Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 332
Filtrar
1.
J Neuroinflammation ; 19(1): 288, 2022 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-36463243

RESUMO

BACKGROUND: Adverse pathophysiological and behavioral outcomes related to mild traumatic brain injury (mTBI), posttraumatic stress disorder (PTSD), and chronic pain are common following blast exposure and contribute to decreased quality of life, but underlying mechanisms and prophylactic/treatment options remain limited. The dynorphin/kappa opioid receptor (KOR) system helps regulate behavioral and inflammatory responses to stress and injury; however, it has yet to be investigated as a potential mechanism in either humans or animals exposed to blast. We hypothesized that blast-induced KOR activation mediates adverse outcomes related to inflammation and affective behavioral response. METHODS: C57Bl/6 adult male mice were singly or repeatedly exposed to either sham (anesthesia only) or blast delivered by a pneumatic shock tube. The selective KOR antagonist norBNI or vehicle (saline) was administered 72 h prior to repetitive blast or sham exposure. Serum and brain were collected 10 min or 4 h post-exposure for dynorphin A-like immunoreactivity and cytokine measurements, respectively. At 1-month post-exposure, mice were tested in a series of behavioral assays related to adverse outcomes reported by humans with blast trauma. RESULTS: Repetitive but not single blast exposure resulted in increased brain dynorphin A-like immunoreactivity. norBNI pretreatment blocked or significantly reduced blast-induced increase in serum and brain cytokines, including IL-6, at 4 h post exposure and aversive/anxiety-like behavioral dysfunction at 1-month post-exposure. CONCLUSIONS: Our findings demonstrate a previously unreported role for the dynorphin/KOR system as a mediator of biochemical and behavioral dysfunction following repetitive blast exposure and highlight this system as a potential prophylactic/therapeutic treatment target.


Assuntos
Traumatismos por Explosões , Dinorfinas , Receptores Opioides kappa , Animais , Masculino , Camundongos , Traumatismos por Explosões/complicações , Traumatismos por Explosões/genética , Traumatismos por Explosões/imunologia , Encéfalo/imunologia , Encéfalo/fisiologia , Dinorfinas/genética , Dinorfinas/imunologia , Qualidade de Vida , Receptores Opioides kappa/genética , Receptores Opioides kappa/imunologia
2.
Neuron ; 110(24): 4125-4143.e6, 2022 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-36202097

RESUMO

Social isolation during opioid withdrawal is a major contributor to the current opioid addiction crisis. We find that sociability deficits during protracted opioid withdrawal in mice require activation of kappa opioid receptors (KORs) in the nucleus accumbens (NAc) medial shell. Blockade of release from dynorphin (Pdyn)-expressing dorsal raphe neurons (DRPdyn), but not from NAcPdyn neurons, prevents these deficits in prosocial behaviors. Conversely, optogenetic activation of DRPdyn neurons reproduced NAc KOR-dependent decreases in sociability. Deletion of KORs from serotonin (5-HT) neurons, but not from NAc neurons or dopamine (DA) neurons, prevented sociability deficits during withdrawal. Finally, measurements with the genetically encoded GRAB5-HT sensor revealed that during withdrawal KORs block the NAc 5-HT release that normally occurs during social interactions. These results define a neuromodulatory mechanism that is engaged during protracted opioid withdrawal to induce maladaptive deficits in prosocial behaviors, which in humans contribute to relapse.


Assuntos
Dinorfinas , Serotonina , Humanos , Camundongos , Animais , Dinorfinas/genética , Dinorfinas/metabolismo , Analgésicos Opioides , Dopamina/fisiologia , Receptores Opioides kappa/genética , Receptores Opioides kappa/metabolismo , Entorpecentes , Núcleo Accumbens/metabolismo
3.
Endocrinology ; 163(12)2022 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-36260530

RESUMO

Classic pharmacological studies suggested that endogenous dynorphin-KOR signaling is important for reproductive neuroendocrine regulation. With the seminal discovery of an interconnected network of hypothalamic arcuate neurons co-expressing kisspeptin, neurokinin B, and dynorphin (KNDy neurons), the KNDy hypothesis was developed to explain how gonadotropin-releasing hormone (GnRH) and luteinizing hormone (LH) pulses are generated. Key to this hypothesis is dynorphin released from KNDy neurons acting in a paracrine manner on other KNDy neurons via kappa opioid receptor (KOR) signaling to terminate neural "pulse" events. While in vitro evidence supports this aspect of the KNDy hypothesis, a direct in vivo test of the necessity of KOR signaling in kisspeptin neurons for proper LH secretion has been lacking. We therefore conditionally knocked out KOR selectively from kisspeptin neurons of male and female mice and tested numerous reproductive measures, including in vivo LH pulse secretion. Surprisingly, despite validating successful knockout of KOR in kisspeptin neurons, we found no significant effect of kisspeptin cell-specific deletion of KOR on any measure of puberty, LH pulse parameters, LH surges, follicle-stimulating hormone (FSH) levels, estrous cycles, or fertility. These outcomes suggest that the KNDy hypothesis, while sufficient normally, may not be the only neural mechanism for sculpting GnRH and LH pulses, supported by recent findings in humans and mice. Thus, besides normally acting via KOR in KNDy neurons, endogenous dynorphin and other opioids may, under some conditions, regulate LH and FSH secretion via KOR in non-kisspeptin cells or perhaps via non-KOR pathways. The current models for GnRH and LH pulse generation should be expanded to consider such alternate mechanisms.


Assuntos
Dinorfinas , Kisspeptinas , Humanos , Feminino , Masculino , Camundongos , Animais , Kisspeptinas/metabolismo , Dinorfinas/genética , Dinorfinas/metabolismo , Receptores Opioides kappa/metabolismo , Núcleo Arqueado do Hipotálamo/metabolismo , Maturidade Sexual , Neurocinina B/metabolismo , Hormônio Luteinizante/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Fertilidade/genética , Hormônio Foliculoestimulante/metabolismo
4.
Mol Cell Endocrinol ; 557: 111752, 2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-35973528

RESUMO

Hypothalamic control of reproduction relies on GnRH and kisspeptin (KP) secretions. KP neurons are sensitive to sex steroids and metabolic status and their distribution overlaps with neurons producing apelin, a metabolic hormone known to decrease LH secretion in rats. Here, we observed neuroanatomical contacts between apelin fibers and both KP and GnRH neurons in the hypothalamus of male rodents. Intracerebroventricular apelin infusion for 2 weeks in male mice did not decrease LH levels nor did it affect gene expression for KP, neurokinin B and dynorphin. Finally, increasing apelin concentrations did not modulate Ca2+ levels of cultured GnRH neurons, while 10 µM apelin infusion on forskolin pretreated GnRH neurons revoked a rhythmic activity in 18% of GnRH neurons. These results suggest that acute apelin effect on LH secretion does not involve modulation of gene expression in KP neurons but may affect the secretory activity of GnRH neurons.


Assuntos
Hormônio Liberador de Gonadotropina , Neurocinina B , Animais , Apelina , Receptores de Apelina , Núcleo Arqueado do Hipotálamo/metabolismo , Colforsina/farmacologia , Dinorfinas/genética , Hormônio Liberador de Gonadotropina/metabolismo , Kisspeptinas/metabolismo , Hormônio Luteinizante/metabolismo , Hormônio Luteinizante/farmacologia , Masculino , Camundongos , Neurocinina B/genética , Neurônios/metabolismo , Ratos , Esteroides/metabolismo
5.
Front Neural Circuits ; 16: 908964, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35937204

RESUMO

The olfactory tubercle (OT) is a striatal region that receives olfactory inputs. mRNAs of prodynorphin (Pdyn) and preproenkephalin (Penk), precursors of dynorphins and enkephalins, respectively, are strongly expressed in the striatum. Both produce opioid peptides with various physiological effects such as pain relief and euphoria. Recent studies have revealed that OT has anatomical and cytoarchitectonic domains that play different roles in odor-induced motivated behavior. Neuronal subtypes of the OT can be distinguished by their expression of the dopamine receptors D1 (Drd1) and D2 (Drd2). Here, we addressed whether and which type of opioid peptide precursors the D1- and D2-expressing neurons in the OT express. We used multiple fluorescence in situ hybridization for mRNAs of the opioid precursors and dopamine receptors to characterize mouse OT neurons. Pdyn was mainly expressed by Drd1-expressing cells in the dense cell layer (DCL) of the OT, whereas Penk was expressed primarily by Drd2-expressing cells in the DCL. We also confirmed the presence of a larger population of Pdyn-Penk-Drd1 co-expressing cells in the DCL of the anteromedial OT compared with the anterolateral OT. These observations will help understand whether and how dynorphins and enkephalins in the OT are involved in diverse odor-induced motivated behaviors.


Assuntos
Dinorfinas , Encefalinas , Neurônios/metabolismo , Tubérculo Olfatório/citologia , Precursores de Proteínas , Animais , Corpo Estriado/metabolismo , Dinorfinas/análise , Dinorfinas/genética , Dinorfinas/metabolismo , Encefalinas/análise , Encefalinas/genética , Encefalinas/metabolismo , Hibridização in Situ Fluorescente , Camundongos , Tubérculo Olfatório/metabolismo , RNA Mensageiro/metabolismo , Receptores de Dopamina D1/metabolismo
6.
Acta Physiol (Oxf) ; 236(3): e13882, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36039689

RESUMO

AIM: Endogenous dynorphin signaling via kappa opioid receptors (KORs) plays a key role in producing the depressive and aversive consequences of stress. We investigated the behavioral effects of the dynorphin/KOR system in the ventral pallidum (VP) and studied the underlying mechanisms. METHODS: To investigate the effects of dynorphin on the VP, we conducted behavioral experiments after microinjection of drugs or shRNA and brain-slice electrophysiological recordings. Histological tracing and molecular biological experiments were used to identify the distribution of KORs and the possible sources of dynorphin projections to the VP. RESULTS: An elevated dynorphin concentration and increased KOR activity were observed in the VP after acute stress. Infusion of dynorphin-A into the VP produced depressive-like phenotypes including anhedonia and despair and anxiety behaviors, but did not alter locomotor behavior. Mechanistically, dynorphin had an inhibitory effect on VP neurons-reducing their firing rate and inhibiting excitatory transmission-through direct activation of KORs and modulation of downstream G-protein-gated inwardly rectifying potassium (GIRK) channels and high-voltage gated calcium channels (VGCCs). Tracing revealed direct innervation of VP neurons by dynorphin-positive projections; potential sources of these dynorphinergic projections include the nucleus accumbens, amygdala, and hypothalamus. Blockade of dynorphin/KOR signaling in the VP by drugs or viral knock-down of KORs significantly reduced despair behavior in rats. CONCLUSIONS: Endogenous dynorphinergic modulation of the VP plays a critical role in mediating depressive reactions to stress.


Assuntos
Prosencéfalo Basal , Dinorfinas , Animais , Camundongos , Ratos , Prosencéfalo Basal/metabolismo , Canais de Cálcio , Dinorfinas/genética , Dinorfinas/metabolismo , Dinorfinas/farmacologia , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Potássio/farmacologia , Receptores Opioides kappa/genética , Receptores Opioides kappa/metabolismo , RNA Interferente Pequeno , Depressão , Comportamento Animal , Estresse Fisiológico
7.
Reprod Biol Endocrinol ; 20(1): 91, 2022 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-35729637

RESUMO

BACKGROUND: Kisspeptin released from Kiss-1 neurons in the hypothalamus plays an essential role in the control of the hypothalamic-pituitary-gonadal axis by regulating the release of gonadotropin-releasing hormone (GnRH). In this study, we examined how androgen supplementation affects the characteristics of Kiss-1 neurons. METHODS: We used a Kiss-1-expressing mHypoA-55 cell model that originated from the arcuate nucleus (ARC) of the mouse hypothalamus. These cells are KNDy neurons that co-express neurokinin B (NKB) and dynorphin A (DynA). We stimulated these cells with androgens and examined them. We also examined the ARC region of the hypothalamus in ovary-intact female rats after supplementation with androgens. RESULTS: Stimulation of mHypoA-55 cells with 100 nM testosterone significantly increased Kiss-1 gene expression by 3.20 ± 0.44-fold; testosterone also increased kisspeptin protein expression. The expression of Tac3, the gene encoding NKB, was also increased by 2.69 ± 0.64-fold following stimulation of mHypoA-55 cells with 100 nM testosterone. DynA gene expression in these cells was unchanged by testosterone stimulation, but it was significantly reduced at the protein level. Dihydrotestosterone (DHT) had a similar effect to testosterone in mHypoA-55 cells; kisspeptin and NKB protein expression was significantly increased by DHT, whereas it significantly reduced DynA expression. In ovary-intact female rats, DTH administration significantly increased the gene expression of Kiss-1 and Tac3, but not DynA, in the arcuate nucleus. Exogenous NKB and DynA stimulation failed to modulate Kiss-1 gene expression in mHypoA-55 cells. Unlike androgen stimulation, prolactin stimulation did not modulate kisspeptin, NKB, or DynA protein expression in these cells. CONCLUSIONS: Our observations imply that hyperandrogenemia affects KNDy neurons and changes their neuronal characteristics by increasing kisspeptin and NKB levels and decreasing DynA levels. These changes might cause dysfunction of the hypothalamic-pituitary-gonadal axis.


Assuntos
Dinorfinas , Hiperandrogenismo , Androgênios/metabolismo , Animais , Dinorfinas/genética , Dinorfinas/metabolismo , Dinorfinas/farmacologia , Feminino , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Hormônio Liberador de Gonadotropina/farmacologia , Hiperandrogenismo/metabolismo , Hipotálamo/metabolismo , Kisspeptinas/genética , Kisspeptinas/metabolismo , Camundongos , Neurocinina B/genética , Neurocinina B/metabolismo , Neurocinina B/farmacologia , Neurônios/metabolismo , Ratos , Taquicininas , Testosterona/metabolismo , Testosterona/farmacologia
8.
Brain Res Bull ; 175: 158-167, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34339779

RESUMO

Fabry disease (FD) is an X-linked inherited disorder characterized by glycosphingolipid accumulation due to deficiency of α-galactosidase A (α-Gal A) enzyme. Chronic pain and mood disorders frequently coexist in FD clinical setting, however underlying pathophysiologic mechanisms are still unclear. Here we investigated the mechanical and thermal sensitivity in α-Gal A (-/0) hemizygous male and the α-Gal A (-/-) homozygous female mice. We also characterized the gene expression of dynorphinergic, nociceptinergic and CRFergic systems, known to be involved in pain control and mood disorders, in the prefrontal cortex, amygdala and thalamus of α-Gal A (-/0) hemizygous male and the α-Gal A (-/-) homozygous female mice. Moreover, KOP receptor protein levels were evaluated in the same areas. Fabry knock-out male, but not female, mice displayed a decreased pain threshold in both mechanical and thermal tests compared to their wild type littermates. In the amygdala and prefrontal cortex, we observed a decrease of pDYN mRNA levels in males, whereas an increase was assessed in females, thus suggesting sex-related dysregulation of stress coping and pain mechanisms. Elevated mRNA levels for pDYN/KOP and CRF/CRFR1 systems were observed in male and female thalamus, a critical crossroad for both painful signals and cognitive/emotional processes. KOP receptor protein level changes assessed in the investigated areas, appeared mostly in agreement with KOP gene expression alterations. Our data suggest that α-Gal A enzyme deficiency in male and female mice is associated with distinct neuropeptide gene and protein expression dysregulations of investigated systems, possibly related to the neuroplasticity underlying the neurological features of FD.


Assuntos
Comportamento Animal , Doença de Fabry/psicologia , Neuropeptídeos/metabolismo , Nociceptividade , Animais , Química Encefálica/genética , Hormônio Liberador da Corticotropina , Dinorfinas/genética , Feminino , Expressão Gênica , Masculino , Camundongos , Camundongos Knockout , Nociceptores , Limiar da Dor , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores Opioides kappa/genética , Caracteres Sexuais
9.
Peptides ; 142: 170546, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33794282

RESUMO

Metabolic stress resulting from either lack or excess of nutrients often causes infertility in both sexes. Kisspeptin-neurokinin B-dynorphin A (KNDy) neurons in the arcuate nucleus (ARC) has been suggested to be a key players in reproduction via direct stimulation of the pulsatile gonadotropin-releasing hormone (GnRH) and subsequent gonadotropin release in mammalian species. In this study, we investigated the effect of high-fat diet (HFD) on hypothalamic KNDy gene expression to examine the pathogenic mechanism underlying obesity-induced infertility in male and female rats. Male and female rats at 7 weeks of age were fed with either a standard or HFD for 4 months. In the male rats, the HFD caused a significant suppression of ARC Kiss1 and Pdyn gene expressions, but did not affect the plasma luteinizing hormone (LH) levels and sizes of the morphology of the testis and epididymis. In the female rats, 58% of the HFD-fed female rats exhibited irregular estrous cycles, whereas the remaining rats showed regular cycles. Two of the 10 rats that showed HFD-induced irregular estrous cycles showed profound suppression of LH pulse frequency and the number of ARC Kiss1-expressing cells, whereas the other females showed normal LH pulses and ARC Kiss1 expression. Our finding shows that suppression of ARC Kiss1 expression might be the initial pathological change of hypogonadotropic hypogonadism in HFD-fed male rats, while the obese-related infertility in the female rats may be mainly induced by KNDy-independent pathways. Taken together, ARC kisspeptin neurons in male rats may be susceptible to HFD-induced obesity compared with those in female rats.


Assuntos
Núcleo Arqueado do Hipotálamo/patologia , Hormônios Esteroides Gonadais/farmacologia , Hipogonadismo/patologia , Hormônio Luteinizante/metabolismo , Doenças Metabólicas/complicações , Neurônios/patologia , Obesidade/complicações , Animais , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/metabolismo , Dieta Hiperlipídica , Dinorfinas/genética , Dinorfinas/metabolismo , Feminino , Hipogonadismo/etiologia , Hipogonadismo/metabolismo , Kisspeptinas/genética , Kisspeptinas/metabolismo , Masculino , Neurocinina B/genética , Neurocinina B/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Ratos , Ratos Wistar
10.
Elife ; 102021 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-33667158

RESUMO

Maintaining stable body temperature through environmental thermal stressors requires detection of temperature changes, relay of information, and coordination of physiological and behavioral responses. Studies have implicated areas in the preoptic area of the hypothalamus (POA) and the parabrachial nucleus (PBN) as nodes in the thermosensory neural circuitry and indicate that the opioid system within the POA is vital in regulating body temperature. In the present study we identify neurons projecting to the POA from PBN expressing the opioid peptides dynorphin and enkephalin. Using mouse models, we determine that warm-activated PBN neuronal populations overlap with both prodynorphin (Pdyn) and proenkephalin (Penk) expressing PBN populations. Here we report that in the PBN Prodynorphin (Pdyn) and Proenkephalin (Penk) mRNA expressing neurons are partially overlapping subsets of a glutamatergic population expressing Solute carrier family 17 (Slc17a6) (VGLUT2). Using optogenetic approaches we selectively activate projections in the POA from PBN Pdyn, Penk, and VGLUT2 expressing neurons. Our findings demonstrate that Pdyn, Penk, and VGLUT2 expressing PBN neurons are critical for physiological and behavioral heat defense.


Assuntos
Encefalinas/metabolismo , Núcleos Parabraquiais/fisiologia , Precursores de Proteínas/metabolismo , Animais , Dinorfinas/genética , Dinorfinas/metabolismo , Encefalinas/genética , Feminino , Temperatura Alta , Masculino , Camundongos , Camundongos Transgênicos , Optogenética , Área Pré-Óptica/fisiologia , Precursores de Proteínas/genética
11.
Acupunct Med ; 39(5): 491-500, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33406849

RESUMO

OBJECTIVE: To explore the effects and mechanism of action of electroacupuncture (EA) in a rat model of pubertal polycystic ovary syndrome (PCOS). METHODS: Female offspring of Sprague-Dawley rats receiving dihydrotestosterone (DHT) during pregnancy (days 16-19), as a model of prenatal androgenization, were divided randomly into three groups: model group (M), EA group, and sham acupuncture (SA) group (n = 8 each). A normal (N) group comprising female offspring of healthy pregnant rats not receiving DHT (n = 8) was added. EA was administered at CV6 and bilateral SP6/ST36 with 2 Hz frequency and 2 mA intensity. SA consisted of superficial needling at different locations without electrical stimulation. RESULTS: EA improved the disturbed estrous cycles, while it could not be concluded that SA was effective in this respect. EA improved ovarian morphology including the number of corpora lutea and area of the ovary, whereas SA did not. However, both EA and SA attenuated the increased luteinizing hormone and decreased estradiol and gonadotropin-releasing hormone levels in the serum of PCOS model rats. Levels of testosterone, follicle-stimulating hormone, and progesterone did not significantly differ between groups. EA and SA alleviated the upregulation of kisspeptin protein and mRNA levels in the hypothalamus and kisspeptin protein level in the arcuate nucleus (ARC). No differences were found between groups in protein or mRNA expression of dynorphin (DYN) or neurokinin B (NKB) in the hypothalamus. Co-expression of kisspeptin, NKB, and DYN were observed in ARC. The GnRH level in the median eminence decreased and could be rescued by EA and SA. Intriguingly, kisspeptin levels in the granulosa cells of the ovary decreased in the model group and could be rescued by EA but not SA. Levels of kisspeptin, NKB, and DYN protein and mRNA in the ovary did not differ between any groups. CONCLUSION: Both EA and SA appeared to improve symptoms of PCOS at puberty by modulating the kisspeptin system in the hypothalamus. EA also had an effect on ovarian kisspeptin expression and a more comprehensive effect with respect to improving PCOS at puberty than SA.


Assuntos
Eletroacupuntura , Kisspeptinas/metabolismo , Síndrome do Ovário Policístico/metabolismo , Síndrome do Ovário Policístico/terapia , Pontos de Acupuntura , Animais , Dinorfinas/genética , Dinorfinas/metabolismo , Ciclo Estral , Feminino , Hormônio Foliculoestimulante/metabolismo , Humanos , Hipotálamo/metabolismo , Kisspeptinas/genética , Hormônio Luteinizante/metabolismo , Neurocinina B/genética , Neurocinina B/metabolismo , Ovário/metabolismo , Síndrome do Ovário Policístico/genética , Síndrome do Ovário Policístico/fisiopatologia , Gravidez , Puberdade/metabolismo , Ratos , Ratos Sprague-Dawley
12.
Pain ; 162(1): 203-218, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33045156

RESUMO

Neonatal tissue damage can have long-lasting effects on nociceptive processing in the central nervous system, which may reflect persistent injury-evoked alterations to the normal balance between synaptic inhibition and excitation in the spinal dorsal horn. Spinal dynorphin-lineage (pDyn) neurons are part of an inhibitory circuit which limits the flow of nociceptive input to the brain and is disrupted by neonatal tissue damage. To identify the potential molecular underpinnings of this disruption, an unbiased single-nucleus RNAseq analysis of adult mouse spinal pDyn cells characterized this population in depth and then identified changes in gene expression evoked by neonatal hindpaw incision. The analysis revealed 11 transcriptionally distinct subpopulations (ie, clusters) of dynorphin-lineage cells, including both inhibitory and excitatory neurons. Investigation of injury-evoked differential gene expression identified 15 genes that were significantly upregulated or downregulated in adult pDyn neurons from neonatally incised mice compared with naive littermate controls, with both cluster-specific and pan-neuronal transcriptional changes observed. Several of the identified genes, such as Oxr1 and Fth1 (encoding ferritin), were related to the cellular stress response. However, the relatively low number of injury-evoked differentially expressed genes also suggests that posttranscriptional regulation within pDyn neurons may play a key role in the priming of developing nociceptive circuits by early-life injury. Overall, the findings reveal novel insights into the molecular heterogeneity of a key population of dorsal horn interneurons that has previously been implicated in the suppression of mechanical pain and itch.


Assuntos
Dinorfinas , Corno Dorsal da Medula Espinal , Animais , Dinorfinas/genética , Interneurônios , Camundongos , Neurônios , Dor , Células do Corno Posterior
13.
Mol Psychiatry ; 26(6): 2187-2199, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-32099099

RESUMO

Excessive alcohol drinking has been shown to modify brain circuitry to predispose individuals for future alcohol abuse. Previous studies have implicated the central nucleus of the amygdala (CeA) as an important site for mediating the somatic symptoms of withdrawal and for regulating alcohol intake. In addition, recent work has established a role for both the Kappa Opioid Receptor (KOR) and its endogenous ligand dynorphin in mediating these processes. However, it is unclear whether these effects are due to dynorphin or KOR arising from within the CeA itself or other input brain regions. To directly examine the role of preprodynorphin (PDYN) and KOR expression in CeA neurons, we performed region-specific conditional knockout of these genes and assessed the effects on the Drinking in the Dark (DID) and Intermittent Access (IA) paradigms. Conditional gene knockout resulted in sex-specific responses wherein PDYN knockout decreased alcohol drinking in both male and female mice, whereas KOR knockout decreased drinking in males only. We also found that neither PDYN nor KOR knockout protected against anxiety caused by alcohol drinking. Lastly, a history of alcohol drinking did not alter synaptic transmission in PDYN neurons in the CeA of either sex, but excitability of PDYN neurons was increased in male mice only. Taken together, our findings indicate that PDYN and KOR signaling in the CeA plays an important role in regulating excessive alcohol consumption and highlight the need for future studies to examine how this is mediated through downstream effector regions.


