Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
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
2.
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
3.
Front Immunol ; 13: 873871, 2022.
Article in English | MEDLINE | ID: mdl-35844556

ABSTRACT

Background: Fluorouracil (FU)-based chemotherapy regimens are indispensable in the comprehensive treatment of colorectal cancer (CRC). However, the heterogeneity of treated individuals and the severe adverse effects of chemotherapy results in limited overall benefit. Methods: Firstly, Weighted gene co-expression network analysis (WGCNA) identified modules tightly associated with chemotherapy response. Then, the in-house cohort and prognostic cohorts from TCGA and GEO were subjected to Cox proportional hazards model and survival analysis to ascertain the predictable function of SCG2 on the prognosis of CRC patients. Finally, we performed In vitro experiments, functional analysis, somatic mutation, and copy number variation research to explore the biological characteristics of SCG2. Results: We identified red and green as the modules most associated with chemotherapy response, in which SCG2 was considered a risky factor with higher expression predicting poorer prognosis. SCG2 expression in the APC non-mutation group was remarkably higher than in the mutation group. The mutation frequencies of amplified genes differed significantly between different SCG2 expression subgroups. Besides, CRC cell lines with SCG2 knockdown have reduced invasive, proliferative, and proliferative capacity. We discovered that the SCG2 high expression subgroup was the immune hot type and considered more suitable for immunotherapy. Conclusion: This study demonstrates the clinical significance and biological characteristics of SCG2, which could serve as a promising biomarker to identify patients who may benefit from chemotherapy and immunotherapy.


Subject(s)
Colorectal Neoplasms , Secretogranin II , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Chemotherapy, Adjuvant , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/therapy , DNA Copy Number Variations , Humans , Immunotherapy , Prognosis , Secretogranin II/genetics , Secretogranin II/immunology
4.
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
5.
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
6.
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
7.
Int J Mol Med ; 45(3): 793-804, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31985029

ABSTRACT

A previous study by our group demonstrated a protective role of the neuropeptide secretoneurin (SN) in DL­isoproterenol hydrochloride (ISO)­induced cardiac hypertrophy in mice. To further characterize the molecular mechanism of SN treatment, an isobaric tags for relative and absolute quantification (iTRAQ)­based quantitative proteomic analysis was applied to identify putative target proteins and molecular pathways. An SN expression vector was injected into the myocardial tissues of mice, and the animals were then subcutaneously injected with ISO (5 mg/kg/day) for 7 days to induce cardiac hypertrophy. The results of echocardiography and hemodynamic measurements indicated that the function of the heart impaired by ISO treatment was significantly ameliorated via SN gene injection. The investigation of heart proteomics was performed by iTRAQ­based liquid chromatography­tandem mass spectrometry analysis. A total of 2,044 quantified proteins and 15 differentially expressed proteins were associated with SN overexpression in mice with cardiac hypertrophy. Functional enrichment analysis demonstrated that these effects were possibly associated with metabolic processes. A protein­protein interaction network analysis was constructed and the data indicated that apolipoprotein C­III (Apoc3) was associated with the positive effect of SN on the induction of cardiac hypertrophy in mice. The present study proposed a potential mechanism of SN action on Apoc3 upregulation that may contribute to the amelioration of cardiac hypertrophy. These findings can aid the clinical application of SN in patients with cardiac hypertrophy.


Subject(s)
Cardiomegaly/chemically induced , Cardiomegaly/therapy , Genetic Therapy/methods , Isoproterenol/toxicity , Neuropeptides/genetics , Neuropeptides/physiology , Proteomics/methods , Secretogranin II/genetics , Secretogranin II/physiology , Animals , Blotting, Western , Cardiomegaly/metabolism , Echocardiography , Hemodynamics/physiology , Hypertrophy, Left Ventricular/chemically induced , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/therapy , Male , Mass Spectrometry , Mice , Mice, Inbred C57BL , Random Allocation
8.
Article in English | MEDLINE | ID: mdl-30818020

ABSTRACT

Secretoneurin (SN) is a novel functional peptide derived from secretogranin II, a protein belonging to the class of chromogranins. Catfish Heteropneustes fossilis is an economically important species of the Asian subcontinent and holds an important place in reproductive physiology study. In the present study, the full-length cDNA encoding secretogranin IIb (SgIIb) was cloned from the brain of catfish H. fossilis. Sequence analysis showed that a 33 amino acid SN peptide (SNb) is present in SgIIb proprotein. The full-length sequence of SgIIb is 2912 bp with open-reading frame 1761 bp long. The 5'UTR is 681 bp long upstream and the 3'UTRis 470 bp long downstream. The ORF encodes 586 amino acid residues comprising signal peptide (24 amino acids) and SNb (33 amino acids), EM66 (66 amino acids). Catfish SgIIb showed 88% nucleotide homology and 90% protein identity with Ictalurus punctatus. Tissue expression showed that it is highly expressed in the brain among all tissues studied. In situ hybridization and quantitative real-time PCR showed significant brain regional distribution with highest transcript abundance in the pituitary. In ovary tissue SgIIb transcripts were localized in the granulosa layer. In the brain, hCG administration stimulated expression highly at 16 h. In ovary, hCG treatment under in vivo and in vitro condition decreased SgIIb expression at all doses. Thus, in the present study in situ localization of SgIIb mRNAs in different regions of brain and pituitary, and ovary suggest its putative role in regulating hypothalamic-pituitary-gonadal (HPG) axis and also suggest that the SgIIb expression pattern in the brain and ovary could be related to reproductive activity and may be involved in the neuroendocrine role of SgIIb in the catfish H. fossilis.


Subject(s)
Brain/metabolism , Catfishes/genetics , Fish Proteins/genetics , Gonadotropins/metabolism , Seasons , Secretogranin II/genetics , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , DNA, Complementary/genetics , Down-Regulation , Female , Male , Ovary/metabolism , Phylogeny , RNA, Messenger/genetics , Sequence Alignment
9.
J Cell Physiol ; 234(4): 3829-3836, 2019 04.
Article in English | MEDLINE | ID: mdl-30132881

ABSTRACT

Colorectal cancer (CRC) ranks as one of the most commonly diagnosed malignancies worldwide. Although mortality rates have been decreasing, the prognosis of CRC patients is still highly dependent on the individual. Therefore, identifying and understanding novel biomarkers for CRC prognosis remains crucial. The gene expression profiles of five-gene expression omnibus (GEO) data sets of CRC were first downloaded. A total of 352 consistent differentially expressed genes (DEGs) were identified for CRC and paired with normal tissues. Functional analysis including gene ontology and Kyoto encyclopedia of genes and genomes pathway enrichment revealed that these DEGs were related to metabolic pathways, tight junctions, and the cell cycle. Ten hub DEGs were identified based on the search tool for the retrieval of interacting genes database and protein-protein interaction networks. By using univariate Cox proportional hazard regression analysis, we found 11 survival-related genes among these DEGs. We finally established a five-gene signature (kinesin family member 15, N-acetyltransferase 2, glutathione peroxidase 3, secretogranin II, and chloride channel accessory 1) with prognostic value in CRC by step multivariate Cox regression analysis. Based on this risk scoring system, patients in the high-risk group had significantly poorer survival results compared with those in the low-risk group (log-rank test, p < 0.0001). Finally, we validated our gene signature scoring system in two independent GEO cohorts (GSE17536 and GSE33113). We found all five of the signature genes to be DEGs in The Cancer Genome Atlas database. In conclusion, our findings suggest that our five DEG-based signature can provide a novel biomarker with useful applications in CRC prognosis.


Subject(s)
Biomarkers, Tumor/genetics , Colorectal Neoplasms/genetics , Gene Expression Profiling , Transcriptome , Arylamine N-Acetyltransferase/genetics , Biomarkers, Tumor/metabolism , Chloride Channels/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/mortality , Colorectal Neoplasms/therapy , Databases, Genetic , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Genetic Predisposition to Disease , Glutathione Peroxidase/genetics , Humans , Kinesins/genetics , Phenotype , Predictive Value of Tests , Prognosis , Protein Interaction Maps , Risk Assessment , Risk Factors , Secretogranin II/genetics , Signal Transduction/genetics
10.
J Mol Cell Biol ; 10(5): 388-401, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29757409

ABSTRACT

Secretoneurin (SN) is a neuropeptide derived from specific proteolytic processing of the precursor secretogranin II (SgII). In zebrafish and other teleosts, there are two paralogs named sgIIa and sgIIb. Our results showed that neurons expressing sgIIb were aligned with central arteries in the hindbrain, demonstrating a close neurovascular association. Both sgIIb-/- and sgIIa-/-/sgIIb-/- mutant embryos were defective in hindbrain central artery development due to impairment of migration and proliferation of central artery cells. Further study revealed that sgIIb is non-cell autonomous and required for central artery development. Hindbrain arterial and venous network identities were not affected in sgIIb-/- mutant embryos, and the mRNA levels of Notch and VEGF pathway-related genes were not altered. However, the activation of MAPK and PI3K/AKT pathways was inhibited in sgIIb-/- mutant embryos. Reactivation of MAPK or PI3K/AKT in endothelial cells could partially rescue the central artery developmental defects in the sgIIb mutants. This study provides the first in vivo evidence that sgIIb plays a critical role in neurovascular modeling of the hindbrain. Targeting the SgII system may, therefore, represent a new avenue for the treatment of vascular defects in the central nervous system.


Subject(s)
Arteries/embryology , Rhombencephalon/blood supply , Secretogranin II/metabolism , Zebrafish Proteins/pharmacology , Zebrafish/embryology , Animals , Animals, Genetically Modified , Arteries/cytology , Cell Movement , Cell Proliferation , Embryo, Nonmammalian , Extracellular Signal-Regulated MAP Kinases/metabolism , Mutation , Neurons/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Notch/metabolism , Rhombencephalon/embryology , Secretogranin II/genetics , Secretogranin II/physiology , Transcription Activator-Like Effector Nucleases , Vascular Endothelial Growth Factor A/metabolism , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/physiology
11.
Endocrinology ; 159(6): 2447-2458, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29648638

ABSTRACT

The luteinizing hormone (LH) surge is essential for ovulation, but the intrafollicular factors induced by LH that mediate ovulatory processes (e.g., angiogenesis) are poorly understood, especially in women. The role of secretogranin II (SCG2) and its cleaved bioactive peptide, secretoneurin (SN), were investigated as potential mediators of ovulation by testing the hypothesis that SCG2/SN is induced in granulosa cells by human chorionic gonadotropin (hCG), via a downstream LH receptor signaling mechanism, and stimulates ovarian angiogenesis. Humans, nonhuman primates, and rodents were treated with hCG in vivo resulting in a significant increase in the messenger RNA and protein levels of SCG2 in granulosa cells collected early during the periovulatory period and just prior to ovulation (humans: 12 to 34 hours; monkeys: 12 to 36 hours; rodents: 4 to 12 hours post-hCG). This induction by hCG was recapitulated in an in vitro culture system utilizing granulosa-lutein cells from in vitro fertilization patients. Using this system, inhibition of downstream LH receptor signaling pathways revealed that the initial induction of SCG2 is regulated, in part, by epidermal growth factor receptor signaling. Further, human ovarian microvascular endothelial cells were treated with SN (1 to 100 ng/mL) and subjected to angiogenesis assays. SN significantly increased endothelial cell migration and new sprout formation, suggesting induction of ovarian angiogenesis. These results establish that SCG2 is increased in granulosa cells across species during the periovulatory period and that SN may mediate ovulatory angiogenesis in the human ovary. These findings provide insight into the regulation of human ovulation and fertility.


Subject(s)
Granulosa Cells/metabolism , Neovascularization, Physiologic/genetics , Ovary/blood supply , Ovulation/genetics , Secretogranin II/genetics , Adult , Animals , Cells, Cultured , Female , Humans , Macaca fascicularis , Mice , Mice, Inbred C57BL , Ovary/metabolism , Rats , Rats, Sprague-Dawley , Secretogranin II/metabolism , Up-Regulation/genetics
12.
Biol Reprod ; 99(3): 565-577, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29635430

ABSTRACT

Gonadotropin-releasing hormone (GNRH) is known as a pivotal upstream regulator of reproduction in vertebrates. However, reproduction is not compromised in the hypophysiotropic Gnrh3 knockout line in zebrafish (gnrh3-/-). In order to determine if Gnrh2, the only other Gnrh isoform in zebrafish brains, is compensating for the loss of Gnrh3, we generated a double Gnrh knockout zebrafish line. Surprisingly, the loss of both Gnrh isoforms resulted in no major impact on reproduction, indicating that a compensatory response, outside of the Gnrh system, was evoked. A plethora of factors acting along the reproductive hypothalamus-pituitary axis were evaluated as possible compensators based on neuroanatomical and differential gene expression studies. In addition, we also examined the involvement of feeding factors in the brain as potential compensators for Gnrh2, which has known anorexigenic effects. We found that the double knockout fish exhibited upregulation of several genes in the brain, specifically gonadotropin-inhibitory hormone (gnih), secretogranin 2 (scg2), tachykinin 3a (tac3a), and pituitary adenylate cyclase-activating peptide 1 (pacap1), and downregulation of agouti-related peptide 1 (agrp1), indicating the compensation occurs outside of Gnrh cells and therefore is a noncell autonomous response to the loss of Gnrh. While the differential expression of gnih and agrp1 in the double knockout line was confined to the periventricular nucleus and hypothalamus, respectively, the upregulation of scg2 corresponded with a broader neuronal redistribution in the lateral hypothalamus and hindbrain. In conclusion, our results demonstrate the existence of a redundant reproductive regulatory system that comes into play when Gnrh2 and Gnrh3 are lost.


Subject(s)
Gene Knockdown Techniques/veterinary , Gonadotropin-Releasing Hormone/genetics , Neuropeptides/administration & dosage , Reproduction/physiology , Zebrafish/genetics , Agouti-Related Protein/genetics , Animals , Brain/metabolism , Down-Regulation , Female , Gonadotropin-Releasing Hormone/deficiency , Gonadotropin-Releasing Hormone/physiology , Hypothalamic Hormones/genetics , Hypothalamus/physiology , Male , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Pituitary Gland/physiology , Secretogranin II/genetics , Tachykinins/genetics , Up-Regulation , Zebrafish/physiology
13.
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
14.
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
15.
Proteomics Clin Appl ; 12(1)2018 01.
Article in English | MEDLINE | ID: mdl-29028155

ABSTRACT

SCOPE: The aim of this study was to develop and evaluate a parallel reaction monitoring mass spectrometry (PRM-MS) assay consisting of a panel of potential protein biomarkers in cerebrospinal fluid (CSF). EXPERIMENTAL DESIGN: Thirteen proteins were selected based on their association with neurodegenerative diseases and involvement in synaptic function, secretory vesicle function, or innate immune system. CSF samples were digested and two to three peptides per protein were quantified using stable isotope-labeled peptide standards. RESULTS: Coefficients of variation were generally below 15%. Clinical evaluation was performed on a cohort of 10 patients with Alzheimer's disease (AD) and 15 healthy subjects. Investigated proteins of the granin family exhibited the largest difference between the patient groups. Secretogranin-2 (p<0.005) and neurosecretory protein VGF (p<0.001) concentrations were lowered in AD. For chromogranin A, two of three peptides had significantly lowered AD concentrations (p<0.01). The concentrations of the synaptic proteins neurexin-1 and neuronal pentraxin-1, as well as neurofascin were also significantly lowered in AD (p<0.05). The other investigated proteins, ß2-microglobulin, cystatin C, amyloid precursor protein, lysozyme C, neurexin-2, neurexin-3, and neurocan core protein, were not significantly altered. CONCLUSION AND CLINICAL RELEVANCE: PRM-MS of protein panels is a valuable tool to evaluate biomarker candidates for neurodegenerative disorders.


Subject(s)
Alzheimer Disease/genetics , C-Reactive Protein/genetics , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules/genetics , Chromogranin A/genetics , Nerve Growth Factors/genetics , Nerve Tissue Proteins/genetics , Secretogranin II/genetics , Aged , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/diagnosis , Alzheimer Disease/pathology , Amino Acid Sequence , Biomarkers/cerebrospinal fluid , C-Reactive Protein/cerebrospinal fluid , Calcium-Binding Proteins , Case-Control Studies , Cell Adhesion Molecules/cerebrospinal fluid , Cell Adhesion Molecules, Neuronal/cerebrospinal fluid , Chromatography, Liquid , Chromogranin A/cerebrospinal fluid , Female , Gene Expression , Humans , Male , Middle Aged , Nerve Growth Factors/cerebrospinal fluid , Nerve Tissue Proteins/cerebrospinal fluid , Neural Cell Adhesion Molecules , Peptide Fragments/cerebrospinal fluid , Pilot Projects , Proteolysis , Proteomics/methods , Secretogranin II/cerebrospinal fluid , Tandem Mass Spectrometry/methods
16.
Sci Rep ; 7(1): 14930, 2017 11 02.
Article in English | MEDLINE | ID: mdl-29097753

ABSTRACT

Radial glial cells (RGCs) are the most abundant macroglia in the teleost brain and have established roles in neurogenesis and neurosteroidogenesis; however, their transcriptome remains uncharacterized, which limits functional understanding of this important cell type. Using cultured goldfish RGCs, RNA sequencing and de novo transcriptome assembly were performed, generating the first reference transcriptome for fish RGCs with 17,620 unique genes identified. These data revealed that RGCs express a diverse repertoire of receptors and signaling molecules, suggesting that RGCs may respond to and synthesize an array of hormones, peptides, cytokines, and growth factors. Building upon neuroanatomical data and studies investigating direct neuronal regulation of RGC physiology, differential gene expression analysis was conducted to identify transcriptional networks that are responsive to the conserved secretogranin II-derived neuropeptide secretoneurin A (SNa). Pathway analysis of the transcriptome indicated that cellular processes related to the central nervous system (e.g., neurogenesis, synaptic plasticity, glial cell development) and immune functions (e.g., immune system activation, leukocyte function, macrophage response) were preferentially modulated by SNa. These data reveal an array of new functions that are proposed to be critical to neuronal-glial interactions through the mediator SNa.


Subject(s)
Fish Proteins/metabolism , Gene Regulatory Networks , Goldfish/physiology , Neuropeptides/metabolism , Secretogranin II/metabolism , Transcriptome , Animals , Cells, Cultured , Ependymoglial Cells/metabolism , Female , Fish Proteins/genetics , Goldfish/genetics , Inflammation/genetics , Neurogenesis , Neuropeptides/genetics , Secretogranin II/genetics , Transcriptional Activation
17.
Elife ; 62017 07 20.
Article in English | MEDLINE | ID: mdl-28726633

ABSTRACT

Sleep loss can severely impair the ability to perform, yet the ability to recover from sleep loss is not well understood. Sleep regulatory processes are assumed to lie exclusively within the brain mainly due to the strong behavioral manifestations of sleep. Whole-body knockout of the circadian clock gene Bmal1 in mice affects several aspects of sleep, however, the cells/tissues responsible are unknown. We found that restoring Bmal1 expression in the brains of Bmal1-knockout mice did not rescue Bmal1-dependent sleep phenotypes. Surprisingly, most sleep-amount, but not sleep-timing, phenotypes could be reproduced or rescued by knocking out or restoring BMAL1 exclusively in skeletal muscle, respectively. We also found that overexpression of skeletal-muscle Bmal1 reduced the recovery response to sleep loss. Together, these findings demonstrate that Bmal1 expression in skeletal muscle is both necessary and sufficient to regulate total sleep amount and reveal that critical components of normal sleep regulation occur in muscle.


Subject(s)
ARNTL Transcription Factors/genetics , Brain/metabolism , Circadian Rhythm/genetics , Gene Expression Regulation , Muscle, Skeletal/metabolism , Sleep/genetics , ARNTL Transcription Factors/deficiency , Animals , Circadian Clocks/genetics , Electrodes, Implanted , Electroencephalography , Electromyography , Male , Mice , Mice, Knockout , Mice, Transgenic , Promoter Regions, Genetic , Secretogranin II/genetics , Secretogranin II/metabolism , Wakefulness/genetics
18.
Endocrinology ; 157(3): 1289-98, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26760117

ABSTRACT

Mechanisms of endocrine secretory granule (SG) formation in thyroid C cells and medullary thyroid cancer (MTC) cells have not been fully elucidated. Here we directly demonstrated that PROX1, a developmental homeobox gene, is transcriptionally involved in SG formation in MTC, which is derived from C cells. Analyses using gene expression databases on web sites revealed that, among thyroid cancer cells, MTC cells specifically and highly express PROX1 as well as several SG-forming molecule genes. Immunohistochemical analyses showed that in vivo MTC and C cells expressed PROX1, although follicular thyroid cancer and papillary thyroid cancer cells, normal follicular cells did not. Knockdown of PROX1 in an MTC cells reduced SGs detected by electron microscopy, and decreased expression of SG-related genes (chromogranin A, chromogranin B, secretogranin II, secretogranin III, synaptophysin, and carboxypeptidase E). Conversely, the introduction of a PROX1 transgene into a papillary thyroid cancer and anaplastic thyroid cancer cells induced the expression of SG-related genes. Reporter assays using the promoter sequence of chromogranin A showed that PROX1 activates the chromogranin A gene in addition to the known regulatory mechanisms, which are mediated via the cAMP response element binding protein and the repressor element 1-silencing transcription factor. Furthermore, chromatin immunoprecipitation-PCR assays demonstrated that PROX1 binds to the transcriptional regulatory element of the chromogranin A gene. In conclusion, PROX1 is an important regulator of endocrine SG formation in MTC cells.


Subject(s)
Adenocarcinoma, Follicular/genetics , Adenoma/genetics , Carcinoma, Neuroendocrine/genetics , Carcinoma/genetics , Gene Expression Regulation, Neoplastic/genetics , Homeodomain Proteins/genetics , Secretory Vesicles/genetics , Thyroid Neoplasms/genetics , Tumor Suppressor Proteins/genetics , Adenocarcinoma, Follicular/metabolism , Adenoma/metabolism , Adult , Aged , Aged, 80 and over , Carboxypeptidase H/genetics , Carcinoma/metabolism , Carcinoma, Neuroendocrine/metabolism , Carcinoma, Papillary , Chromatin Immunoprecipitation , Chromogranin A/genetics , Chromogranin B/genetics , Chromogranins/genetics , Female , Gene Knock-In Techniques , Gene Knockdown Techniques , Humans , Immunohistochemistry , Male , Middle Aged , Secretogranin II/genetics , Synaptophysin/genetics , Thyroid Cancer, Papillary , Thyroid Gland/metabolism , Thyroid Neoplasms/metabolism
19.
Cardiovasc Res ; 105(1): 96-106, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25377726

ABSTRACT

AIMS: Hypercholesterolaemia is a major risk factor for cardiovascular diseases and has been shown to influence angiogenesis in the hind limb ischaemia (HLI) model. The impaired up-regulation of angiogenic factors seems to be one of the underlying mechanisms for reduced vessel formation. Since we found that secretoneurin (SN) is up-regulated in hypoxic skeletal muscle cells and exerts beneficial effects in myocardial and HLI, we hypothesized that SN therapy might improve neovascularization in hypercholesterolaemic Apo E(-/-) (Apo E knockout) mice suffering from an impaired vascular response. METHODS AND RESULTS: For in vitro experiments, endothelial cells (ECs) were incubated with oxidized low-density lipoprotein (oxLDL) to mimic hypercholesterolaemia. EC function was impaired by oxLDL, but SN induced EC proliferation and in vitro tube formation under these conditions. In the HLI model, injection of SN plasmid resulted in a significant better outcome regarding blood flow recovery, amputation rate, and vessel density. In the myocardial infarction (MI) model, the SN group showed improvement in cardiac parameters. Aortic plaque area was not influenced by local SN injection. Interestingly, SN-induced recruitment of angiogenic monocytic cells was abolished under hypercholesterolaemia. CONCLUSIONS: SN gene therapy exerts beneficial effects in cardiovascular animal models in Apo E(-/-) mice without influencing atherosclerosis and might qualify as a promising therapy for cardiovascular disorders.


Subject(s)
Apolipoproteins E/deficiency , Genetic Therapy , Ischemia/therapy , Myocardial Ischemia/therapy , Neuropeptides/genetics , Neuropeptides/therapeutic use , Secretogranin II/genetics , Secretogranin II/therapeutic use , Animals , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/physiopathology , Atherosclerosis/therapy , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/pathology , Endothelial Cells/physiology , Hindlimb/blood supply , Human Umbilical Vein Endothelial Cells , Humans , Hypercholesterolemia/genetics , Hypercholesterolemia/physiopathology , Hypercholesterolemia/therapy , Ischemia/genetics , Ischemia/physiopathology , Lipoproteins, LDL/administration & dosage , Lipoproteins, LDL/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Cardiovascular , Myocardial Infarction/genetics , Myocardial Infarction/physiopathology , Myocardial Infarction/therapy , Myocardial Ischemia/genetics , Myocardial Ischemia/physiopathology , Neovascularization, Physiologic/genetics , Neovascularization, Physiologic/physiology , Neuropeptides/physiology , Secretogranin II/physiology
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...