Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Apoptosis ; 29(1-2): 121-141, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37848672

RESUMO

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.


Assuntos
NF-kappa B , Neoplasias da Bexiga Urinária , Animais , Humanos , Camundongos , Apoptose , Cromograninas/uso terapêutico , Camundongos Nus , Quinases de Proteína Quinase Ativadas por Mitógeno , NF-kappa B/genética , NF-kappa B/metabolismo , Secretogranina II/genética , Secretogranina II/metabolismo , Secretogranina II/uso terapêutico , Microambiente Tumoral , Neoplasias da Bexiga Urinária/genética , Neoplasias da Bexiga Urinária/tratamento farmacológico , Neoplasias da Bexiga Urinária/metabolismo
2.
Nan Fang Yi Ke Da Xue Xue Bao ; 43(10): 1657-1664, 2023 Oct 20.
Artigo em Chinês | MEDLINE | ID: mdl-37933640

RESUMO

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.


Assuntos
Neoplasias Colorretais , Resistencia a Medicamentos Antineoplásicos , Secretogranina II , Humanos , Linhagem Celular Tumoral , Neoplasias Colorretais/patologia , Oxaliplatina/farmacologia , Oxaliplatina/uso terapêutico , Secretogranina II/metabolismo
3.
Int Immunopharmacol ; 118: 110025, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36933488

RESUMO

The present study investigated whether bone marrow-derived mesenchymal stem cells (BMMSCs) facilitate angiogenesis and improve outcomes of pregnancy with obstetric deep venous thrombosis (DVT) and explored the underlying mechanism. A pregnant DVT rat model was established using a "stenosis" method on the lower segment of the inferior vena cava (IVC). The extent of vascularization in thrombosed IVC was examined by immunohistochemistry. In addition, the effect of BMMSCs on DVT pregnancy outcomes was evaluated. We also characterized the effect of BMMSC-derived conditioned medium (BM-CM) on the impaired human umbilical vein endothelial cells (HUVECs). Thereafter, transcriptome sequencing was employed to identify the differentially expressed genes in thrombosed IVC tissues of DVT and DVT plus BMMSCs (thrice) groups. Lastly, the candidate gene's role in the promotion of angiogenesis was demonstrated in vitro and in vivo. The DVT model was successfully established using IVC stenosis. The injection of three consecutive BMMSC doses into pregnant SD rats with DVT was demonstrated to be the most effective treatment, which significantly reduced the length and weight of the thrombus, induced the highest level of angiogenesis, and ameliorated the embryo absorption rate. In vitro, BM-CM efficiently increased the abilities of impaired endothelial cells to proliferate, migrate, invade, and form vessel-like tubes, while inhibiting their apoptosis. Transcriptome sequencing revealed that BMMSCs induced a prominent upregulation of a variety of pro-angiogenic genes, including secretogranin II (SCG2). When SCG2 expression was knocked down by lentivirus, the BMMSCs' and BM-CM-induced pro-angiogenic effects on pregnant DVT rats and HUVECs were markedly attenuated. In conclusion, the study results suggest that BMMSCs enhance angiogenesis via up-regulation of SCG2, providing an effective alternative regenerative agent and novel target for the therapy of obstetric DVT.


Assuntos
Células-Tronco Mesenquimais , Trombose Venosa , Ratos , Humanos , Animais , Gravidez , Feminino , Regulação para Cima , Trombose Venosa/terapia , Ratos Sprague-Dawley , Secretogranina II/metabolismo , Medula Óssea , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células-Tronco Mesenquimais/metabolismo
4.
J Histochem Cytochem ; 70(5): 335-356, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35400231

RESUMO

Secretogranin II (SgII) and III (SgIII) function within peptide hormone-producing cells and are involved in secretory granule formation. However, their function in active amine-producing cells is not fully understood. In this study, we analyzed the expression profiles of SgII and SgIII in canine adrenal medulla and pheochromocytomas by immunohistochemical staining. In normal adrenal tissues, the intensity of coexpression of these two secretogranins (Sgs) differed from each chromaffin cell, although a complete match was not observed. The coexpression of vesicular monoamine transporter 2 (VMAT2) with SgIII was similar to that with chromogranin A, but there was a subpopulation of VMAT2-expressing cells that were negative or hardly detectable for SgII. These results are the first to indicate that there are distinct expression patterns for SgII and SgIII in adrenal chromaffin cells. Furthermore, the expression of these two Sgs varied in intensity among pheochromocytomas and did not necessarily correlate with clinical plasma catecholamine levels in patients. However, compared with SgIII, the expression of SgII was shown to be strong at the single-cell level in some tumor tissues. These findings provide a fundamental understanding of the expression differences between SgII and SgIII in normal adrenal chromaffin cells and pheochromocytomas.


Assuntos
Neoplasias das Glândulas Suprarrenais , Células Cromafins , Feocromocitoma , Neoplasias das Glândulas Suprarrenais/metabolismo , Neoplasias das Glândulas Suprarrenais/patologia , Neoplasias das Glândulas Suprarrenais/veterinária , Animais , Células Cromafins/metabolismo , Células Cromafins/patologia , Cromograninas/metabolismo , Cães , Humanos , Feocromocitoma/metabolismo , Feocromocitoma/patologia , Feocromocitoma/veterinária , Secretogranina II/metabolismo
5.
Nature ; 590(7844): 115-121, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33299180

RESUMO

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.


Assuntos
Rede Nervosa/citologia , Rede Nervosa/fisiologia , Inibição Neural , Plasticidade Neuronal/fisiologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Animais , Região CA1 Hipocampal/metabolismo , Colecistocinina/metabolismo , Comportamento Exploratório/fisiologia , Feminino , Ritmo Gama , Interneurônios/metabolismo , Masculino , Consolidação da Memória , Camundongos , Parvalbuminas/metabolismo , Células Piramidais/metabolismo , Secretogranina II/genética , Secretogranina II/metabolismo , Navegação Espacial/fisiologia , Ritmo Teta
6.
J Toxicol Sci ; 45(5): 271-280, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32404559

RESUMO

Environmental neurotoxins such as paraquat (PQ), manganese, and 1-1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) are associated with a higher risk of Parkinson's disease (PD). These parkinsonian toxins exert certain common toxicological effects on astroglia; however, their role in the regulatory functions of astroglial secretory proteins remains unclear. In a previous study, we observed that secretogranin II (SCG2) and secretogranin III (SCG3), which are important components of the regulated secretory pathway, were elevated in PQ-activated U118 astroglia. In the current study, we used the parkinsonian toxins dopamine (DA), active metabolite of MPTP (MPP+), MnCl2, and lipopolysaccharide (LPS) as inducers, and studied the potential regulation of SCG2 and SCG3. Our results showed that all the parkinsonian toxins except LPS affected astroglial viability but did not cause apoptosis. Exposure to DA, MPP+, and MnCl2 upregulated glial fibrillary acidic protein (GFAP), a marker for astrocyte activation, and stimulated the levels of several astrocytic-derived factors. Further, DA, MPP+, and MnCl2 exposure impeded astroglial cell cycle progression. Moreover, the expression of SCG3 was elevated, while its exosecretion was inhibited in astroglia activated by parkinsonian toxins. The level of SCG2 remained unchanged. In combination with our previous findings, the results of this study indicate that SCG3 may act as a cofactor in astrocyte activation stimulated by various toxins, and the regulation of SCG3 could be involved in the toxicological mechanism by which parkinsonian toxins affect astroglia.


Assuntos
Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Cromograninas/fisiologia , Intoxicação por MPTP/complicações , Neurotoxinas/toxicidade , Doença de Parkinson Secundária/etiologia , Ciclo Celular/efeitos dos fármacos , Cloretos/efeitos adversos , Cloretos/toxicidade , Cromograninas/metabolismo , Dopamina/administração & dosagem , Dopamina/toxicidade , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Compostos de Manganês/efeitos adversos , Paraquat/toxicidade , Secretogranina II/metabolismo , Secretogranina II/fisiologia , Células Tumorais Cultivadas , Regulação para Cima/efeitos dos fármacos
7.
Proc Natl Acad Sci U S A ; 117(23): 12772-12783, 2020 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-32467166

RESUMO

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.


Assuntos
Fertilidade , Preferência de Acasalamento Animal , Mutação , Secretogranina II/genética , Proteínas de Peixe-Zebra/genética , Animais , Feminino , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Hormônio Luteinizante/metabolismo , Masculino , Neuropeptídeos/metabolismo , Oviposição , Ovulação , Hipófise/metabolismo , Secretogranina II/metabolismo , Peixe-Zebra , Proteínas de Peixe-Zebra/metabolismo
8.
Theranostics ; 10(8): 3779-3792, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32206122

RESUMO

Healing of the chronic diabetic ulceration and large burns remains a clinical challenge. Therapeutic fasting has been shown to improve health. Our study tested whether fasting facilitates diabetic and burn wound healing and explored the underlying mechanism. Methods: The effects of fasting on diabetic and burn wound healing were evaluated by analyzing the rates of wound closure, re-epithelialization, scar formation, collagen deposition, skin cell proliferation and neovascularization using histological analyses and immunostaining. In vitro functional assays were conducted to assess fasting and refeeding on the angiogenic activities of endothelial cells. Transcriptome sequencing was employed to identify the differentially expressed genes in endothelial cells after fasting treatment and the role of the candidate genes in the fasting-induced promotion of angiogenesis was demonstrated. Results: Two times of 24-h fasting in a week after but especially before wound injury efficiently induced faster wound closure, better epidermal and dermal regeneration, less scar formation and higher level of angiogenesis in mice with diabetic or burn wounds. In vitro, fasting alone by serum deprivation did not increase, but rather reduced the abilities of endothelial cell to proliferate, migrate and form vessel-like tubes. However, subsequent refeeding did not merely rescue, but further augmented the angiogenic activities of endothelial cells. Transcriptome sequencing revealed that fasting itself, but not the following refeeding, induced a prominent upregulation of a variety of pro-angiogenic genes, including SMOC1 (SPARC related modular calcium binding 1) and SCG2 (secretogranin II). Immunofluorescent staining confirmed the increase of SMOC1 and SCG2 expression in both diabetic and burn wounds after fasting treatment. When the expression of SMOC1 or SCG2 was down-regulated, the fasting/refeeding-induced pro-angiogenic effects were markedly attenuated. Conclusion: This study suggests that fasting combined with refeeding, but not fasting solely, enhance endothelial angiogenesis through the activation of SMOC1 and SCG2, thus facilitating neovascularization and rapid wound healing.


Assuntos
Diabetes Mellitus Experimental/dietoterapia , Jejum , Neovascularização Fisiológica , Osteonectina/metabolismo , Reepitelização , Secretogranina II/metabolismo , Animais , Queimaduras/terapia , Linhagem Celular , Proliferação de Células , Cicatriz/metabolismo , Células Endoteliais , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pele/metabolismo , Pele/patologia
9.
Circ Arrhythm Electrophysiol ; 12(4): e007045, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30943765

RESUMO

BACKGROUND: Circulating SN (secretoneurin) concentrations are increased in patients with myocardial dysfunction and predict poor outcome. Because SN inhibits CaMKIIδ (Ca2+/calmodulin-dependent protein kinase IIδ) activity, we hypothesized that upregulation of SN in patients protects against cardiomyocyte mechanisms of arrhythmia. METHODS: Circulating levels of SN and other biomarkers were assessed in patients with catecholaminergic polymorphic ventricular tachycardia (CPVT; n=8) and in resuscitated patients after ventricular arrhythmia-induced cardiac arrest (n=155). In vivo effects of SN were investigated in CPVT mice (RyR2 [ryanodine receptor 2]-R2474S) using adeno-associated virus-9-induced overexpression. Interactions between SN and CaMKIIδ were mapped using pull-down experiments, mutagenesis, ELISA, and structural homology modeling. Ex vivo actions were tested in Langendorff hearts and effects on Ca2+ homeostasis examined by fluorescence (fluo-4) and patch-clamp recordings in isolated cardiomyocytes. RESULTS: SN levels were elevated in patients with CPVT and following ventricular arrhythmia-induced cardiac arrest. In contrast to NT-proBNP (N-terminal pro-B-type natriuretic peptide) and hs-TnT (high-sensitivity troponin T), circulating SN levels declined after resuscitation, as the risk of a new arrhythmia waned. Myocardial pro-SN expression was also increased in CPVT mice, and further adeno-associated virus-9-induced overexpression of SN attenuated arrhythmic induction during stress testing with isoproterenol. Mechanistic studies mapped SN binding to the substrate binding site in the catalytic region of CaMKIIδ. Accordingly, SN attenuated isoproterenol induced autophosphorylation of Thr287-CaMKIIδ in Langendorff hearts and inhibited CaMKIIδ-dependent RyR phosphorylation. In line with CaMKIIδ and RyR inhibition, SN treatment decreased Ca2+ spark frequency and dimensions in cardiomyocytes during isoproterenol challenge, and reduced the incidence of Ca2+ waves, delayed afterdepolarizations, and spontaneous action potentials. SN treatment also lowered the incidence of early afterdepolarizations during isoproterenol; an effect paralleled by reduced magnitude of L-type Ca2+ current. CONCLUSIONS: SN production is upregulated in conditions with cardiomyocyte Ca2+ dysregulation and offers compensatory protection against cardiomyocyte mechanisms of arrhythmia, which may underlie its putative use as a biomarker in at-risk patients.


Assuntos
Parada Cardíaca/metabolismo , Neuropeptídeos/metabolismo , Secretogranina II/metabolismo , Taquicardia Ventricular/metabolismo , Animais , Biomarcadores/metabolismo , Cálcio/metabolismo , Sinalização do Cálcio , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Parada Cardíaca/fisiopatologia , Humanos , Camundongos , Miócitos Cardíacos/metabolismo , Peptídeo Natriurético Encefálico/metabolismo , Técnicas de Patch-Clamp , Fragmentos de Peptídeos/metabolismo , Fosforilação , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Taquicardia Ventricular/fisiopatologia , Troponina T/metabolismo , Regulação para Cima
10.
J Mol Cell Biol ; 10(5): 388-401, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-29757409

RESUMO

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.


Assuntos
Artérias/embriologia , Rombencéfalo/irrigação sanguínea , Secretogranina II/metabolismo , Proteínas de Peixe-Zebra/farmacologia , Peixe-Zebra/embriologia , Animais , Animais Geneticamente Modificados , Artérias/citologia , Movimento Celular , Proliferação de Células , Embrião não Mamífero , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Mutação , Neurônios/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores Notch/metabolismo , Rombencéfalo/embriologia , Secretogranina II/genética , Secretogranina II/fisiologia , Nucleases dos Efetores Semelhantes a Ativadores de Transcrição , Fator A de Crescimento do Endotélio Vascular/metabolismo , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/fisiologia
11.
Biochem Biophys Res Commun ; 497(2): 675-682, 2018 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-29454966

RESUMO

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.


Assuntos
Astrócitos/efeitos dos fármacos , Herbicidas/toxicidade , Paraquat/toxicidade , Secretogranina II/genética , Regulação para Cima/efeitos dos fármacos , Astrócitos/citologia , Astrócitos/metabolismo , Astrócitos/patologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos , Mapas de Interação de Proteínas , Secretogranina II/metabolismo
12.
Peptides ; 78: 42-50, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26860475

RESUMO

Secretoneurin (SN) is a conserved peptide derived by proteolytic processing from the middle domain of the ∼600 amino acid precursor secretogranin-II (SgII). Secretoneurin is widely distributed in secretory granules of endocrine cells and neurons and has important roles in reproduction as it stimulates luteinizing hormone release from the pituitary. A potential new role of SN in goldfish feeding is the subject of this study. Firstly, we established that acute (26 h; p<0.0001) and short-term (72 h; p=0.016) fasting increased SgIIa precursor mRNA levels 1.25-fold in the telencephalon, implicating SN in the control of feeding. Secondly, we determined that intracerebroventricular injections of the type A SN (SNa; 0.2 and 1 ng/g BW) increased food intake and locomotor behavior by 60 min. Fish injected with the lower and higher doses of SNa (0.2 and 1 ng/g) respectively exhibited significant 1.77- and 2.58-fold higher food intake (p<0.0001) than the saline-injected control fish. Locomotor behavior was increased by 1.35- and 2.26-fold for 0.2 ng/g SNa (p=0.0001) and 1 ng/g SNa (p<0.0001), respectively. Injection of 1 ng/g SNa increased mRNA levels of hypothalamic neuropeptide Y 1.36-fold (p=0.038) and decreased hypothalamic cocaine-and amphetamine-regulated transcript by 33% (p=0.01) at 2h and 5h post-injection, respectively. These data suggest interactions of SNa with stimulatory and inhibitory pathways of food intake control in fish.


Assuntos
Ingestão de Alimentos/efeitos dos fármacos , Hipotálamo/efeitos dos fármacos , Locomoção/efeitos dos fármacos , Neuropeptídeos/farmacologia , Peptídeos/farmacologia , Secretogranina II/metabolismo , Telencéfalo/efeitos dos fármacos , Animais , Ingestão de Alimentos/fisiologia , Jejum/fisiologia , Feminino , Regulação da Expressão Gênica , Carpa Dourada , Hipotálamo/fisiologia , Injeções Intraventriculares , Locomoção/fisiologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neuropeptídeo Y/genética , Neuropeptídeo Y/metabolismo , Neuropeptídeos/metabolismo , Peptídeos/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Secretogranina II/farmacologia , Transdução de Sinais , Técnicas Estereotáxicas , Telencéfalo/fisiologia
13.
J Virol ; 89(23): 11954-64, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26378181

RESUMO

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.


Assuntos
Regulação da Expressão Gênica/fisiologia , Herpesvirus Humano 1/metabolismo , Orthoreovirus de Mamíferos/metabolismo , Secretogranina II/metabolismo , Fator de Transcrição AP-1/metabolismo , Liberação de Vírus/fisiologia , Animais , Chlorocebus aethiops , Células HEK293 , Humanos , Immunoblotting , Camundongos , Microscopia de Fluorescência , Fosforilação , Células Vero
14.
Gen Comp Endocrinol ; 220: 124-32, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-25435278

RESUMO

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.


Assuntos
Hormônio do Crescimento Humano/metabolismo , Neuropeptídeos/metabolismo , Células Ganglionares da Retina/metabolismo , Secretogranina II/metabolismo , Somatotrofos/metabolismo , Animais , Hormônio Liberador de Hormônio do Crescimento/metabolismo , Humanos , Hipófise/metabolismo , Codorniz , Células Ganglionares da Retina/citologia
15.
Eur J Cancer ; 50(17): 3039-49, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25307750

RESUMO

AIM: In prostate cancer (PCa), neuroendocrine differentiation (NED) is commonly observed in relapsing, hormone therapy-resistant tumours after androgen deprivation. However, the molecular mechanisms involved in the NED of PCa cells remain poorly understood. In this study, we investigated the expression of the neuroendocrine secretory protein secretogranin II (SgII) in PCa, and its potential involvement in the progression of this cancer as a granulogenic factor promoting NED. METHODS: We have examined SgII immunoreactivity in 25 benign prostate hyperplasia and 32 PCa biopsies. In vitro experiments were performed to investigate the involvement of SgII in the neuroendocrine differentiation and the proliferation of PCa cell lines. RESULTS: We showed that immunoreactive SgII intensity correlates with tumour grade in PCa patients. Using the androgen-dependent lymph node cancer prostate cells (LNCaP) cells, we found that NED triggered by androgen deprivation is associated with the induction of SgII expression. In addition, forced expression of SgII in LNCaP cells implemented a regulated secretory pathway by triggering the formation of secretory granule-like structures competent for hormone storage and regulated release. Finally, we found that SgII promotes prostate cancer (CaP) cell proliferation. CONCLUSION: The present data show that SgII is highly expressed in advanced PCa and may contribute to the neuroendocrine differentiation by promoting the formation of secretory granules and the proliferation of PCa cells.


Assuntos
Neoplasias da Próstata/metabolismo , Secretogranina II/metabolismo , Antagonistas de Androgênios/farmacologia , Androgênios/farmacologia , Linhagem Celular Tumoral , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Meios de Cultura/farmacologia , Progressão da Doença , Humanos , Masculino , Neuropeptídeo Y/farmacologia , Antígeno Prostático Específico/metabolismo , Neoplasias da Próstata/patologia , Esteroides/farmacologia
16.
Carcinogenesis ; 35(8): 1798-806, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24710625

RESUMO

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.


Assuntos
Fator de Crescimento Insulin-Like II/metabolismo , Miofibroblastos/patologia , Sistemas Neurossecretores/patologia , Secretogranina II/metabolismo , Neoplasias Gástricas/patologia , Western Blotting , Estudos de Casos e Controles , Células Cultivadas , Progressão da Doença , Exocitose/fisiologia , Mucosa Gástrica/metabolismo , Humanos , Técnicas Imunoenzimáticas , Marcação por Isótopo , Miofibroblastos/metabolismo , Sistemas Neurossecretores/metabolismo , Fenótipo , RNA Interferente Pequeno/genética , Secretogranina II/antagonistas & inibidores , Secretogranina II/genética , Neoplasias Gástricas/metabolismo , Espectrometria de Massas em Tandem
17.
PLoS One ; 9(2): e89491, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24558503

RESUMO

During development and progression of malignant melanoma, an important role has been attributed to alterations of cell-cell adhesions, in particular, to a "cadherin switch" from E- to N-cadherin. We have previously shown that a subtype of melanoma cells express the desmosomal cadherin desmoglein 2 as non-junction-bound cell surface protein in addition to classical cadherins. To study the role of desmoglein 2 in melanoma cells, melanoma lines containing high endogenous amounts of desmoglein 2 were depleted of the protein by RNA interference. Transwell migration and scratch wounding assays showed markedly increased migration upon desmoglein 2 suppression whereas proliferation and viability remained unaltered. In gene expression profiles, desmoglein 2 depletion was associated with overexpression of migration-related genes. Strongest overexpression was found for secretogranin II which has not been reported in melanoma cells before. The bioactive peptide derived from secretogranin II, secretoneurin, is known to exert chemoattractive functions and was demonstrated here to stimulate melanoma cell migration. In summary, we show that desmoglein 2 expression attenuates migration of melanoma cells. The mechanism of desmoglein 2 impaired cell migration is mediated by downregulation of secretogranin II. Loss of desmoglein 2 increases expression of secretogranin II, followed by an enhanced migratory activity of melanoma cells. Our data add a new pathway of regulating melanoma cell migration related to a desmoglein 2-secretogranin II axis.


Assuntos
Movimento Celular/fisiologia , Desmogleína 2/metabolismo , Regulação da Expressão Gênica/fisiologia , Melanoma/fisiopatologia , Bromodesoxiuridina , Linhagem Celular Tumoral , Movimento Celular/genética , Desmogleína 2/deficiência , Impedância Elétrica , Perfilação da Expressão Gênica , Humanos , Immunoblotting , Imuno-Histoquímica , Microscopia de Fluorescência , Interferência de RNA , Radioimunoensaio , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Secretogranina II/metabolismo
18.
Cell Physiol Biochem ; 33(2): 446-56, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24556756

RESUMO

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.


Assuntos
Fator de Crescimento Epidérmico/metabolismo , Mucina-5AC/metabolismo , Muco/metabolismo , Neuropeptídeos/metabolismo , Neuropilina-1/metabolismo , Mucosa Respiratória/metabolismo , Secretogranina II/metabolismo , Linhagem Celular Transformada , Fator de Crescimento Epidérmico/genética , Humanos , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Mucina-5AC/genética , Neuropeptídeos/genética , Neuropilina-1/genética , Ligação Proteica , Secretogranina II/genética
19.
J Neurochem ; 128(2): 233-45, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24111984

RESUMO

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.


Assuntos
Dopamina/metabolismo , Glutamina/metabolismo , Neurotransmissores/metabolismo , Secretogranina II/metabolismo , Animais , Bucladesina/farmacologia , Cálcio/metabolismo , Carbazóis/farmacologia , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Hipocampo/citologia , Hipocampo/metabolismo , Ionomicina/farmacologia , Camundongos , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Pirróis/farmacologia , RNA Mensageiro/metabolismo , Secretogranina II/genética , Transdução de Sinais
20.
PLoS One ; 8(3): e57321, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23526941

RESUMO

Neuropeptide- and hormone-containing secretory granules (SGs) are synthesized at the trans-Golgi network (TGN) as immature secretory granules (ISGs) and complete their maturation in the F-actin-rich cell cortex. This maturation process is characterized by acidification-dependent processing of cargo proteins, condensation of the SG matrix and removal of membrane and proteins not destined to mature secretory granules (MSGs). Here we addressed a potential role of Rab3 isoforms in these maturation steps by expressing their nucleotide-binding deficient mutants in PC12 cells. Our data show that the presence of Rab3D(N135I) decreases the restriction of maturing SGs to the F-actin-rich cell cortex, blocks the removal of the endoprotease furin from SGs and impedes the processing of the luminal SG protein secretogranin II. This strongly suggests that Rab3D is implicated in the subcellular localization and maturation of ISGs.


Assuntos
Células Neuroendócrinas/metabolismo , Vesículas Secretórias/metabolismo , Proteínas rab3 de Ligação ao GTP/metabolismo , Actinas/metabolismo , Animais , Furina/metabolismo , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Células PC12 , Ratos , Secretogranina II/metabolismo , Proteínas rab3 de Ligação ao GTP/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA