Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.359
Filter
1.
Cells ; 10(11)2021 10 26.
Article in English | MEDLINE | ID: mdl-34831120

ABSTRACT

Medullary thyroid carcinoma contributes to about 3-4% of thyroid cancers and affects C cells rather than follicular cells. Thyroid C cell differentiation from human pluripotent stem cells has not been reported. We report the stepwise differentiation of human embryonic stem cells into thyroid C cell-like cells through definitive endoderm and anterior foregut endoderm and ultimobranchial body-like intermediates in monolayer and 3D Matrigel culture conditions. The protocol involved sequential treatment with interferon/transferrin/selenium/pyruvate, foetal bovine serum, and activin A, then IGF-1 (Insulin-like growth factor 1), on the basis of embryonic thyroid developmental sequence. As well as expressing C cell lineage relative to follicular-lineage markers by qPCR (quantitative polymerase chain reaction) and immunolabelling, these cells by ELISA (enzyme-linked immunoassay) exhibited functional properties in vitro of calcitonin storage and release of calcitonin on calcium challenge. This method will contribute to developmental studies of the human thyroid gland and facilitate in vitro modelling of medullary thyroid carcinoma and provide a valuable platform for drug screening.


Subject(s)
Pluripotent Stem Cells/cytology , Thyroid Gland/cytology , Tissue Scaffolds/chemistry , Biomarkers/metabolism , Calcitonin/metabolism , Calcium/metabolism , Cell Differentiation/drug effects , Collagen/pharmacology , Drug Combinations , Endoderm/cytology , Gastrointestinal Tract/cytology , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/drug effects , Human Embryonic Stem Cells/metabolism , Humans , Laminin/pharmacology , Neurosecretory Systems/cytology , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Proteoglycans/pharmacology
2.
Nat Commun ; 11(1): 1266, 2020 03 09.
Article in English | MEDLINE | ID: mdl-32152276

ABSTRACT

Endophilins-A are conserved endocytic adaptors with membrane curvature-sensing and -inducing properties. We show here that, independently of their role in endocytosis, endophilin-A1 and endophilin-A2 regulate exocytosis of neurosecretory vesicles. The number and distribution of neurosecretory vesicles were not changed in chromaffin cells lacking endophilin-A, yet fast capacitance and amperometry measurements revealed reduced exocytosis, smaller vesicle pools and altered fusion kinetics. The levels and distributions of the main exocytic and endocytic factors were unchanged, and slow compensatory endocytosis was not robustly affected. Endophilin-A's role in exocytosis is mediated through its SH3-domain, specifically via a direct interaction with intersectin-1, a coordinator of exocytic and endocytic traffic. Endophilin-A not able to bind intersectin-1, and intersectin-1 not able to bind endophilin-A, resulted in similar exocytic defects in chromaffin cells. Altogether, we report that two endocytic proteins, endophilin-A and intersectin-1, are enriched on neurosecretory vesicles and regulate exocytosis by coordinating neurosecretory vesicle priming and fusion.


Subject(s)
Acyltransferases/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Cytoplasmic Vesicles/metabolism , Endocytosis/physiology , Neurosecretory Systems/metabolism , Acyltransferases/genetics , Animals , Chromaffin Cells/metabolism , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurosecretory Systems/cytology
3.
Sci Rep ; 10(1): 1892, 2020 02 05.
Article in English | MEDLINE | ID: mdl-32024913

ABSTRACT

Ascidians are the sister group of vertebrates and occupy a critical position in explorations of the evolution of the endocrine and nervous systems of chordates. Here, we describe the complete ventral peptidergic system in adult transgenic Ciona robusta (Ciona intestinalis Type A) which expresses the Kaede reporter gene driven by the prohormone convertase 2 (PC2) gene promoter. Numerous PC2 promoter-driven fluorescent (Kaede-positive) non-neural cells were distributed in the blood sinus located at the anterior end of the pharynx, suggesting the acquisition of a peptidergic circulatory system in Ciona. Kaede-positive ciliated columnar cells, rounded cells, and tall ciliated cells were observed in the alimentary organs, including the endostyle, pharynx, esophagus, stomach, and intestine, suggesting that digestive functions are regulated by multiple peptidergic systems. In the heart, Kaede-positive neurons were located in the ring-shaped plexus at both ends of the myocardium. Nerve fiber-like tracts ran along the raphe and appeared to be connected with the plexuses. Such unique structures suggest a role for the peptidergic system in cardiac function. Collectively, the present anatomic analysis revealed the major framework of the ventral peptidergic system of adult Ciona, which could facilitate investigations of peptidergic regulation of the pharynx, endostyle, alimentary tissues, and heart.


Subject(s)
Ciona intestinalis/physiology , Neuropeptides/metabolism , Neurosecretory Systems/metabolism , Animals , Animals, Genetically Modified , Esophagus/innervation , Esophagus/metabolism , Gastric Mucosa/innervation , Gastric Mucosa/metabolism , Genes, Reporter/genetics , Heart/innervation , Intestinal Mucosa/innervation , Intestinal Mucosa/metabolism , Luminescent Proteins/chemistry , Luminescent Proteins/genetics , Myocardium/metabolism , Neurons/metabolism , Neurosecretory Systems/cytology , Pharynx/innervation , Pharynx/metabolism
4.
Sci Rep ; 10(1): 1216, 2020 01 27.
Article in English | MEDLINE | ID: mdl-31988304

ABSTRACT

Advanced and therapy-resistant prostate tumors often display neural or neuroendocrine behavior. We assessed the consequences of prostate cancer cell interaction with neural cells, which are rich in the human prostate and resident of the prostate tumor. In 3-dimensional co-culture with neurospheres, red fluorescent human LNCaP cells formed agglomerates on the neurosphere surface. Upon induced neural differentiation, some red fluorescent cells showed morphology of fully differentiated neural cells, indicating fusion between the cancer and neural stem cells. These fusion hybrids survived for extended times in a quiescent state. A few eventually restarted cell division and propagated to form derivative hybrid progenies. Clones of the hybrid progenies were highly heterogeneous; most had lost prostatic and epithelial markers while some had acquired neural marker expression. These results indicate that cancer cells can fuse with bystander neural cells in the tumor microenvironment; and cancer cell fusion is a direct route to tumor cell heterogeneity.


Subject(s)
Neural Stem Cells/metabolism , Neuroendocrine Cells/metabolism , Prostatic Neoplasms/metabolism , Animals , Cell Communication/physiology , Cell Differentiation/physiology , Cell Fusion/methods , Cell Line, Tumor , Cell Proliferation/physiology , Coculture Techniques/methods , Humans , Male , Neural Stem Cells/physiology , Neurosecretory Systems/cytology , Prostate/cytology , Prostatic Neoplasms/immunology , Rats , Stromal Cells/cytology , Tumor Microenvironment/physiology
5.
Nihon Yakurigaku Zasshi ; 154(3): 97-102, 2019.
Article in Japanese | MEDLINE | ID: mdl-31527367

ABSTRACT

Among voltage-gated Ca2+ channels, T-type Ca2+ channels, which are activated by low voltages, regulate neuronal excitability, spontaneous neurotransmitter release, hormone secretion, etc. and also participate in proliferation of distinct cancer cells. Among three isoforms of T-type Ca2+ channels, Cav3.2 is detectable in 100% of biopsy samples from prostate cancer patients. In general, prostate cancer cells are highly sensitive to androgen deprivation therapy, but often acquire hormone-therapy resistance. The androgen deprivation may trigger neuroendocrine (NE)-like differentiation of some prostate cancer cells. We have analyzed the expression and function of Cav3.2 in human prostate cancer LNCaP cells during NE-like differentiation. NE-like LNCaP cells overexpress Cav3.2 through the CREB/Egr-1 pathway and also cystathionine-γ-lyase (CSE), which generates H2S that enhances the channel activity of Cav3.2. H2S generated by upregulated CSE appears to enhance the activity of upregulated Cav3.2 after the differentiation. The enhanced Cav3.2 activity in NE-like cells may contribute to increased secretion of mitogenic factors essential for androgen-independent proliferation of surrounding prostate cancer cells. It is known that increased extracellular glucose levels enhance Cav3.2 activity through asparagine (N)-linked glycosylation of Cav3.2, which might contribute to diabetic neuropathy. We then found that high glucose accelerates the enhanced channel function and overexpression of Cav3.2 in NE-like LNCaP cells, which might be associated with clinical evidence for diabetes-related poor prognosis of prostate cancer and development of hormone therapy resistance. Thus, Cav3.2 is considered to play a role in the pathophysiology of prostate cancer, and may serve as a therapeutic target.


Subject(s)
Calcium Channels, T-Type/physiology , Neurosecretory Systems/cytology , Prostatic Neoplasms/pathology , Androgen Antagonists/pharmacology , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Cystathionine gamma-Lyase/physiology , Humans , Hydrogen Sulfide , Male
6.
PLoS One ; 14(3): e0213927, 2019.
Article in English | MEDLINE | ID: mdl-30917148

ABSTRACT

The normal function of the mammalian reproductive axis is strongly influenced by physiological, metabolic and environmental factors. Kisspeptin neuropeptides, encoded by the Kiss1 gene, are potent regulators of the mammalian reproductive axis by stimulating gonadodropin releasing hormone secretion from the hypothalamus. To understand how the reproductive axis is modulated by higher order neuronal inputs we have mapped the afferent circuits into arcuate (ARC) Kiss1 neurons. We used a transgenic mouse that expresses the CRE recombinase in Kiss1 neurons for conditional viral tracing with genetically modified viruses. CRE-mediated activation of these viruses in Kiss1 neurons allows the virus to move transynaptically to label neurons with primary or secondary afferent inputs into the Kiss1 neurons. Several regions of the brain showed synaptic connectivity to arcuate Kiss1 neurons including proopiomelanocortin neurons in the ARC itself, kisspeptin neurons in the anteroventral periventricular nucleus, vasopressin neurons in the supraoptic and suprachiasmatic nuclei, thyrotropin releasing neurons in the paraventricular nucleus and unidentified neurons in other regions including the subfornical organ, amygdala, interpeduncular nucleus, ventral premammilary nucleus, basal nucleus of stria terminalis and the visual, somatosensory and piriform regions of the cortex. These data provide an insight into how the activity of Kiss1 neurons may be regulated by metabolic signals and provide a detailed neuroanatomical map for future functional studies.


Subject(s)
Arcuate Nucleus of Hypothalamus/cytology , Arcuate Nucleus of Hypothalamus/metabolism , Kisspeptins/metabolism , Neurons/metabolism , Animals , Brain Mapping , Female , Kisspeptins/genetics , Male , Mice , Mice, Transgenic , Neural Pathways/cytology , Neural Pathways/metabolism , Neurons/cytology , Neurosecretory Systems/cytology , Neurosecretory Systems/metabolism , Optogenetics , Reproduction/physiology , Synapses/metabolism
7.
Curr Top Dev Biol ; 132: 67-89, 2019.
Article in English | MEDLINE | ID: mdl-30797518

ABSTRACT

While the lung is commonly known for its gas exchange function, it is exposed to signals in the inhaled air and responds to them by collaborating with other systems including immune cells and the neural circuit. This important aspect of lung physiology led us to consider the lung as a sensory organ. Among different cell types within the lung that mediate this role, several recent studies have renewed attention on pulmonary neuroendocrine cells (PNECs). PNECs are a rare, innervated airway epithelial cell type that accounts for <1% of the lung epithelium population. They are enriched at airway branch points. Classical in vitro studies have shown that PNECs can respond to an array of aerosol stimuli such as hypoxia, hypercapnia and nicotine. Recent in vivo evidence suggests an essential role of PNECs at neuroimmunomodulatory sites of action, releasing neuropeptides, neurotransmitters and facilitating asthmatic responses to allergen. In addition, evidence supports that PNECs can function both as progenitor cells and progenitor niches following airway epithelial injury. Increases in PNECs have been documented in a large array of chronic lung diseases. They are also the cells-of-origin for small cell lung cancer. A better understanding of the specificity of their responses to distinct insults, their impact on normal lung function and their roles in the pathogenesis of pulmonary ailments will be the next challenge toward designing therapeutics targeting the neuroendocrine system in lung.


Subject(s)
Epithelial Cells/metabolism , Lung/embryology , Neuroendocrine Cells/metabolism , Neurosecretory Systems/embryology , Animals , Cell Lineage/genetics , Epithelial Cells/cytology , Epithelium/embryology , Epithelium/metabolism , Gene Expression Regulation, Developmental , Humans , Lung/cytology , Lung/metabolism , Neuroendocrine Cells/cytology , Neurosecretory Systems/cytology , Neurosecretory Systems/metabolism , Stem Cells/cytology , Stem Cells/metabolism
8.
Cell Tissue Res ; 375(1): 23-39, 2019 Jan.
Article in English | MEDLINE | ID: mdl-29869716

ABSTRACT

The neuroendocrine system consists of a heterogeneous collection of (mostly) neuropeptidergic neurons found in four hypothalamic nuclei and sharing the ability to secrete neurohormones (all of them neuropeptides except dopamine) into the bloodstream. There are, however, abundant hypothalamic non-neuroendocrine neuropeptidergic neurons developing in parallel with the neuroendocrine system, so that both cannot be entirely disentangled. This heterogeneity results from the workings of a network of transcription factors many of which are already known. Olig2 and Fezf2 expressed in the progenitors, acting through mantle-expressed Otp and Sim1, Sim2 and Pou3f2 (Brn2), regulate production of magnocellular and anterior parvocellular neurons. Nkx2-1, Rax, Ascl1, Neurog3 and Dbx1 expressed in the progenitors, acting through mantle-expressed Isl1, Dlx1, Gsx1, Bsx, Hmx2/3, Ikzf1, Nr5a2 (LH-1) and Nr5a1 (SF-1) are responsible for tuberal parvocellular (arcuate nucleus) and other neuropeptidergic neurons. The existence of multiple progenitor domains whose progeny undergoes intricate tangential migrations as one source of complexity in the neuropeptidergic hypothalamus is the focus of much attention. How neurosecretory cells target axons to the medial eminence and posterior hypophysis is gradually becoming clear and exciting progress has been made on the mechanisms underlying neurovascular interface formation. While rat neuroanatomy and targeted mutations in mice have yielded fundamental knowledge about the neuroendocrine system in mammals, experiments on chick and zebrafish are providing key information about cellular and molecular mechanisms. Looking forward, data from every source will be necessary to unravel the ways in which the environment affects neuroendocrine development with consequences for adult health and disease.


Subject(s)
Hypothalamus/cytology , Mammals/metabolism , Neurons/cytology , Neurosecretory Systems/cytology , Animals , Cell Movement , Gene Regulatory Networks
9.
Mech Ageing Dev ; 177: 74-79, 2019 01.
Article in English | MEDLINE | ID: mdl-29729230

ABSTRACT

Aging is characterized by a progressive loss of several physiological functions that can cause various age-related disorders. Several factors have been identified as causes of aging to elucidate the decline in functions. Various aspects of physiological deterioration are controlled by the hypothalamus, a critical brain region that connects the neuroendocrine system to physiological functions. In addition, functional alterations in a set of agouti-related peptide/neuropeptide Y (AgRP/NPY) and pro-opiomelanocortin (POMC) neurons, a set of growth hormone-releasing hormone (GHRH) and somatostatin (SST) neurons, a set of arginine vasopressin (AVP) and vasoactive intestinal peptide (VIP) neurons, and a set of gonadotropin-releasing hormone (GnRH) and kisspeptin/neurokinin B/dynorphin (KNDy) neurons contribute to age-related physiological decline in energy metabolism, hormone regulation, circadian rhythm, and reproduction, respectively. The underlying cellular mechanism for the hypothalamus-mediated aging progression comprises dysregulation of nutrient sensing, altered intercellular communication, stem cell exhaustion, loss of proteostasis, and epigenetic alterations. Furthermore, mammalian target of rapamycin (mTOR), NF-kB, hypothalamic stem cell, autophagy, and SIRT1 have been recognized as critical factors or pathways mediating the mechanism. Perhaps, further dissection of these pathways or components could provide the potential for developing a therapeutic intervention for age-related diseases or the extension of healthy lifespan.


Subject(s)
Aging/metabolism , Hypothalamus/metabolism , Nerve Tissue Proteins/metabolism , Neural Stem Cells/metabolism , Neurosecretory Systems/metabolism , Peptide Hormones/metabolism , Animals , Humans , Hypothalamus/cytology , Neural Stem Cells/cytology , Neurosecretory Systems/cytology
10.
Cancer Lett ; 433: 43-52, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29944905

ABSTRACT

Long non-coding RNAs (lncRNAs) are emerging as novel diagnostic markers of prostate cancer (PCa) and new determinants of castration-resistant PCa (CRPC), an aggressive and metastatic form of PCa. In addition to androgen receptor (AR) signaling, neuroendocrine differentiation (NED) is associated with CRPC. Recent reports demonstrate that the downregulation of repressor element-1 silencing transcription factor (REST) protein is a key step in NED of PCa cells. Here, we report HOTAIR as a novel REST-repressed lncRNA that is upregulated in NED PCa cells and in CRPC. HOTAIR overexpression is sufficient to induce, whereas knockdown of HOTAIR suppressed NED of PCa cells. Gene ontology (GO) analysis of differentially expressed genes under HOTAIR overexpression and in CRPC versus benign prostatic hyperplasia (BPH) suggests that HOTAIR may participate in PCa progression. Taken together, our results provide the first evidence of lncRNA HOTAIR as a driver for NED of PCa cells.


Subject(s)
Neurosecretory Systems/cytology , Prostatic Neoplasms, Castration-Resistant/genetics , RNA, Long Noncoding/genetics , Repressor Proteins/genetics , Up-Regulation , Cell Differentiation , Cell Line, Tumor , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Male , Prostatic Neoplasms, Castration-Resistant/metabolism , Repressor Proteins/metabolism , Sequence Analysis, RNA
11.
Cell Tissue Res ; 373(2): 487-498, 2018 08.
Article in English | MEDLINE | ID: mdl-29549435

ABSTRACT

The morphology and neurophysin expression of the magnocellular accessory neuroendocrine system located in the rostral human hypothalamus is investigated in a series of brains obtained at autopsy. The hypothalami were fixed in formalin and embedded in paraffin, or after cryoprotection, frozen for cryostat sectioning. Paraffin sections were either stained with Luxol Fast blue or immunoreacted for neurophysin I or neurophysin II, the precursor molecule for oxytocin and vasopressin. Further, 50-µm-thick serial cryostat sections were immunoreacted with the same antibodies. Both the paraventricular and supraoptic nuclei as well as the hypothalamo-hypophysial tracts exhibited strong immunoreactivity for the neurophysin antibodies. In addition, large collections of immunoreactive accessory magnocellular nuclei and single scattered neurophysin-positive neurons were located in the preoptic region between the paraventricular and supraoptic nucleus among the hypothalamo-hypophysial nerve fibers. In addition, smaller collections of neurophysin-immunoreactive neurons were located in the basal part of this region. Among the accessory magnocellular nuclei, the classical circular nucleus was identified. Accessory magnocellular neurons were often located along the blood vessels and projections of some of these neurons penetrated the vascular endothelium. The accessory magnocellular cell bodies expressed either neurophysin I or neurophysin II immunoreactivity. Summarizing, the accessory magnocellular system in the human brain is large and differs in morphology compared to the system seen in other vertebrates. The neurons of this system contain both vasopressin and oxytocin. Some neurons of the accessory neuronal systems might secrete vasopressin or oxytocin directly into the blood stream.


Subject(s)
Hypothalamus/cytology , Magnetic Phenomena , Neurosecretory Systems/cytology , Adult , Aged , Female , Humans , Male , Middle Aged , Young Adult
12.
Gen Comp Endocrinol ; 261: 9-22, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29355533

ABSTRACT

A neuromodulatory role for glutamate has been reported for magnocellular neuroendocrine cells in mammalian hypothalamus. We examined the potential role of glutamate as a local intercellular messenger in the neuroendocrine Dahlgren cell population of the caudal neurosecretory system (CNSS) in the euryhaline flounder Paralichthys olivaceus. In pharmacological experiments in vitro, glutamate (Glu) caused an increase in electrical activity of Dahlgren cells, recruitment of previously silent cells, together with a greater proportion of cells showing phasic (irregular) activity. The glutamate substrate, glutamine (Gln), led to increased firing frequency, cell recruitment and enhanced bursting activity. The glutamate effect was not blocked by the N-methyl-D-aspartate (NMDA) receptor antagonist MK-801, or the GluR1/GluR3 (AMPA) receptor antagonist IEm1795-2HBr, but was blocked by the broad-spectrum α-amino-3-hydroxy- 5- methyl-4-isoxazo-lepropionic acid (AMPA) receptor antagonist ZK200775. Our transcriptome sequencing study revealed three AMPA receptor (GluR1, GluR2 and GluR3) in the olive flounder CNSS. Quantitative RT-PCR revealed that GluR2 receptor mRNA expression was significant increased following dose-dependent superfusion with glutamate in the CNSS. GluR1 and GluR3 receptor mRNA expression were decreased following superfusion with glutamate. L-type Ca2+ channel mRNA expression had a significant dose-dependent decrease following superfusion with glutamate, compared to the control. In the salinity challenge experiment, acute transfer from SW to FW, GluR2 receptor mRNA expression was significantly higher than the control at 2 h. These findings suggest that GluR2 is one of the mechanisms which can medicate glutamate action within the CNSS, enhancing electrical activity and hence secretory output.


Subject(s)
Flounder/metabolism , Neurosecretory Systems/cytology , Neurosecretory Systems/metabolism , Receptors, AMPA/metabolism , Animals , Calcium Channels, L-Type/metabolism , Glutamic Acid/pharmacology , Glutamine/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, AMPA/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Sequence Analysis, RNA , Software , Transcriptome/genetics
13.
Development ; 145(2)2018 01 22.
Article in English | MEDLINE | ID: mdl-29358214

ABSTRACT

Oocyte meiotic maturation is crucial for sexually reproducing animals, and its core cytoplasmic regulators are highly conserved between species. By contrast, the few known maturation-inducing hormones (MIHs) that act on oocytes to initiate this process are highly variable in their molecular nature. Using the hydrozoan jellyfish species Clytia and Cladonema, which undergo oocyte maturation in response to dark-light and light-dark transitions, respectively, we deduced amidated tetrapeptide sequences from gonad transcriptome data and found that synthetic peptides could induce maturation of isolated oocytes at nanomolar concentrations. Antibody preabsorption experiments conclusively demonstrated that these W/RPRPamide-related neuropeptides account for endogenous MIH activity produced by isolated gonads. We show that the MIH peptides are synthesised by neural-type cells in the gonad, are released following dark-light/light-dark transitions, and probably act on the oocyte surface. They are produced by male as well as female jellyfish and can trigger both sperm and egg release, suggesting a role in spawning coordination. We propose an evolutionary link between hydrozoan MIHs and the neuropeptide hormones that regulate reproduction upstream of MIHs in bilaterian species.


Subject(s)
Hydrozoa/growth & development , Hydrozoa/physiology , Neuropeptides/physiology , Oocytes/growth & development , Oogenesis/physiology , Amino Acid Sequence , Animals , Darkness , Female , Gene Expression Profiling , Gonadal Steroid Hormones/genetics , Gonadal Steroid Hormones/pharmacology , Gonadal Steroid Hormones/physiology , Hydrozoa/genetics , Light , Male , Neuropeptides/genetics , Neuropeptides/pharmacology , Neurosecretory Systems/cytology , Oligopeptides/genetics , Oligopeptides/pharmacology , Oligopeptides/physiology , Oocytes/drug effects , Oogenesis/drug effects , Oogenesis/genetics , Species Specificity
14.
Nat Rev Endocrinol ; 14(1): 25-44, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29076504

ABSTRACT

Natural fluctuations in physiological conditions require adaptive responses involving rapid and reversible structural and functional changes in the hypothalamic neuroendocrine circuits that control homeostasis. Here, we discuss the data that implicate hypothalamic glia in the control of hypothalamic neuroendocrine circuits, specifically neuron-glia interactions in the regulation of neurosecretion as well as neuronal excitability. Mechanistically, the morphological plasticity displayed by distal processes of astrocytes, pituicytes and tanycytes modifies the geometry and diffusion properties of the extracellular space. These changes alter the relationship between glial cells of the hypothalamus and adjacent neuronal elements, especially at specialized intersections such as synapses and neurohaemal junctions. The structural alterations in turn lead to functional plasticity that alters the release and spread of neurotransmitters, neuromodulators and gliotransmitters, as well as the activity of discrete glial signalling pathways that mediate feedback by peripheral signals to the hypothalamus. An understanding of the contributions of these and other non-neuronal cell types to hypothalamic neuroendocrine function is thus critical both to understand physiological processes such as puberty, the maintenance of bodily homeostasis and ageing and to develop novel therapeutic strategies for dysfunctions of these processes, such as infertility and metabolic disorders.


Subject(s)
Cell Communication/physiology , Hypothalamus/physiology , Neuroglia/physiology , Neurons/physiology , Neurosecretory Systems/physiology , Sexual Maturation/physiology , Animals , Humans , Hypothalamus/cytology , Neurosecretory Systems/cytology
15.
Sci Rep ; 7(1): 13580, 2017 10 19.
Article in English | MEDLINE | ID: mdl-29051571

ABSTRACT

Telocytes, a peculiar cell type, were recently found in vertebrates. Hence this cell system has been reported as ubiquitous in the bodies of mammals and interpreted as an important player in innate immunity and tissue regeneration, it is reasonable to look for it also in invertebrates, that rely their integrity solely by innate immunity. Here we describe, at morphological and functional level, invertebrate telocytes from the body of leech Hirudo medicinalis (Annelida), suggesting how these cells, forming a resident stromal 3D network, can influence or participate in different events. These findings support the concepts that leech telocytes: i) are organized in a cellular dynamic and versatile 3D network likewise the vertebrate counterpart; ii) are an evolutionarily conserved immune-neuroendocrine system; iii) form an immuno-surveillance system of resident cells responding faster than migrating immunocytes recruited in stimulated area; iv) communicate with neighbouring cells directly and indirectly, via cell-cell contacts and soluble molecules secreted by multivesicular bodies; v) present within neo-vessels, share with immunocytes the mesodermal lineage; vi) are involved in regenerative processes. In conclusion, we propose that HmTCs, integrating so different functions, might explain the innate immune memory and can be associated with several aged related diseases.


Subject(s)
Hirudo medicinalis/cytology , Hirudo medicinalis/physiology , Animals , Biomarkers/metabolism , Immunity, Cellular , Microscopy, Electron, Transmission , Neurosecretory Systems/cytology , Wound Healing
16.
Dev Biol ; 431(2): 226-238, 2017 11 15.
Article in English | MEDLINE | ID: mdl-28916169

ABSTRACT

Split ends (SPEN) is the founding member of a well conserved family of nuclear proteins with critical functions in transcriptional regulation and the post-transcriptional processing and nuclear export of transcripts. In animals, the SPEN proteins fall into two size classes that perform either complementary or antagonistic functions in different cellular contexts. Here, we show that the two Drosophila representatives of this family, SPEN and Spenito (NITO), regulate metamorphic remodeling of the CCAP/bursicon neurosecretory cells. CCAP/bursicon cell-targeted overexpression of SPEN had no effect on the larval morphology or the pruning back of the CCAP/bursicon cell axons at the onset of metamorphosis. During the subsequent outgrowth phase of metamorphic remodeling, overexpression of either SPEN or NITO strongly inhibited axon extension, axon branching, peripheral neuropeptide accumulation, and soma growth. Cell-targeted loss-of-function alleles for both spen and nito caused similar reductions in axon outgrowth, indicating that the absolute levels of SPEN and NITO activity are critical to support the developmental plasticity of these neurons. Although nito RNAi did not affect SPEN protein levels, the phenotypes produced by SPEN overexpression were suppressed by nito RNAi. We propose that SPEN and NITO function additively or synergistically in the CCAP/bursicon neurons to regulate multiple aspects of neurite outgrowth during metamorphic remodeling.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Multigene Family , Neuronal Outgrowth , Neurosecretory Systems/cytology , Neurosecretory Systems/metabolism , Animals , Larva/metabolism , Neurons/metabolism , Neurosecretion , Presynaptic Terminals/metabolism , RNA Interference , Wings, Animal/metabolism
17.
World J Gastroenterol ; 23(28): 5068-5085, 2017 Jul 28.
Article in English | MEDLINE | ID: mdl-28811704

ABSTRACT

Inflammatory bowel disease (IBD) is a chronic recurrent condition whose etiology is unknown, and it includes ulcerative colitis, Crohn's disease, and microscopic colitis. These three diseases differ in clinical manifestations, courses, and prognoses. IBD reduces the patients' quality of life and is an economic burden to both the patients and society. Interactions between the gastrointestinal (GI) neuroendocrine peptides/amines (NEPA) and the immune system are believed to play an important role in the pathophysiology of IBD. Moreover, the interaction between GI NEPA and intestinal microbiota appears to play also a pivotal role in the pathophysiology of IBD. This review summarizes the available data on GI NEPA in IBD, and speculates on their possible role in the pathophysiology and the potential use of this information when developing treatments. GI NEPA serotonin, the neuropeptide Y family, and substance P are proinflammatory, while the chromogranin/secretogranin family, vasoactive intestinal peptide, somatostatin, and ghrelin are anti-inflammatory. Several innate and adaptive immune cells express these NEPA and/or have receptors to them. The GI NEPA are affected in patients with IBD and in animal models of human IBD. The GI NEPA are potentially useful for the diagnosis and follow-up of the activity of IBD, and are candidate targets for treatments of this disease.


Subject(s)
Gastrointestinal Microbiome , Gastrointestinal Tract/immunology , Inflammatory Bowel Diseases/immunology , Neurosecretory Systems/immunology , Amines/immunology , Animals , Chromogranins/immunology , Chromogranins/metabolism , Disease Models, Animal , Gastrointestinal Tract/metabolism , Ghrelin/immunology , Ghrelin/metabolism , Humans , Inflammatory Bowel Diseases/diagnosis , Inflammatory Bowel Diseases/epidemiology , Inflammatory Bowel Diseases/therapy , Neuroendocrine Cells/immunology , Neuroendocrine Cells/metabolism , Neuropeptide Y/antagonists & inhibitors , Neuropeptide Y/immunology , Neuropeptide Y/metabolism , Neurosecretory Systems/cytology , Prevalence , Quality of Life , Recurrence , Serotonin/immunology , Serotonin/metabolism , Serotonin Antagonists/therapeutic use , Somatostatin/immunology , Somatostatin/metabolism , Substance P/antagonists & inhibitors , Substance P/immunology , Substance P/metabolism , Vasoactive Intestinal Peptide/immunology , Vasoactive Intestinal Peptide/metabolism
18.
PLoS One ; 12(7): e0182001, 2017.
Article in English | MEDLINE | ID: mdl-28753616

ABSTRACT

The structure and regeneration of the digestive system in the crinoid Himerometra robustipinna (Carpenter, 1881) were studied. The gut comprises a spiral tube forming radial lateral processes, which gives it a five-lobed shape. The digestive tube consists of three segments: esophagus, intestine, and rectum. The epithelia of these segments have different cell compositions. Regeneration of the gut after autotomy of the visceral mass progresses very rapidly. Within 6 h after autotomy, an aggregation consisting of amoebocytes, coelomic epithelial cells and juxtaligamental cells (neurosecretory neurons) forms on the inner surface of the skeletal calyx. At 12 h post-autotomy, transdifferentiation of the juxtaligamental cells starts. At 24 h post-autotomy these cells undergo a mesenchymal-epithelial-like transition, resulting in the formation of the luminal epithelium of the gut. Specialization of the intestinal epithelial cells begins on day 2 post-autotomy. At this stage animals acquire the mouth and anal opening. On day 4 post-autotomy the height of both the enterocytes and the visceral mass gradually increases. Proliferation does not play any noticeable role in gut regeneration. The immersion of animals in a 10-7 M solution of colchicine neither stopped formation of the lost structures nor caused accumulation of mitoses in tissues. Weakly EdU-labeled nuclei were observed in the gut only on day 2 post-autotomy and were not detected at later regeneration stages. Single mitotically dividing cells were recorded during the same period. It is concluded that juxtaligamental cells play a major role in gut regeneration in H. robustipinna. The main mechanisms of morphogenesis are cell migration and transdifferentiation.


Subject(s)
Cell Transdifferentiation , Echinodermata/cytology , Echinodermata/physiology , Gastrointestinal Tract/physiology , Neurosecretory Systems/cytology , Regeneration/physiology , Animals , Cell Transdifferentiation/drug effects , Colchicine/pharmacology , DNA/biosynthesis , Echinodermata/drug effects , Epithelium/drug effects , Epithelium/ultrastructure , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/ultrastructure , Imaging, Three-Dimensional , Mitosis/drug effects , Neurosecretory Systems/drug effects , Regeneration/drug effects , Solutions
19.
Sci Rep ; 7(1): 5899, 2017 07 19.
Article in English | MEDLINE | ID: mdl-28724969

ABSTRACT

Three-dimensional tissue-structural relationships are not well captured by typical thin-section histology, posing challenges for the study of tissue physiology and pathology. Moreover, while recent progress has been made with intact methods for clearing, labeling, and imaging whole organs such as the mature brain, these approaches are generally unsuitable for soft, irregular, and heterogeneous tissues that account for the vast majority of clinical samples and biopsies. Here we develop a biphasic hydrogel methodology, which along with automated analysis, provides for high-throughput quantitative volumetric interrogation of spatially-irregular and friable tissue structures. We validate and apply this approach in the examination of a variety of developing and diseased tissues, with specific focus on the dynamics of normal and pathological pancreatic innervation and development, including in clinical samples. Quantitative advantages of the intact-tissue approach were demonstrated compared to conventional thin-section histology, pointing to broad applications in both research and clinical settings.


Subject(s)
Disease , Imaging, Three-Dimensional/methods , Organogenesis , Animals , Female , Humans , Hydrogels/chemistry , Mice, Inbred C57BL , Neural Crest/cytology , Neurosecretory Systems/cytology , Pancreas/cytology
20.
Physiol Behav ; 176: 195-206, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28095318

ABSTRACT

Periods of caloric deficit substantially attenuate many centrally mediated responses to acute stress, including neural drive to the hypothalamic-pituitary-adrenal (HPA) axis, anxiety-like behavior, and stress-induced suppression of food intake (i.e., stress hypophagia). It is posited that this stress response plasticity supports food foraging and promotes intake during periods of negative energy balance, even in the face of other internal or external threats, thereby increasing the likelihood that energy stores are repleted. The mechanisms by which caloric deficit alters central stress responses, however, remain unclear. The caudal brainstem contains two distinct populations of stress-recruited neurons [i.e., noradrenergic neurons of the A2 cell group that co-express prolactin-releasing peptide (PrRP+ A2 neurons), and glucagon-like peptide 1 (GLP-1) neurons] that also are responsive to interoceptive feedback about feeding and metabolic status. A2/PrRP and GLP-1 neurons have been implicated anatomically and functionally in the central control of the HPA axis, anxiety-like behavior, and stress hypophagia. The current review summarizes a growing body of evidence that caloric deficits attenuate physiological and behavioral responses to acute stress as a consequence of reduced recruitment of PrRP+ A2 and hindbrain GLP-1 neurons, accompanied by reduced signaling to their brainstem, hypothalamic, and limbic forebrain targets.


Subject(s)
Eating/physiology , Emotions/physiology , Interoception/physiology , Neurosecretory Systems/metabolism , Stress, Psychological , Animals , Humans , Inhibition, Psychological , Neurons/physiology , Neurosecretory Systems/cytology , Stress, Psychological/metabolism , Stress, Psychological/physiopathology , Stress, Psychological/psychology
SELECTION OF CITATIONS
SEARCH DETAIL
...