Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
1.
Cell Mol Gastroenterol Hepatol ; 14(5): 1025-1051, 2022.
Article in English | MEDLINE | ID: mdl-35835391

ABSTRACT

BACKGROUND & AIMS: Efforts to characterize the signaling mechanisms that underlie gastroenteropancreatic neoplasms (GEP-NENs) are precluded by a lack of comprehensive models that recapitulate pathogenesis. Investigation into a potential cell-of-origin for gastrin-secreting NENs revealed a non-cell autonomous role for loss of menin in neuroendocrine cell specification, resulting in an induction of gastrin in enteric glia. Here, we investigated the hypothesis that cell autonomous Men1 inactivation in glial fibrillary acidic protein (GFAP)-expressing cells induced neuroendocrine differentiation and tumorigenesis. METHODS: Transgenic GFAPΔMen1 mice were generated by conditional GFAP-directed Men1 deletion in GFAP-expressing cells. Cre specificity was confirmed using a tdTomato reporter. GFAPΔMen1 mice were evaluated for GEP-NEN development and neuroendocrine cell hyperplasia. Small interfering RNA-mediated Men1 silencing in a rat enteric glial cell line was performed in parallel. RESULTS: GFAPΔMen1 mice developed pancreatic NENs, in addition to pituitary prolactinomas that phenocopied the human MEN1 syndrome. GFAPΔMen1 mice exhibited gastric neuroendocrine hyperplasia that coincided with a significant loss of GFAP expression. Men1 deletion induced loss of glial-restricted progenitor lineage markers and an increase in neuroendocrine genes, suggesting a reprogramming of GFAP+ cells. Deleting Kif3a, a mediator of Hedgehog signaling, in GFAP-expressing cells attenuated neuroendocrine hyperplasia by restricting the neuroendocrine cell fate. Similar results in the pancreas were observed when Sox10 was used to delete Men1. CONCLUSIONS: GFAP-directed Men1 inactivation exploits glial cell plasticity in favor of neuroendocrine differentiation.


Subject(s)
Cell Plasticity , Neuroglia , Animals , Mice , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Plasticity/genetics , Cell Plasticity/physiology , Gastrins , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Hedgehog Proteins , Hyperplasia/pathology , Multiple Endocrine Neoplasia Type 1/genetics , Multiple Endocrine Neoplasia Type 1/metabolism , Multiple Endocrine Neoplasia Type 1/pathology , Neuroendocrine Cells/metabolism , Neuroendocrine Cells/pathology , Neuroendocrine Cells/physiology , Neuroglia/metabolism , Proto-Oncogene Proteins , RNA, Small Interfering
2.
STAR Protoc ; 2(3): 100787, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34485946

ABSTRACT

The hypothalamic magnocellular neuroendocrine cells (MNCs) project to the posterior pituitary (PPi), regulating reproduction and fluid homeostasis. It has been challenging to selectively label and manipulate MNCs, as they are intermingled with parvocellular neuroendocrine cells projecting to the median eminence. Here, we provide a step-by-step protocol for specifically targeting the MNCs by infusing retrograde viral tracers into the PPi. When combined with optogenetics, chemogenetics, and transgenic animals, this approach allows cell-type-specific manipulation of MNCs in multiple sites for functional dissection. For complete details on the use and execution of this protocol, please refer to Zhang et al. (2021) and Tang et al. (2020).


Subject(s)
Hypothalamus/cytology , Neuroendocrine Cells , Optogenetics/methods , Pituitary Gland, Posterior/cytology , Animals , Animals, Genetically Modified , Male , Median Eminence/cytology , Nerve Net/cytology , Nerve Net/physiology , Neuroendocrine Cells/cytology , Neuroendocrine Cells/physiology , Rats , Rats, Sprague-Dawley
4.
Neuron ; 109(19): 3119-3134.e5, 2021 10 06.
Article in English | MEDLINE | ID: mdl-34411513

ABSTRACT

Transformation of flat membrane into round vesicles is generally thought to underlie endocytosis and produce speed-, amount-, and vesicle-size-specific endocytic modes. Visualizing depolarization-induced exocytic and endocytic membrane transformation in live neuroendocrine chromaffin cells, we found that flat membrane is transformed into Λ-shaped, Ω-shaped, and O-shaped vesicles via invagination, Λ-base constriction, and Ω-pore constriction, respectively. Surprisingly, endocytic vesicle formation is predominantly from not flat-membrane-to-round-vesicle transformation but calcium-triggered and dynamin-mediated closure of (1) Ω profiles formed before depolarization and (2) fusion pores (called kiss-and-run). Varying calcium influxes control the speed, number, and vesicle size of these pore closures, resulting in speed-specific slow (more than ∼6 s), fast (less than ∼6 s), or ultrafast (<0.6 s) endocytosis, amount-specific compensatory endocytosis (endocytosis = exocytosis) or overshoot endocytosis (endocytosis > exocytosis), and size-specific bulk endocytosis. These findings reveal major membrane transformation mechanisms underlying endocytosis, diverse endocytic modes, and exocytosis-endocytosis coupling, calling for correction of the half-a-century concept that the flat-to-round transformation predominantly mediates endocytosis after physiological stimulation.


Subject(s)
Chromaffin Cells/physiology , Chromaffin Cells/ultrastructure , Endocytosis/physiology , Neuroendocrine Cells/physiology , Neuroendocrine Cells/ultrastructure , Animals , Calcium Signaling , Cattle , Cell Fusion , Cell Membrane/physiology , Cell Membrane/ultrastructure , Computer Systems , Dynamins/physiology , Exocytosis/physiology , Membrane Fusion , Primary Cell Culture , Synaptic Vesicles/metabolism
5.
Science ; 371(6524): 52-57, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33384370

ABSTRACT

Neuroendocrine (NE) cells are epithelial cells that possess many of the characteristics of neurons, including the presence of secretory vesicles and the ability to sense environmental stimuli. The normal physiologic functions of solitary airway NE cells remain a mystery. We show that mouse and human airway basal stem cells sense hypoxia. Hypoxia triggers the direct differentiation of these stem cells into solitary NE cells. Ablation of these solitary NE cells during hypoxia results in increased epithelial injury, whereas the administration of the NE cell peptide CGRP rescues this excess damage. Thus, we identify stem cells that directly sense hypoxia and respond by differentiating into solitary NE cells that secrete a protective peptide that mitigates hypoxic injury.


Subject(s)
Cell Differentiation , Hypoxia/pathology , Neuroendocrine Cells/physiology , Oxygen/physiology , Stem Cells/physiology , Trachea/cytology , Anaerobiosis , Animals , Calcitonin Gene-Related Peptide/metabolism , Calcitonin Gene-Related Peptide/pharmacology , Calcitonin Receptor-Like Protein/metabolism , Cell Count , Gene Deletion , Humans , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, Mutant Strains , Neuroendocrine Cells/cytology , Prolyl Hydroxylases/metabolism , Stem Cells/cytology , Stem Cells/drug effects , Trans-Activators/genetics
6.
J Endocrinol Invest ; 43(9): 1259-1269, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32157664

ABSTRACT

PURPOSE: Nowadays, no human neuroendocrine cell models derived from the neural crest are available. In this study, we present non-transformed long-term primary Neural Crest Cells (NCCs) isolated from the trunk region of the neural crest at VIII-XII gestational weeks of human foetuses obtained from voluntary legal abortion. METHODS AND RESULTS: In NCC, quantitative real-time RT PCR demonstrated the expression of neural crest specifier genes, such as Snail1, Snail2/SLUG, Sox10, FoxD3, c-Myc, and p75NTR. Moreover, these cell populations expressed stemness markers (such as Nanog and nestin), as well as markers of motility and invasion (TAGLN, MMP9, CXCR4, and CXCR7), and of neuronal/glial differentiation (MAP2, GFAP, SYP, and TAU). Functional analysis demonstrated that these cells not only possessed high migration properties, but most importantly, they expressed markers of sympatho-adrenal lineage, such as ASCL1 and tyrosine hydroxylase (TH). Moreover, the expression of TH increased after the induction with two different protocols of differentiation towards neuronal and sympatho-adrenal phenotypes. Finally, exposure to conditioned culture media from NCC induced a mature phenotype in a neuronal cell model (namely SH-SY5Y), suggesting that NCC may also act like Schwann precursors. CONCLUSION: This unique human cell model provides a solid tool for future studies addressing the bases of human neural crest-derived neuroendocrine tumours.


Subject(s)
Cell Separation , Fetus/cytology , Neural Crest/cytology , Neuroendocrine Cells/cytology , Cell Differentiation , Cell Line , Cell Movement , Cell Separation/methods , Female , Humans , Neural Crest/embryology , Neural Crest/physiology , Neuroendocrine Cells/physiology , Phenotype , Pregnancy , Primary Cell Culture
7.
Article in English | MEDLINE | ID: mdl-32122885

ABSTRACT

Advances in single-cell RNA-seq (scRNA-seq) and computational analysis have enabled the systematic interrogation of the cellular composition of tissues. Combined with tools from developmental biology, cell biology, and genetics, these approaches are revealing fundamental aspects of tissue geometry and physiology, including the distribution, origins, and inferred functions of specialized cell types, and the dynamics of cellular turnover and differentiation. By comparing different tissues, such studies can delineate shared and specialized features of cell types and their lineage. Here, we compare two developmentally related murine epithelia, the airway and the small intestinal epithelia, which are both derived from the embryonic endodermal gut tube. We examine how airway and intestine generate and functionalize common archetypal cell types to fulfill similar shared physiologic functionalities. We point to cases in which similar cell types are repurposed to accommodate each tissue's unique physiologic role, and highlight tissue-specific cells whose specializations contribute to the distinct functional roles of each organ. We discuss how archetypal and unique cell types are incorporated within a cellular lineage, and how the regulation of the proportions of these cell types enables tissue-level organization to meet functional demands and maintain homeostasis.


Subject(s)
Cell Differentiation/physiology , Enteroendocrine Cells/physiology , Intestinal Mucosa/cytology , Neuroendocrine Cells/physiology , Respiratory Mucosa/cytology , Animals , Intestinal Mucosa/growth & development , Mice , Respiratory Mucosa/growth & development
8.
Gen Comp Endocrinol ; 288: 113345, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31812531

ABSTRACT

Natural and synthetic estrogens and progestins are widely used in human and veterinary medicine and are detected in waste and surface waters. Our previous studies have clearly shown that a number of these substances targets the brain to induce the estrogen-regulated brain aromatase expression but the consequences on brain development remain virtually unexplored. The aim of the present study was therefore to investigate the effect of estradiol (E2), progesterone (P4) and norethindrone (NOR), a 19-nortestosterone progestin, on zebrafish larval neurogenesis. We first demonstrated using real-time quantitative PCR that nuclear estrogen and progesterone receptor brain expression is impacted by E2, P4 and NOR. We brought evidence that brain proliferative and apoptotic activities were differentially affected depending on the steroidal hormone studied, the concentration of steroids and the region investigated. Our findings demonstrate for the first time that steroid compounds released in aquatic environment have the capacity to disrupt key cellular events involved in brain development in zebrafish embryos further questioning the short- and long-term consequences of this disruption on the physiology and behavior of organisms.


Subject(s)
Estradiol Congeners/pharmacology , Estrogens/pharmacology , Nervous System/drug effects , Neurogenesis/drug effects , Progesterone Congeners/pharmacology , Progesterone/pharmacology , Zebrafish/embryology , Animals , Embryo, Nonmammalian , Embryonic Development/drug effects , Endocrine Disruptors/pharmacology , Estradiol/pharmacology , Estrogens/analogs & derivatives , Estrogens/chemical synthesis , Humans , Ligands , Nandrolone/pharmacology , Nervous System/embryology , Neuroendocrine Cells/drug effects , Neuroendocrine Cells/physiology , Norethindrone/pharmacology , Progesterone/analogs & derivatives , Progesterone/chemical synthesis , Receptors, Estrogen/metabolism , Receptors, Progesterone/agonists , Receptors, Progesterone/metabolism , Zebrafish/growth & development
9.
Psychoneuroendocrinology ; 113: 104549, 2020 03.
Article in English | MEDLINE | ID: mdl-31884322

ABSTRACT

Endogenous cannabinoids (endocannabinoids, eCB) are expressed throughout the body and contribute to regulation of the hypothalamo-pituitary-adrenal (HPA) axis and general stress reactivity. This study assessed the contributions of CB1 receptors (CB1R) in the modulation of basal and stress-induced neural and HPA axis activities. Catheterized adult male rats were placed in chambers to acclimate overnight, with their catheters connected and exteriorized from the chambers for relatively stress-free remote injections. The next morning, the CB1R antagonist AM251 (1 or 2 mg/kg) or vehicle was administered, and 30 min later, rats were exposed to loud noise stress (30 min) or no noise (basal condition). Blood, brains, pituitary and adrenal glands were collected immediately after the procedures for analysis of c-fos and CB1R mRNAs, corticosterone (CORT) and adrenocorticotropin hormone (ACTH) plasma levels. Basally, CB1R antagonism induced c-fos mRNA in the basolateral amygdala (BLA) and auditory cortex (AUD) and elevated plasma CORT, indicating disruption of eCB-mediated constitutive inhibition of activity. CB1R blockade also potentiated stress-induced hormone levels and c-fos mRNA in several regions such as the bed nucleus of the stria terminalis (BST), lateral septum (LS), and basolateral amygdala (BLA) and the paraventricular nucleus of the hypothalamus (PVN). CB1R mRNA was detected in all central tissues investigated, and the adrenal cortex, but at very low levels in the anterior pituitary gland. Interestingly, CB1R mRNA was rapidly and bidirectionally regulated in response to stress and/or antagonist treatment in some regions. eCBs therefore modulate the HPA axis by regulating both constitutive and activity-dependent inhibition at multiple levels.


Subject(s)
Neuroendocrine Cells/physiology , Receptor, Cannabinoid, CB1/physiology , Adrenal Cortex/metabolism , Adrenal Glands/metabolism , Adrenocorticotropic Hormone/blood , Animals , Corticosterone/blood , Endocannabinoids/pharmacology , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/metabolism , Hypothalamus/metabolism , Male , Neuroendocrine Cells/drug effects , Neuroendocrine Cells/metabolism , Neurosecretory Systems/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Piperidines/pharmacology , Pituitary-Adrenal System/metabolism , Proto-Oncogene Proteins c-fos/blood , Pyrazoles/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/drug effects , Receptor, Cannabinoid, CB1/metabolism , Restraint, Physical/psychology , Stress, Physiological/physiology , Stress, Psychological/physiopathology
10.
Physiol Rev ; 100(2): 869-943, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31625459

ABSTRACT

In humans, as in the other mammals, the neuroendocrine control of reproduction is ensured by the brain-pituitary gonadotropic axis. Multiple internal and environmental cues are integrated via brain neuronal networks, ultimately leading to the modulation of the activity of gonadotropin-releasing hormone (GnRH) neurons. The decapeptide GnRH is released into the hypothalamic-hypophysial portal blood system and stimulates the production of pituitary glycoprotein hormones, the two gonadotropins luteinizing hormone and follicle-stimulating hormone. A novel actor, the neuropeptide kisspeptin, acting upstream of GnRH, has attracted increasing attention in recent years. Other neuropeptides, such as gonadotropin-inhibiting hormone/RF-amide related peptide, and other members of the RF-amide peptide superfamily, as well as various nonpeptidic neuromediators such as dopamine and serotonin also provide a large panel of stimulatory or inhibitory regulators. This paper addresses the origin and evolution of the vertebrate gonadotropic axis. Brain-pituitary neuroendocrine axes are typical of vertebrates, the pituitary gland, mediator and amplifier of brain control on peripheral organs, being a vertebrate innovation. The paper reviews, from molecular and functional perspectives, the evolution across vertebrate radiation of some key actors of the vertebrate neuroendocrine control of reproduction and traces back their origin along the vertebrate lineage and in other metazoa before the emergence of vertebrates. A focus is given on how gene duplications, resulting from either local events or from whole genome duplication events, and followed by paralogous gene loss or conservation, might have shaped the evolutionary scenarios of current families of key actors of the gonadotropic axis.


Subject(s)
Evolution, Molecular , Gene Duplication , Genome, Human , Gonadotropins/genetics , Gonads/physiology , Hypothalamo-Hypophyseal System/physiology , Neuroendocrine Cells/physiology , Reproduction/genetics , Animals , Gonadotropins/metabolism , Gonads/metabolism , Humans , Hypothalamo-Hypophyseal System/metabolism , Neuroendocrine Cells/metabolism , Phylogeny , Species Specificity
11.
J Neurosci ; 39(50): 9900-9913, 2019 12 11.
Article in English | MEDLINE | ID: mdl-31676600

ABSTRACT

Nonselective cation channels promote persistent spiking in many neurons from a diversity of animals. In the hermaphroditic marine-snail, Aplysia californica, synaptic input to the neuroendocrine bag cell neurons triggers various cation channels, causing an ∼30 min afterdischarge of action potentials and the secretion of egg-laying hormone. During the afterdischarge, protein kinase C is also activated, which in turn elevates hydrogen peroxide (H2O2), likely by stimulating nicotinamide adenine dinucleotide phosphate oxidase. The present study investigated whether H2O2 regulates cation channels to drive the afterdischarge. In single, cultured bag cell neurons, H2O2 elicited a prolonged, concentration- and voltage-dependent inward current, associated with an increase in membrane conductance and a reversal potential of ∼+30 mV. Compared with normal saline, the presence of Ca2+-free, Na+-free, or Na+/Ca2+-free extracellular saline, lowered the current amplitude and left-shifted the reversal potential, consistent with a nonselective cationic conductance. Preventing H2O2 reduction with the glutathione peroxidase inhibitor, mercaptosuccinate, enhanced the H2O2-induced current, while boosting glutathione production with its precursor, N-acetylcysteine, or adding the reducing agent, dithiothreitol, lessened the response. Moreover, the current generated by the alkylating agent, N-ethylmaleimide, occluded the effect of H2O2 The H2O2-induced current was inhibited by tetrodotoxin as well as the cation channel blockers, 9-phenanthrol and clotrimazole. In current-clamp, H2O2 stimulated burst firing, but this was attenuated or prevented altogether by the channel blockers. Finally, H2O2 evoked an afterdischarge from whole bag cell neuron clusters recorded ex vivo by sharp-electrode. H2O2 may regulate a cation channel to influence long-term changes in activity and ultimately reproduction.SIGNIFICANCE STATEMENT Hydrogen peroxide (H2O2) is often studied in a pathological context, such as ischemia or inflammation. However, H2O2 also physiologically modulates synaptic transmission and gates certain transient receptor potential channels. That stated, the effect of H2O2 on neuronal excitability remains less well defined. Here, we examine how H2O2 influences Aplysia bag cell neurons, which elicit ovulation by releasing hormones during an afterdischarge. These neuroendocrine cells are uniquely identifiable and amenable to recording as individual cultured neurons or a cluster from the nervous system. In both culture and the cluster, H2O2 evokes prolonged, afterdischarge-like bursting by gating a nonselective voltage-dependent cationic current. Thus, H2O2, which is generated in response to afterdischarge-associated second messengers, may prompt the firing necessary for hormone secretion and procreation.


Subject(s)
Hydrogen Peroxide/pharmacology , Membrane Potentials/drug effects , Neuroendocrine Cells/drug effects , Synaptic Transmission/drug effects , Animals , Aplysia , Calcium/metabolism , Cells, Cultured , Dithiothreitol/pharmacology , Neuroendocrine Cells/physiology , Patch-Clamp Techniques , Protein Kinase C/metabolism , Synaptic Transmission/physiology
12.
Am J Physiol Lung Cell Mol Physiol ; 317(4): L486-L495, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31313615

ABSTRACT

This article traces the beginnings of the various areas of physiological research on airway epithelium. First mentioned in 1600, it was not until 1834 that it was found to be ciliated. Goblet and basal cells were described in 1852, to be followed by ~10 other epithelial cell types (the most recent in 2018). It also contains nerve endings and resident leukocytes. Mucociliary clearance was documented in 1835, but the first studies on the ciliary beat cycle did not appear until 1890, and a definitive description was not published until 1981. It was established in 1932 that goblet cells in the cat trachea were unresponsive to cholinergic agents; but only since 1980 or so has any significant progress been made on what does cause them to degranulate. Active transfer of salts across epithelia creates local osmotic gradients that drive transepithelial water flows. Vectorial salt transport was first described for airway epithelium in 1968, and the associated volume flows were measured in 1981. Evidence that airway epithelium releases signaling molecules first appeared in 1981. Since then, scores of molecules have been identified. The pace of research in most areas increased dramatically after the development of confluent, polarized cultures of airway epithelium in the early 1980s.


Subject(s)
Alveolar Epithelial Cells/physiology , Cilia/physiology , Goblet Cells/physiology , Leukocytes/physiology , Neuroendocrine Cells/physiology , Alveolar Epithelial Cells/cytology , Animals , Biological Transport , Cats , Cell Communication/physiology , Cilia/ultrastructure , Goblet Cells/cytology , History, 17th Century , History, 19th Century , History, 20th Century , History, 21st Century , Humans , Leukocytes/cytology , Mucociliary Clearance/physiology , Neuroendocrine Cells/cytology , Respiratory System/anatomy & histology , Respiratory System/cytology , Respiratory System/metabolism , Sheep
13.
Physiol Rep ; 7(8): e14047, 2019 04.
Article in English | MEDLINE | ID: mdl-31008554

ABSTRACT

Hypothalamic magnocellular neurosecretory cells (MNCs) undergo dramatic structural reorganization during lactation in female rats that is thought to contribute to the pulsatile secretion of oxytocin critical for milk ejection. MNCs from male rats generate robust bursts of GABAergic synaptic currents, a subset of which are onset-synchronized between MNC pairs, but the functional role of the IPSC bursts is not known. To determine the physiological relevance of IPSC bursts, we compared MNCs from lactating and non-lactating female rats using whole-cell recordings in brain slices. We recorded a sixfold increase in the incidence of IPSC bursts in oxytocin (OT)-MNCs from lactating rats compared to non-lactating rats, whereas there was no change in IPSC bursts in vasopressin (VP)-MNCs. Synchronized bursts of IPSCs were observed in pairs of MNCs in slices from lactating rats. Our data indicate, therefore, that IPSC bursts are upregulated specifically in OT-MNCs during lactation, and may, therefore, contribute via rebound depolarization to the spike trains in OT neurons that lead to reflex milk ejection.


Subject(s)
Inhibitory Postsynaptic Potentials , Lactation/physiology , Neuroendocrine Cells/physiology , Oxytocin/metabolism , Animals , Female , Hypothalamus/cytology , Hypothalamus/metabolism , Hypothalamus/physiology , Lactation/metabolism , Neuroendocrine Cells/metabolism , Rats , Rats, Wistar , Vasopressins/metabolism
15.
Oncogene ; 38(24): 4875-4884, 2019 06.
Article in English | MEDLINE | ID: mdl-30770901

ABSTRACT

Castration-resistant prostate cancer (CRPC) with neuroendocrine differentiation (NED) is a lethal disease for which effective therapies are urgently needed. The mechanism underlying development of CRPC with NED, however, remains largely uncharacterized. In this study, we explored and characterized the functional role of neurotensin (NTS) in cell line and animal models of CRPC with NED. NTS was acutely induced by androgen deprivation in animal models of prostate cancer (PCa) and activated downstream signaling leading to NED through activation of neurotensin receptor 1 (NTSR1) and neurotensin receptor 3 (NTSR3), but not neurotensin receptor 2 (NTSR2). Our findings also revealed the existence of a CK8+/CK14+ subpopulation in the LNCaP cell line that expresses high levels of both NTSR1 and NTSR3, and displays an enhanced susceptibility to develop neuroendocrine-like phenotypes upon treatment with NTS. More importantly, NTSR1 pathway inhibition prevented the development of NED and castration resistance in vivo. We propose a novel role of NTS in the development of CRPC with NED, and a possible strategy to prevent the onset of NED by targeting the NTS signaling pathway.


Subject(s)
Cell Transdifferentiation/genetics , Neuroendocrine Cells/physiology , Neurotensin/physiology , Prostate/pathology , Prostatic Neoplasms, Castration-Resistant/pathology , Receptors, Neurotensin/physiology , Adaptor Proteins, Vesicular Transport/physiology , Animals , Cell Line, Tumor , Humans , Male , Mice , Mice, Inbred BALB C , Prostate/metabolism , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/physiopathology
16.
Psychoneuroendocrinology ; 97: 111-119, 2018 11.
Article in English | MEDLINE | ID: mdl-30015006

ABSTRACT

This study investigated neuroendocrine, autonomic, and cardiovascular changes evoked by daily exposure to the same type of stressor (homotypic) or different aversive stressor stimuli (heterotypic) in 60-days-old female normotensive Wistar rats and female spontaneously hypertensive rats (SHR). Both strains of rats were exposed for 10 consecutive days to either the homotypic stressor repeated restraint stress (RRS) or the heterotypic stressor chronic unpredictable stress (CUS). As expected, SHR had higher baseline blood pressure values and impaired baroreflex activity in relation to normotensive animals. Besides, SHR presented higher plasma corticosterone levels and decreased thymus weight. Both RRS and CUS increased baseline plasma corticosterone concentration and decreased body weight gain in both normotensive and SHR rats. In addition, both stress protocols caused hypertrophy of adrenal glands in normotensive rats. Regarding the cardiovascular effects, RRS increased basal heart rate in both rat strains, which was mediated by an increase in sympathetic tone to the heart. Besides, RRS increased baroreflex-mediated tachycardia in SHR animals, while CUS increased cardiac parasympathetic activity and pacemaker activity in normotensive rats. Taken together, these results indicate a stress type-specific effect, as identified by a vulnerability of both strains to the deleterious cardiovascular effects evoked by the homotypic stressor and a resilience to the impact of the heterotypic stressor. Vulnerability of hypertensive rats was evidenced by the absence of CUS-evoked adaptive cardiovascular responses and an increase of baroreflex tachycardia in SHR animals subjected to RRS. The somatic and HPA axis changes were overall independent of the chronic stress regimen and pre-existing hypertension.


Subject(s)
Hypertension/physiopathology , Stress, Psychological/physiopathology , Animals , Blood Pressure/physiology , Cardiovascular Physiological Phenomena , Cardiovascular System/physiopathology , Chronic Disease/psychology , Corticosterone/analysis , Female , Heart Rate/physiology , Hypertension/complications , Hypothalamo-Hypophyseal System , Neuroendocrine Cells/physiology , Neurosecretory Systems/physiopathology , Pituitary-Adrenal System , Preexisting Condition Coverage , Rats , Rats, Inbred SHR , Rats, Wistar
17.
Nan Fang Yi Ke Da Xue Xue Bao ; 38(6): 723-730, 2018 Jun 20.
Article in Chinese | MEDLINE | ID: mdl-29997096

ABSTRACT

OBJECTIVE: To investigate the effect of infiltrating mast cells on neuroendocrine differentiation (NED) and docetaxel sensitivity of prostate cancer (PCa) cells in vitro. METHODS: Human PCa cell lines (LNCaP and C4-2) were co-cultured with human mast cell line (HMC-1) in Transwell chambers. Androgen receptor (AR) was silenced in C4-2 cells using sh-AR lentivirus, and p21 was knocked down and overexpressed by transfecting C4-2 cells with pLKO.1-sh-p21 and pCMV-p21, respectively. The morphological changes of LNCaP and C4-2 cells were observed. MTT assay and colony formation assay were used to assess the proliferation of LNCaP and C4-2 cells. CCK8 assay was used to detect the cell viability of C4-2 cells following docetaxel trreatment. RT-qPCR and Western blotting were performed to determine the mRNA and protein expressions of neuroendocrine markers, AR and p21 in the cells. RESULTS: Co-culture with HMC-1 cells enhanced the neuroendocrine phenotypes, inhibited the proliferation and up-regulated the expression of p21 in LNCaP and C4-2 cells. P21 positively regulated NED through a non-AR-dependent signaling pathway, while p21 knockdown partially reversed NED promoted by the mast cells. PCa cells co-cultured with HMC-1 cells showed increased resistance to docetaxel, and silencing p21 partially reversed docetaxel resistance in PCa cells. CONCLUSION: Infiltrating mast cells up-regulates p21 to promote NED and increase docetaxel resistance in PCa cells in vitro.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Differentiation/physiology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Docetaxel/pharmacology , Mast Cells/physiology , Neuroendocrine Cells/cytology , Prostatic Neoplasms/drug therapy , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cyclin-Dependent Kinase Inhibitor p21/genetics , Drug Resistance, Neoplasm/physiology , Gene Expression Regulation, Neoplastic , Humans , Male , Neuroendocrine Cells/physiology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Signal Transduction , Up-Regulation/physiology
18.
Vitam Horm ; 107: 27-66, 2018.
Article in English | MEDLINE | ID: mdl-29544634

ABSTRACT

Gonadotropin-releasing hormone (GnRH) is recognized as the central regulator of the functions of the pituitary-gonadal axis. The increasing knowledge on the mechanisms controlling the development and the function of GnRH-producing neurons is leading to a better diagnostic and therapeutic approach for hypogonadotropic hypogonadisms and for alterations of the puberty onset. During female life span, the function of the GnRH pulse generator may be affected by a number of inputs from other neuronal systems, offering alternative strategies for diagnostic and therapeutic interventions. Moreover, the identification of a GnRH/GnRH receptor system in both human ovary and endometrium has widened the spectrum of action of the peptide outside its hypothalamic functions. The pharmacological use of GnRH itself or its synthetic analogs (agonists and antagonists) provides a valid tool to either stimulate or block gonadotropin secretion and to modulate the female fertility in several reproductive disorders and in assisted reproduction technology. The use of GnRH agonists in young female patients undergoing chemotherapy is also considered a promising therapeutic approach to counteract iatrogenic ovarian failure.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Hypothalamo-Hypophyseal System/physiology , Models, Biological , Neuroendocrine Cells/physiology , Ovary/physiology , Protein Precursors/metabolism , Receptors, LHRH/agonists , Reproduction , Animals , Endometrium/drug effects , Endometrium/growth & development , Endometrium/physiology , Endometrium/physiopathology , Female , Fertility Agents, Female/pharmacology , Fertility Agents, Female/therapeutic use , Fertility Preservation/trends , Gonadotropin-Releasing Hormone/chemistry , Humans , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/growth & development , Hypothalamo-Hypophyseal System/physiopathology , Infertility, Female/drug therapy , Infertility, Female/pathology , Infertility, Female/physiopathology , Infertility, Female/therapy , Menstrual Cycle/drug effects , Neuroendocrine Cells/cytology , Neuroendocrine Cells/metabolism , Neuroendocrine Cells/pathology , Ovary/drug effects , Ovary/growth & development , Ovary/physiopathology , Pregnancy , Primary Ovarian Insufficiency/drug therapy , Primary Ovarian Insufficiency/pathology , Primary Ovarian Insufficiency/physiopathology , Protein Isoforms/agonists , Protein Isoforms/metabolism , Protein Precursors/chemistry , Puberty/drug effects , Receptors, LHRH/antagonists & inhibitors , Receptors, LHRH/metabolism , Receptors, LHRH/therapeutic use , Reproduction/drug effects , Signal Transduction/drug effects
19.
J Neurosci ; 38(11): 2796-2808, 2018 03 14.
Article in English | MEDLINE | ID: mdl-29440551

ABSTRACT

By synchronizing neuronal activity, electrical transmission influences the coordination, pattern, and/or frequency of firing. In the hemaphroditic marine-snail, Aplysia calfornica, the neuroendocrine bag cell neurons use electrical synapses to synchronize a 30 min afterdischarge of action potentials for the release of reproductive hormone. During the afterdischarge, protein kinase C (PKC) is activated, although its impact on bag cell neuron electrical transmission is unknown. This was investigated here by monitoring electrical synapses between paired cultured bag cell neurons using dual whole-cell recording. Voltage clamp revealed a largely voltage-independent junctional current, which was enhanced by treating with a PKC activator, PMA, before recording. We also examined the transfer of presynaptic action potential-like waveforms (generated in voltage clamp) to the postsynaptic cell (measured in current clamp). For control pairs, the presynaptic spike-like waveforms mainly evoked electrotonic potentials; however, when PKC was triggered, these stimuli consistently produced postsynaptic action potentials. To assess whether this involved changes to postsynaptic responsiveness, single bag cell neurons were injected with junctional-like current mimicking that evoked by a presynaptic action potential. Unlike control neurons, which were less likely to spike, cells in PMA always fired action potentials to the junctional-like current. Furthermore, PKC activation increased a postsynaptic voltage-gated Ca2+ current, which was recruited even by modest depolarization associated with an electrotonic potential. Whereas PKC inhibits gap junctions in most systems, bag cell neurons are rather unique, as the kinase potentiates the electrical synapse; in turn, this synergizes with augmented postsynaptic Ca2+ current to promote synchronous firing.SIGNIFICANCE STATEMENT Electrical coupling is a fundamental form of communication. For the bag cell neurons of Aplysia, electrical synapses coordinate a prolonged burst of action potentials known as the afterdischarge. We looked at how protein kinase C, which is upregulated with the afterdischarge, influences information transfer across the synapse. The kinase activation increased junctional current, a remarkable finding given that this enzyme is largely considered inhibitory for gap junctions. There was also an augmentation in the ability of a presynaptic neuron to provoke postsynaptic action potentials. This increased excitability was, in part, due to enhanced postsynaptic voltage-dependent Ca2+ current. Thus, protein kinase C improves the fidelity of electrotonic transmission and promotes synchronous firing by modulating both junctional and membrane conductances.


Subject(s)
Aplysia/physiology , Calcium Channels/physiology , Protein Kinase C/physiology , Synapses/drug effects , Synaptic Transmission/physiology , Action Potentials/physiology , Animals , Electrical Synapses/drug effects , Enzyme Activation , Excitatory Postsynaptic Potentials/physiology , Neuroendocrine Cells/physiology , Neurons/drug effects , Patch-Clamp Techniques , Synaptic Potentials
20.
Sci Rep ; 8(1): 2760, 2018 02 09.
Article in English | MEDLINE | ID: mdl-29426889

ABSTRACT

Engineered silica nanoparticles (NPs) have attracted increasing interest in several applications, and particularly in the field of nanomedicine, thanks to the high biocompatibility of this material. For their optimal and controlled use, the understanding of the mechanisms elicited by their interaction with the biological target is a prerequisite, especially when dealing with cells particularly vulnerable to environmental stimuli like neurons. Here we have combined different electrophysiological approaches (both at the single cell and at the population level) with a genomic screening in order to analyze, in GT1-7 neuroendocrine cells, the impact of SiO2 NPs (50 ± 3 nm in diameter) on electrical activity and gene expression, providing a detailed analysis of the impact of a nanoparticle on neuronal excitability. We find that 20 µg mL-1 NPs induce depolarization of the membrane potential, with a modulation of the firing of action potentials. Recordings of electrical activity with multielectrode arrays provide further evidence that the NPs evoke a temporary increase in firing frequency, without affecting the functional behavior on a time scale of hours. Finally, NPs incubation up to 24 hours does not induce any change in gene expression.


Subject(s)
Action Potentials/drug effects , Nanoparticles , Neuroendocrine Cells/drug effects , Neurons/metabolism , Silicon Dioxide/pharmacology , Animals , Cell Line , Gene Expression/drug effects , Hypothalamus/cytology , Mice , Neuroendocrine Cells/physiology , Neurons/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...