Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
J Periodontal Res ; 58(3): 483-492, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36942454

RESUMO

OBJECTIVE: To investigate the effect of Advanced Platelet-Rich Fibrin (A-PRF+), Leukocyte Platelet-Rich Fibrin (L-PRF), and injectable Platelet-Rich Fibrin (i-PRF) on osteogenesis of a human osteoblast-like cell line in vitro. BACKGROUND: Different PRF protocols are used in clinical dentistry in the last years. Recent literature documented the positive impact of PRF derivatives in vivo and in vitro, on different types of cells. However, hardly any literature comparing the new protocols for PRF (the A-PRF+ and i-PRF) with the original protocol of PRF (L-PRF) is present for osteoblast-like cells. MATERIALS AND METHODS: A-PRF+, L-PRF, and i-PRF were prepared from six male donors and pre-cultured with 10 mL culture medium for 6 days. 5 x 103  cells/ml osteoblasts from the osteoblast cell line (U2OS) were seeded and cultured either with conditioned medium derived from the different PRF conditions or with regular culture medium. At five different time points (0, 7, 14, 21, 28 days), the osteogenic capacity of the cells was assessed with Alizarin Red S to visualize mineralization. Also in these cells, the calcium concentration and alkaline phosphatase activity were investigated. Using qPCR, the expression of alkaline phosphatase, osteocalcin, osteonectin, ICAM-1, RUNX-2, and collagen 1a was assessed. RESULTS: In osteoblast-like cells cultured with conditioned medium, the A-PRF+ conditioned medium induced more mineralization and calcium production after 28 days of culturing compared with the control (p < .05). No significant differences were found in the extent of cell proliferation between the different conditions. RUNX-2 and osteonectin mRNA expression in the cells were lower in all PRF-stimulated cultures compared with control at different time points. The i-PRF-conditioned medium induced more ALP activity (p < .05) compared with control and osteoblasts-like cells differentiated more compared with osteoblasts cultured with L-PRF. CONCLUSIONS: The three PRF preparations seem to have the capacity to increase the osteogenic potential of osteoblast-like cells. A-PRF+ seems to have the highest potential for mineralization, while i-PRF seems to have the potential to enhance early cell differentiation.


Assuntos
Osteogênese , Osteonectina , Masculino , Humanos , Osteonectina/metabolismo , Osteonectina/farmacologia , Meios de Cultivo Condicionados/farmacologia , Fosfatase Alcalina/metabolismo , Cálcio , Plaquetas , Proliferação de Células , Diferenciação Celular , Osteoblastos , Células Cultivadas
2.
Peptides ; 163: 170974, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36775021

RESUMO

Bone cells express the glucagon-like peptide 1 receptor (GLP-1R). However, its presence and role in human dental pulp derived stem cells (hDPSCs) remains elusive. Hence, in the current study, we isolated hDPSCs and differentiated them into osteoblasts, where GLP-1R expression was found to be upregulated during osteoblast differentiation. GLP-1 receptor agonist, liraglutide peptide treatment, increased osteoblast differentiation in hDPSCs by increasing calcium deposition, ALP activity, and osteoblast marker genes, Runx2, type 1 col, osteonectin, and osteocalcin. Furthermore, activation of long non-coding RNA (LncRNA) LINC00968 and microRNA-3658 signalling increased Runx2 expression. Specifically, liraglutide increased LncRNA-LINC00968 expression while decreasing miR-3658 expression. LINC00968 targets miR-3658, and miR-3658 targets Runx2. Additionally, in an in-vivo study, zebrafish scale regeneration model, liraglutide promoted calcium deposition, osteoblastic cell count, collagen 1α, osteonectin, osteocalcin, runx2a MASNA isoform expression (transcribed from promoter P1), and Ca/P ratio in scales. Overall, GLP-1R activation promotes osteoblast differentiation via Runx2/LncRNA-LINC00968/miR-3658 signalling in hDPSCs and promotes bone formation in zebrafish scale regeneration.


Assuntos
MicroRNAs , RNA Longo não Codificante , Animais , Humanos , Osteogênese/genética , Receptor do Peptídeo Semelhante ao Glucagon 1/genética , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Osteonectina/metabolismo , Osteonectina/farmacologia , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/farmacologia , Osteocalcina/genética , Liraglutida/farmacologia , Cálcio/metabolismo , Polpa Dentária/metabolismo , Diferenciação Celular/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Células-Tronco , Osteoblastos/metabolismo
3.
J Dermatol Sci ; 109(1): 2-11, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36642579

RESUMO

BACKGROUND: Keloid, an aggressive fibroproliferative disease of the skin, is usually caused by infectious skin diseases, burns, and trauma. OBJECTIVE: This study aimed to assess the effect of SPARC on the keloid pathogenesis. METHODS: In normal skin and keloid scar tissues, changes in SPARC expression were analysed by qRT-PCR, western blotting, and immunohistochemistry. Keloid fibroblasts were isolated from human keloid tissue. GSEA was performed to investigate the signalling pathways related to SPARC. Cell Counting Kit-8, 5-Ethynyl-2'-deoxyuridine, transwell assay, and scratching assays were used to assess fibroblast proliferation and migration. Changes in α-SMA, fibronectin, collagen I, and collagen III levels were examined in fibroblasts by western blotting. RESULTS: SPARC expression was upregulated in keloid scar tissues. In fibroblasts, cell proliferation, migration, collagen production, and extracellular matrix (ECM) synthesis were promoted by SPARC overexpression, whereas SPARC knockdown resulted a converse result. GSEA showed that SPARC regulates the p53 pathway. In keloid scar tissues, there was a negative correlation between SPARC and p53 expression. p53 expression was decreased by SPARC overexpression, whereas SPARC knockdown increased p53 expression. Furthermore, the effects of SPARC on the fibroblast phenotype were reversed by p53 overexpression. CONCLUSIONS: Fibroblast proliferation, migration, and ECM synthesis were promoted by SPARC overexpression, which was achieved by regulating the p53 pathway. Our findings provide new therapeutic targets for keloids.


Assuntos
Queloide , Humanos , Queloide/patologia , Proteína Supressora de Tumor p53/metabolismo , Colágeno/metabolismo , Fibroblastos/metabolismo , Proliferação de Células , Células Cultivadas , Osteonectina/genética , Osteonectina/metabolismo , Osteonectina/farmacologia
4.
Cell Biol Int ; 47(1): 250-259, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36229930

RESUMO

Secreted protein, acidic and rich in cysteine (SPARC) has been characterized as an oncoprotein in esophageal squamous cell carcinoma (ESCC), but its involvement in the pathological development of esophageal adenocarcinoma (ESAD) remains poorly understood. In this study, we aimed to explore the sources of SPARC in the tumor microenvironment (TME) and its functional role in ESAD. Bioinformatic analysis was conducted using data from The Cancer Genome Atlas (TCGA)-esophageal cancer (ESCA) and Genotype-Tissue Expression (GTEx). ESAD tumor cell line OE33 and OE19 cells were used as in vitro cell models. Results showed that SPARC upregulation was associated with unfavorable disease-specific survival (DSS) in ESAD. ESAD tumor cells (OE33 and OE19) had no detectable SPARC protein expression. In contrast, IHC staining in ESAD tumor tissues suggested that peritumoral stromal cells (tumor-associated fibroblasts and macrophages) were the dominant SPARC source in TME. Exogenous SPARC induced partial epithelial-to-mesenchymal transition of ESAD cells, reflected by reduced CDH1 and elevated ZEB1/VIM expression at both mRNA and protein levels. Besides, exogenous SPARC enhanced tumor cell invasion. When TGFBR2 expression was inhibited, the activation of TGF-ß signaling induced by exogenous SPARC was impaired. However, the activating effects were rescued by overexpressing mutant TGFBR2 resistant to the shRNA sequence. Copresence of exogenous SPARC and TGF-ß1 induced higher expression of mesenchymal markers and enhanced the invading capability of ESAD cells than TGF-ß1 alone. In conclusion, this study suggests a potential cross-talk between ESAD tumor stromal cells and cancer cells via a SPARC-TGF-ß1 paracrine network.


Assuntos
Adenocarcinoma , Transição Epitelial-Mesenquimal , Neoplasias Esofágicas , Carcinoma de Células Escamosas do Esôfago , Osteonectina , Fator de Crescimento Transformador beta1 , Microambiente Tumoral , Humanos , Adenocarcinoma/patologia , Linhagem Celular Tumoral , Neoplasias Esofágicas/patologia , Osteonectina/genética , Osteonectina/metabolismo , Osteonectina/farmacologia , Receptor do Fator de Crescimento Transformador beta Tipo II/metabolismo , Fator de Crescimento Transformador beta1/metabolismo
5.
BMC Genomics ; 23(1): 682, 2022 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-36192692

RESUMO

BACKGROUND: The brown planthopper (Nilaparvata lugens Stål)is a notorious rice pest in many areas of Asia. Study on the molecular mechanisms underlying its development and reproduction will provide scientific basis for effective control. SPARC (Secreted Protein, Acidic and Rich in Cysteine) is one of structural component of the extracellular matrix, which influences a diverse array of biological functions. In this study, the gene for SPARC was identified and functionally analysed from N.lugens. RESULTS: The result showed that the NlSPARC mRNA was highly expressed in fat body, hemolymph and early embryo. The mortality increased significantly when NlSPARC was downregulated after RNA interference (RNAi) in 3 ~ 4th instar nymphs. Downregulation of NlSPARC in adults significantly reduced the number of eggs and offspring, as well as the transcription level of NlSPARC in newly hatched nymphs and survival rate in progeny. The observation with microanatomy on individuals after NlSPARC RNAi showed smaller and less abundant fat body than that in control. No obvious morphological abnormalities in the nymphal development and no differences in development of internal reproductive organ were observed when compared with control. CONCLUSION: NlSPARC is required for oviposition and nymphal development mainly through regulating the tissue of fat body in N.lugens. NlSPARC could be a new candidate target for controlling the rapid propagation of N.lugens population. Our results also demonstrated that the effect of NlSPARC RNAi can transfer to the next generation in N.lugens.


Assuntos
Hemípteros , Oviposição , Animais , Cisteína/metabolismo , Feminino , Hemípteros/fisiologia , Ninfa/genética , Ninfa/metabolismo , Osteonectina/genética , Osteonectina/metabolismo , Osteonectina/farmacologia , Oviposição/genética , Interferência de RNA , RNA Mensageiro/metabolismo
6.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 30(5): 1508-1514, 2022 Oct.
Artigo em Chinês | MEDLINE | ID: mdl-36208257

RESUMO

OBJECTIVE: To investigate the effect of SPARC gene overexpression on the chemotherapeutic sensitivity of AML-MDS cell line SKM-1 to Ara-C and to further explore its mechanism. METHODS: Subjects were divided into 6 groups: SKM-1 cells (Control), Negative control (LV-NC), SPARC overexpression (LV-SPARC), SKM-1 cells+30 ng/ml Ara-C (30 ng/ml Ara-C), LV-NC+30 ng/ml Ara-C and LV-SPARC+30 ng/ml Ara-C. Cell activity was detected by CCK-8 assay, cell cycle distribution and apoptosis were detected by flow cytometry, mRNA expression levels of SPARC, CPBP and MLKL were detected by RT-qPCR, and the expression levels of related protein were detected by Western blot. RESULTS: After co-treatment with SPARC overexpression and Ara-C, the cell viability decreased and apoptosis increased significantly, with obvious up-regulation of Bax and down-regulation of BCL-2 (P<0.05). Compared with the control group, the cell cycle of LV-SPARC+30 ng/ml Ara-C group was significantly arrested in S phase with obvious down-regulation of CDK2 and up-regulation of p27KIP1 (P<0.05). Compared with LV-SPARC group and 30 ng/ml Ara-C group, the mRNA and protein expression levels of CPBP and MLKL (p-MLKL) were significantly elevated in LV-SPARC+30 ng/ml Ara-C group (P<0.05). In addition, after co-treatment with SPARC overexpression and Ara-C, the protein expression level of p-AKT decreased and the protein expression level of p53 increased (P<0.05). CONCLUSION: SPARC overexpression enhanced the sensitivity of SKM-1 cells to Ara-C and promoted cell cycle arrest and apoptosis, the mechanism of which may be related to the regulation of CPBP/MLKL pathway.


Assuntos
Citarabina , Proteína Supressora de Tumor p53 , Apoptose , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Fator 6 Semelhante a Kruppel/metabolismo , Osteonectina/farmacologia , Proteínas Quinases/metabolismo , Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA Mensageiro , Proteína X Associada a bcl-2/metabolismo , Proteína X Associada a bcl-2/farmacologia
7.
J Adv Res ; 40: 95-107, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36100336

RESUMO

INTRODUCTION: Basic fibroblast growth factor (bFGF) plays a critical role in odontoblast differentiation and dentin matrix deposition, thereby aiding pulpo-dentin repair and regeneration. OBJECTIVES: The purpose of this study was to clarify the effects of bFGF on plasminogen activation factors, TIMP-1), ALP; and SPARC (osteonectin) expression/production of stem cells from apical papilla (SCAP) in vitro; and the involvement of MEK/ERK, p38, Akt, and TAK1 signaling. METHODS: SCAP were exposed to bFGF with/without pretreatment and co-incubation with various signal transduction inhibitors (U0126, SB203580, LY294002, and 5Z-7-oxozeaenol). The expression of FGF receptors (FGFRs), PAI-1, uPA, p-ERK, p-TAK1, and p-p38 was analyzed via immunofluorescent staining. The gene expression and protein secretion of SCAP were determined via real-time PCR and ELISA. ALP activity was evaluated via ALP staining. RESULTS: SCAP expressed FGFR1, 2, 3, and 4. bFGF stimulated the PAI-1, uPA, uPAR, and TIMP-1 mRNA expression (p < 0.05). bFGF induced PAI-1, uPA, and soluble uPAR production (p < 0.05) but suppressed the ALP activity and SPARC production (p < 0.05) of SCAP. bFGF stimulated ERK, TAK1, and p38 phosphorylation of SCAP. U0126 (a MEK/ERK inhibitor) and 5Z-7-oxozeaenol (a TAK1 inhibitor) attenuated the bFGF-induced PAI-1, uPA, uPAR, and TIMP-1 expression and production of SCAP, but SB203580 (a p38 inhibitor) did not. LY294002, SB203580, and 5Z-7oxozeaenol could not reverse the inhibition of ALP activity caused by bFGF. Interestingly, U0126 and 5Z-7-oxozeaenol prevented the bFGF-induced decline of SPARC production (p < 0.05). CONCLUSION: bFGF may regulate fibrinolysis and matrix turnover via modulation of PAI-1, uPA, uPAR, and TIMP-1, but bFGF inhibited the differentiation (ALP, SPARC) of SCAP. These events are mainly regulated by MEK/ERK, p38, and TAK1. Combined use of bFGF and SCAP may facilitate pulpal/root repair and regeneration via regulation of the plasminogen activation system, migration, matrix turnover, and differentiation of SCAP.


Assuntos
Fosfatase Alcalina , Fator 2 de Crescimento de Fibroblastos , Fosfatase Alcalina/metabolismo , Fosfatase Alcalina/farmacologia , Butadienos , Fator 2 de Crescimento de Fibroblastos/metabolismo , Fator 2 de Crescimento de Fibroblastos/farmacologia , Lactonas , Sistema de Sinalização das MAP Quinases , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/farmacologia , Nitrilas , Osteonectina/metabolismo , Osteonectina/farmacologia , Plasminogênio/metabolismo , Plasminogênio/farmacologia , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Inibidor 1 de Ativador de Plasminogênio/farmacologia , Resorcinóis , Transdução de Sinais , Células-Tronco/metabolismo , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Inibidor Tecidual de Metaloproteinase-1/farmacologia , Zearalenona/administração & dosagem
8.
Int J Biol Sci ; 18(3): 911-922, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35173526

RESUMO

Pancreatic cancer is a dismal malignancy with poor prognosis. In spite of progress in surgical technology, chemotherapy is still the cornerstone in the multi-disciplinary treatment. Albumin-bound paclitaxel is a first-line treatment for PDAC patients. Yet the response rate of the drug is far from satisfying. SOX8 is a member of the sex determining region Y-boxes family, which is potentially related to the chemoresistance of tumor. Patient with high expression of SOX8 were insensitive to albumin-bound paclitaxel. SOX8 reduced apoptosis and G2/M cell cycle arrest caused by albumin-bound paclitaxel. SOX8 transcriptionally regulated EZH2, which reduced expression of SPARC by promoting the methylation of SPARC, thereby reducing the transport of albumin-bound paclitaxel in pancreatic cancer cells. EZH2 inhibitor, UNC1999, can reverse the effect of SOX8 on chemo-resistance of albumin-bound paclitaxel. Collectively, our data revealed SOX8/EZH2/SPARC signaling induced primary chemo-resistance of albumin-bound paclitaxel in pancreatic ductal adenocarcinoma.


Assuntos
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Paclitaxel Ligado a Albumina/metabolismo , Paclitaxel Ligado a Albumina/farmacologia , Paclitaxel Ligado a Albumina/uso terapêutico , Apoptose/genética , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patologia , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Humanos , Osteonectina/genética , Osteonectina/farmacologia , Osteonectina/uso terapêutico , Paclitaxel/farmacologia , Paclitaxel/uso terapêutico , Neoplasias Pancreáticas/metabolismo , Fatores de Transcrição SOXE/metabolismo , Neoplasias Pancreáticas
9.
Elife ; 102021 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-33904394

RESUMO

Cortical interneurons establish inhibitory microcircuits throughout the neocortex and their dysfunction has been implicated in epilepsy and neuropsychiatric diseases. Developmentally, interneurons migrate from a distal progenitor domain in order to populate the neocortex - a process that occurs at a slower rate in humans than in mice. In this study, we sought to identify factors that regulate the rate of interneuron maturation across the two species. Using embryonic mouse development as a model system, we found that the process of initiating interneuron migration is regulated by blood vessels of the medial ganglionic eminence (MGE), an interneuron progenitor domain. We identified two endothelial cell-derived paracrine factors, SPARC and SerpinE1, that enhance interneuron migration in mouse MGE explants and organotypic cultures. Moreover, pre-treatment of human stem cell-derived interneurons (hSC-interneurons) with SPARC and SerpinE1 prior to transplantation into neonatal mouse cortex enhanced their migration and morphological elaboration in the host cortex. Further, SPARC and SerpinE1-treated hSC-interneurons also exhibited more mature electrophysiological characteristics compared to controls. Overall, our studies suggest a critical role for CNS vasculature in regulating interneuron developmental maturation in both mice and humans.


Assuntos
Movimento Celular/efeitos dos fármacos , Córtex Cerebral/metabolismo , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Interneurônios/efeitos dos fármacos , Eminência Mediana/irrigação sanguínea , Células-Tronco Neurais/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Osteonectina/farmacologia , Inibidor 1 de Ativador de Plasminogênio/farmacologia , Potenciais de Ação , Animais , Córtex Cerebral/embriologia , Córtex Cerebral/cirurgia , Células Endoteliais/metabolismo , Células HEK293 , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/transplante , Interneurônios/metabolismo , Interneurônios/transplante , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Eminência Mediana/embriologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Knockout , Neovascularização Fisiológica , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/transplante , Osteonectina/metabolismo , Comunicação Parácrina , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Transdução de Sinais
10.
Stem Cells Dev ; 30(6): 301-308, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33487117

RESUMO

Secreted protein acidic and rich in cysteine (SPARC) shows a specific colocalization with limbal epithelial stem cells (LESCs) in vivo; however, the inherent relationship between SPARC and LESCs is still unclear. This study investigated the effects of SPARC on the maintenance of LESC stemness and corneal wound healing. To test the influence of different concentration of exogenous SPARC on the proliferation of LESCs, cell counting kit-8 assay and 5-ethynyl-2'-deoxyuridine staining were performed and the results indicated that 1 µg/mL SPARC was the optimum concentration for enhanced LESC proliferation. Compared with a control group, SPARC-treated group showed a higher expression of LESC-positive markers p63α, ABCG-2, and Bmi-1, and a lower level of differentiation marker cytokeratin-3 (CK3), thereby suggesting that SPARC could maintain LESC characteristic phenotype and suppress spontaneous epithelial differentiation in vitro. In vivo, exogenous SPARC accelerated the wound-healing process by both the enhancement of LESC proliferation and promoting the migration of the proliferating cells. However, the intact epithelium impaired this function of SPARC by contact inhibition.


Assuntos
Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Epitélio Corneano/efeitos dos fármacos , Osteonectina/farmacologia , Células-Tronco/efeitos dos fármacos , Cicatrização/efeitos dos fármacos , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Western Blotting , Células Cultivadas , Células Epiteliais/citologia , Epitélio Corneano/fisiopatologia , Humanos , Limbo da Córnea/citologia , Camundongos , Camundongos Endogâmicos C57BL , Células NIH 3T3 , Coelhos , Células-Tronco/citologia , Células-Tronco/metabolismo
11.
Mol Cell Endocrinol ; 506: 110757, 2020 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-32057945

RESUMO

SPARC, also known as osteonectin, is well known for its physiological roles in bone formation and tissue remodeling, as well as in cancer pathology; however, evidence regarding its function in adipocytes is lacking. The present study explored the physiological role of SPARC in cultured 3T3-L1 white and HIB1B brown adipocytes of murine cell lines. Treatment of recombinant SPARC upregulated the fat browning marker proteins and genes in white adipocytes and activated brown adipocytes. Conversely, knockdown of Sparc markedly reduced these genes and proteins in both cell lines. In addition, recombinant SPARC inhibited expression of adipogenic and lipogenic proteins but elevated lipolytic and fatty acid oxidation proteins. Furthermore, in silico analysis revealed that SPARC directly interacted and regulated VEGF in adipocytes. In conclusion, SPARC acts as a regulatory protein in both white and brown adipocytes by controlling thermogenesis and is thus regarded as a possible therapeutic target for treatment of obesity.


Assuntos
Adipócitos Marrons/fisiologia , Adipócitos Brancos/fisiologia , Osteonectina/fisiologia , Termogênese/genética , Células 3T3-L1 , Adipócitos Marrons/efeitos dos fármacos , Adipócitos Brancos/efeitos dos fármacos , Adipogenia/efeitos dos fármacos , Adipogenia/genética , Animais , Transdiferenciação Celular/efeitos dos fármacos , Células Cultivadas , Metabolismo dos Lipídeos/efeitos dos fármacos , Metabolismo dos Lipídeos/genética , Lipogênese/efeitos dos fármacos , Lipogênese/genética , Camundongos , Osteonectina/farmacologia , Proteínas Recombinantes/farmacologia , Termogênese/efeitos dos fármacos
12.
Neurosci Lett ; 712: 134485, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31499136

RESUMO

Secreted protein acidic and rich in cysteine (SPARC) was widely expressed in VSMCs of human IAs and could reduce the capability of self-repair. This indicates that SPARC may play a role in the promotion of IAs formation and progression, but the mechanism remains unclear. In this study, we further investigated whether SPARC could induce phenotypic modulation of Human Brain Vascular Smooth Muscle Cells (HBVSMCs) and sought to elucidate the role of SPARC-mediated autophagy involved in it. The results demonstrated that SPARC inhibited the expression of contractile genes in HBVSMCs and induced a synthetic phenotype. More importantly, SPARC significantly up-regulated multiple proteins including autophagy marker microtubule-associated protein light chain 3-II (LC3-II), Beclin-1, and autophagy-related gene 5(ATG5). Furthermore, SPARC could promote p62 degradation. The autophagy inhibitor 3- methyladenine (3-MA) significantly blocked SPARC-induced phenotypic modulation of HBVSMCs. We further sought to elucidate the molecular mechanism involved in SPARC-induced autophagy, and found that SPARC could activate the AMPK/mTOR signaling pathway in HBVSMCs. AMPK could be pharmacologically inhibited by Compound C (CC), which significantly decreased the phosphorylation of AMPK into p-AMPK, increased the phosphorylation of mTOR into p-mTOR, and decreased LC3-II, Beclin-1 and ATG5 levels. This suggested that activated AMPK/ mTOR signaling is related to SPARC-mediated autophagy. These results indicated that SPARC plays a role in the phenotypic modulation of HBVSMCs through autophagy activation by AMPK/mTOR signaling pathway.


Assuntos
Adenilato Quinase/metabolismo , Autofagia/fisiologia , Encéfalo/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Osteonectina/farmacologia , Serina-Treonina Quinases TOR/metabolismo , Autofagia/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Células Cultivadas , Humanos , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Fenótipo , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
13.
Int J Mol Sci ; 18(7)2017 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-28718842

RESUMO

Secreted protein acidic and rich in cysteine (SPARC) is a secreted protein which is involved in various biological processes. SPARC expression is associated with tumor metastasis and poor prognosis in several types of cancer. However, the SPARC-induced signaling pathway was not fully understood in head and neck cancer. In this study, our results showed that SPARC treatment promoted cell proliferation and migration in head and neck cancer cell lines FaDu and Detroit 562. In addition, SPARC induced expression of epithelial mesenchymal transition (EMT) regulators, including Slug, Snail, and Twist in Detroit 562. The results of phospho-kinase array analysis showed that SPARC treatment increased phosphorylation of some molecules including protein kinase B (PKB/AKT), ribosomal S6 kinase (RSK), and extracellular signal-regulated kinases (ERK). The expression of SPARC-induced EMT regulator Slug was suppressed by AKT inhibitor, but not ERK and RSK inhibitors. The SPARC expression in grade IV tumor samples is higher when compared to that in grade I-III tumor samples. Our results suggest that SPARC treatment enhances the EMT signaling pathway via activation of AKT, and exogenous SPARC and tumor expressing SPARC might be associated with tumor progression in head and neck cancers.


Assuntos
Movimento Celular/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Neoplasias de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/patologia , Osteonectina/genética , Osteonectina/farmacologia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Transição Epitelial-Mesenquimal/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Modelos Biológicos , Gradação de Tumores , Osteonectina/metabolismo , Fenótipo , Transdução de Sinais/efeitos dos fármacos
14.
J Neuroinflammation ; 13(1): 225, 2016 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-27581191

RESUMO

BACKGROUND: SPARC (secreted protein acidic and rich in cysteine) is a nonstructural, cell-matrix modulating protein involved in angiogenesis and endothelial barrier function, yet its potential role in cerebrovascular development, inflammation, and repair in the central nervous system (CNS) remains undetermined. METHODS: This study examines SPARC expression in cultured human cerebral microvascular endothelial cells (hCMEC/D3)-an in vitro model of the blood-brain barrier (BBB)-as they transition between proliferative and barrier phenotypes and encounter pro-inflammatory stimuli. SPARC protein levels were quantified by Western blotting and immunocytochemistry and messenger RNA (mRNA) by RT-PCR. RESULTS: Constitutive SPARC expression by proliferating hCMEC/D3s is reduced as cells mature and establish a confluent monolayer. SPARC expression positively correlated with the proliferation marker Ki-67 suggesting a role for SPARC in cerebrovascular development. The pro-inflammatory molecules tumor necrosis factor-α (TNF-α) and endotoxin lipopolysaccharide (LPS) increased SPARC expression in cerebral endothelia. Interferon gamma (IFN-γ) abrogated SPARC induction observed with TNF-α alone. Barrier function assays show recombinant human (rh)-SPARC increased paracellular permeability and decreased transendothelial electrical resistance (TEER). This was paralleled by reduced zonula occludens-1 (ZO-1) and occludin expression in hCMEC/D3s exposed to rh-SPARC (1-10 µg/ml) compared with cells in media containing a physiological dose of SPARC. CONCLUSIONS: Together, these findings define a role for SPARC in influencing cerebral microvascular properties and function during development and inflammation at the BBB such that it may mediate processes of CNS inflammation and repair.


Assuntos
Barreira Hematoencefálica/metabolismo , Circulação Cerebrovascular/fisiologia , Células Endoteliais/metabolismo , Microvasos/metabolismo , Osteonectina/biossíntese , Barreira Hematoencefálica/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Circulação Cerebrovascular/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Expressão Gênica , Humanos , Microvasos/efeitos dos fármacos , Osteonectina/genética , Osteonectina/farmacologia
15.
J Mol Neurosci ; 60(4): 509-516, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27561928

RESUMO

Secreted protein acidic and rich in cysteine (SPARC) is widely expressed in the vascular smooth muscle cells (VSMCs) of human intracranial aneurysms (IAs), but the effect and underlying mechanism of SPARC on VSMCs during the formation and progression of IAs needs to be probed. Human umbilical arterial smooth muscle cells (HUASMCs) were treated with a gradient concentrations of SPARC in vitro for different time. Cell counting kit-8 (CCK-8) assay, cell cycle, and cell apoptosis were used to investigate the effect of SPARC on HUASMCs. After exposure to 2 and 4 µg/ml SPARC, cell viability were 89.3 ± 2.00 %, and 87.57 ± 2.17 % (P < 0.05 vs. control), respectively. Induced by 2 µg/ml SPARC, the proportion of cells in G0/G1 phase was 74.77 ± 1.33 % (P < 0.05 vs. control), and the early and late apoptosis ratio were 7.38 ± 1.25 % and 4.86 ± 0.81 % (P < 0.01 vs. control), respectively. After exposure to 2 µg/ml SPARC for 2, 6, 12, 24, and 48 h, Western blot analysis showed that the protein level of p21 was upregulated significantly at 2-12 h (P < 0.05 vs. control), while the expression of p53 remained stable within 48 h. The expression of Bax protein increased markedly and peaked at 24 (P < 0.01 vs. control), while Bcl2 protein decreased significantly at 48 h (P < 0.01 vs. control). Cleaved caspase3 was also upregulated dramatically and peaked at 24 h (P < 0.05 vs. control). The protein level of MMP2 increased significantly and peaked at 24 h (P < 0.01 vs. control), while TIMP2 remained stable and even reduced at 48 h (P < 0.05 vs. control). Taken together, SPARC could arrest HUASMCs in G0/G1 phase by overexpression of p21 and induce mitochondria-mediated apoptosis in vitro, which could result in the decreased cell viability. Besides, SPARC might also lead to the activation of MMP2 instead of MMP9. These results indicated SPARC could reduce the self-repair capability and increase injury of media layer and internal elastic lamina of intracranial artery, which would disrupt the normal homeostatic mechanism controlling vascular repair, thus promoting the formation and progression of IAs.


Assuntos
Artérias/metabolismo , Homeostase , Miócitos de Músculo Liso/efeitos dos fármacos , Osteonectina/farmacologia , Apoptose , Artérias/citologia , Caspase 3/genética , Caspase 3/metabolismo , Linhagem Celular , Humanos , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/metabolismo , Fase de Repouso do Ciclo Celular , Inibidor Tecidual de Metaloproteinase-2/genética , Inibidor Tecidual de Metaloproteinase-2/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
16.
Matrix Biol ; 31(5): 299-307, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22525512

RESUMO

There is considerable interest in understanding prostate cancer metastasis to bone and the interaction of these cells with the bone microenvironment. Osteonectin/SPARC/BM-40 is a collagen binding matricellular protein that is enriched in bone. Its expression is increased in prostate cancer metastases, and it stimulates the migration of prostate carcinoma cells. However, the presence of osteonectin in cancer cells and the stroma may limit prostate tumor development and progression. To determine how bone matrix osteonectin affects the behavior of prostate cancer cells, we modeled prostate cancer cell-bone interactions using the human prostate cancer cell line PC-3, and mineralized matrices synthesized by wild type and osteonectin-null osteoblasts in vitro. We developed this in vitro system because the structural complexity of collagen matrices in vivo is not mimicked by reconstituted collagen scaffolds or by more complex substrates, like basement membrane extracts. Second harmonic generation imaging demonstrated that the wild type matrices had thick collagen fibers organized into longitudinal bundles, whereas osteonectin-null matrices had thinner fibers in random networks. Importantly, a mouse model of prostate cancer metastases to bone showed a collagen fiber phenotype similar to the wild type matrix synthesized in vitro. When PC-3 cells were grown on the wild type matrices, they displayed decreased cell proliferation, increased cell spreading, and decreased resistance to radiation-induced cell death, compared to cells grown on osteonectin-null matrix. Our data support the idea that osteonectin can suppress prostate cancer pathogenesis, expanding this concept to the microenvironment of skeletal metastases.


Assuntos
Matriz Óssea/metabolismo , Raios gama , Osteoblastos/efeitos dos fármacos , Osteonectina/metabolismo , Neoplasias da Próstata/patologia , Animais , Western Blotting , Matriz Óssea/efeitos dos fármacos , Matriz Óssea/patologia , Matriz Óssea/efeitos da radiação , Comunicação Celular , Morte Celular/efeitos da radiação , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Forma Celular , Sobrevivência Celular , Masculino , Camundongos , Osteoblastos/metabolismo , Osteoblastos/patologia , Osteoblastos/efeitos da radiação , Osteonectina/genética , Osteonectina/farmacologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Microambiente Tumoral
17.
Aging (Albany NY) ; 4(1): 40-8, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22289652

RESUMO

Aging causes phenotypic changes in skeletal muscle progenitor cells (SMPCs) that lead to the loss of myogenicity and adipogenesis. Secreted protein acidic and rich in cysteine (SPARC), which is secreted from SMPCs, stimulates myogenesis and inhibits adipogenesis. The present study aimed to examine whether changes in SPARC expression, its signaling pathway, or both are involved in age-related phenotypic changes in SMPCs. SPARC expression levels were comparable in SMPCs derived from young and old rats. However, when SPARC expression was reduced by a SPARC-specific siRNA, SMPCs from young rats showed reduced myogenesis and increased adipogenesis. In striking contrast, old rats showed little changes in these functions. Recombinant SPARC was effective in inhibiting adipogenesis and promoting myogenesis of SMPCs from young rats but had no effect on SMPCs from old rats when endogenous SPARC levels were reduced by the SPARC-siRNA. Further, the level of integrin α5, a subunit of the putative SPARC receptor, was decreased in SMPCs from old rats, and its inhibition in SMPCs from young rats by siRNA reduced adipogenesis in response to SPARC. These results suggest that, although SPARC plays a role in regulating SMPC function, SMPCs become refractory to the action of SPARC with age. Our data may explain an age-related shift from myogenesis to adipogenesis, associated with sarcopenia.


Assuntos
Adipogenia/fisiologia , Envelhecimento/fisiologia , Desenvolvimento Muscular/fisiologia , Músculo Esquelético/citologia , Músculo Esquelético/fisiologia , Osteonectina/metabolismo , Animais , Regulação da Expressão Gênica/fisiologia , Integrinas/genética , Integrinas/metabolismo , Osteonectina/genética , Osteonectina/farmacologia , Interferência de RNA , RNA Interferente Pequeno , Ratos , Transdução de Sinais
18.
Oral Oncol ; 48(2): 102-9, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21996542

RESUMO

In order to investigate the role of myoepithelial cell and tumor microenvironment in salivary gland neoplasma, we have performed a study towards the effect of different extracellular matrix proteins (basement membrane matrix, type I collagen and fibronectin) on morphology and differentiation of benign myoepithelial cells from pleomorphic adenoma cultured with malignant cell culture medium from squamous cell carcinoma. We have also analyzed the expression of α-smooth muscle actin (α-SMA) and FGF-2 by immunofluorescence and qPCR. Our immunofluorescence results, supported by qPCR analysis, demonstrated that α-SMA and FGF-2 were upregulated in the benign myoepithelial cells from pleomorphic adenoma in all studied conditions on fibronectin substratum. However, the myoepithelial cells on fibronectin substratum did not alter their morphology under malignant conditioned medium stimulation and exhibited a stellate morphology and, occasionally focal adhesions with the substratum. In summary, our data demonstrated that the extracellular matrix exerts an important role in the morphology of the benign myoepithelial cells by the presence of focal adhesions and also inducing increase FGF-2 and α-SMA expression by these cells, especially in the fibronectin substratum.


Assuntos
Adenoma Pleomorfo/metabolismo , Carcinoma de Células Escamosas/metabolismo , Células Epiteliais/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Neoplasias das Glândulas Salivares/metabolismo , Actinas/metabolismo , Colágeno Tipo I/farmacologia , Meios de Cultivo Condicionados , Fator 2 de Crescimento de Fibroblastos/metabolismo , Fibronectinas/farmacologia , Imunofluorescência , Humanos , Osteonectina/farmacologia , Reação em Cadeia da Polimerase , Regulação para Cima
19.
Mol Cancer ; 9: 138, 2010 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-20525313

RESUMO

BACKGROUND: New, more effective strategies are needed to treat highly aggressive neuroblastoma. Our laboratory has previously shown that full-length Secreted Protein Acidic and Rich in Cysteine (SPARC) and a SPARC peptide corresponding to the follistatin domain of the protein (FS-E) potently block angiogenesis and inhibit the growth of neuroblastoma tumors in preclinical models. Peptide FS-E is structurally complex and difficult to produce, limiting its potential as a therapeutic in the clinic. RESULTS: In this study, we synthesized two smaller and structurally more simple SPARC peptides, FSEN and FSEC, that respectively correspond to the N-and C-terminal loops of peptide FS-E. We show that both peptides FSEN and FSEC have anti-angiogenic activity in vitro and in vivo, although FSEC is more potent. Peptide FSEC also significantly inhibited the growth of neuroblastoma xenografts. Histologic examination demonstrated characteristic features of tumor angiogenesis with structurally abnormal, tortuous blood vessels in control neuroblastoma xenografts. In contrast, the blood vessels observed in tumors, treated with SPARC peptides, were thin walled and structurally more normal. Using a novel method to quantitatively assess blood vessel abnormality we demonstrated that both SPARC peptides induced changes in blood vessel architecture that are consistent with blood vessel normalization. CONCLUSION: Our results demonstrate that SPARC peptide FSEC has potent anti-angiogenic and anti-tumorigenic effects in neuroblastoma. Its simple structure and ease of production indicate that it may have clinical utility in the treatment of high-risk neuroblastoma and other types of pediatric and adult cancers, which depend on angiogenesis.


Assuntos
Inibidores da Angiogênese/farmacologia , Neuroblastoma/tratamento farmacológico , Osteonectina/farmacologia , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Progressão da Doença , Células Endoteliais/efeitos dos fármacos , Imunofluorescência , Humanos , Camundongos , Camundongos Nus , Neovascularização Patológica/tratamento farmacológico , Neuroblastoma/irrigação sanguínea , Peptídeos , Ensaios Antitumorais Modelo de Xenoenxerto
20.
Carcinogenesis ; 31(5): 812-9, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20164124

RESUMO

Cellular senescence is another mechanism that can be exploited to achieve better chemosensitivity and greater tumor regression. Unlike apoptosis, cellular senescence can be induced at much lower concentrations of chemotherapy that are better tolerated by patients. We previously revealed that secreted protein acidic and rich in cysteine (SPARC), a matricellular protein, may function as a modulator of chemotherapy sensitivity by enhancing apoptosis. Here, we examine the effects of SPARC on cellular senescence in the presence of chemotherapy. Cellular senescence is induced only in sensitive colorectal cancer (CRC) cells with low concentrations of irinotecan (CPT-11). However, CPT-11-resistant cells exposed to endogenous or exogenous SPARC can also be triggered into cellular senescence. This induction is associated with higher levels of p16(INK4A) and phosphorylated p53. Knock down of p16(INK4A) reduces drug-induced senescence in all cells, but knock down and overexpression of p53 modulates senescence only in cells exposed to SPARC. Furthermore, treatment of mice with SPARC and CPT-11 leads to significantly increased cellular senescence and tumor regression. The chemosensitizing effects of SPARC in CRCs are, therefore, probably mediated in part by activating cellular senescence.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Camptotecina/análogos & derivados , Senescência Celular/efeitos dos fármacos , Neoplasias Colorretais/tratamento farmacológico , Osteonectina/farmacologia , Proteína Supressora de Tumor p53/fisiologia , Animais , Camptotecina/farmacologia , Neoplasias Colorretais/patologia , Inibidor p16 de Quinase Dependente de Ciclina/fisiologia , Dano ao DNA , Humanos , Irinotecano , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...