Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
1.
Nature ; 590(7844): 115-121, 2021 02.
Article in English | MEDLINE | ID: mdl-33299180

ABSTRACT

Behavioural experiences activate the FOS transcription factor in sparse populations of neurons that are critical for encoding and recalling specific events1-3. However, there is limited understanding of the mechanisms by which experience drives circuit reorganization to establish a network of Fos-activated cells. It is also not known whether FOS is required in this process beyond serving as a marker of recent neural activity and, if so, which of its many gene targets underlie circuit reorganization. Here we demonstrate that when mice engage in spatial exploration of novel environments, perisomatic inhibition of Fos-activated hippocampal CA1 pyramidal neurons by parvalbumin-expressing interneurons is enhanced, whereas perisomatic inhibition by cholecystokinin-expressing interneurons is weakened. This bidirectional modulation of inhibition is abolished when the function of the FOS transcription factor complex is disrupted. Single-cell RNA-sequencing, ribosome-associated mRNA profiling and chromatin analyses, combined with electrophysiology, reveal that FOS activates the transcription of Scg2, a gene that encodes multiple distinct neuropeptides, to coordinate these changes in inhibition. As parvalbumin- and cholecystokinin-expressing interneurons mediate distinct features of pyramidal cell activity4-6, the SCG2-dependent reorganization of inhibitory synaptic input might be predicted to affect network function in vivo. Consistent with this prediction, hippocampal gamma rhythms and pyramidal cell coupling to theta phase are significantly altered in the absence of Scg2. These findings reveal an instructive role for FOS and SCG2 in establishing a network of Fos-activated neurons via the rewiring of local inhibition to form a selectively modulated state. The opposing plasticity mechanisms acting on distinct inhibitory pathways may support the consolidation of memories over time.


Subject(s)
Nerve Net/cytology , Nerve Net/physiology , Neural Inhibition , Neuronal Plasticity/physiology , Proto-Oncogene Proteins c-fos/metabolism , Animals , CA1 Region, Hippocampal/metabolism , Cholecystokinin/metabolism , Exploratory Behavior/physiology , Female , Gamma Rhythm , Interneurons/metabolism , Male , Memory Consolidation , Mice , Parvalbumins/metabolism , Pyramidal Cells/metabolism , Secretogranin II/genetics , Secretogranin II/metabolism , Spatial Navigation/physiology , Theta Rhythm
2.
Apoptosis ; 29(1-2): 121-141, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37848672

ABSTRACT

Bladder cancer (BLCA) is ranked among the top ten most prevalent cancers worldwide and is the second most common malignant tumor within the field of urology. The limited effectiveness of immune targeted therapy in treating BLCA, due to its high metastasis and recurrence rates, necessitates the identification of new therapeutic targets. Secretogranin II (SCG2), a member of the chromaffin granin/secreted granin family, plays a crucial role in the regulated release of peptides and hormones. The role of SCG2 in the tumor microenvironment (TME) of lung adenocarcinoma and colon cancer has been established, but its functional significance in BLCA remains uncertain. This study aimed to investigate SCG2 expression in 15 bladder cancer tissue samples and their corresponding adjacent control tissues. The potential involvement of SCG2 in BLCA progression was assessed using various techniques, including analysis of public databases, immunohistochemistry, Western Blotting, immunofluorescence, wound-healing assay, Transwell assay, and xenograft tumor formation experiments in nude mice. This study provided novel evidence indicating that SCG2 plays a pivotal role in facilitating the proliferation, migration, and invasion of BLCA by activating the MEK/Erk and MEK/IKK/NF-κB signaling pathways, as well as by promoting M2 macrophage polarization. These findings propose the potential of SCG2 as a molecular target for immunotherapy in human BLCA.


Subject(s)
NF-kappa B , Urinary Bladder Neoplasms , Animals , Humans , Mice , Apoptosis , Chromogranins/therapeutic use , Mice, Nude , Mitogen-Activated Protein Kinase Kinases , NF-kappa B/genetics , NF-kappa B/metabolism , Secretogranin II/genetics , Secretogranin II/metabolism , Secretogranin II/therapeutic use , Tumor Microenvironment , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/metabolism
3.
Proc Natl Acad Sci U S A ; 117(23): 12772-12783, 2020 06 09.
Article in English | MEDLINE | ID: mdl-32467166

ABSTRACT

The luteinizing hormone surge is essential for fertility as it triggers ovulation in females and sperm release in males. We previously reported that secretoneurin-a, a neuropeptide derived from the processing of secretogranin-2a (Scg2a), stimulates luteinizing hormone release, suggesting a role in reproduction. Here we provide evidence that mutation of the scg2a and scg2b genes using TALENs in zebrafish reduces sexual behavior, ovulation, oviposition, and fertility. Large-scale spawning within-line crossings (n = 82 to 101) were conducted. Wild-type (WT) males paired with WT females successfully spawned in 62% of the breeding trials. Spawning success was reduced to 37% (P = 0.006), 44% (P = 0.0169), and 6% (P < 0.0001) for scg2a-/- , scg2b-/- , and scg2a-/-;scg2b-/- mutants, respectively. Comprehensive video analysis indicates that scg2a-/-;scg2b-/- mutation reduces all male courtship behaviors. Spawning success was 47% in saline-injected WT controls compared to 11% in saline-injected scg2a-/-;scg2b-/- double mutants. For these mutants, spawning success increased 3-fold following a single intraperitoneal (i.p.) injection of synthetic secretoneurin-a (P = 0.0403) and increased 3.5-fold with injection of human chorionic gonadotropin (hCG). Embryonic survival at 24 h remained on average lower in scg2a-/-;scg2b-/- fish compared to WT injected with secretoneurin-a (P < 0.001). Significant reductions in the expression of gonadotropin-releasing hormone 3 in the hypothalamus, and luteinizing hormone beta and glycoprotein alpha subunits in the pituitary provide evidence for disrupted hypothalamo-pituitary function in scg2a and scg2b mutant fish. Our results indicate that secretogranin-2 is required for optimal reproductive function and support the hypothesis that secretoneurin is a reproductive hormone.


Subject(s)
Fertility , Mating Preference, Animal , Mutation , Secretogranin II/genetics , Zebrafish Proteins/genetics , Animals , Female , Gonadotropin-Releasing Hormone/metabolism , Hypothalamus/metabolism , Luteinizing Hormone/metabolism , Male , Neuropeptides/metabolism , Oviposition , Ovulation , Pituitary Gland/metabolism , Secretogranin II/metabolism , Zebrafish , Zebrafish Proteins/metabolism
4.
Mol Vis ; 26: 576-587, 2020.
Article in English | MEDLINE | ID: mdl-32863706

ABSTRACT

Purpose: The cornea is highly enriched in sensory neurons expressing the thermal TRP channels TRPV1, TRPA1, and TRPM8, and is an accessible tissue for study and experimental manipulation. The aim of this work was to provide a concise characterization of the expression patterns of various TRP channels and vesicular proteins in the mammalian cornea. Methods: Immunohistochemistry (IHC) was performed using wholemount and cryostat tissue preparations of mouse and monkey corneas. The expression patterns of TRPV1 and TRPA1 were determined using specific antisera, and further colocalization was performed with antibodies directed against calcitonin-related gene protein (CGRP), neurofilament protein NF200, and the secretogranins ScgII and SCG3. The expression of TRPM8 was determined using corneas from mice expressing EGFP under the direction of a TRPM8 promoter (TRPM8EGFP mice). Laser scanning confocal microscopy and image analysis were performed. Results: In the mouse cornea, TRPV1 and TRPM8 were expressed in distinct populations of small diameter C fibers extending to the corneal surface and ending either as simple or ramifying terminals, or in the case of TRPM8, as complex terminals. TRPA1 was expressed in large-diameter NF200-positive Aδ axons. TRPV1 and TRPA1 appeared to localize to separate intracellular vesicular structures and were primarily found in axons containing components of large dense vesicles with TRPV1 colocalizing with CGRP and ScgII, and TRPA1 colocalizing with SCG3. Monkey corneas showed similar colocalization of CGRP and TRPV1 on small-diameter axons extending to the epithelial surface. Conclusions: The mouse cornea is abundant in sensory neurons expressing TRPV1, TRPM8, and TRPA1, and provides an accessible tissue source for implementing a live tissue preparation useful for further exploration of the molecular mechanisms of hyperalgesia. This study showed that surprisingly, these TRP channels localize to separate neurons in the mouse cornea and likely have unique physiological functions. The similar TRPV1 expression pattern we observed in the mouse and monkey corneas suggests that mice provide a reasonable initial model for understanding the role of these ion channels in higher mammalian corneal physiology.


Subject(s)
Axons/metabolism , Cornea/metabolism , Sensory Receptor Cells/metabolism , TRPA1 Cation Channel/genetics , TRPM Cation Channels/genetics , TRPV Cation Channels/genetics , Animals , Axons/ultrastructure , Chromogranins/genetics , Chromogranins/metabolism , Conserved Sequence , Cornea/anatomy & histology , Cornea/ultrastructure , Gene Expression , Hyperalgesia/genetics , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Immunohistochemistry , Macaca nemestrina , Mice , Receptors, Calcitonin Gene-Related Peptide/genetics , Receptors, Calcitonin Gene-Related Peptide/metabolism , Secretogranin II/genetics , Secretogranin II/metabolism , Sensory Receptor Cells/ultrastructure , Synaptic Transmission/genetics , TRPA1 Cation Channel/metabolism , TRPM Cation Channels/metabolism , TRPV Cation Channels/metabolism
5.
Gen Comp Endocrinol ; 299: 113588, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32828813

ABSTRACT

Secretogranin-2 (SCG2) is a large precursor protein that is processed into several potentially bioactive peptides, with the 30-43 amino acid central domain called secretoneurin (SN) being clearly evolutionary conserved in vertebrates. Secretoneurin exerts a diverse array of biological functions including regulating nervous, endocrine, and immune systems in part due to its wide tissue distribution. Expressed in some neuroendocrine neurons and pituitary cells, SN is a stimulator of the synthesis and release of luteinizing hormone from both goldfish pituitary cells and the mouse LßT2 cell line. Neuroendocrine, paracrine and autocrine signaling pathways for the stimulation of luteinizing hormone release indicate hormone-like activities to regulate reproduction. Mutation of the scg2a and scg2b genes using TALENs in zebrafish reduces sexual behavior, ovulation, oviposition, and fertility. A single injection of the SNa peptide enhanced reproductive outcomes in scg2a/scg2b double mutant zebrafish. Evidence in goldfish suggests a new role for SN to stimulate food intake by actions on other feeding-related neuropeptides. Expression and regulation of the Scg2a precursor mRNA in goldfish gut also supports a role in feeding. In rodent models, SN has trophic-like properties promoting both neuroprotection and neuronal plasticity and has chemoattractant properties that regulate neuroinflammation. Data obtained from several cellular models suggest that SN binds to and activates a G-protein coupled receptor (GPCR), but a bona fide SN receptor protein needs to be identified. Other signaling pathways for SN have been reported which provides alternatives to the GPCR hypothesis. These include AMP-activated protein kinase (AMPK), extracellular signal-regulated kinases (ERK), mitogen-activated protein kinase (MAPK)and calcium/calmodulin-dependent protein kinase II in cardiomyocytes, phosphatidylinositol 3-kinase (PI3K) and Akt/Protein Kinase B (AKT, and MAPK in endothelial cells and Janus kinase 2/signal transducer and activator of transcription protein (JAK2-STAT) signaling in neurons. Some studies in cardiac cells provide evidence for cellular internalization of SN by an unknown mechanism. Many of the biological functions of SN remain to be fully characterized, which could lead to new and exciting applications.


Subject(s)
Neuropeptides/metabolism , Secretogranin II/metabolism , Amino Acid Sequence , Animals , Female , Goldfish , Humans , Male , Mice , Zebrafish
6.
FASEB J ; 32(9): 4815-4823, 2018 09.
Article in English | MEDLINE | ID: mdl-29913555

ABSTRACT

The pathogenesis of diabetic neuropathy remains enigmatic. Damage to the vasa nervorum may be responsible for this disorder. Recently, we showed that secretoneurin (SN) induces angiogenesis in hindlimb and myocardial ischemia. Moreover, beneficial effects were observed in wound healing. We therefore hypothesized that SN therapy may ameliorate diabetic neuropathy. We used db/db mice as animal model for neuropathy. Gene therapy was accomplished by intramuscular injection of SN plasmid along the sciatic nerve. Sciatic nerve motor and sensory conduction velocities were then measured for 9 wk. Nerve conduction velocities showed normal values in heterozygous mice for the observational period, but were severely reduced in homozygous mice in which velocities were significantly improved by SN, but not by control plasmid gene therapy. The reaction time in the tail-flick test improved significantly in SN-treated animals. The induction of growth of vasa nervorum seems to be part of the underlying mechanism. In addition, SN positively affected Schwann cell function in vitro and induced activation of important signaling pathways. Our observations suggest that SN exerts beneficial effects on nerve function in vivo and on Schwann cells in vitro. It therefore may be a promising treatment option for diabetic neuropathy.-Theurl, M., Lener, D., Albrecht-Schgoer, K., Beer, A., Schgoer, W., Liu, Y., Stanzl, U., Fischer-Colbrie, R., Kirchmair, R. Gene therapy with the angiogenic neuropeptide secretoneurin ameliorates experimental diabetic neuropathy.


Subject(s)
Diabetes Mellitus, Experimental/therapy , Diabetic Neuropathies/therapy , Genetic Therapy , Neuropeptides/therapeutic use , Secretogranin II/therapeutic use , Animals , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetic Neuropathies/genetics , Disease Models, Animal , Humans , Mice , Myocardial Ischemia/genetics , Myocardial Ischemia/metabolism , Neovascularization, Physiologic/genetics , Neuropeptides/metabolism , Schwann Cells/metabolism , Secretogranin II/metabolism
7.
Biochem Biophys Res Commun ; 497(2): 675-682, 2018 03 04.
Article in English | MEDLINE | ID: mdl-29454966

ABSTRACT

It has been extensively characterized that paraquat (PQ) selectively targets to the substantia nigra and exerts neurotoxic actions on dopaminergic neurons. However, a little knowledge is available about astroglia in PQ exposure, especially its complex secretory machinery. To explore this point, we built up a PQ-induced model in cultural U118 astrocyte. Since the granin family is considered as a master regulator of cargo sorting and large dense core vesicles (LDCVs) biogenesis in the regulated secretory pathway of nervous and neuroendocrine cells, the current study focused on one member, secretogranin II (SCG2) and investigated its alternation and potential relationship with other astrocyte-derived factors under PQ insult. We found that PQ upregulated SCG2 expression on both RNA and protein levels and stimulated the mRNA expression of neurotrophic factors, cytokines and glutamine synthetase (GS) simultaneously. RNAi knockdown of SCG2 did not rescue the cell cycle arrest induced by PQ but affected expressions of IL-6 and GS on mRNA and protein levels. Further studies on subcellular location showed that SCG2-positive secretory granules were partially colocalized with IL-6 but not GS in PQ exposure astrocyte. Taken together, our findings indicate that the expression alternation of SCG2 under astroglial activation by PQ may be necessary compensation for cargo sorting and LDCV biogenesis. The involvement of the IL-6 and GS suggests that the SCG2 may potentially regulate inflammatory factors and excitatory neurotransmitter to the cytotoxicity of PQ on astroglia.


Subject(s)
Astrocytes/drug effects , Herbicides/toxicity , Paraquat/toxicity , Secretogranin II/genetics , Up-Regulation/drug effects , Astrocytes/cytology , Astrocytes/metabolism , Astrocytes/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Humans , Protein Interaction Maps , Secretogranin II/metabolism
8.
Gen Comp Endocrinol ; 261: 115-126, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29486146

ABSTRACT

Secretoneurin (SN) is an important stimulator of pituitary luteinizing hormone (LH) synthesis and secretion in goldfish. It is unknown whether this neuropeptide performs the same role in other fish species. In this study, the full-length cDNAs encoding Secretogranin IIa (SgIIa) and b (SgIIb) were cloned from the brain of orange-spotted grouper. Sequence analysis showed that a 34-amino acid SN peptide (SNa) is present in SgIIa proprotein, and a 33-amino acid SN peptide (SNb) is present in SgIIb proprotein. The two SN peptides share a low degree of similarity but contain the signature YTPQ-X-LA-X7-EL sequence. Real-time PCR showed that two SgII genes are mainly expressed in the brain and pituitary. During ovarian development, the expression levels of two SgII genes in the hypothalamus and pituitary were significantly reduced at the stage when the ovary contained full-grown oocytes. The biological functions of the two SN peptides were further investigated in vitro and in vivo. Both SN peptides could significantly elevate the mRNA levels of Gonadotropin-Releasing Hormone 1 (GnRH1) and 3 (GnRH3) in the hypothalamic fragments and upregulated the expression of Follicle-Stimulating Hormone beta (FSHb) and Luteinizing Hormone beta (LHb) in the pituitary cells. The stimulatory effects on the expression of GnRHs and Gonadotropins were also observed after intraperitoneal injection of SN peptides. Our study indicated that the SgII/SN system has stimulatory effects on the reproductive axis of orange-spotted grouper.


Subject(s)
Bass/genetics , Reproduction/genetics , Secretogranin II/genetics , Secretogranin II/physiology , Amino Acid Sequence , Animals , Bass/metabolism , Cloning, Molecular , DNA, Complementary/genetics , DNA, Complementary/isolation & purification , Female , Gene Expression Profiling , Male , Secretogranin II/isolation & purification , Secretogranin II/metabolism , Sequence Analysis, DNA
9.
Bratisl Lek Listy ; 118(10): 609-612, 2017.
Article in English | MEDLINE | ID: mdl-29198128

ABSTRACT

BACKGROUND: Pro-protein processing mechanism plays an important role in neuron injury. OBJECTIVE: To study the protein convertase 1 (PC1) mediated processing mechanism, the ischemic cellular or tissue proPC1/PC1 or proCgA/CgA (pro-chromogranin A) was analyzed. METHODS: NS20Y differentiated cells were stressed by 0-6 h of oxygen and glucose deprivation (OGD) in glucose-free DMEM and an anaerobic jar environment. Ischemic C57BL/J mouse model was established by performing 60-min of middle cerebral artery occlusion (MCAO) operation and subsequent 4 or 24-h reperfusion. The TUNEL, immunochemistry, and Western blot methods were used to detect protein expression in ischemic cells or tissues. RESULTS: The OGD or MCAO stress caused substantial cell death in a dose-dependent manner (p < 0.05 or 0.01). With the increasing OGD dose, proPC1 and PC1 proteins gradually increased (p < 0.05 or 0.01) whereas proCgA and CgA proteins decreased (p < 0.05). In vivo the proPC1 and PC1 expressions presented with a peak at 4-h and then decreased at 24-h reperfusion (p < 0.05 or 0.01). The tissue proCgA and CgA proteins decreased with the increasing reperfusion time (p < 0.05). CONCLUSIONS: The results suggest that the increasing PC1 expression promoted the transformation of proCgA into CgA or smaller peptides, i.e. Pancreastatin or Secretoneurin, and the PC1 mediated processing plays a critical role (Fig. 4, Ref. 15).


Subject(s)
Cell Death , Chromogranin A/metabolism , Infarction, Middle Cerebral Artery/metabolism , Neurons/metabolism , Proprotein Convertase 1/metabolism , Animals , Blotting, Western , Brain Ischemia/metabolism , Cell Differentiation , Cell Hypoxia , Glucose , In Situ Nick-End Labeling , Mice , Mice, Inbred C57BL , Neuropeptides/metabolism , Secretogranin II/metabolism
10.
J Neurosci ; 35(44): 14872-84, 2015 Nov 04.
Article in English | MEDLINE | ID: mdl-26538656

ABSTRACT

RE-1 silencing transcription factor (REST), a master negative regulator of neuronal differentiation, controls neurogenesis by preventing the differentiation of neural stem cells. Here we focused on the role of REST in the early steps of differentiation and maturation of adult hippocampal progenitors (AHPs). REST knockdown promoted differentiation and affected the maturation of rat AHPs. Surprisingly, REST knockdown cells enhanced the differentiation of neighboring wild-type AHPs, suggesting that REST may play a non-cell-autonomous role. Gene expression analysis identified Secretogranin II (Scg2) as the major secreted REST target responsible for the non-cell-autonomous phenotype. Loss-of-function of Scg2 inhibited differentiation in vitro, and exogenous SCG2 partially rescued this phenotype. Knockdown of REST in neural progenitors in mice led to precocious maturation into neurons at the expense of mushroom spines in vivo. In summary, we found that, in addition to its cell-autonomous function, REST regulates differentiation and maturation of AHPs non-cell-autonomously via SCG2. SIGNIFICANCE STATEMENT: Our results reveal that REST regulates differentiation and maturation of neural progenitor cells in vitro by orchestrating both cell-intrinsic and non-cell-autonomous factors and that Scg2 is a major secretory target of REST with a differentiation-enhancing activity in a paracrine manner. In vivo, REST depletion causes accelerated differentiation of newborn neurons at the expense of spine defects, suggesting a potential role for REST in the timing of the maturation of granule neurons.


Subject(s)
Cell Differentiation/physiology , Neural Stem Cells/physiology , Neurons/physiology , Repressor Proteins/physiology , Secretogranin II/metabolism , Animals , Cells, Cultured , Female , Hippocampus/cytology , Hippocampus/growth & development , Hippocampus/metabolism , Mice , Mice, Inbred C57BL , Neural Stem Cells/metabolism , Neurogenesis/physiology , Rats, Wistar
11.
J Virol ; 89(23): 11954-64, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26378181

ABSTRACT

UNLABELLED: Viruses utilize host cell machinery for propagation and manage to evade cellular host defense mechanisms in the process. Much remains unknown regarding how the host responds to viral infection. We recently performed global proteomic screens of mammalian reovirus TIL- and T3D-infected and herpesvirus (herpes simplex virus 1 [HSV-1])-infected HEK293 cells. The nonenveloped RNA reoviruses caused an upregulation, whereas the enveloped DNA HSV-1 caused a downregulation, of cellular secretogranin II (SCG2). SCG2, a member of the granin family that functions in hormonal peptide sorting into secretory vesicles, has not been linked to virus infections previously. We confirmed SCG2 upregulation and found SCG2 phosphorylation by 18 h postinfection (hpi) in reovirus-infected cells. We also found a decrease in the amount of reovirus secretion from SCG2 knockdown cells. Similar analyses of cells infected with HSV-1 showed an increase in the amount of secreted virus. Analysis of the stress-activated protein kinase (SAPK)/Jun N-terminal protein kinase (JNK) pathway indicated that each virus activates different pathways leading to activator protein 1 (AP-1) activation, which is the known SCG2 transcription activator. We conclude from these experiments that the negative correlation between SCG2 quantity and virus secretion for both viruses indicates a virus-specific role for SCG2 during infection. IMPORTANCE: Mammalian reoviruses affect the gastrointestinal system or cause respiratory infections in humans. Recent work has shown that all mammalian reovirus strains (most specifically T3D) may be useful oncolytic agents. The ubiquitous herpes simplex viruses cause common sores in mucosal areas of their host and have coevolved with hosts over many years. Both of these virus species are prototypical representatives of their viral families, and investigation of these viruses can lead to further knowledge of how they and the other more pathogenic members of their respective families interact with the host. Here we show that secretogranin II (SCG2), a protein not previously studied in the context of virus infections, alters virus output in a virus-specific manner and that the quantity of SCG2 is inversely related to amounts of infectious-virus secretion. Herpesviruses may target this protein to facilitate enhanced virus release from the host.


Subject(s)
Gene Expression Regulation/physiology , Herpesvirus 1, Human/metabolism , Orthoreovirus, Mammalian/metabolism , Secretogranin II/metabolism , Transcription Factor AP-1/metabolism , Virus Release/physiology , Animals , Chlorocebus aethiops , HEK293 Cells , Humans , Immunoblotting , Mice , Microscopy, Fluorescence , Phosphorylation , Vero Cells
12.
Traffic ; 14(2): 205-18, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23171199

ABSTRACT

Prior to secretion, regulated peptide hormones are selectively sorted to secretory granules (SGs) at the trans-Golgi network (TGN) in endocrine cells. Secretogranin III (SgIII) appears to facilitate SG sorting process by tethering of protein aggregates containing chromogranin A (CgA) and peptide hormones to the cholesterol-rich SG membrane (SGM). Here, we evaluated the role of SgIII in SG sorting in AtT-20 cells transfected with small interfering RNA targeting SgIII. In the SgIII-knockdown cells, the intracellular retention of CgA was greatly impaired, and only a trace amount of CgA was localized within the vacuoles formed in the TGN, confirming the significance of SgIII in both the tethering of CgA-containing aggregates and the establishment of the proper SG morphology. Although the intracellular retention of proopiomelanocortin (POMC) was considerably impaired in SgIII-knockdown cells, residual adrenocorticotropic hormone (ACTH)/POMC was still localized to some few remaining SGs together with another granin protein, secretogranin II (SgII), and was secreted in a regulated manner. Biochemical analyses indicated that SgII bound directly to the SGM in a cholesterol-dependent manner and was able to retain the aggregated form of POMC, revealing a latent redundancy in the SG sorting and retention mechanisms, that ensures the regulated secretion of bioactive peptides.


Subject(s)
Adrenocorticotropic Hormone/metabolism , Exocytosis , Pro-Opiomelanocortin/metabolism , Secretory Vesicles/metabolism , Animals , Cell Line , Cholesterol/metabolism , Chromogranin A/metabolism , Chromogranins/genetics , Chromogranins/metabolism , Intracellular Membranes/metabolism , Mice , PC12 Cells , Protein Binding , Protein Transport , RNA, Small Interfering , Secretogranin II/metabolism , Secretory Vesicles/ultrastructure , Vacuoles/metabolism , trans-Golgi Network/metabolism
13.
Gen Comp Endocrinol ; 222: 158-66, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26141148

ABSTRACT

Secretoneurin (SN) in the preoptic area and pituitary of mammals and fish has a conserved close association with the vasopressin and oxytocin systems, members of a peptide family that are key in the modulation of sexual and social behaviors. Here we show the presence of SN-immunoreactive cells and projections in the brain of the electric fish, Brachyhypopomus gauderio. Secretoneurin colocalized with vasotocin (AVT) and isotocin in cells and fibers of the preoptic area. In the rostral pars distalis of the pituitary, many cells were both SN and prolactin-positive. In the hindbrain, at the level of the command nucleus of the electric behavior (pacemaker nucleus; PN), some of SN-positive fibers colocalized with AVT. We also explored the potential neuromodulatory role of SN on electric behavior, specifically on the rate of the electric organ discharge (EOD) that signals arousal, dominance and subordinate status. Each EOD is triggered by the command discharge of the PN, ultimately responsible for the basal EOD rate. SN modulated diurnal basal EOD rate in freely swimming fish in a context-dependent manner; determined by the initial value of EOD rate. In brainstem slices, SN partially mimicked the in vivo behavioral effects acting on PN firing rate. Taken together, our results suggest that SN may regulate electric behavior, and that its effect on EOD rate may be explained by direct action of SN at the PN level through either neuroendocrine and/or endocrine mechanisms.


Subject(s)
Electric Fish/genetics , Neuropeptides/metabolism , Secretogranin II/metabolism , Vasotocin/metabolism , Animals
14.
Gen Comp Endocrinol ; 220: 124-32, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-25435278

ABSTRACT

It is well established that growth hormone (GH) and granins are co-stored and co-secreted from pituitary somatotrophs. In this work we demonstrate for the first time that GH- and secretoneurin (SN) immunoreactivity (the secretogranin II (SgII) fragment) are similarly present in retinal ganglion cells (RGCs), which is an extrapituitary site of GH expression, and in quail QNR/D cells, which provide an experimental RGC model. The expression of SgII and chromogranin A in the pituitary gland, neuroretina and QNR/D cells was confirmed by RT-PCR analysis. Western blotting also showed that the SN-immunoreactivity in somatotrophs and QNR/D cells was associated with multiple protein bands (24, 35, 48, 72, 78, 93 and 148kDa) of which the 72kDa and 148kDa bands were most abundant. Secretoneurin was constitutively secreted from QNR/D cells as 35kDa and 37kDa proteins and unlike GH, was not increased by exogenous GH-releasing hormone (GHRH). Intracellular analysis by EM showed co-localization of GH and SN in cell bodies and neurites in QNR/D cells. This co-localization was associated with small dark bodies in the neurites. In addition, co-localization of GH and SNAP-25 in the cell surface of QNR/D's plasma membranes suggests GH-release involves specific vesicle-membrane recognition in QNR/D cells. As SN is a marker for secretory granules, GH secretion from RGCs is thus likely to be in secretory granules, as in somatotrophs.


Subject(s)
Human Growth Hormone/metabolism , Neuropeptides/metabolism , Retinal Ganglion Cells/metabolism , Secretogranin II/metabolism , Somatotrophs/metabolism , Animals , Growth Hormone-Releasing Hormone/metabolism , Humans , Pituitary Gland/metabolism , Quail , Retinal Ganglion Cells/cytology
15.
Carcinogenesis ; 35(8): 1798-806, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24710625

ABSTRACT

Stromal cells influence cancer progression. Myofibroblasts are an important stromal cell type, which influence the tumour microenvironment by release of extracellular matrix (ECM) proteins, proteases, cytokines and chemokines. The mechanisms of secretion are poorly understood. Here, we describe the secretion of marker proteins in gastric cancer and control myofibroblasts in response to insulin-like growth factor (IGF) stimulation and, using functional genomic approaches, we identify proteins influencing the secretory response. IGF rapidly increased myofibroblast secretion of an ECM protein, TGFßig-h3. The secretory response was not blocked by inhibition of protein synthesis and was partially mediated by increased intracellular calcium (Ca(2+)). The capacity for evoked secretion was associated with the presence of dense-core secretory vesicles and was lost in cells from patients with advanced gastric cancer. In cells responding to IGF-II, the expression of neuroendocrine marker proteins, including secretogranin-II and proenkephalin, was identified by gene array and LC-MS/MS respectively, and verified experimentally. The expression of proenkephalin was decreased in cancers from patients with advanced disease. Inhibition of secretogranin-II expression decreased the secretory response to IGF, and its over-expression recovered the secretory response consistent with a role in secretory vesicle biogenesis. We conclude that normal and some gastric cancer myofibroblasts have a neuroendocrine-like phenotype characterized by Ca(2+)-dependent regulated secretion, dense-core secretory vesicles and expression of neuroendocrine marker proteins; loss of the phenotype is associated with advanced cancer. A failure to regulate myofibroblast protein secretion may contribute to cancer progression.


Subject(s)
Insulin-Like Growth Factor II/metabolism , Myofibroblasts/pathology , Neurosecretory Systems/pathology , Secretogranin II/metabolism , Stomach Neoplasms/pathology , Blotting, Western , Case-Control Studies , Cells, Cultured , Disease Progression , Exocytosis/physiology , Gastric Mucosa/metabolism , Humans , Immunoenzyme Techniques , Isotope Labeling , Myofibroblasts/metabolism , Neurosecretory Systems/metabolism , Phenotype , RNA, Small Interfering/genetics , Secretogranin II/antagonists & inhibitors , Secretogranin II/genetics , Stomach Neoplasms/metabolism , Tandem Mass Spectrometry
16.
J Neurochem ; 128(2): 233-45, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24111984

ABSTRACT

Cooperative gene regulation by different neurotransmitters likely underlies the long-term forms of associative learning and memory, but this mechanism largely remains to be elucidated. Following cDNA microarray analysis for genes regulated by Ca(2+) or cAMP, we found that the secretogranin II gene (Scg2) was cooperatively activated by glutamate and dopamine in primary cultured mouse hippocampal neurons. The Ca(2+) chelator 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid acetoxymethyl ester (BAPTA-AM) and the mitogen-activated protein kinase (MAPK) kinase (MEK) inhibitor PD98059 prevented Scg2 activation by glutamate or dopamine; thus, the Ca(2+) /MEK pathway is predicted to include a convergence point(s) of glutamatergic and dopaminergic signaling. Unexpectedly, the protein kinase A inhibitor KT5720 enhanced Scg2 activation by dopamine. The protein-synthesis inhibitor cycloheximide also enhanced Scg2 activation, and the proteasome inhibitor ZLLLH diminished the KT5720-mediated augmentation of Scg2 activation. These results are concordant with the notion that dopaminergic input leads to accumulation of a KT5720-sensitive transcriptional repressor, which is short-lived because of rapid degradation by proteasomes. This repression pathway may effectively limit the time window permissive to Scg2 activation by in-phase glutamate and dopamine inputs via the Ca(2+) /MEK pathway. We propose that the regulatory system of Scg2 expression is equipped with machinery that is refined for the signal integration of in-phase synaptic inputs. We proposed hypothetical mechanism for the regulation of the secretogranin II gene as a signal integrator of glutamate and dopamine inputs. Glutamate or dopamine activates the Ca(2+) /MEK/ERK pathway, which thus contributes to the signal integration. Concurrently, activation of the PKA inhibitor KT5720-sensitive pathway by dopamine leads to accumulation of the repressor protein X that is otherwise susceptible to proteasome degradation. This repression system may determine the time window permissive to the cooperative activation by in-phase glutamate and dopamine inputs.


Subject(s)
Dopamine/metabolism , Glutamine/metabolism , Neurotransmitter Agents/metabolism , Secretogranin II/metabolism , Animals , Bucladesine/pharmacology , Calcium/metabolism , Carbazoles/pharmacology , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Hippocampus/cytology , Hippocampus/metabolism , Ionomycin/pharmacology , Mice , Neuroglia/drug effects , Neuroglia/metabolism , Neurons/drug effects , Neurons/metabolism , Oligonucleotide Array Sequence Analysis , Pyrroles/pharmacology , RNA, Messenger/metabolism , Secretogranin II/genetics , Signal Transduction
17.
Cell Physiol Biochem ; 33(2): 446-56, 2014.
Article in English | MEDLINE | ID: mdl-24556756

ABSTRACT

AIMS: Secretoneurin(SN), a neuropeptide, has been considered a reliable marker of allergenic stimulation. However, the relationship between SN and the secretion of airway mucin remains unclear. In this study, we aimed to examine the in vitro relationship between SN and airway mucin over synthesis, as well as the signaling pathways involved. METHODS AND RESULTS: Exogenous SN was added to two human airway epithelial cell lines (16HBE and NCI-H292). Measured by real-time quantitative polymerase chain reaction (qPCR) and enzyme-linked immuno sorbent assay (ELISA) respectively, the intracellular mucin(MUC)5AC mRNA and MUC5AC protein of culture supernates exhibited a time- and dose-dependent increase after stimulation of SN. Based on the evidence of an increased phosphorylation of ERK1/2 induced by exogenous SN, we performed the radioactive binding assay. We failed to find direct binding of SN to either epidermal growth factor receptor (EGFR) or Neuropilin-1(NRP1), the co-receptor of EGFR. But we detected an enhanced binding of EGF to NRP1 in the two airway epithelial cell lines induced by exogenous SN. Either EGF neutralizing antibody or MEK specific inhibitor (PD-98059) could attenuate the over synthesis of MUC5AC induced by exogenous SN, indicating an endogenous EGF dependent mechanism in MUC5AC over synthesis induced by SN. CONCLUSIONS: We conclude that SN induces MUC5AC hypersecretion in a dose- and time-dependent manner; moreover, the MUC5AC over synthesis induced by SN is strongly associated with the enhanced binding of EGF to NRP1 and the activation of EGFR and ERK1/2 subsequently.


Subject(s)
Epidermal Growth Factor/metabolism , Mucin 5AC/metabolism , Mucus/metabolism , Neuropeptides/metabolism , Neuropilin-1/metabolism , Respiratory Mucosa/metabolism , Secretogranin II/metabolism , Cell Line, Transformed , Epidermal Growth Factor/genetics , Humans , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Mucin 5AC/genetics , Neuropeptides/genetics , Neuropilin-1/genetics , Protein Binding , Secretogranin II/genetics
18.
Neurochem Int ; 178: 105770, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38761854

ABSTRACT

Transient global cerebral ischemia (GCI) results in delayed neuronal death, primarily apoptosis, in the hippocampal CA1 subregion, which leads to severe cognitive deficits. While therapeutic hypothermia is an approved treatment for patients following cardiac arrest, it is associated with various adverse effects. Secretoneurin (SN) is an evolutionarily conserved neuropeptide generated in the brain, adrenal medulla and other endocrine tissues. In this study, SN was infused into the rat brain by intracerebroventricular injection 1 day after GCI, and we demonstrated that SN could significantly preserve spatial learning and memory in the Barnes maze tasks examined on days 14-17 after GCI. To further investigate underlying pathways involved, we demonstrated that, on day 5 after GCI, SN could significantly inhibit GCI-induced expression levels of Apoptosis Inducing Factor (AIF) and cleaved-PARP1, as well as neuronal apoptosis and synaptic loss in the hippocampal CA1 region. Additionally, SN could attenuate GCI-induced activation of both caspase-1 and caspase-3, and the levels of pro-inflammatory cytokines IL-1ß and IL-18 in the CA1 region. Mechanically, we observed that treatment with SN effectively inhibited NLRP3 protein elevation and the bindings of NLRP3-ASC and ASC-caspase-1 in hippocampal neurons after GCI. In summary, our data indicate that SN could effectively attenuate NLRP3 inflammasome formation, as well as the activation of caspase-1 and -3, the production of pro-inflammatory cytokines, and ultimately the neuronal apoptotic loss induced by GCI. Potential neuronal pyroptosis, or caspase-1-dependent cell death, could also be involved in ischemic neuronal death, which needs further investigation.


Subject(s)
Apoptosis , Brain Ischemia , Memory , NLR Family, Pyrin Domain-Containing 3 Protein , Neurons , Neuropeptides , Rats, Sprague-Dawley , Animals , Apoptosis/drug effects , Male , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Brain Ischemia/metabolism , Brain Ischemia/drug therapy , Brain Ischemia/pathology , Neuropeptides/administration & dosage , Neuropeptides/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Rats , Memory/drug effects , Secretogranin II/administration & dosage , Secretogranin II/metabolism , Infusions, Intraventricular , Maze Learning/drug effects , Maze Learning/physiology , Signal Transduction/drug effects , Signal Transduction/physiology
19.
Nan Fang Yi Ke Da Xue Xue Bao ; 43(10): 1657-1664, 2023 Oct 20.
Article in Zh | MEDLINE | ID: mdl-37933640

ABSTRACT

OBJECTIVE: To investigate the expression of secretogranin II (SCG2) in colorectal cancer (CRC) tissues and its impact on oxaliplatin resistance of CRC cells. METHODS: We performed immunohistochemistry to detect the expression level of SCG2 on a tissue microarray containing 96 CRC and 84 adjacent tissues and analyzed the association of SCG2 expression with the clinical features of the CRC patients. SCG2 expression was also measured in DLD1 cells treated with oxaliplatin using immunoblotting and RT-qPCR analyses. The effects of SCG2 expression on oxaliplatin sensitivity and cell viability were evaluated in a DLD1 cell model of SCG2 knockout established using CRISPR-cas9 technique, and the expressions of apoptosis-related proteins were detected using Western blotting and RT-qPCR. We further examined SCG2 expression levels in an oxaliplatin-resistant DLD1 cell line and its parental DLD1 cells. RESULTS: SCG2 expression was significantly increased in CRC tissues as compared with the adjacent tissues (1.932±0.816 vs 1), and the tumor tissues in advanced stages showed higher SCG2 expression levels. In DLD1 cells, treatment with oxaliplatin significantly increased SCG2 expression, and SCG2 knockout obviously increased oxaliplatin sensitivity of the cells and enhanced the expressions of apoptosis-related proteins. Compared with the parental cells, oxaliplatin-resistant DLD1 cells showed a significant increase of SCG2 expression by 3.901±0.471 folds. CONCLUSION: SCG2 may serve as a risk gene in CRC, and its high expression increases oxaliplatin resistance of CRC cells.


Subject(s)
Colorectal Neoplasms , Drug Resistance, Neoplasm , Secretogranin II , Humans , Cell Line, Tumor , Colorectal Neoplasms/pathology , Oxaliplatin/pharmacology , Oxaliplatin/therapeutic use , Secretogranin II/metabolism
20.
Article in English | MEDLINE | ID: mdl-37028701

ABSTRACT

Secretoneurin (SN), a conserved peptide derived from secretogranin-2 (scg2), also known as secretogranin II or chromogranin C, plays an important role in regulating gonadotropin in the pituitary, which affects the reproductive system. This study aimed to clarify the mode of action of scg2 in regulating gonad development and maturation and the expression of mating behavior-related genes. Two scg2 cDNAs were cloned from the ovoviviparity teleost black rockfish (Sebastes schlegelii). In situ hybridization detected positive scg2 mRNA signals in the telencephalon and hypothalamus, where sgnrh and kisspeptin neurons were reported to be located and potentially regulated by scg2. In vivo, intracerebral ventricular injections of synthetic black rockfish SNa affected brain cgnrh, sgnrh, kisspeptin1, pituitary lh and fsh and gonad steroidogenesis-related gene expression levels with sex dimorphism. In vitro, a similar effect was found in primary cultured brain and pituitary cells. Thus, SN could contribute to the regulation of gonadal development, as well as reproductive behaviors, including mating and parturition.


Subject(s)
Perciformes , Secretogranin II , Animals , Secretogranin II/genetics , Secretogranin II/metabolism , Ovoviviparity/physiology , Reproduction/physiology , Perciformes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL