied by anxiolysis and antidepressant effects. Very likely, these effects of CeA Enk neurons are result of the activation of vlPAG, a brain region that is essential not only for descending inhibition of pain but it is also a core element in the resilience to stress.


Assuntos
Núcleo Central da Amígdala/fisiologia , Interneurônios/fisiologia , Neuralgia/fisiopatologia , Animais , Ansiedade/fisiopatologia , Dor Crônica/fisiopatologia , Dor Crônica/psicologia , Encefalinas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neuralgia/psicologia
7.
Neuroscience ; 390: 187-197, 2018 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-30170157

RESUMO

The encoding, consolidation and retrieval of memories is a multifaceted process that depends strongly on the optimal level of arousal but high levels of arousal may trigger anxiety, which negatively impacts the memory processing by the brain. We investigated the role of CRH neurons in the central amygdala (CeA) for their capacity to modulate both, the anxiety-like behavior and hippocampus-dependent memory. First, we activated the CRH neurons in CeA using cre-dependent AAV-DREADD in CRH-cre mice. The activation of CeA CRH neurons increased the anxiety-like behavior in Elevated-O maze (O-maze) and Light-Dark box (LDB). The activation of the CeA CRH also decreased Y-maze memory performance and the discrimination index in novel object recognition test (NOR). The inhibition of CeA CRH neurons with AAV-DREADD had the opposite effects on the anxiety-like behavior and the memory tests. Next, we used a combination of retrograde cre virus injected into locus ceruleus (LC) and cre-dependent AAV-DREADD injected into the CeA. While the excitation of the CeA neurons that project to LC increased the anxiety-like behavior, it also led to a better performance on the memory tests. The behavioral and memory effects were accompanied by increased c-Fos expression in the LC region. Pretreatment with CRH1 receptor antagonist antalarmin hydrochloride blocked the effects that were observed after the activation of the CeA projections to LC. Our findings highlight the role of CeA CRH neuronal population not only as a generator of anxiety but also demonstrate their role in the control of hippocampus-dependent memory.


Assuntos
Ansiedade , Núcleo Central da Amígdala/fisiologia , Hormônio Liberador da Corticotropina/fisiologia , Hipocampo/fisiologia , Memória , Neurônios/fisiologia , Animais , Locus Cerúleo/fisiologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Vias Neurais/fisiologia
8.
Exp Neurol ; 298(Pt A): 1-12, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28830762

RESUMO

We investigated the role of amygdala corticotropin-releasing factor (CRF) neurons in the perturbations of descending pain inhibition caused by neuropathic pain. Forced swim increased the tail-flick response latency in uninjured mice, a phenomenon known as stress-induced analgesia (SIA) but did not change the tail-flick response latency in mice with neuropathic pain caused by sciatic nerve constriction. Neuropathic pain also increased the expression of CRF in the central amygdala (CeAmy) and ΔFosB in the dorsal horn of the spinal cord. Next, we injected the CeAmy of CRF-cre mice with cre activated AAV-DREADD (Designer Receptors Exclusively Activated by Designer Drugs) vectors. Activation of CRF neurons by DREADD/Gq did not affect the impaired SIA but inhibition of CRF neurons by DREADD/Gi restored SIA and decreased allodynia in mice with neuropathic pain. The possible downstream circuitry involved in the regulation of SIA was investigated by combined injections of retrograde cre-virus (CAV2-cre) into the locus ceruleus (LC) and cre activated AAV-diphtheria toxin (AAV-FLEX-DTX) virus into the CeAmy. The viral injections were followed by a sciatic nerve constriction ipsilateral or contralateral to the injections. Ablation of amygdala projections to the LC on the side of injury but not on the opposite side, completely restored SIA, decreased allodynia and decreased ΔFosB expression in the spinal cord in mice with neuropathic pain. The possible lateralization of SIA impairment to the side of injury was confirmed by an experiment in which unilateral inhibition of the LC decreased SIA even in uninjured mice. The current view in the field of pain research attributes the process of pain chronification to abnormal functioning of descending pain inhibition. Our results demonstrate that the continuous activity of CRF neurons brought about by persistent pain leads to impaired SIA, which is a symptom of dysregulation of descending pain inhibition. Therefore, an over-activation of amygdala CRF neurons is very likely an important contributing factor for pain chronification.


Assuntos
Tonsila do Cerebelo/metabolismo , Dor Crônica/metabolismo , Hormônio Liberador da Corticotropina/biossíntese , Neurônios/metabolismo , Medição da Dor/métodos , Tonsila do Cerebelo/patologia , Animais , Dor Crônica/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/patologia , Limiar da Dor/fisiologia
9.
Neuroscientist ; 22(5): 521-33, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27621368

RESUMO

Chronic pain is frequently associated with anxiety, depression, and cognitive dysfunction. This review discusses recent work in rodents that contributes to the understanding of their neurobiological links. Brain regions that contain circuits that mediate persistent changes in behavior that are caused by nerve injury or joint inflammation include the rostral anterior cingulate and other parts of the medial prefrontal cortex, the basolateral and central nucleus of the amygdala, and the nucleus accumbens. Functional changes, including increases in the activity within specific neuronal pathways and in the levels of specific synaptic components, that are associated with the behavior changes, or are in some cases necessary for them, have recently been identified. Broadly projecting modulatory systems and widely expressed factors such as cytokines and growth factors also contribute to pain-associated behavior. Integrating these observations and determining their causal relationships is now critical for the identification of therapeutic targets and the design of appropriate interventions.


Assuntos
Tonsila do Cerebelo/fisiopatologia , Comportamento/fisiologia , Hipocampo/fisiopatologia , Vias Neurais/fisiopatologia , Dor/fisiopatologia , Animais , Humanos , Córtex Pré-Frontal/fisiopatologia
10.
J Neurosci ; 34(37): 12304-12, 2014 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-25209272

RESUMO

Pain and depression are frequently associated with and often persist after resolution of an initial injury. Identifying the extent to which depression remains causally associated with ongoing physical discomfort during chronic pain, or becomes independent of it, is an important problem for basic neuroscience and psychiatry. Difficulty in distinguishing between effects of ongoing aversive sensory input and its long-term consequences is a significant roadblock, especially in animal models. To address this relationship between localized physical discomfort and its more global consequences, we investigated cellular and behavioral changes during and after reversing a mouse model of neuropathic pain. Tactile allodynia produced by placing a plastic cuff around the sciatic nerve resolved within several days when the cuff was removed. In contrast, the changes in elevated O-maze, forced-swim, Y-maze spontaneous alternation and novel-object recognition test performance that developed after nerve cuff placement remained for at least 3 weeks after the nerve cuffs were removed, or 10-15 d following complete normalization of mechanical sensitivity. Hippocampal neurogenesis, measured by doublecortin and proliferating cell nuclear antigen expression, was also suppressed after nerve cuff placement and remained suppressed 3 weeks after cuff removal. FosB expression was elevated in the central nucleus of the amygdala and spinal cord dorsal horn only in mice with ongoing allodynia. In contrast, FosB remained elevated in the basolateral amygdala of mice with resolved nociception and persisting behavioral effects. These observations suggest that different processes control tactile hypersensitivity and the behavioral changes and impaired neurogenesis that are associated with neuropathic allodynia.


Assuntos
Ansiedade/fisiopatologia , Depressão/fisiopatologia , Hiperalgesia/fisiopatologia , Transtornos Mentais/fisiopatologia , Neuralgia/fisiopatologia , Neurogênese/fisiologia , Neuropatia Ciática/fisiopatologia , Animais , Ansiedade/etiologia , Comportamento Animal , Depressão/etiologia , Hiperalgesia/complicações , Masculino , Transtornos Mentais/etiologia , Camundongos , Camundongos Endogâmicos C57BL , Neuralgia/complicações , Plasticidade Neuronal , Neuropatia Ciática/complicações , Tato
11.
Proc Natl Acad Sci U S A ; 110(32): 13156-61, 2013 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-23878240

RESUMO

Nociceptive information is modulated by a large number of endogenous signaling agents that change over the course of recovery from injury. This plasticity makes understanding regulatory mechanisms involved in descending inhibition of pain scientifically and clinically important. Neurons that synthesize the neuropeptide TIP39 project to many areas that modulate nociceptive information. These areas are enriched in its receptor, the parathyroid hormone 2 receptor (PTH2R). We previously found that TIP39 affects several acute nociceptive responses, leading us to now investigate its potential role in chronic pain. Following nerve injury, both PTH2R and TIP39 knockout mice developed less tactile and thermal hypersensitivity than controls and returned to baseline sensory thresholds faster. Effects of hindpaw inflammatory injury were similarly decreased in knockout mice. Blockade of α-2 adrenergic receptors increased the tactile and thermal sensitivity of apparently recovered knockout mice, returning it to levels of neuropathic controls. Mice with locus coeruleus (LC) area injection of lentivirus encoding a secreted PTH2R antagonist had a rapid, α-2 reversible, apparent recovery from neuropathic injury similar to the knockout mice. Ablation of LC area glutamatergic neurons led to local PTH2R-ir loss, and barley lectin was transferred from local glutamatergic neurons to GABA interneurons that surround the LC. These results suggest that TIP39 signaling modulates sensory thresholds via effects on glutamatergic transmission to brainstem GABAergic interneurons that innervate noradrenergic neurons. TIP39's normal role may be to inhibit release of hypoalgesic amounts of norepinephrine during chronic pain. The neuropeptide may help maintain central sensitization, which could serve to enhance guarding behavior.


Assuntos
Inflamação/fisiopatologia , Neuralgia/fisiopatologia , Neuropeptídeos/fisiologia , Receptor Tipo 2 de Hormônio Paratireóideo/fisiologia , Animais , Feminino , Neurônios GABAérgicos/metabolismo , Transportador de Glucose Tipo 2/genética , Transportador de Glucose Tipo 2/metabolismo , Ácido Glutâmico/metabolismo , Membro Posterior/patologia , Membro Posterior/fisiopatologia , Hiperalgesia/genética , Hiperalgesia/fisiopatologia , Inflamação/genética , Locus Cerúleo/citologia , Locus Cerúleo/metabolismo , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Neuralgia/genética , Neurônios/metabolismo , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Medição da Dor , Receptor Tipo 2 de Hormônio Paratireóideo/genética , Receptor Tipo 2 de Hormônio Paratireóideo/metabolismo , Receptores Adrenérgicos alfa 2/genética , Receptores Adrenérgicos alfa 2/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
12.
J Comp Neurol ; 521(10): 2373-97, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23296594

RESUMO

The noradrenergic locus coeruleus (LC) regulates arousal, memory, sympathetic nervous system activity, and pain. Forebrain projections to LC have been characterized in rat, cat, and primates, but not systematically in mouse. We surveyed mouse forebrain LC-projecting neurons by examining retrogradely labeled cells following LC iontophoresis of Fluoro-Gold and anterograde LC labeling after forebrain injection of biotinylated dextran amine or viral tracer. Similar to other species, the central amygdalar nucleus (CAmy), anterior hypothalamus, paraventricular nucleus, and posterior lateral hypothalamic area (PLH) provide major LC inputs. By using mice expressing green fluorescent protein in γ-aminobutyric acid (GABA)ergic neurons, we found that more than one-third of LC-projecting CAmy and PLH neurons are GABAergic. LC colocalization of biotinylated dextran amine, following CAmy or PLH injection, with either green fluorescent protein or glutamic acid decarboxylase (GAD)65/67 immunoreactivity confirmed these GABAergic projections. CAmy injection of adeno-associated virus encoding channelrhodopsin-2-Venus showed similar fiber labeling and association with GAD65/67-immunoreactive (ir) and tyrosine hydroxylase (TH)-ir neurons. CAmy and PLH projections were densest in a pericoerulear zone, but many fibers entered the LC proper. Close apposition between CAmy GABAergic projections and TH-ir processes suggests that CAmy GABAergic neurons may directly inhibit noradrenergic principal neurons. Direct LC neuron targeting was confirmed by anterograde transneuronal labeling of LC TH-ir neurons following CAmy or PLH injection of a herpes virus that expresses red fluorescent protein following activation by Cre recombinase in mice that express Cre recombinase in GABAergic neurons. This description of GABAergic projections from the CAmy and PLH to the LC clarifies important forebrain sources of inhibitory control of central nervous system noradrenergic activity.


Assuntos
Vias Aferentes/fisiologia , Neurônios GABAérgicos/fisiologia , Locus Cerúleo/fisiologia , Prosencéfalo/citologia , Animais , Biotina/análogos & derivados , Biotina/metabolismo , Dextranos/metabolismo , Glutamato Descarboxilase/genética , Glutamato Descarboxilase/metabolismo , Proteínas de Fluorescência Verde/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Confocal , Proteínas do Tecido Nervoso/metabolismo , Proteínas/genética , Proteínas/metabolismo , RNA não Traduzido , Estilbamidinas/metabolismo , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/genética , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/metabolismo
13.
Artigo em Inglês | MEDLINE | ID: mdl-23060860

RESUMO

The G-protein coupled parathyroid hormone 2 receptor (PTH2R) is concentrated in endocrine and limbic regions in the forebrain. Its endogenous ligand, tuberoinfundibular peptide of 39 residues (TIP39), is synthesized in only two brain regions, within the posterior thalamus and the lateral pons. TIP39-expressing neurons have a widespread projection pattern, which matches the PTH2R distribution in the brain. Neuroendocrine centers including the preoptic area, the periventricular, paraventricular, and arcuate nuclei contain the highest density of PTH2R-positive networks. The administration of TIP39 and an antagonist of the PTH2R as well as the investigation of mice that lack functional TIP39 and PTH2R revealed the involvement of the PTH2R in a variety of neural and neuroendocrine functions. TIP39 acting via the PTH2R modulates several aspects of the stress response. It evokes corticosterone release by activating corticotropin-releasing hormone-containing neurons in the hypothalamic paraventricular nucleus. Block of TIP39 signaling elevates the anxiety state of animals and their fear response, and increases stress-induced analgesia. TIP39 has also been suggested to affect the release of additional pituitary hormones including arginine-vasopressin and growth hormone. A role of the TIP39-PTH2R system in thermoregulation was also identified. TIP39 may play a role in maintaining body temperature in a cold environment via descending excitatory pathways from the preoptic area. Anatomical and functional studies also implicated the TIP39-PTH2R system in nociceptive information processing. Finally, TIP39 induced in postpartum dams may play a role in the release of prolactin during lactation. Potential mechanisms leading to the activation of TIP39 neurons and how they influence the neuroendocrine system are also described. The unique TIP39-PTH2R neuromodulator system provides the possibility for developing drugs with a novel mechanism of action to control neuroendocrine disorders.

14.
J Neurosci ; 31(49): 18166-79, 2011 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-22159128

RESUMO

Euthermia is critical for mammalian homeostasis. Circuits within the preoptic hypothalamus regulate temperature, with fine control exerted via descending GABAergic inhibition of presympathetic motor neurons that control brown adipose tissue (BAT) thermogenesis and cutaneous vascular tone. The thermoregulatory role of hypothalamic excitatory neurons is less clear. Here we report peptidergic regulation of preoptic glutamatergic neurons that contributes to temperature regulation. Tuberoinfundibular peptide of 39 residues (TIP39) is a ligand for the parathyroid hormone 2 receptor (PTH2R). Both peptide and receptor are abundant in the preoptic hypothalamus. Based on PTH2R and vesicular glutamate transporter 2 (VGlut2) immunolabeling in animals with retrograde tracer injection, PTH2R-containing glutamatergic fibers are presynaptic to neurons projecting from the median preoptic nucleus (MnPO) to the dorsomedial hypothalamus. Transneuronal retrograde pathway tracing with pseudorabies virus revealed connectivity between MnPO VGlut2 and PTH2R neurons and BAT. MnPO injection of TIP39 increased body temperature by 2°C for several hours. Mice lacking TIP39 signaling, either because of PTH2R-null mutation or brain delivery of a PTH2R antagonist had impaired heat production upon cold exposure, but no change in basal temperature and no impairment in response to a hot environment. Thus, TIP39 appears to act on PTH2Rs present on MnPO glutamatergic terminals to regulate their activation of projection neurons and subsequent sympathetic BAT activation. This excitatory mechanism of heat production appears to be activated on demand, during cold exposure, and parallels the tonic inhibitory GABAergic control of body temperature.


Assuntos
Regulação da Temperatura Corporal/efeitos dos fármacos , Temperatura Baixa , Hipotálamo/efeitos dos fármacos , Neuropeptídeos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tecido Adiposo Marrom/efeitos dos fármacos , Tecido Adiposo Marrom/metabolismo , Antagonistas Adrenérgicos beta/farmacologia , Análise de Variância , Animais , Temperatura Corporal/efeitos dos fármacos , Temperatura Corporal/genética , Regulação da Temperatura Corporal/genética , Relação Dose-Resposta a Droga , Interações Medicamentosas , Glutamato Descarboxilase/metabolismo , Proteínas de Fluorescência Verde/genética , Hipotálamo/citologia , Hipotálamo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microinjeções/métodos , Proteínas Associadas aos Microtúbulos/metabolismo , Nadolol/farmacologia , Vias Neurais/fisiologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neuropeptídeos/farmacologia , RNA Mensageiro/metabolismo , Receptor Tipo 2 de Hormônio Paratireóideo/deficiência , Transdução de Sinais/genética , Estilbamidinas/metabolismo , Proteína Vesicular 2 de Transporte de Glutamato/genética , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo , beta-Galactosidase/genética , beta-Galactosidase/metabolismo
15.
Behav Genet ; 41(5): 724-33, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21484271

RESUMO

N-formylpeptide receptor 1 (FPR1) is a G protein-coupled receptor that mediates pro-inflammatory chemotactic responses by phagocytic leukocytes to N-formylpeptides produced by bacteria or mitochondria. Mice lacking Fpr1 (Fpr1 (-/-) mice) have increased susceptibility to challenge with certain bacteria. FPR1 is also a receptor for annexin-1, which mediates the anti-inflammatory effects of glucocorticoids as well as negative feedback by glucocorticoids of the hypothalamic-pituitary-adrenocortical axis. However, homeostatic functions of FPR1 in the neuroendocrine system have not previously been defined. Here we show that in systematic behavioral testing Fpr1 (-/-) mice exhibited increased exploratory activity, reduced anxiety-like behavior, and impaired fear memory, but normal spatial memory and learning capacity. Consistent with this, the homeostatic serum level of corticosterone in Fpr1 (-/-) mice was significantly lower compared with wild-type mice. The data implicate Fpr1 in modulation of anxiety-like behavior and fear memory by regulating glucocorticoid production.


Assuntos
Ansiedade/genética , Medo , Memória , Receptores de Formil Peptídeo/genética , Corticosteroides/metabolismo , Animais , Ansiedade/metabolismo , Comportamento Animal , Modelos Animais de Doenças , Glucocorticoides/metabolismo , Homeostase , Inflamação , Masculino , Aprendizagem em Labirinto , Camundongos , Camundongos Knockout , Mitocôndrias/metabolismo , N-Formilmetionina Leucil-Fenilalanina/metabolismo , Fagócitos/metabolismo
16.
J Comp Neurol ; 518(21): 4375-94, 2010 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-20853513

RESUMO

Neurons in the subparafascicular area at the caudal border of the thalamus that contain the neuropeptide tuberoinfundibular peptide of 39 residues (TIP39) densely innervate several hypothalamic areas, including the paraventricular nucleus (PVN). These areas contain a matching distribution of TIP39's receptor, the parathyroid hormone receptor 2 (PTH2R). Frequent PTH2R coexpression with a vesicular glutamate transporter (VGlut2) suggests that TIP39 could presynaptically regulate glutamate release. By using immunohistochemistry we found CRH-ir neurons surrounded by PTH2R-ir fibers and TIP39-ir axonal projections in the PVN area of the mouse brain. Labeling hypothalamic neuroendocrine neurons by peripheral injection of fluorogold in PTH2R-lacZ knock-in mice showed that most PTH2Rs are on PVN and peri-PVN interneurons and not on neuroendocrine cells. Double fluorescent in situ hybridization revealed a high level of coexpression between PTH2R and VGlut2 mRNA by cells located in the PVN and nearby brain areas. Local TIP39 infusion (100 pmol) robustly increased pCREB-ir in the PVN and adjacent perinuclear zone. It also increased plasma corticosterone and decreased plasma prolactin. These effects of TIP39 on pCREB-ir, corticosterone, and prolactin were abolished by coinfusion of the ionotropic glutamate receptor antagonists 6-cyano-7-nitroquinoxaline-2,3-dione (CNQX) and DL-2-amino-5-phosphonopentanoic acid (AP-5; 30 pmol each) and were absent in PTH2R knockout mice. Basal plasma corticosterone was slightly decreased in TIP39 knockout mice just before onset of their active phase. The present data indicate that the TIP39 ligand/PTH2 receptor system provides facilitatory regulation of the hypothalamic-pituitary-adrenal axis via hypothalamic glutamatergic neurons and that it may regulate other neuroendocrine systems by a similar mechanism.


Assuntos
Ácido Glutâmico/metabolismo , Sistema Hipotálamo-Hipofisário/metabolismo , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Núcleo Hipotalâmico Paraventricular/citologia , Sistema Hipófise-Suprarrenal/metabolismo , Animais , Corticosterona/metabolismo , Hormônio Liberador da Corticotropina/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/citologia , Neuropeptídeos/genética , Receptor Tipo 2 de Hormônio Paratireóideo/genética , Receptor Tipo 2 de Hormônio Paratireóideo/metabolismo , Proteína Vesicular 2 de Transporte de Glutamato/genética , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo
17.
Exp Neurol ; 226(1): 68-83, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20696160

RESUMO

Tuberoinfundibular peptide of 39 residues (TIP39) synthesizing neurons at the caudal border of the thalamus and in the lateral pons project to areas rich in its receptor, the parathyroid hormone 2 receptor (PTH2R). These areas include many involved in processing nociceptive information. Here we examined the potential role of TIP39 signaling in nociception using a PTH2R antagonist (HYWH) and mice with deletion of TIP39's coding sequence or PTH2R null mutation. Intracerebroventricular (icv) infusion of HYWH significantly inhibited nociceptive responses in tail-flick and hot-plate tests and attenuated the nociceptive response to hindpaw formalin injection. TIP39-KO and PTH2R-KO had increased response latency in the 55°C hot-plate test and reduced responses in the hindpaw formalin test. The tail-flick test was not affected in either KO line. Thermal hypoalgesia in KO mice was dose-dependently reversed by systemic administration of the cannabinoid receptor 1 (CB1) antagonist rimonabant, which did not affect nociception in wild-type (WT). Systemic administration of the cannabinoid agonist CP 55,940 did not affect nociception in KO mice at a dose effective in WT. WT mice administered HYWH icv, and both KOs, had significantly increased stress-induced analgesia (SIA). Rimonabant blocked the increased SIA in TIP39-KO, PTH2R-KO or after HYWH infusion. CB1 and FAAH mRNA were decreased and increased, respectively, in the basolateral amygdala of TIP39-KO mice. These data suggest that TIP39 signaling modulates nociception, very likely by inhibiting endocannabinoid circuitry at a supraspinal level. We infer a new central mechanism for endocannabinoid regulation, via TIP39 acting on the PTH2R in discrete brain regions.


Assuntos
Neuropeptídeos/farmacologia , Nociceptores/fisiologia , Dor/fisiopatologia , Transdução de Sinais/fisiologia , Amidoidrolases/metabolismo , Animais , Moduladores de Receptores de Canabinoides/metabolismo , Formaldeído , Hibridização In Situ , Injeções Intraventriculares , Injeções Espinhais , Masculino , Camundongos , Camundongos Knockout , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Neuropeptídeos/administração & dosagem , Nociceptores/efeitos dos fármacos , Dor/patologia , Medição da Dor/efeitos dos fármacos , Piperidinas/farmacologia , Pirazóis/farmacologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptor CB1 de Canabinoide/antagonistas & inibidores , Receptor CB1 de Canabinoide/biossíntese , Receptor Tipo 2 de Hormônio Paratireóideo/biossíntese , Receptor Tipo 2 de Hormônio Paratireóideo/genética , Rimonabanto , Estresse Psicológico/psicologia , Sinapses/fisiologia , Proteína Vesicular 2 de Transporte de Glutamato/biossíntese , Proteína Vesicular 2 de Transporte de Glutamato/genética
18.
Endocrinology ; 148(8): 3666-73, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17463058

RESUMO

The neuroendocrine parvocellular CRH neurons in the paraventricular nucleus (PVN) of the hypothalamus are the main integrators of neural inputs that initiate hypothalamic-pituitary-adrenal (HPA) axis activation. Neuropeptide Y (NPY) expression is prominent within the PVN, and previous reports indicated that NPY stimulates CRH mRNA levels. The purpose of these studies was to examine the participation of NPY receptors in HPA axis activation and determine whether neuroendocrine CRH neurons express NPY receptor immunoreactivity. Infusion of 0.5 nmol NPY into the third ventricle increased plasma corticosterone levels in conscious rats, with the peak of hormone levels occurring 30 min after injection. This increase was prevented by pretreatment with the Y1 receptor antagonist BIBP3226. Immunohistochemistry showed that CRH-immunoreactive neurons coexpressed Y1 receptor immunoreactivity (Y1r-ir) in the PVN, and a majority of these neurons (88.8%) were neuroendocrine as determined by ip injections of FluoroGold. Bilateral infusion of the Y1/Y5 agonist, [leu(31)pro(34)]NPY (110 pmol), into the PVN increased c-Fos and phosphorylated cAMP response element-binding protein expression and elevated plasma corticosterone levels. Increased expression of c-Fos and phosphorylated cAMP response element-binding protein was observed in populations of CRH/Y1r-ir cells. The current findings present a comprehensive study of NPY Y1 receptor distribution and activation with respect to CRH neurons in the PVN. The expression of NPY Y1r-ir by neuroendocrine CRH cells suggests that alterations in NPY release and subsequent activation of NPY Y1 receptors plays an important role in the regulation of the HPA.


Assuntos
Hormônio Liberador da Corticotropina/metabolismo , Neurônios/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Receptores de Neuropeptídeo Y/metabolismo , Animais , Ansiolíticos/farmacologia , Arginina/análogos & derivados , Arginina/farmacologia , Biomarcadores/metabolismo , Proteína de Ligação a CREB/metabolismo , Imuno-Histoquímica , Injeções Intraventriculares , Masculino , Neuropeptídeo Y/análogos & derivados , Neuropeptídeo Y/farmacologia , Núcleo Hipotalâmico Paraventricular/citologia , Fosforilação , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Neuropeptídeo Y/agonistas , Terceiro Ventrículo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA