Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 31
Filtrer
1.
Front Immunol ; 15: 1338585, 2024.
Article de Anglais | MEDLINE | ID: mdl-38994359

RÉSUMÉ

Regular assessment of disease activity in relapsing-remitting multiple sclerosis (RRMS) is required to optimize clinical outcomes. Biomarkers can be a valuable tool for measuring disease activity in multiple sclerosis (MS) if they reflect the pathological processes underlying MS pathogenicity. In this pilot study, we combined multiple biomarkers previously analyzed in RRMS patients into an MS disease activity (MSDA) score to evaluate their ability to predict relapses and treatment response to glatiramer acetate (GA). Response Gene to Complement 32 (RGC-32), FasL, IL-21, SIRT1, phosphorylated SIRT1 (p-SIRT1), and JNK1 p54 levels were used to generate cut-off values for each biomarker. Any value below the cutoff for RGC-32, FasL SIRT1, or p-SIRT1 or above the cutoff for IL-21 or JNK1 p54 was given a +1 value, indicating relapse or lack of response to GA. Any value above the cutoff value for RGC-32, FasL, SIRT1, p-SIRT1 or below that for IL-21 or JNK1 p54 was given a -1 value, indicating clinical stability or response to GA. An MSDA score above +1 indicated a relapse or lack of response to treatment. An MSDA score below -1 indicated clinical stability or response to treatment. Our results showed that the MSDA scores generated using either four or six biomarkers had a higher sensitivity and specificity and significantly correlated with the expanded disability status scale. Although these results suggest that the MSDA test can be useful for monitoring therapeutic response to biologic agents and assessing clinically challenging situations, the present findings need to be confirmed in larger studies.


Sujet(s)
Marqueurs biologiques , Acétate de glatiramère , Sirtuine-1 , Humains , Mâle , Adulte , Femelle , Sirtuine-1/métabolisme , Acétate de glatiramère/usage thérapeutique , Adulte d'âge moyen , Sclérose en plaques récurrente-rémittente/traitement médicamenteux , Sclérose en plaques récurrente-rémittente/diagnostic , Ligand de Fas/métabolisme , Résultat thérapeutique , Projets pilotes , Mitogen-Activated Protein Kinase 8/métabolisme , Interleukines , Sclérose en plaques/traitement médicamenteux , Sclérose en plaques/diagnostic , Indice de gravité de la maladie , Immunosuppresseurs/usage thérapeutique
2.
Aging (Albany NY) ; 162024 May 27.
Article de Anglais | MEDLINE | ID: mdl-38809518

RÉSUMÉ

BACKGROUND: The role and clinical significance of the response gene to complement 32 (RGC32) in various cancers have been documented, yet its implications in clear cell Renal Cell Carcinoma (ccRCC) remain underexplored. METHODS: This study investigated RGC32's diagnostic and prognostic relevance in ccRCC using bioinformatics methods with data from The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO). The impact of RGC32 on ccRCC progression was assessed through nude mouse tumor assays. Immunohistochemistry evaluated RGC32 levels in ccRCC and adjacent normal tissues, while cell proliferation, migration, and invasion capabilities were analyzed using CCK-8, monoclonal proliferation assays, Transwell, and wound healing assays, respectively. Western blotting measured relevant protein expressions. RESULTS: Bioinformatics analysis highlighted RGC32's significant role in ccRCC pathogenesis. Elevated RGC32 expression in ccRCC tissues was linked to disease progression. Functionally, RGC32 was found to enhance the expression of proteins such as p-PI3K, CyclinA1, CyclinD1, p-STAT3, MMP2, MMP3, MMP9, p-SMAD2/3, Snail, Slug, and N-Cadherin via the NF-κB/SHP2/EGFR pathway, while decreasing E-cadherin levels. Moreover, RGC32 facilitated ccRCC cell proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT). CONCLUSION: RGC32 is a pivotal factor in ccRCC development, primarily through the activation of the NF-κB/SHP2/EGFR signaling pathway.

3.
Biosci Rep ; 43(8)2023 08 31.
Article de Anglais | MEDLINE | ID: mdl-37527500

RÉSUMÉ

BACKGROUND: The regular use of gold nanoparticles (Au-NPs) may increase the likelihood of human exposure to these nanoparticles (NPs) and raises concerns about toxicity. AIM: This study investigated the short-term impact of exposure to Au-NPs on inducing cerebellar pathology in rats, and whether the dose or duration of exposure was more important. METHODOLOGY: The study used two concentrations of Au-NPs (25 and 50 particles per million) and 18 rats were randomly assigned to three groups. Assessments of the animals were done via behavioral, gene expression, histological, and immunohistochemistry analyses. RESULTS: Both concentrations of Au-NPs caused cerebellar pathology, as assessed through the investigation test battery. The Au-NPs50 group displayed more injury and decreased mobility compared with the control and the Au-NPs25 group. The Au-NPs25 group showed an increase in supported rearing and significant up-regulation of the Rgc32 gene compared with the control. The Trkb gene was insignificantly up-regulated in both Au-NPs groups compared with the control. CONCLUSION: The study indicates that exposure to Au-NPs can cause cerebellar pathology in rats and that the toxicity is more dependent on dose than the duration of exposure. These findings have significant implications for the safe use of Au-NPs in various applications.


Sujet(s)
Nanoparticules métalliques , Humains , Mâle , Rats , Animaux , Nanoparticules métalliques/toxicité , Nanoparticules métalliques/composition chimique , Or/toxicité , Or/composition chimique
4.
Journal of Clinical Hepatology ; (12): 2396-2405, 2023.
Article de Chinois | WPRIM (Pacifique Occidental) | ID: wpr-998307

RÉSUMÉ

ObjectiveTo investigate the expression and role of response gene to complement 32 (RGC32) in liver regeneration after partial hepatectomy (PH). MethodsA total of 42 male C57BL/6 mice, aged 10 weeks, were randomly divided into control group, postoperative day 1 group (1-d group), postoperative day 2 group (2-d group), postoperative day 4 group (4-d group), postoperative day 6 group (6-d group), postoperative day 8 group (8-d group), and postoperative day 10 group (10-d group), with 6 mice in each group. In the control group, the complete liver of the mice was resected for weighing and photography as the normal control group (sham group); further, the left and middle lobes of the liver were resected for weighing and photography as the surgical control group (0-day group); the sham group and the 0-day group shared the same group of mice. After successful modeling by PH, the mice were sacrificed on days 1, 2, 4, 6, 8, and 10 after surgery, and the liver was collected to measure the change in size. HE staining and oil red O staining were used to evaluate liver histomorphological changes; serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) were measured to evaluate the changes in liver function; immunohistochemical staining was used to measure the expression of proliferating cell nuclear antigen (PCNA) and Ki67 and analyze the change in cell proliferation during liver regeneration; quantitatie real-time PCR and immunohistochemical staining were uused to measure the expression and subcellular distribution of RGC32 during liver regeneration; EdU cell proliferation assay was used to analyze the effect of RGC32 overexpression or knocknout on hepatocyte proliferation in L02 cells. For continuous data, comparison between multiple groups was made by analysis of variance, and further pairwise comparisons were conducted using the LSD-t test. The independent samples t-test was used for comparison of continuous data between two groups. A Pearson correlation analysis was performed. ResultsThe liver gradually enlarged after PH, and the liver/body weight ratio rose to the peak from days 0 to 6, with significant differences between different time points (all P<0.05), while there was no significant change in liver size from days 6 to 10. The number of liver lipid droplets significantly increased after PH surgery and gradually decreased with liver regeneration, with a significant difference between the portal vein region and the central vein region (all P<0.05). Compared with the sham group, the 1d group had significant increases in the serum levels of ALT and AST (all P<0.05), which gradually returned to the levels of the sham group on day 6 and day 2 after surgery, respectively (P>0.05). Immunohistochemical staining showed that there were rapid increases in the numbers of PCNA- and Ki67-positive liver parenchymal cells after PH surgery, with the highest numbers of 86±5 and 89±5, respectively, on day 2, which then gradually decreased; however, there were gradual increases in the numbers of PCNA- and Ki67-positive nonparenchymal cells, with the peak numbers of 34±5 and 25±3, respectively, on day 6, which then gradually decreased. The total expression of RGC32 increased to the highest level on day 2 after PH surgery and then gradually decreased, and the changing trend of RGC32 expression in cytoplasm was consistent with that of total RGC32 expression; however, the expression of RGC32 in nucleus decreased to the lowest level on day 2 after PH surgery and then increased gradually. The correlation analysis showed that the expression of RGC32 in nucleus was negatively correlated with the proliferation of liver parenchymal cells (R2=0.308 3, P=0.016 7), and the expression of RGC32 in cytoplasm was positively correlated with the proliferation of liver parenchymal cells (R2=0.808 6, P<0.000 1). Cell experiments showed that compared with the control group, the EdU-positive rate was reduced by 15.6% after RGC32 overexpression (P<0.01) and was increased by 19.2% after RGC32 knockdown (P<0.01). ConclusionLiver parenchymal cells and nonparenchymal cells show asynchronous proliferation and participate in liver regeneration together. During liver regeneration after hepatectomy, there are differences in the expression of RGC32 between nucleus and cytoplasm, and RGC32 in nucleus may inhibit hepatocyte proliferation.

5.
Front Immunol ; 13: 979414, 2022.
Article de Anglais | MEDLINE | ID: mdl-36172382

RÉSUMÉ

Recent advances in understanding the pathogenesis of multiple sclerosis (MS) have brought into the spotlight the major role played by reactive astrocytes in this condition. Response Gene to Complement (RGC)-32 is a gene induced by complement activation, growth factors, and cytokines, notably transforming growth factor ß, that is involved in the modulation of processes such as angiogenesis, fibrosis, cell migration, and cell differentiation. Studies have uncovered the crucial role that RGC-32 plays in promoting the differentiation of Th17 cells, a subtype of CD4+ T lymphocytes with an important role in MS and its murine model, experimental autoimmune encephalomyelitis. The latest data have also shown that RGC-32 is involved in regulating major transcriptomic changes in astrocytes and in favoring the synthesis and secretion of extracellular matrix components, growth factors, axonal growth molecules, and pro-astrogliogenic molecules. These results suggest that RGC-32 plays a major role in driving reactive astrocytosis and the generation of astrocytes from radial glia precursors. In this review, we summarize recent advances in understanding how RGC-32 regulates the behavior of Th17 cells and astrocytes in neuroinflammation, providing insight into its role as a potential new biomarker and therapeutic target.


Sujet(s)
Protéines du cycle cellulaire , Sclérose en plaques , Protéines du muscle , Protéines de tissu nerveux , Animaux , Marqueurs biologiques , Protéines du cycle cellulaire/génétique , Protéines du système du complément , Cytokines , Humains , Souris , Protéines du muscle/génétique , Protéines de tissu nerveux/génétique , Maladies neuro-inflammatoires , Protéines nucléaires/génétique , Facteur de croissance transformant bêta/métabolisme
6.
Front Oncol ; 12: 875888, 2022.
Article de Anglais | MEDLINE | ID: mdl-35664796

RÉSUMÉ

Background: Bevacizumab is the representative drug in antiangiogenic therapy for lung cancer. However, it induced resistance in some neoplasm. Anlotinib, a novel multi-target tyrosine kinase inhibitor which has an inhibitory action on both angiogenesis and malignancy, is possible to reverse the resistance. Methods: Transwell migration and invasion experiments of bevacizumab with or without anlotinib were conducted to verify the activated/inhibited ability of lung adenocarcinoma cells. We sequenced A549 cells with enhanced migration and invasion abilities after bevacizumab treatment, screened out the differentially expressed gene and further confirmed by western blot and q-PCR assays. We also investigated immunohistochemical staining of tumor tissue in mice and human lung adenocarcinoma. Results: Bevacizumab facilitated migration and invasion of lung adenocarcinoma cells. Differentially expressed gene RGC32 was screened out. Bevacizumab upregulated the expression of RGC32, N-cadherin, and MMP2 through ERK-MAPK and PI3K-AKT pathways. Anlotinib downregulated their expression and reversed the effect of bevacizumab on A549 cells. In vivo experiments confirmed that higher-dose bevacizumab facilitated metastasis in tumor-bearing nude mice and upregulated the expression of RGC32, N-cadherin, and MMP2, whereas anlotinib abrogated its effect. Expression of both RGC32 and N-cadherin positively correlated with lymph node metastasis and stage in lung adenocarcinoma was found. Survival analysis revealed that higher expressions of RGC32 and N-cadherin were associated with poor progression-free survival and overall survival. Conclusions: Bevacizumab may promote invasion and metastasis of lung adenocarcinoma cells by upregulating RGC32 through ERK-MAPK and PI3K-AKT pathways to promote epithelial-mesenchymal transition, whereas anlotinib reverses the effect. RGC32 and N-cadherin are independent prognostic factors in lung adenocarcinoma.

7.
Clin Immunol ; 238: 109020, 2022 05.
Article de Anglais | MEDLINE | ID: mdl-35462050

RÉSUMÉ

Proliferation of endothelial cells (EC) and smooth muscle cells (SMC) is a critical process in atherosclerosis. Here, we investigated the involvement of sublytic C5b-9 effector Response Gene to Complement 32 (RGC-32) in cell cycle activation, phenotypic switch, and production of extracellular matrix (ECM) in SMC. Overexpression of RGC-32 augmented C5b-9-induced cell cycle activation and proliferation of SMC in an ERK1-dependent manner and silencing of RGC-32 inhibited C5b-9-induced cell cycle activation. C5b-9-induced cell cycle activation also required phosphorylation of RGC-32 at threonine 91. We found that ECM components fibronectin and collagens I-V were expressed by SMC in human aortic atherosclerotic tissue. Silencing of RGC-32 in cultured SMC was followed by a significant reduction in TGF-ß-induced expression of SMC differentiation markers myocardin, SM22 and α-SMA, and that of collagens I, IV and V. These data suggest that RGC-32 participates in both sublytic C5b-9-induced cell cycle activation and TGF-ß-induced ECM production.


Sujet(s)
Athérosclérose , Protéines du cycle cellulaire , Complexe d'attaque membranaire du complément , Protéines du muscle , Protéines de tissu nerveux , Protéines du cycle cellulaire/génétique , Protéines du cycle cellulaire/métabolisme , Cellules cultivées , Complexe d'attaque membranaire du complément/métabolisme , Protéines du système du complément , Cellules endothéliales , Humains , Protéines du muscle/génétique , Protéines du muscle/métabolisme , Myocytes du muscle lisse/métabolisme , Protéines de tissu nerveux/génétique , Protéines de tissu nerveux/métabolisme , Facteur de croissance transformant bêta
8.
Front Immunol ; 12: 705308, 2021.
Article de Anglais | MEDLINE | ID: mdl-34394104

RÉSUMÉ

Response Gene to Complement 32 (RGC-32) is an important mediator of the TGF-ß signaling pathway, and an increasing amount of evidence implicates this protein in regulating astrocyte biology. We showed recently that spinal cord astrocytes in mice lacking RGC-32 display an immature phenotype reminiscent of progenitors and radial glia, with an overall elongated morphology, increased proliferative capacity, and increased expression of progenitor markers when compared to their wild-type (WT) counterparts that make them incapable of undergoing reactive changes during the acute phase of experimental autoimmune encephalomyelitis (EAE). Here, in order to decipher the molecular networks underlying RGC-32's ability to regulate astrocytic maturation and reactivity, we performed next-generation sequencing of RNA from WT and RGC-32 knockout (KO) neonatal mouse brain astrocytes, either unstimulated or stimulated with the pleiotropic cytokine TGF-ß. Pathway enrichment analysis showed that RGC-32 is critical for the TGF-ß-induced up-regulation of transcripts encoding proteins involved in brain development and tissue remodeling, such as axonal guidance molecules, transcription factors, extracellular matrix (ECM)-related proteins, and proteoglycans. Our next-generation sequencing of RNA analysis also demonstrated that a lack of RGC-32 results in a significant induction of WD repeat and FYVE domain-containing protein 1 (Wdfy1) and stanniocalcin-1 (Stc1). Immunohistochemical analysis of spinal cords isolated from normal adult mice and mice with EAE at the peak of disease showed that RGC-32 is necessary for the in vivo expression of ephrin receptor type A7 in reactive astrocytes, and that the lack of RGC-32 results in a higher number of homeodomain-only protein homeobox (HOPX)+ and CD133+ radial glia cells. Collectively, these findings suggest that RGC-32 plays a major role in modulating the transcriptomic changes in astrocytes that ultimately lead to molecular programs involved in astrocytic differentiation and reactive changes during neuroinflammation.


Sujet(s)
Astrocytes/métabolisme , Gliose/génétique , Maladies neuro-inflammatoires/génétique , Protéines nucléaires/physiologie , Transcriptome , Animaux , Guidage axonal/génétique , Encéphale/anatomopathologie , Encéphalomyélite auto-immune expérimentale/génétique , Encéphalomyélite auto-immune expérimentale/immunologie , Encéphalomyélite auto-immune expérimentale/anatomopathologie , Femelle , Régulation de l'expression des gènes , Gene Ontology , Réseaux de régulation génique , Gliose/étiologie , Gliose/métabolisme , Souris , Souris de lignée C57BL , Souris knockout , Protéines de tissu nerveux/biosynthèse , Protéines de tissu nerveux/génétique , Cellules souches neurales/métabolisme , Neurogenèse , Maladies neuro-inflammatoires/métabolisme , Protéines nucléaires/déficit , Organismes exempts d'organismes pathogènes spécifiques , Moelle spinale/anatomopathologie
9.
Cell Biochem Funct ; 38(7): 943-954, 2020 Oct.
Article de Anglais | MEDLINE | ID: mdl-32325539

RÉSUMÉ

Growing data have recognized the significance of Response Gene to Complement (RGC)-32 in numerous tumour developments. Notwithstanding, the functional role and underlying mechanism of it in tongue squamous cell carcinoma (TSCC) remain enigmatic. Here, to identify the impact of RGC-32 in TSCC, its expression in multiple TSCC cells was measured and loss-of-function experiments in cell lines were performed to illuminate the function of it induced TSCC progression, via si-RNA knockdown, CCK-8, colony formation, wound-healing, transwell, flow cytometry and western blot assays. To clarify potential mechanism, expressions of hallmarks in epithelial-mesenchymal transition (EMT) process and PI3K/AKT signalling were assessed, and the upstream miR regulator of RGC-32 was predicted and verified by applying bioinformatic approaches and dual-luciferase reporter assay, respectively. Finally, the rescue experiments were applied to better elucidate the effect of miR-26b/RGC-32 axis in TSCC behaviours. As a result, RGC-32 was upregulated in TSCC cells and knocking down of it abrogated cell proliferation, trans-migration and invasion, whilst promoted apoptosis in TSCC, which was regulated through repressing EMT and inactivation of PI3K/AKT signalling. Subsequently, miR-26b was predicted and identified as an upstream regulator of RGC-32, and the pro-tumorigenic effect of RGC-32 was reversed by miR-26b overexpression. Collectively, our results demonstrated that RGC-32 facilitated TSCC progression, which was modulated by activations of PI3K/AKT pathway and EMT process, and reduction of its negative regulator of miR-26b. These findings highlight a novel role of miR-26b/RGC-32 axis in TSCC and underlying mechanism, encouraging a potent usage in TSCC treatment. SIGNIFICANCE OF THE STUDY: We first uncovered that Response Gene to Complement-32 played a significantly pro-tumorigenic role in tongue squamous cell carcinoma (TSCC), which was closely regulated by downregulation of miR-26b and activations of epithelial-mesenchymal transition process and PI3K/AKT signalling. These findings contribute to better understand the molecular mechanism in carcinogenesis of TSCC, and shed some light on promising strategy for TSCC therapeutics.


Sujet(s)
Protéines du cycle cellulaire/métabolisme , Transition épithélio-mésenchymateuse , microARN/métabolisme , Protéines du muscle/métabolisme , Protéines de tissu nerveux/métabolisme , Transduction du signal , Antagomirs/métabolisme , Cadhérines/métabolisme , Carcinome épidermoïde/métabolisme , Carcinome épidermoïde/anatomopathologie , Protéines du cycle cellulaire/antagonistes et inhibiteurs , Protéines du cycle cellulaire/génétique , Lignée cellulaire tumorale , Mouvement cellulaire , Prolifération cellulaire , Humains , microARN/antagonistes et inhibiteurs , microARN/génétique , Protéines du muscle/antagonistes et inhibiteurs , Protéines du muscle/génétique , Protéines de tissu nerveux/antagonistes et inhibiteurs , Protéines de tissu nerveux/génétique , Phosphatidylinositol 3-kinases/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Interférence par ARN , Petit ARN interférent/métabolisme , Tumeurs de la langue/métabolisme , Tumeurs de la langue/anatomopathologie , Régulation positive
10.
Immunol Res ; 67(2-3): 267-279, 2019 06.
Article de Anglais | MEDLINE | ID: mdl-31250246

RÉSUMÉ

The response gene to complement (RGC)-32 acts as a cell cycle regulator and mediator of TGF-ß effects. However, recent studies have revealed other functions for RGC-32 in diverse processes such as cellular migration, differentiation, and fibrosis. In addition to its induction by complement activation and the C5b-9 terminal complement complex, RGC-32 expression is also stimulated by growth factors, hormones, and cytokines. RGC-32 is induced by TGF-ß through Smad3 and RhoA signaling and plays an important role in cell differentiation. In particular, RGC-32 is essential for the differentiation of Th17 cells. RGC-32-/- mice display an attenuated experimental autoimmune encephalomyelitis phenotype that is accompanied by decreased central nervous system inflammation and reductions in IL-17- and GM-CSF-producing CD4+ T cells. Accumulating evidence has drawn attention to the deregulated expression of RGC-32 in human cancers, atherogenesis, metabolic disorders, and autoimmune disease. Furthermore, RGC-32 is a potential therapeutic target in multiple sclerosis and other Th17-mediated autoimmune diseases. A better understanding of the mechanism(s) by which RGC-32 contributes to the pathogenesis of all these diseases will provide new insights into its therapeutic potential.


Sujet(s)
Protéines du cycle cellulaire/génétique , Prédisposition aux maladies , Protéines du muscle/génétique , Protéines de tissu nerveux/génétique , Animaux , Marqueurs biologiques , Protéines du cycle cellulaire/métabolisme , Différenciation cellulaire/génétique , Prolifération cellulaire , Régulation de l'expression des gènes , Humains , Protéines du muscle/métabolisme , Protéines de tissu nerveux/métabolisme , Transduction du signal
11.
Front Immunol ; 10: 1054, 2019.
Article de Anglais | MEDLINE | ID: mdl-31156630

RÉSUMÉ

The complement system represents an effective arsenal of innate immunity as well as an interface between innate and adaptive immunity. Activation of the complement system culminates with the assembly of the C5b-9 terminal complement complex on cell membranes, inducing target cell lysis. Translation of this sequence of events into a malignant setting has traditionally afforded C5b-9 a strict antitumoral role, in synergy with antibody-dependent tumor cytolysis. However, in recent decades, a plethora of evidence has revised this view, highlighting the tumor-promoting properties of C5b-9. Sublytic C5b-9 induces cell cycle progression by activating signal transduction pathways (e.g., Gi protein/ phosphatidylinositol 3-kinase (PI3K)/Akt kinase and Ras/Raf1/ERK1) and modulating the activation of cancer-related transcription factors, while shielding malignant cells from apoptosis. C5b-9 also induces Response Gene to Complement (RGC)-32, a gene that contributes to cell cycle regulation by activating the Akt and CDC2 kinases. RGC-32 is expressed by tumor cells and plays a dual role in cancer, functioning as either a tumor promoter by endorsing malignancy initiation, progression, invasion, metastasis, and angiogenesis, or as a tumor suppressor. In this review, we present recent data describing the versatile, multifaceted roles of C5b-9 and its effector, RGC-32, in cancer.


Sujet(s)
Protéines du cycle cellulaire/génétique , Protéines du cycle cellulaire/métabolisme , Complexe d'attaque membranaire du complément/immunologie , Complexe d'attaque membranaire du complément/métabolisme , Prédisposition aux maladies , Protéines du muscle/génétique , Protéines du muscle/métabolisme , Tumeurs/étiologie , Tumeurs/métabolisme , Protéines de tissu nerveux/génétique , Protéines de tissu nerveux/métabolisme , Apoptose/génétique , Apoptose/immunologie , Prolifération cellulaire , Activation du complément/immunologie , Cytotoxicité immunologique , Régulation de l'expression des gènes tumoraux , Humains , Tumeurs/anatomopathologie , Néovascularisation pathologique/génétique , Néovascularisation pathologique/immunologie , Néovascularisation pathologique/métabolisme , Transduction du signal , Transcription génétique
12.
Exp Mol Pathol ; 108: 97-104, 2019 06.
Article de Anglais | MEDLINE | ID: mdl-30986397

RÉSUMÉ

There is increasing awareness that in addition to the metabolic crisis of diabetic ketoacidosis (DKA) caused by severe insulin deficiency, the immune inflammatory response is likely an active multicomponent participant in both the acute and chronic insults of this medical crisis, with strong evidence of activation for both the cytokine and complement system. Recent studies report that the matrix metalloproteinase enzymes and their inhibitors are systemically activated in young Type 1 diabetes mellitus (T1D) patients during DKA and speculate on their involvement in blood-brain barrier (BBB) disruption. Based on our previous studies, we address the question if matrix metalloproteinase 9 (MMP9) is expressed in the brain in the fatal brain edema (BE) of DKA. Our data show significant expression of MMP9 on the cells present in brain intravascular areas. The presence of MMP9 in intravascular cells and that of MMP+ cells seen passing the BBB indicates a possible role in tight junction protein disruption of the BBB, possibly leading to neurological complications including BE. We have also shown that MMP9 is expressed on neurons in the hippocampal areas of both BE/DKA cases investigated, while expression of tissue inhibitor of metalloproteinases 1 (TIMP1) was reduced in the same areas. We can speculate that intraneuronal MMP9 can be a sign of neurodegeneration. Further studies are necessary to determine the role of MMP9 in the pathogenesis of the neurologic catastrophe of the brain edema of DKA. Inhibition of MMP9 expression might be helpful in preserving neuronal function and BBB integrity during DKA.


Sujet(s)
Acidocétose diabétique/métabolisme , Matrix metalloproteinase 9/métabolisme , Adolescent , Barrière hémato-encéphalique/métabolisme , Encéphale/métabolisme , Oedème cérébral/génétique , Oedème cérébral/métabolisme , Acidocétose diabétique/mortalité , Femelle , Hippocampe/métabolisme , Humains , Matrix metalloproteinases/métabolisme , Neurones/métabolisme , Jonctions serrées/métabolisme , Inhibiteur tissulaire de métalloprotéinase-1/génétique , Inhibiteur tissulaire de métalloprotéinase-1/métabolisme , Transcriptome/génétique
13.
Article de Chinois | WPRIM (Pacifique Occidental) | ID: wpr-823440

RÉSUMÉ

Response gene to complement 32 (RGC-32) as an important response gene to complement was widely expressed in a lot of tissues and organs and participated in many biological processes such as cell proliferation and differentiation,cell cycle regulation,inflammation,immune regulation,and tumor,etc.As a cell cycle regulator,RGC-32 affected the development of numerous diseases by regulating the cell cycle.In recent years,many studies have shown that RGC-32 may be involved in the renal tubular injury and repair,and its role in the renal tubular injury and repair may be related to its regulation of cell cycle especially the G2/M.This article will make a brief review on the progress of the mechanism of RGC-32 regulating the renal injury and repair.

14.
Article de Chinois | WPRIM (Pacifique Occidental) | ID: wpr-801552

RÉSUMÉ

Response gene to complement 32(RGC-32)as an important response gene to complement was widely expressed in a lot of tissues and organs and participated in many biological processes such as cell proliferation and differentiation, cell cycle regulation, inflammation, immune regulation, and tumor, etc.As a cell cycle regulator, RGC-32 affected the development of numerous diseases by regulating the cell cycle.In recent years, many studies have shown that RGC-32 may be involved in the renal tubular injury and repair, and its role in the renal tubular injury and repair may be related to its regulation of cell cycle especially the G2/M.This article will make a brief review on the progress of the mechanism of RGC-32 regulating the renal injury and repair.

15.
Int J Mol Sci ; 19(11)2018 Nov 17.
Article de Anglais | MEDLINE | ID: mdl-30453650

RÉSUMÉ

Diabetic retinopathy (DR) is a severe and recurrent microvascular complication in diabetes. The multifunctional response gene to complement 32 (RGC-32) is involved in the regulation of cell cycle, proliferation, and apoptosis. To investigate the role of RGC-32 in the development of DR, we used human retinal microvascular endothelial cells under high-glucose conditions and type 2 diabetes (T2D) mice (+Leprdb/ + Leprdb, db/db). The results showed that RGC-32 expression increased moderately in human retinal endothelial cells under hyperglycemic conditions. Histopathology and RGC-32 expression showed no significant changes between T2D and control mice retina at 16 and 24 weeks of age. However, RGC-32 expression was significantly decreased in T2D mouse retina compared to the control group at 32 weeks of age, which develop features of the early clinical stages of DR, namely reduced retinal thickness and increased ganglion cell death. Moreover, immunohistochemistry showed that RGC-32 was predominantly expressed in the photoreceptor inner segments of control mice, while the expression was dramatically lowered in the T2D retinas. Furthermore, we found that the level of anti-apoptotic protein Bcl-2 was decreased (approximately 2-fold) with a concomitant increase in cleaved caspase-3 (approximately 3-fold) in T2D retina compared to control. In summary, RGC-32 may lose its expression in T2D retina with features of DR, suggesting that it plays a critical role in DR pathogenesis.


Sujet(s)
Diabète expérimental/métabolisme , Rétinopathie diabétique/métabolisme , Protéines nucléaires/métabolisme , Rétine/métabolisme , Animaux , Apoptose , Humains , Hyperglycémie/métabolisme , Hyperglycémie/anatomopathologie , Traitement d'image par ordinateur , Mâle , Souris , Facteurs temps
16.
Immunol Res ; 66(4): 445-461, 2018 08.
Article de Anglais | MEDLINE | ID: mdl-30006805

RÉSUMÉ

Extracellular matrix (ECM) deposition in active demyelinating multiple sclerosis (MS) lesions may impede axonal regeneration and can modify immune reactions. Response gene to complement (RGC)-32 plays an important role in the mediation of TGF-ß downstream effects, but its role in gliosis has not been investigated. To gain more insight into the role played by RGC-32 in gliosis, we investigated its involvement in TGF-ß-induced ECM expression and the upregulation of the reactive astrocyte markers α-smooth muscle actin (α-SMA) and nestin. In cultured neonatal rat astrocytes, collagens I, IV, and V, fibronectin, α-SMA, and nestin were significantly induced by TGF-ß stimulation, and RGC-32 silencing resulted in a significant reduction in their expression. Using astrocytes isolated from RGC-32 knock-out (KO) mice, we found that the expression of TGF-ß-induced collagens I, IV, and V, fibronectin, and α-SMA was significantly reduced in RGC-32 KO mice when compared with wild-type (WT) mice. SIS3 inhibition of Smad3 phosphorylation was also associated with a significant reduction in RGC-32 nuclear translocation and TGF-ß-induced collagen I expression. In addition, during experimental autoimmune encephalomyelitis (EAE), RGC-32 KO mouse astrocytes displayed an elongated, bipolar phenotype, resembling immature astrocytes and glial progenitors whereas those from WT mice had a reactive, hypertrophied phenotype. Taken together, our data demonstrate that RGC-32 plays an important role in mediating TGF-ß-induced reactive astrogliosis in EAE. Therefore, RGC-32 may represent a new target for therapeutic intervention in MS.


Sujet(s)
Astrocytes/physiologie , Encéphalomyélite auto-immune expérimentale/métabolisme , Gliose/métabolisme , Sclérose en plaques/métabolisme , Protéines nucléaires/métabolisme , Actines/métabolisme , Animaux , Cellules cultivées , Collagène/métabolisme , Modèles animaux de maladie humaine , Matrice extracellulaire/métabolisme , Femelle , Collagènes associés aux fibrilles , Humains , Souris , Souris knockout , Nestine/métabolisme , Protéines nucléaires/génétique , Petit ARN interférent/génétique , Rats , Facteur de croissance transformant bêta/métabolisme
17.
Pancreatology ; 18(5): 572-576, 2018 Jul.
Article de Anglais | MEDLINE | ID: mdl-29886073

RÉSUMÉ

OBJECTIVES: This study was undertaken to investigate the induction of transition of pancreatic cancer to epithelial mesenchyme by RGC-32. METHODS: Primary human pancreatic cancer cell line BXPC-3 was transfected with lentivirus overexpressing the response gene to complement-32 gene (RGC-32) and used to induce tumor in mice. The tumor sizes were measured and the expression of cytokeratin, e-cadherin and vimentin at mRNA using real time PCR and at protein levels by Western blot. RESULTS: Compared with the control, mice inoculated with the cells transfected with empty vector had similar tumor size while those inoculated with the cells transfected with RGC-32 expressing virus had significantly greater tumor size. HE staining showed that tumors were formed in all treatments. Molecular analyses showed that there was no difference in the expression of the cytokeratin, e-cadherin and vimentin genes at mRNA and protein levels between control and empty vector groups. However, mice derived from cells transfected with RGC-32 expressing virus had reduced cytokeratin and e-cadherin expression and increased vimentin expression. CONCLUSIONS: These data suggest that RGC-32 promotes the proliferation of pancreatic cancer and induces the epithelial-mesenchymal transition (EMT). It would be a future direction of research to investigate the regulatory mechanism of signal molecules downstream RGC-32 on EMT-related transcription factors and deliberate the role of RGC-32 in tumorigenicity. As a result, RGC-32 may become a new therapeutic target for cancers.

18.
Am J Transl Res ; 10(3): 847-856, 2018.
Article de Anglais | MEDLINE | ID: mdl-29636874

RÉSUMÉ

This study aimed to investigate the exact function of RGC-32 in kidney diseases and explore the potential mechanism of RGC-32 in regulating cell cycle. RGC-32 knockout (RGC-32-/-) mice were generated from C57BL/6 embryonic stem cells. Differentially expressed proteins in the kidney were investigated with the isobaric tags for relative and absolute quantification (iTRAQ) technique. Gene ontology analyses (GO), Kyoto encyclopedia of genes and genomes (KEGG) pathway mapping analysis and functional network analysis were also performed. The expressions of Smc3, Smad 2-3, DNA-PK were further confirmed by qPCR. Results showed that 4690 proteins were quantified on the basis of 25165 unique peptides. Comparative proteomic analysis revealed 361 differentially expressed proteins in RGC-32-/- mice (knockout/wild ratio >+/- 1.2 and P<0.05). GO and KEGG pathway mapping analyses showed differentially expressed proteins were involved in spliceosome, fluid shear stress and atherosclerosis protein processing in endoplasmic reticulum, pathways in cancer, viral carcinogenesis, epithelial cell signaling in Helicobacter pylori infection, HTLV-I infection, PI3K-Akt signaling pathway, ubiquitin mediated proteolysis, Parkinson's disease, MAPK signaling pathway, carbon metabolism, Alzheimer's disease, NOD-like receptor signaling pathway, tight junction, Proteoglycans in cancer, phagosome, ribosome, mTOR signaling pathway, and AMPK signaling pathway. Differentially expressed proteins Smc3 (0.821), DNA-PK (0.761), Smad 2-3 (0.631) were involved in cell cycle regulation. mRNA expression of Smad2-3, DNA-PK, and Smc3 was consistent with that from iTRAQ. It is concluded that RGC-32 may affect the expression of many proteins (76 up-regulated and 285 down-regulated) in the kidney, and may regulate the expression of Smc3, DNA-PK and Smad 2-3 to affect the cell cycle.

19.
FASEB J ; 32(9): 4836-4847, 2018 09.
Article de Anglais | MEDLINE | ID: mdl-29579398

RÉSUMÉ

Our previous studies have shown that response gene to complement (RGC)-32 deficiency (Rgc32-/-) protects mice from diet-induced obesity and increases thermogenic gene expression in adipose tissues. However, the underlying mechanisms by which RGC-32 regulates thermogenic gene expression remain to be determined. In the present study, RGC-32 expression in white adipose tissue (WAT) was suppressed during cold exposure-induced WAT browning. Rgc32-/- significantly increased thermogenic gene expression in the differentiated stromal vascular fraction (SVF) of inguinal (i)WAT and interscapular brown adipose tissue (BAT). Rgc32-/- and cold exposure regulated a common set of genes in iWAT, as shown by RNA sequencing data. Pathway enrichment analyses showed that Rgc32-/- down-regulated PI3K/Akt signaling-related genes. Akt phosphorylation was also consistently decreased in Rgc32-/- iWAT, which led to an increase in ß3-adrenergic receptor (ß3-AR) expression and subsequent activation of mammalian target of rapamycin complex (mTORC)-1. ß3-AR antagonist SR 59230A and mTORC1 inhibitor rapamycin blocked Rgc32-/--induced thermogenic gene expression in both iWAT and interscapular BAT. These results indicate that RGC-32 suppresses adipose tissue thermogenic gene expression through down-regulation of ß3-AR expression and mTORC1 activity via a PI3K/Akt-dependent mechanism.-Chen, S., Mei, X., Yin, A., Yin, H., Cui, X.-B., Chen, S.-Y. Response gene to complement 32 suppresses adipose tissue thermogenic genes through inhibiting ß3-adrenergic receptor/mTORC1 signaling.


Sujet(s)
Tissu adipeux brun/métabolisme , Complexe-1 cible mécanistique de la rapamycine/métabolisme , Protéines nucléaires/déficit , Récepteurs bêta-3 adrénergiques/métabolisme , Thermogenèse/génétique , Tissu adipeux/métabolisme , Tissu adipeux blanc/métabolisme , Animaux , Différenciation cellulaire/génétique , Protéines du système du complément/métabolisme , Souris knockout , Protéines nucléaires/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Transduction du signal/génétique
20.
FASEB J ; 32(5): 2574-2586, 2018 05.
Article de Anglais | MEDLINE | ID: mdl-29279353

RÉSUMÉ

Preeclampsia (PE), a hypertensive disorder of pregnancy, is a leading cause of maternal and fetal morbidity and mortality. Although the etiology is unknown, PE is thought to be caused by defective implantation and decidualization in pregnancy. Pregnant blood pressure high (BPH)/5 mice spontaneously develop placentopathies and maternal features of human PE. We hypothesized that BPH/5 implantation sites have transcriptomic alterations. Next-generation RNA sequencing of implantation sites at peak decidualization, embryonic day (E)7.5, revealed complement gene up-regulation in BPH/5 vs. controls. In BPH/5, expression of complement factor 3 was increased around the decidual vasculature of E7.5 implantation sites and in the trophoblast giant cell layer of E10.5 placentae. Altered expression of VEGF pathway genes in E5.5 BPH/5 implantation sites preceded complement dysregulation, which correlated with abnormal vasculature and increased placental growth factor mRNA and VEGF164 expression at E7.5. By E10.5, proangiogenic genes were down-regulated, whereas antiangiogenic sFlt-1 was up-regulated in BPH/5 placentae. We found that early local misexpression of VEGF genes and abnormal decidual vasculature preceded sFlt-1 overexpression and increased complement deposition in BPH/5 placentae. Our findings suggest that abnormal decidual angiogenesis precedes complement activation, which in turn contributes to the aberrant trophoblast invasion and poor placentation that underlie PE.-Sones, J. L., Merriam, A. A., Seffens, A., Brown-Grant, D.-A., Butler, S. D., Zhao, A. M., Xu, X., Shawber, C. J., Grenier, J. K., Douglas, N. C. Angiogenic factor imbalance precedes complement deposition in placentae of the BPH/5 model of preeclampsia.


Sujet(s)
Caduques , Régulation de l'expression des gènes , Néovascularisation pathologique/métabolisme , Pré-éclampsie/métabolisme , Trophoblastes/métabolisme , Facteur de croissance endothéliale vasculaire de type A/biosynthèse , Animaux , Protéines du système du complément/génétique , Protéines du système du complément/métabolisme , Caduques/vascularisation , Caduques/métabolisme , Caduques/anatomopathologie , Modèles animaux de maladie humaine , Femelle , Humains , Souris , Néovascularisation pathologique/génétique , Néovascularisation pathologique/anatomopathologie , Pré-éclampsie/génétique , Pré-éclampsie/anatomopathologie , Grossesse , Trophoblastes/anatomopathologie , Facteur de croissance endothéliale vasculaire de type A/génétique , Récepteur-1 au facteur croissance endothéliale vasculaire/biosynthèse , Récepteur-1 au facteur croissance endothéliale vasculaire/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE