Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
1.
Biomed Pharmacother ; 140: 111746, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34062412

ABSTRACT

BACKGROUND/AIMS: Asthma is a common chronic respiratory disease. It has been reported that Pingchuan formula (PCF) can control asthma attacks by reducing airway inflammation, muscle spasm and mucus secretion. However, PCF's mechanism for reducing airway mucus hypersecretion remains unclear. This study aimed to investigate the effect of PCF on airway mucus secretion in asthmatic mice and to explore changes in the PNEC-GABA-IL13-Muc5ac axis. METHODS: Male Babl/c mice were used to establish the asthma model via sensitisation with OVA. Mice were randomly divided into Normal, OVA, DEX, and PCF groups. After treatment, lung histopathology was observed with H&E and PAS staining. BALF levels of IL-5 and IL-13 were detected using ELISA. The levels of mRNA and protein expression for GAD1, GABAARß1, GABAARα1 and Muc5ac in the lung tissue were measured by RT-PCR and Western blot assays. PNECs were observed with AgNOR staining. RESULTS: PCF treatment effectively reduced goblet cell (P < 0.01) and PNEC (P < 0.05) proliferation, lung tissue inflammation and airway mucus hypersecretion. In addition, PCF also markedly downregulated mRNA and protein expression of GAD1, GABAARß1, GABAARα1 and Muc5ac (P < 0.05, compared with OVA), thus inhibiting the GABA-IL-13 pathway in the lung tissue of asthmatic mice. CONCLUSION: These findings suggest that PCF controls asthma attacks by reducing airway inflammation and mucus hypersecretion via the PNEC-GABA-IL13-Muc5ac axis.


Subject(s)
Anti-Asthmatic Agents/therapeutic use , Asthma/drug therapy , Drugs, Chinese Herbal/therapeutic use , Animals , Anti-Asthmatic Agents/pharmacology , Asthma/immunology , Asthma/metabolism , Asthma/pathology , Bronchoalveolar Lavage Fluid/immunology , Cell Proliferation/drug effects , Cytokines/immunology , Drugs, Chinese Herbal/pharmacology , Goblet Cells/drug effects , Interleukin-13/metabolism , Lung/drug effects , Lung/pathology , Male , Mice, Inbred BALB C , Mucin 5AC/metabolism , Mucus/metabolism , Neuroendocrine Cells/drug effects , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism
2.
Genes Dev ; 35(11-12): 870-887, 2021 06.
Article in English | MEDLINE | ID: mdl-34016692

ABSTRACT

Small cell lung carcinoma (SCLC) is among the most lethal of all solid tumor malignancies. In an effort to identify novel therapeutic approaches for this recalcitrant cancer type, we applied genome-scale CRISPR/Cas9 inactivation screens to cell lines that we derived from a murine model of SCLC. SCLC cells were particularly sensitive to the deletion of NEDD8 and other neddylation pathway genes. Genetic suppression or pharmacological inhibition of this pathway using MLN4924 caused cell death not only in mouse SCLC cell lines but also in patient-derived xenograft (PDX) models of pulmonary and extrapulmonary small cell carcinoma treated ex vivo or in vivo. A subset of PDX models were exceptionally sensitive to neddylation inhibition. Neddylation inhibition suppressed expression of major regulators of neuroendocrine cell state such as INSM1 and ASCL1, which a subset of SCLC rely upon for cell proliferation and survival. To identify potential mechanisms of resistance to neddylation inhibition, we performed a genome-scale CRISPR/Cas9 suppressor screen. Deletion of components of the COP9 signalosome strongly mitigated the effects of neddylation inhibition in small cell carcinoma, including the ability of MLN4924 to suppress neuroendocrine transcriptional program expression. This work identifies neddylation as a regulator of neuroendocrine cell state and potential therapeutic target for small cell carcinomas.


Subject(s)
Carcinoma, Small Cell/therapy , Cyclopentanes , Lung Neoplasms/therapy , NEDD8 Protein/metabolism , Pyrimidines , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , COP9 Signalosome Complex/genetics , Carcinoma, Small Cell/physiopathology , Cell Death/drug effects , Cell Line, Tumor , Cyclopentanes/pharmacology , Cyclopentanes/therapeutic use , Disease Models, Animal , Gene Expression Regulation, Neoplastic/drug effects , Heterografts , Humans , Lung Neoplasms/physiopathology , Mice , NEDD8 Protein/genetics , Neuroendocrine Cells/cytology , Neuroendocrine Cells/drug effects , Proteins/genetics , Proteins/metabolism , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Repressor Proteins/genetics , Sequence Deletion
4.
PLoS One ; 15(10): e0240107, 2020.
Article in English | MEDLINE | ID: mdl-33045023

ABSTRACT

Somatostatin analogs, which are used to treat neuroendocrine tumors, inhibit hormone secretion or promote tumor shrinkage; however, their efficacy varies between patients, possibly because of differential expression of somatostatin receptors (SSTRs) in tumors. In this study, we evaluated the regulatory mechanism underlying the expression of SSTR2, the main octreotide target. Thirty miRNAs were found to be dysregulated in neuroendocrine cells (INS-1 cells) incubated with octreotide compared to that in placebo-treated cells. Among the upregulated miRNAs, miR-16-5p was elevated after short-term octreotide treatment. We conducted in vitro experiments to determine whether the expression of miR-16-5p was associated with the regulation of SSTR2 expression and affected octreotide sensitivity in INS-1 cells. Overexpression of miR-16-5p by transfected mimics induced upregulation of SSTR2 expression. Additionally, the expression of miR-16-5p further enhanced octreotide-induced reduction in cell proliferation in both two- and three-dimensional culture of INS-1 cells. Thus, our results reveal the mechanism underlying SSTR2 expression regulation and may aid in developing therapeutic approaches for enhancing the response to octreotide, particularly in patients unresponsive to SSTR2-targeted somatostatin analog treatment.


Subject(s)
Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Neuroendocrine Cells/pathology , Neuroendocrine Tumors/genetics , Receptors, Somatostatin/genetics , Animals , Antineoplastic Agents, Hormonal/pharmacology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , HeLa Cells , Humans , Neuroendocrine Cells/drug effects , Neuroendocrine Cells/metabolism , Neuroendocrine Tumors/drug therapy , Neuroendocrine Tumors/pathology , Octreotide/pharmacology , Rats , Transcriptome/drug effects , Up-Regulation/drug effects
5.
J Neuroendocrinol ; 32(11): e12881, 2020 11.
Article in English | MEDLINE | ID: mdl-32803906

ABSTRACT

The secretion of prolactin from the pituitary is negatively controlled by tuberoinfundibular dopamine (TIDA) neurones. The electrical properties of TIDA cells have recently been identified as a modulatory target of neurotransmitters and hormones in the lactotrophic axis. The role of the GABAB receptor in this control has received little attention, yet is of particular interest because it may act as a TIDA neurone autoreceptor. Here, this issue was explored in a spontaneously active rat TIDA in vitro slice preparation using whole-cell recordings. Application of the GABAB receptor agonist, baclofen, dose-dependently slowed down or abolished the network oscillations typical of this preparation. Pharmacological manipulations identify the underlying mechanism as an outward current mediated by G-protein-coupled inwardly rectifying K+ -like channels. In addition to this postsynaptic modulation, we describe a presynaptic modulation where GABAB receptors restrain the release of glutamate and GABA onto TIDA neurones. Our data identify both pre- and postsynaptic modulation of TIDA neurones by GABAB receptors that may play a role in the neuronal network control of pituitary prolactin secretion and lactation.


Subject(s)
Dopaminergic Neurons/metabolism , Neuroendocrine Cells/metabolism , Receptors, GABA-B/metabolism , Receptors, Presynaptic/metabolism , Synapses/metabolism , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Baclofen/pharmacology , Dopaminergic Neurons/drug effects , Dose-Response Relationship, Drug , Electrophysiological Phenomena , G Protein-Coupled Inwardly-Rectifying Potassium Channels/drug effects , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , GABA Agonists/pharmacology , Male , Neuroendocrine Cells/drug effects , Patch-Clamp Techniques , Potassium Channels, Inwardly Rectifying/drug effects , Potassium Channels, Inwardly Rectifying/metabolism , Rats , Rats, Sprague-Dawley , Receptors, GABA-B/drug effects , Receptors, Presynaptic/drug effects , Synapses/drug effects
6.
Cell Calcium ; 87: 102184, 2020 05.
Article in English | MEDLINE | ID: mdl-32151786

ABSTRACT

Neuroendocrine adrenal chromaffin cells release neurohormones catecholamines in response to Ca2+ entry via voltage-gated Ca2+ channels (VGCCs). Adrenal chromaffin cells also express non-voltage-gated channels, which may conduct Ca2+ at negative membrane potentials, whose role in regulation of exocytosis is poorly understood. We explored how modulation of Ca2+ influx at negative membrane potentials affects basal cytosolic Ca2+ concentration ([Ca2+]i) and exocytosis in metabolically intact voltage-clamped bovine adrenal chromaffin cells. We found that in these cells, Ca2+ entry at negative membrane potentials is balanced by Ca2+ extrusion by the Na+/Ca2+ exchanger and that this balance can be altered by membrane hyperpolarization or stimulation with an inflammatory hormone bradykinin. Membrane hyperpolarization or application of bradykinin augmented Ca2+-carrying current at negative membrane potentials, elevated basal [Ca2+]i, and facilitated synchronous exocytosis evoked by the small amounts of Ca2+ injected into the cell via VGCCs (up to 20 pC). Exocytotic responses evoked by the injections of the larger amounts of Ca2+ via VGCCs (> 20 pC) were suppressed by preceding hyperpolarization. In the absence of Ca2+ entry via VGCCs and Ca2+ extrusion via the Na+/Ca2+ exchanger, membrane hyperpolarization induced a significant elevation in [Ca2+]i and asynchronous exocytosis. Our results indicate that physiological interferences, such as membrane hyperpolarization and/or activation of non-voltage-gated Ca2+ channels, modulate basal [Ca2+]i and, consequently, segregation of exocytotic vesicles and their readiness to be released spontaneously and in response to Ca2+ entry via VGCCs. These mechanisms may play role in homeostatic plasticity of neuronal and endocrine cells.


Subject(s)
Calcium/metabolism , Exocytosis , Membrane Potentials , Neuroendocrine Cells/metabolism , Animals , Bradykinin/pharmacology , Cattle , Exocytosis/drug effects , Membrane Potentials/drug effects , Models, Biological , Neuroendocrine Cells/drug effects , Receptors, Bradykinin/metabolism , Solutions
7.
Int J Mol Sci ; 21(2)2020 Jan 08.
Article in English | MEDLINE | ID: mdl-31936301

ABSTRACT

Oxaliplatin (OXAL) is regarded as a platinum-based anti-neoplastic agent. However, its perturbations on membrane ionic currents in neurons and neuroendocrine or endocrine cells are largely unclear, though peripheral neuropathy has been noted during its long-term administration. In this study, we investigated how the presence of OXAL and other related compounds can interact with two types of inward currents; namely, hyperpolarization-activated cation current (Ih) and membrane electroporation-induced current (IMEP). OXAL increased the amplitude or activation rate constant of Ih in a concentration-dependent manner with effective EC50 or KD values of 3.2 or 6.4 µM, respectively, in pituitary GH3 cells. The stimulation by this agent of Ih could be attenuated by subsequent addition of ivabradine, protopine, or dexmedetomidine. Cell exposure to OXAL (3 µM) resulted in an approximately 11 mV rightward shift in Ih activation along the voltage axis with minimal changes in the gating charge of the curve. The exposure to OXAL also effected an elevation in area of the voltage-dependent hysteresis elicited by long-lasting triangular ramp. Additionally, its application resulted in an increase in the amplitude of IMEP elicited by large hyperpolarization in GH3 cells with an EC50 value of 1.3 µM. However, in the continued presence of OXAL, further addition of ivabradine, protopine, or dexmedetomidine always resulted in failure to attenuate the OXAL-induced increase of IMEP amplitude effectively. Averaged current-voltage relation of membrane electroporation-induced current (IMEP) was altered in the presence of OXAL. In pituitary R1220 cells, OXAL-stimulated Ih remained effective. In Rolf B1.T olfactory sensory neurons, this agent was also observed to increase IMEP in a concentration-dependent manner. In light of the findings from this study, OXAL-mediated increases of Ih and IMEP may coincide and then synergistically act to increase the amplitude of inward currents, raising the membrane excitability of electrically excitable cells, if similar in vivo findings occur.


Subject(s)
Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Neuroendocrine Cells/drug effects , Oxaliplatin/adverse effects , Sensory Receptor Cells/drug effects , Animals , Cations/pharmacology , Electroporation , Humans , Mice , Neoplasms/complications , Neoplasms/drug therapy , Oxaliplatin/pharmacology , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/pathology
8.
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
9.
Invest New Drugs ; 38(3): 746-754, 2020 06.
Article in English | MEDLINE | ID: mdl-31312936

ABSTRACT

Prostate cancer is the second leading cause of cancer-related deaths among men in developed countries. Neuroendocrine prostate cancer, in particular, is associated with an aggressive phenotype and a poor prognosis. Neuroendocrine cells produce and secrete peptide hormones and growth factors in a paracrine/autocrine manner which promote the progression of the disease. Recent studies have demonstrated that extracellular vesicles or exosomes are released by prostate cancer cells, supporting the spread of prostate cancer. Hence, the aim of this study was to investigate the effect of growth hormone-releasing hormone (GHRH) on neuroendocrine differentiation (NED) in the androgen-dependent prostate cancer cell line LNCaP and the molecular mechanisms underlying these effects. GHRH induced an increase in the percentage of neurite-bearing cells and in the protein levels of Neuron-Specific Enolase. Both effects were blocked by the GHRH receptor antagonist MIA-690. In addition, pretreatment of these cells with the calcium chelator BAPTA, the EGFR inhibitor AG-1478 or the HER2 inhibitor AG-825 reduced the effect of GHRH, suggesting that the GHRH-induced stimulation of NED involves calcium channel activation and EGFR/HER2 transactivation. Finally, PC3-derived exosomes led to an increase in NED, cell proliferation and cell adhesion. Altogether, these findings suggest that GHRH antagonists should be considered for in the management of neuroendocrine prostate cancer.


Subject(s)
Cell Differentiation/drug effects , Growth Hormone-Releasing Hormone/antagonists & inhibitors , Neuroendocrine Cells/drug effects , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Androgens/pharmacology , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Male , Neuroendocrine Cells/metabolism , PC-3 Cells , Quinazolines/pharmacology , Receptor, ErbB-2/metabolism , Tyrphostins/pharmacology
10.
Eur Neuropsychopharmacol ; 31: 86-99, 2020 02.
Article in English | MEDLINE | ID: mdl-31831204

ABSTRACT

Second-line depression treatment involves augmentation with one (rarely two) additional drugs, of chronic administration of a selective serotonin reuptake inhibitor (SSRI), which is the first-line depression treatment. Unfortunately, many depressed patients still fail to respond even after months to years of searching to find an effective combination. To aid in the identification of potentially effective antidepressant combinations, we created a computational model of the monoaminergic neurotransmitter (serotonin, norepinephrine, and dopamine), stress-hormone (cortisol), and male sex hormone (testosterone) systems. The model was trained via machine learning to represent a broad range of empirical observations. Neuroadaptation to chronic drug administration was simulated through incremental adjustments in model parameters that corresponded to key regulatory components of the neurotransmitter and neurohormone systems. Analysis revealed that neuroadaptation in the model depended on all of the regulatory components in complicated ways, and did not reveal any one or a few specific components that could be targeted in the design of antidepressant treatments. We used large sets of neuroadapted states of the model to screen 74 different drug and hormone combinations and identified several combinations that could potentially be therapeutic for a higher proportion of male patients than SSRIs by themselves.


Subject(s)
Antidepressive Agents/administration & dosage , Biogenic Monoamines/metabolism , Neural Networks, Computer , Neuroendocrine Cells/metabolism , Neurotransmitter Agents/metabolism , Testosterone/metabolism , Drug Administration Schedule , Humans , Hydrocortisone/metabolism , Male , Neuroendocrine Cells/drug effects , Selective Serotonin Reuptake Inhibitors/administration & dosage
11.
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
12.
Sci Rep ; 9(1): 19076, 2019 12 13.
Article in English | MEDLINE | ID: mdl-31836808

ABSTRACT

Neuroendocrine prostate cancer (NEPC) is thought to arise as prostate adenocarcinoma cells transdifferentiate into neuroendocrine (NE) cells to escape potent anti-androgen therapies however, the exact molecular events accompanying NE transdifferentiation and their plasticity remain poorly defined. Cell fate regulator ASCL1/hASH1's expression was markedly induced in androgen deprived (AD) LNCaP cells and prominent nuclear localisation accompanied acquisition of the NE-like morphology and expression of NE markers (NSE). By contrast, androgen-insensitive PC3 and DU145 cells displayed clear nuclear hASH1 localisation under control conditions that was unchanged by AD, suggesting AR signalling negatively regulated hASH1 expression and localisation. Synthetic androgen (R1881) prevented NE transdifferentiation of AD LNCaP cells and markedly suppressed expression of key regulators of lineage commitment and neurogenesis (REST and ASCL1/hASH1). Post-AD, NE LNCaP cells rapidly lost NE-like morphology following R1881 treatment, yet ASCL1/hASH1 expression was resistant to R1881 treatment and hASH1 nuclear localisation remained evident in apparently dedifferentiated LNCaP cells. Consequently, NE cells may not fully revert to an epithelial state and retain key NE-like features, suggesting a "hybrid" phenotype. This could fuel greater NE transdifferentiation, therapeutic resistance and NEPC evolution upon subsequent androgen deprivation. Such knowledge could facilitate CRPC tumour stratification and identify targets for more effective NEPC management.


Subject(s)
Androgens/pharmacology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Nucleus/metabolism , Cell Transdifferentiation , Neuroendocrine Cells/pathology , Prostatic Neoplasms/pathology , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Shape/drug effects , Cell Transdifferentiation/drug effects , Humans , Male , Neuroendocrine Cells/drug effects , Neurogenesis/drug effects , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Protein Transport/drug effects
13.
Sci Rep ; 9(1): 18471, 2019 12 05.
Article in English | MEDLINE | ID: mdl-31804600

ABSTRACT

FTY-720 (Fingolimod) was one of the first compounds authorized for the treatment of multiple sclerosis. Among its other activities, this sphingosine analogue enhances exocytosis in neuroendocrine chromaffin cells, altering the quantal release of catecholamines. Surprisingly, the size of chromaffin granules is reduced within few minutes of treatment, a process that is paralleled by the homotypic fusion of granules and their heterotypic fusion with mitochondria, as witnessed by dynamic confocal and TIRF microscopy. Electron microscopy studies support these observations, revealing the fusion of several vesicles with individual mitochondria to form large, round mixed organelles. This cross-fusion is SNARE-dependent, being partially prevented by the expression of an inactive form of SNAP-25. Fused mitochondria exhibit an altered redox potential, which dramatically enhances cell death. Therefore, the cross-fusion of intracellular organelles appears to be a new mechanism to be borne in mind when considering the effect of FTY-720 on the survival of neuroendocrine cells.


Subject(s)
Chromaffin Granules/drug effects , Fingolimod Hydrochloride/toxicity , Multiple Sclerosis/drug therapy , Neuroendocrine Cells/drug effects , Animals , Cattle , Cells, Cultured , Chromaffin Granules/metabolism , Chromaffin Granules/pathology , Humans , Microscopy, Electron, Transmission , Mitochondria/drug effects , Mitochondria/pathology , Mitochondrial Dynamics/drug effects , Neuroendocrine Cells/cytology , Neuroendocrine Cells/metabolism , Primary Cell Culture , Synaptosomal-Associated Protein 25/metabolism , Toxicity Tests
14.
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
15.
PLoS One ; 14(9): e0213488, 2019.
Article in English | MEDLINE | ID: mdl-31536510

ABSTRACT

In treating patients with castration resistant prostate cancer (CRPC), enzalutamide, the second-generation androgen receptor (AR) antagonist, is an accepted standard of care. However, clinical benefits are limited to a median time of 4.8 months because resistance inevitably emerges. To determine the mechanism of treatment resistance, we carried out a RNA sequence analysis and found increased expression levels of neuroendocrine markers in the enzalutamide-resistant LNCaP human prostate cancer (CaP) cell line when compared to the parental cell line. Subsequent studies demonstrated that Transcription Factor-4 (TCF4), a transcription factor implicated in WNT signaling, mediated neuroendocrine differentiation (NED) in response to enzalutamide treatment and was elevated in the enzalutamide-resistant LNCaP. In addition, we observed that PTHrP mediated enzalutamide resistance in tissue culture and inducible TCF4 overexpression resulted in enzalutamide-resistance in a mouse xenograft model. Finally, small molecule inhibitors of TCF4 or PTHrP partially reversed enzalutamide resistance in CaP cells. When tissues obtained from men who died of metastatic CaP were examined, a positive correlation was found between the expression levels of TCF4 and PTHrP. Taken together, the current results indicate that TCF4 induces enzalutamide resistance via NED in CaP.


Subject(s)
Drug Resistance, Neoplasm/genetics , Neuroendocrine Cells/metabolism , Neuroendocrine Cells/pathology , Phenylthiohydantoin/analogs & derivatives , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Transcription Factor 7-Like 2 Protein/genetics , Animals , Benzamides , Cell Line, Tumor , Disease Models, Animal , Humans , Male , Mice , Mice, Knockout , Neuroendocrine Cells/drug effects , Nitriles , Phenylthiohydantoin/pharmacology , Protein Binding , Protein Transport , Transcription Factor 7-Like 2 Protein/antagonists & inhibitors , Transcription Factor 7-Like 2 Protein/metabolism , Xenograft Model Antitumor Assays , beta Catenin/metabolism
16.
Nat Commun ; 10(1): 2571, 2019 06 12.
Article in English | MEDLINE | ID: mdl-31189930

ABSTRACT

While the antiandrogen enzalutamide (Enz) extends the castration resistant prostate cancer (CRPC) patients' survival an extra 4.8 months, it might also result in some adverse effects via inducing the neuroendocrine differentiation (NED). Here we found that lncRNA-p21 is highly expressed in the NEPC patients derived xenograft tissues (NEPC-PDX). Results from cell lines and human clinical sample surveys also revealed that lncRNA-p21 expression is up-regulated in NEPC and Enz treatment could increase the lncRNA-p21 to induce the NED. Mechanism dissection revealed that Enz could promote the lncRNA-p21 transcription via altering the androgen receptor (AR) binding to different androgen-response-elements, which switch the EZH2 function from histone-methyltransferase to non-histone methyltransferase, consequently methylating the STAT3 to promote the NED. Preclinical studies using the PDX mouse model proved that EZH2 inhibitor could block the Enz-induced NED. Together, these results suggest targeting the Enz/AR/lncRNA-p21/EZH2/STAT3 signaling may help urologists to develop a treatment for better suppression of the human CRPC progression.


Subject(s)
Androgen Antagonists/adverse effects , Gene Expression Regulation, Neoplastic/drug effects , Neuroendocrine Tumors/pathology , Phenylthiohydantoin/analogs & derivatives , Prostatic Neoplasms, Castration-Resistant/pathology , RNA, Long Noncoding/metabolism , Animals , Benzamides , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line, Tumor , Disease Progression , Enhancer of Zeste Homolog 2 Protein/metabolism , Humans , Male , Mice , Mice, SCID , Neuroendocrine Cells/drug effects , Neuroendocrine Cells/pathology , Neuroendocrine Tumors/drug therapy , Neuroendocrine Tumors/genetics , Nitriles , Phenylthiohydantoin/adverse effects , Prostate/cytology , Prostate/drug effects , Prostate/pathology , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
17.
Int J Mol Sci ; 20(9)2019 May 01.
Article in English | MEDLINE | ID: mdl-31052388

ABSTRACT

Bisphenol A (BPA) is one of the most widely used chemicals worldwide, e.g., as a component of plastic containers for food and water. It is considered to exert an estrogenic effect, by mimicking estradiol (E2) action. Because of this widespread presence, it has attracted the interest and concern of researchers and regulators. Despite the vast amount of related literature, the potential adverse effects of environmentally significant doses of BPA are still object of controversy, and the mechanisms by which it can perturb endocrine functions, and particularly the neuroendocrine axis, are not adequately understood. One of the ways by which endocrine disruptors (EDCs) can exert their effects is the perturbation of calcium signaling mechanisms. In this study, we addressed the issue of the impact of BPA on the neuroendocrine system with an in vitro approach, using a consolidated model of immortalized Gonadotropin-Releasing Hormone (GnRH) expressing neurons, the GT1-7 cell line, focusing on the calcium signals activated by the endocrine disruptor. The investigation was limited to biologically relevant doses (nM-µM range). We found that BPA induced moderate increases in intracellular calcium concentration, comparable with those induced by nanomolar doses of E2, without affecting cell survival and with only a minor effect on proliferation.


Subject(s)
Benzhydryl Compounds/pharmacology , Calcium/metabolism , Endocrine Disruptors/pharmacology , Neuroendocrine Cells/drug effects , Phenols/pharmacology , Animals , Benzhydryl Compounds/toxicity , Cell Line , Endocrine Disruptors/toxicity , Gonadotropin-Releasing Hormone/metabolism , Ion Transport , Mice , Neuroendocrine Cells/metabolism , Phenols/toxicity
18.
Neuron ; 102(5): 1053-1065.e4, 2019 06 05.
Article in English | MEDLINE | ID: mdl-31006556

ABSTRACT

How general anesthesia (GA) induces loss of consciousness remains unclear, and whether diverse anesthetic drugs and sleep share a common neural pathway is unknown. Previous studies have revealed that many GA drugs inhibit neural activity through targeting GABA receptors. Here, using Fos staining, ex vivo brain slice recording, and in vivo multi-channel electrophysiology, we discovered a core ensemble of hypothalamic neurons in and near the supraoptic nucleus, consisting primarily of neuroendocrine cells, which are persistently and commonly activated by multiple classes of GA drugs. Remarkably, chemogenetic or brief optogenetic activations of these anesthesia-activated neurons (AANs) strongly promote slow-wave sleep and potentiates GA, whereas conditional ablation or inhibition of AANs led to diminished slow-wave oscillation, significant loss of sleep, and shortened durations of GA. These findings identify a common neural substrate underlying diverse GA drugs and natural sleep and reveal a crucial role of the neuroendocrine system in regulating global brain states. VIDEO ABSTRACT.


Subject(s)
Anesthetics, General/pharmacology , Hypnotics and Sedatives/pharmacology , Neuroendocrine Cells/drug effects , Sleep, Slow-Wave/drug effects , Supraoptic Nucleus/drug effects , Anesthesia, General , Animals , Dexmedetomidine/pharmacology , Electroencephalography , Electromyography , Electrophysiological Phenomena , Hypothalamus/cytology , Hypothalamus/drug effects , Hypothalamus/metabolism , Isoflurane/pharmacology , Ketamine/pharmacology , Mice , Neuroendocrine Cells/metabolism , Neurons/drug effects , Neurons/metabolism , Optogenetics , Patch-Clamp Techniques , Propofol/pharmacology , Proto-Oncogene Proteins c-fos/metabolism , Sleep/drug effects , Sleep/physiology , Sleep, Slow-Wave/physiology , Supraoptic Nucleus/cytology , Supraoptic Nucleus/metabolism
19.
Prostate ; 79(1): 21-30, 2019 01.
Article in English | MEDLINE | ID: mdl-30106164

ABSTRACT

BACKGROUND: Following androgen deprivation for the treatment of advanced adenocarcinoma of the prostate, tumors can progress to neuroendocrine prostate cancer (NEPC). This transdifferentiation process is poorly understood, but trafficking of transcriptional factors and/or cytoskeletal rearrangements may be involved. We observed the role of geranylgeranylation in this process by treatment with digeranyl bisphosphonate (DGBP), a selective inhibitor of geranylgeranyl pyrophosphate synthase which blocks the prenylation of small GTPases such as Rho and Rab family proteins, including Cdc42 and Rac1. METHODS: We examined the therapeutic potential of DGBP in LNCaP, C4-2B4, and 22Rv1 cell culture models. Cell morphology and protein expression were quantified to observe the development of the neuroendocrine phenotype in androgen-deprivation and abiraterone-treated LNCaP models of NEPC development. Luciferase reporter assays were utilized to examine AR activity, and immunofluorescence visualized the localization of AR within the cell. RESULTS: Essential genes in the isoprenoid pathway, such as HMGCR, MVK, GGPS1, and GGT1, were highly expressed in a subset of castration resistant prostate cancers reported by Beltran et al. Under treatment with DGBP, nuclear localization of AR decreased in LNCaP, 22Rv1, and C4-2B4 cell lines, luciferase reporter activity was reduced in LNCaP and 22Rv1, and AR target gene transcription also decreased in LNCaP. Conversely, nuclear localization of AR was enhanced by the addition of GGOH. Finally, induction of the NEPC structural and molecular phenotype via androgen deprivation in LNCaP cells was inhibited by DGBP in a GGOH-dependent manner. CONCLUSIONS: DGBP is a novel compound with the potential to reduce AR transcriptional activity and inhibit PCa progression to NEPC phenotype. These results suggest that DGBP may be used to block cell growth and metastasis in both hormone therapy sensitive and resistant paradigms.


Subject(s)
Cell Nucleus/metabolism , Diphosphonates/pharmacology , Neuroendocrine Cells/metabolism , Polyisoprenyl Phosphates/antagonists & inhibitors , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Terpenes/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Line, Tumor , Cell Nucleus/drug effects , Diphosphonates/therapeutic use , Dose-Response Relationship, Drug , Humans , Male , Neuroendocrine Cells/drug effects , Polyisoprenyl Phosphates/biosynthesis , Prostatic Neoplasms/drug therapy , Signal Transduction/drug effects , Signal Transduction/physiology , Terpenes/therapeutic use
20.
Mol Neurobiol ; 56(6): 4086-4101, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30267375

ABSTRACT

Several cues including pituitary adenylate cyclase-activating polypeptide (PACAP), which acts through cAMP stimulation, specify the conversion of sympathoadrenal (SA) precursors toward different cell phenotypes by promoting their survival and differentiation. Selenoprotein T (SELENOT) is a PACAP-stimulated ER oxidoreductase that exerts an essential antioxidant activity and whose up-regulation is associated with SA cell differentiation. In the present study, we investigated the transcriptional cascade elicited by PACAP/cAMP to trigger SELENOT gene transcription during the conversion of PC12 cells from SA progenitor-like cells toward a neuroendocrine phenotype. Unexpectedly, we found that PACAP/cAMP recruits the canonical pathway that regulates mitochondrial function in order to elicit SELENOT gene transcription and the consequent antioxidant response during PC12 cell differentiation. This cascade involves LKB1-mediated AMPK activation in order to stimulate SELENOT gene transcription through the PGC1-α/NRF-1 complex, thus allowing SELENOT to promote PACAP-stimulated neuroendocrine cell survival and differentiation. Our data reveal that a PACAP and cAMP-activated AMPK-PGC-1α/NRF-1 cascade is critical for the coupling of oxidative stress tolerance, via SELENOT gene expression, and mitochondrial biogenesis in order to achieve PC12 cell differentiation. The data further highlight the essential role of SELENOT in cell metabolism during differentiation.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Cell Differentiation/drug effects , Neuroendocrine Cells/cytology , Nuclear Respiratory Factor 1/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Selenoproteins/genetics , Transcription, Genetic/drug effects , Animals , Cell Survival/drug effects , Enzyme Activation/drug effects , HEK293 Cells , Humans , Models, Biological , Neuroendocrine Cells/drug effects , Neuroendocrine Cells/metabolism , Oxidative Stress/drug effects , PC12 Cells , Protein Binding/drug effects , Rats , Selenoproteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...