Assuntos
Alcoolismo , Núcleo Central da Amígdala , Consumo de Bebidas Alcoólicas/genética , Animais , Núcleo Central da Amígdala/metabolismo , Dinorfinas/genética , Dinorfinas/metabolismo , Feminino , Masculino , Camundongos , Receptores Opioides kappa/genética , Receptores Opioides kappa/metabolismo
14.
Sci Rep ; 10(1): 3821, 2020 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-32123224

RESUMO

The neuronal regulation of metabolic and behavioral responses to different diets and feeding regimens is an important research area. Herein, we investigated if the opioid peptide dynorphin modulates feeding behavior and metabolism. Mice lacking dynorphin peptides (KO) were exposed to either a normal diet (ND) or a high-fat diet (HFD) for a period of 12 weeks. Additionally, mice had either time-restricted (TR) or ad libitum (AL) access to food. Body weight, food intake and blood glucose levels were monitored throughout the 12-week feeding schedule. Brain samples were analyzed by immunohistochemistry to detect changes in the expression levels of hypothalamic peptides. As expected, animals on HFD or having AL access to food gained more weight than mice on ND or having TR access. Unexpectedly, KO females on TR HFD as well as KO males on AL ND or AL HFD demonstrated a significantly increased body weight gain compared to the respective WT groups. The calorie intake differed only marginally between the genotypes: a significant difference was present in the female ND AL group, where dynorphin KO mice ate more than WT mice. Although female KO mice on a TR feeding regimen consumed a similar amount of food as WT controls, they displayed significantly higher levels of blood glucose. We observed significantly reduced levels of hypothalamic orexigenic peptides neuropeptide Y (NPY) and orexin-A in KO mice. This decrease became particularly pronounced in the HFD groups and under AL condition. The kappa opiod receptor (KOR) levels were higher after HFD compared to ND feeding in the ventral pallidum of WT mice. We hypothesize that HFD enhances dynorphin signaling in this hedonic center to maintain energy homeostasis, therefore KO mice have a more pronounced phenotype in the HFD condition due to the lack of it. Our data suggest that dynorphin modulates metabolic changes associated with TR feeding regimen and HFD consumption. We conclude that the lack of dynorphin causes uncoupling between energy intake and body weight gain in mice; KO mice maintained on HFD become overweight despite their normal food intake. Thus, using kappa opioid receptor agonists against obesity could be considered as a potential treatment strategy.


Assuntos
Encéfalo/metabolismo , Dinorfinas/metabolismo , Comportamento Alimentar , Animais , Glicemia/metabolismo , Peso Corporal , Dieta Hiperlipídica/efeitos adversos , Dinorfinas/deficiência , Dinorfinas/genética , Feminino , Técnicas de Inativação de Genes , Masculino , Camundongos
15.
Endocrinology ; 161(4)2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32031594

RESUMO

Polycystic ovary syndrome (PCOS), a common reproductive disorder in women, is characterized by hyperandrogenemia, chronic anovulation, cystic ovarian follicles, and luteinizing hormone (LH) hyper-pulsatility, but the pathophysiology isn't completely understood. We recently reported a novel mouse model of PCOS using chronic letrozole (LET; aromatase inhibitor). Letrozole-treated females demonstrate multiple PCOS-like phenotypes, including polycystic ovaries, anovulation, and elevated circulating testosterone and LH, assayed in "one-off" measures. However, due to technical limitations, in vivo LH pulsatile secretion, which is elevated in PCOS women, was not previously studied, nor were the possible changes in reproductive neurons. Here, we used recent technical advances to examine in vivo LH pulse dynamics of freely moving LET female mice versus control and ovariectomized (OVX) mice. We also determined whether neural gene expression of important reproductive regulators such as kisspeptin, neurokinin B (NKB), and dynorphin, is altered in LET females. Compared to controls, LET females exhibited very rapid, elevated in vivo LH pulsatility, with increased pulse frequency, amplitude, and basal levels, similar to PCOS women. Letrozole-treated mice also had markedly elevated Kiss1, Tac2, and Pdyn expression and increased Kiss1 neuronal activation in the hypothalamic arcuate nucleus. Notably, the hyperactive LH pulses and increased kisspeptin neuron measures of LET mice were not as elevated as OVX females. Our findings indicate that LET mice, like PCOS women, have markedly elevated LH pulsatility, which likely drives increased androgen secretion. Increased hypothalamic kisspeptin and NKB levels may be fundamental contributors to the hyperactive LH pulse secretion in the LET PCOS-like condition and, perhaps, in PCOS women.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Kisspeptinas/metabolismo , Hormônio Luteinizante/sangue , Neurocinina B/metabolismo , Síndrome do Ovário Policístico/metabolismo , Animais , Inibidores da Aromatase , Modelos Animais de Doenças , Dinorfinas/genética , Dinorfinas/metabolismo , Feminino , Expressão Gênica , Kisspeptinas/genética , Letrozol , Camundongos , Neurocinina B/genética , Neurônios/metabolismo , Síndrome do Ovário Policístico/sangue , Síndrome do Ovário Policístico/induzido quimicamente
16.
J Chem Neuroanat ; 102: 101705, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31669432

RESUMO

Damaraland mole rats (Fukomys damarensis) are cooperatively breeding, subterranean mammals, which exhibit high reproductive skew. Reproduction is monopolized by the dominant female of the group, while subordinates are physiologically suppressed. The blockade of reproduction results from an inhibition of ovulation, which is caused by inadequate secretion of luteinizing hormone (LH) from the pituitary, which in turn might be brought about by a disruption of the normal GnRH secretion from the hypothalamus. The neuropeptides dynorphin and neurokinin B are expressed together with kisspeptin in a subpopulation of neurons in the arcuate nucleus (ARC). This neuron population is termed KNDy neurons and is considered to constitute the GnRH pulse generator. To assess whether dynorphin (encoded by the Pdyn gene) and neurokinin B (NKB, encoded by the Tac3 gene) are involved in the mechanism of reproductive suppression we investigated the distribution and gene expression of Pdyn and Tac3 by means of in situ hybridisation in wild-caught female Damaraland mole-rats with different reproductive status. In both reproductive phenotypes, substantial Pdyn expression was found in several brain regions of the telencephalon including the cerebral cortex, the striatum, the hippocampus, the amygdala and the olfactory tubercle. Within the hypothalamus Pdyn expression occurred in the paraventricular nucleus, the dorsomedial nucleus, the supraoptic nucleus, the ventromedial nucleus and the ARC. Prominent Tac3 expression was found in the habenula, the bed nucleus of the stria terminalis, the cerebral cortex, the striatum, the hippocampus, the amygdala, the dorsomedial nucleus, the ARC and the lateral mammillary nucleus. Quantification of the gene expression levels in the ARC revealed decreased Pdyn and increased Tac3 expression in breeding compared to nonbreeding females. This suggests that both neuropeptides play a role in the regulation of reproduction in Damaraland mole-rats. Their exact role in mediating the inhibition of GnRH release in nonbreeding females remains to be determined.


Assuntos
Encéfalo/metabolismo , Dinorfinas/metabolismo , Regulação da Expressão Gênica , Neurocinina B/metabolismo , Reprodução/fisiologia , Animais , Dinorfinas/genética , Feminino , Ratos-Toupeira , Neurocinina B/genética , Neurônios/metabolismo , Fosforilação
17.
EMBO Mol Med ; 11(10): e9963, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31486590

RESUMO

Focal epilepsy represents one of the most common chronic CNS diseases. The high incidence of drug resistance, devastating comorbidities, and insufficient responsiveness to surgery pose unmet medical challenges. In the quest of novel, disease-modifying treatment strategies of neuropeptides represent promising candidates. Here, we provide the "proof of concept" that gene therapy by adeno-associated virus (AAV) vector transduction of preprodynorphin into the epileptogenic focus of well-accepted mouse and rat models for temporal lobe epilepsy leads to suppression of seizures over months. The debilitating long-term decline of spatial learning and memory is prevented. In human hippocampal slices obtained from epilepsy surgery, dynorphins suppressed seizure-like activity, suggestive of a high potential for clinical translation. AAV-delivered preprodynorphin expression is focally and neuronally restricted and release is dependent on high-frequency stimulation, as it occurs at the onset of seizures. The novel format of "release on demand" dynorphin delivery is viewed as a key to prevent habituation and to minimize the risk of adverse effects, leading to long-term suppression of seizures and of their devastating sequel.


Assuntos
Dinorfinas/farmacologia , Epilepsia do Lobo Temporal/terapia , Expressão Gênica , Terapia Genética/métodos , Neurotransmissores/farmacologia , Animais , Dependovirus/genética , Modelos Animais de Doenças , Dinorfinas/genética , Regulação da Expressão Gênica , Vetores Genéticos , Humanos , Camundongos , Modelos Teóricos , Neurotransmissores/genética , Técnicas de Cultura de Órgãos , Ratos , Transdução Genética , Resultado do Tratamento
18.
Vitam Horm ; 111: 17-47, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31421700

RESUMO

Dynorphin is a neuropeptide involved in pain, addiction and mood regulation. It exerts its activity by binding to the kappa opioid receptor (KOP) which belongs to the large family of G protein-coupled receptors. The dynorphin peptide was discovered in 1975, while its receptor was cloned in 1993. This review will describe: (a) the activities and physiological functions of dynorphin and its receptor, (b) early structure-activity relationship studies performed before cloning of the receptor (mostly pharmacological and biophysical studies of peptide analogues), (c) structure-activity relationship studies performed after cloning of the receptor via receptor mutagenesis and the development of recombinant receptor expression systems, (d) structural biology of the opiate receptors culminating in X-ray structures of the four opioid receptors in their inactive state and structures of MOP and KOP receptors in their active state. X-ray and EM structures are combined with NMR data, which gives complementary insight into receptor and peptide dynamics. Molecular modeling greatly benefited from the availability of atomic resolution 3D structures of receptor-ligand complexes and an example of the strategy used to model a dynorphin-KOP receptor complex using NMR data will be described. These achievements have led to a better understanding of the complex dynamics of KOP receptor activation and to the development of new ligands and drugs.


Assuntos
Dinorfinas/química , Dinorfinas/fisiologia , Receptores Opioides/química , Receptores Opioides/fisiologia , Sequência de Aminoácidos , Animais , Clonagem Molecular , Dinorfinas/genética , Humanos , Modelos Moleculares , Estrutura Molecular , Mutagênese Sítio-Dirigida , Dor , Ligação Proteica , Receptores Opioides/genética , Relação Estrutura-Atividade , Transtornos Relacionados ao Uso de Substâncias
19.
Pain ; 160(10): 2380-2397, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31166300

RESUMO

Mounting evidence suggests that the spinal dorsal horn (SDH) contains multiple subpopulations of inhibitory interneurons that play distinct roles in somatosensory processing, as exemplified by the importance of spinal dynorphin-expressing neurons for the suppression of mechanical pain and chemical itch. Although it is clear that GABAergic transmission in the SDH undergoes significant alterations during early postnatal development, little is known about the maturation of discrete inhibitory "microcircuits" within the region. As a result, the goal of this study was to elucidate the gene expression profile of spinal dynorphin (pDyn)-lineage neurons throughout life. We isolated nuclear RNA specifically from pDyn-lineage SDH interneurons at postnatal days 7, 21, and 80 using the Isolation of Nuclei Tagged in Specific Cell Types (INTACT) technique, followed by RNA-seq analysis. Over 650 genes were ≥2-fold enriched in adult pDyn nuclei compared with non-pDyn spinal cord nuclei, including targets with known relevance to pain such as galanin (Gal), prepronociceptin (Pnoc), and nitric oxide synthase 1 (Nos1). In addition, the gene encoding a membrane-bound guanylate cyclase, Gucy2d, was identified as a novel and highly selective marker of the pDyn population within the SDH. Differential gene expression analysis comparing pDyn nuclei across the 3 ages revealed sets of genes that were significantly upregulated (such as Cartpt, encoding cocaine- and amphetamine-regulated transcript peptide) or downregulated (including Npbwr1, encoding the receptor for neuropeptides B/W) during postnatal development. Collectively, these results provide new insight into the potential molecular mechanisms underlying the known age-dependent changes in spinal nociceptive processing and pain sensitivity.


Assuntos
Dinorfinas/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Perfil Genético , Corno Dorsal da Medula Espinal/crescimento & desenvolvimento , Transcrição Gênica/genética , Animais , Feminino , Masculino , Camundongos , Camundongos Transgênicos
20.
J Clin Endocrinol Metab ; 104(10): 4304-4318, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31132118

RESUMO

CONTEXT: Kisspeptin-neurokinin B (NKB)-dynorphin neurons are critical regulators of the hypothalamic-pituitary-gonadal axis. NKB and dynorphin are hypothesized to influence the frequency of GnRH pulses, whereas kisspeptin is hypothesized to be a generator of the GnRH pulse. How these neuropeptides interact remains unclear. OBJECTIVE: To probe the role of NKB in GnRH pulse generation and to determine the interactions between NKB, kisspeptin, and dynorphin in humans and mice with a complete absence of NKB. DESIGN: Case/control. SETTING: Academic medical center. PARTICIPANTS: Members of a consanguineous family bearing biallelic loss-of-function mutations in the gene encoding NKB and NKB-deficient mice. INTERVENTIONS: Frequent blood sampling to characterize neuroendocrine profile and administration of kisspeptin, GnRH, and naloxone, a nonspecific opioid receptor antagonist used to block dynorphin. MAIN OUTCOME MEASURES: LH pulse characteristics. RESULTS: Humans lacking NKB demonstrate slow LH pulse frequency, which can be increased by opioid antagonism. Mice lacking NKB also demonstrate impaired LH secretion, which can be augmented with an identical pharmacologic manipulation. Both mice and humans with NKB deficiency respond to exogenous kisspeptin. CONCLUSION: The preservation of LH pulses in the absence of NKB and dynorphin signaling suggests that both peptides are dispensable for GnRH pulse generation and kisspeptin responsiveness. However, NKB and dynorphin appear to have opposing roles in the modulation of GnRH pulse frequency.


Assuntos
Dinorfinas/genética , Hipogonadismo/genética , Kisspeptinas/genética , Hormônio Luteinizante/administração & dosagem , Neurocinina B/genética , Transdução de Sinais/efeitos dos fármacos , Centros Médicos Acadêmicos , Adolescente , Adulto , Animais , Estudos de Casos e Controles , Criança , Modelos Animais de Doenças , Feminino , Hormônio Liberador de Gonadotropina/administração & dosagem , Humanos , Hipogonadismo/sangue , Hipogonadismo/tratamento farmacológico , Camundongos , Camundongos Knockout , Antagonistas de Entorpecentes/administração & dosagem , Neurônios/efeitos dos fármacos , Substância P/metabolismo , Resultado do Tratamento , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...