Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
1.
FEBS Lett ; 598(8): 935-944, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38553249

RESUMO

Chondrocyte differentiation is crucial for cartilage formation. However, the complex processes and mechanisms coordinating chondrocyte proliferation and differentiation remain incompletely understood. Here, we report a novel function of the adaptor protein Gulp1 in chondrocyte differentiation. Gulp1 expression is upregulated during chondrogenic differentiation. Gulp1 knockdown in chondrogenic ATDC5 cells reduces the expression of chondrogenic and hypertrophic marker genes during differentiation. Furthermore, Gulp1 knockdown impairs cell growth arrest during chondrocyte differentiation and reduces the expression of the cyclin-dependent kinase inhibitor p21. The activation of the TGF-ß/SMAD2/3 pathway, which is associated with p21 expression in chondrocytes, is impaired in Gulp1 knockdown cells. Collectively, these results demonstrate that Gulp1 contributes to cell growth arrest and chondrocyte differentiation by modulating the TGF-ß/SMAD2/3 pathway.


Assuntos
Diferenciação Celular , Condrócitos , Condrogênese , Inibidor de Quinase Dependente de Ciclina p21 , Transdução de Sinais , Proteína Smad2 , Proteína Smad3 , Fator de Crescimento Transformador beta , Animais , Camundongos , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Pontos de Checagem do Ciclo Celular/genética , Linhagem Celular , Proliferação de Células , Condrócitos/metabolismo , Condrócitos/citologia , Condrogênese/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/genética , Técnicas de Silenciamento de Genes , Proteína Smad2/metabolismo , Proteína Smad2/genética , Proteína Smad3/metabolismo , Proteína Smad3/genética , Fator de Crescimento Transformador beta/metabolismo
2.
J Cell Physiol ; 239(2): e31173, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38214103

RESUMO

Obesity and metabolic disorders caused by alterations in lipid metabolism are major health issues in developed, affluent societies. Adipose tissue is the only organ that stores lipids and prevents lipotoxicity in other organs. Mature adipocytes can affect themselves and distant metabolism-related tissues by producing various adipokines, including adiponectin and leptin. The engulfment adaptor phosphotyrosine-binding domain-containing 1 (GULP1) regulates intracellular trafficking of glycosphingolipids and cholesterol, suggesting its close association with lipid metabolism. However, the role of GULP1 in adipocytes remains unknown. Therefore, this study aimed to investigate the function of GULP1 in adipogenesis, glucose uptake, and the insulin signaling pathway in adipocytes. A 3T3-L1 cell line with Gulp1 knockdown (shGulp1) and a 3T3-L1 control group (U6) were established. Changes in shGulp1 cells due to GULP1 deficiency were examined and compared to those in U6 cells using microarray analysis. Glucose uptake was monitored via insulin stimulation in shGulp1 and U6 cells using a 2-NBDG glucose uptake assay, and the insulin signaling pathway was investigated by western blot analysis. Adipogenesis was significantly delayed, lipid metabolism was altered, and several adipogenesis-related genes were downregulated in shGulp1 cells compared to those in U6 cells. Microarray analysis revealed significant inhibition of peroxisome proliferator-activated receptor signaling in shGulp1 cells compared with U6 cells. The production and secretion of adiponectin as well as the expression of adiponectin receptor were decreased in shGulp1 cells. In particular, compared with U6 cells, glucose uptake via insulin stimulation was significantly decreased in shGulp1 cells through the disturbance of ERK1/2 phosphorylation. This is the first study to identify the role of GULP1 in adipogenesis and insulin-stimulated glucose uptake by adipocytes, thereby providing new insights into the differentiation and functions of adipocytes and the metabolism of lipids and glucose, which can help better understand metabolic diseases.


Assuntos
Adipogenia , Insulina , Transdução de Sinais , Animais , Camundongos , Células 3T3-L1 , Adipogenia/genética , Adiponectina/genética , Adiponectina/metabolismo , Diferenciação Celular , Regulação para Baixo , Glucose/metabolismo , Insulina/metabolismo , Lipídeos , Receptores Ativados por Proliferador de Peroxissomo/genética , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , PPAR gama/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo
3.
J Cell Physiol ; 238(5): 1006-1019, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36870066

RESUMO

The engulfment adaptor phosphotyrosine-binding domain containing 1 (GULP1) is an adaptor protein involved in the engulfment of apoptotic cells via phagocytosis. Gulp1 was first found to promote the phagocytosis of apoptotic cells by macrophages, and its role in various tissues, including neurons and ovaries, has been well studied. However, the expression and function of GULP1 in bone tissue are poorly understood. Consequently, to determine whether GULP1 plays a role in the regulation of bone remodeling in vitro and in vivo, we generated Gulp1 knockout (KO) mice. Gulp1 was expressed in bone tissue, mainly in osteoblasts, while its expression is very low in osteoclasts. Microcomputed tomography and histomorphometry analysis in 8-week-old male Gulp1 KO mice revealed a high bone mass in comparison with male wild-type (WT) mice. This was a result of decreased osteoclast differentiation and function in vivo and in vitro as confirmed by a reduced actin ring and microtubule formation in osteoclasts. Gas chromatography-mass spectrometry analysis further showed that both 17ß-estradiol (E2) and 2-hydroxyestradiol levels, and the E2/testosterone metabolic ratio, reflecting aromatase activity, were also higher in the bone marrow of male Gulp1 KO mice than in male WT mice. Consistent with mass spectrometry analysis, aromatase enzymatic activity was significantly higher in the bone marrow of male Gulp1 KO mice. Altogether, our results suggest that GULP1 deficiency decreases the differentiation and function of osteoclasts themselves and increases sex steroid hormone-mediated inhibition of osteoclast differentiation and function, rather than affecting osteoblasts, resulting in a high bone mass in male mice. To the best of our knowledge, this is the first study to explore the direct and indirect roles of GULP1 in bone remodeling, providing new insights into its regulation.


Assuntos
Aromatase , Estradiol , Osteoclastos , Animais , Masculino , Camundongos , Aromatase/genética , Aromatase/metabolismo , Osso e Ossos , Diferenciação Celular , Camundongos Knockout , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Microtomografia por Raio-X , Estradiol/metabolismo
4.
Curr Mol Med ; 22(8): 747-754, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34789124

RESUMO

OBJECTIVE: The relative balance of osteoblasts in bone formation and osteoclasts in bone resorption is crucial for maintaining bone health. With age, this balance between osteoblasts and osteoclasts is broken, resulting in bone loss. Anabolic drugs are continuously being developed to counteract this low bone mass. Recombinant proteins are used as biotherapeutics due to being relatively easy to produce on a large scale and are cost-effective through various expression systems. This study aimed to develop a recombinant protein that would positively impact osteoblast differentiation and mineralized nodule formation using unique cartilage matrix-associated protein (UCMA). METHODS: A recombinant glutathione-S-transferase (GST)-UCMA fusion protein was generated in an E.coli system, and purified by affinity chromatography. MC3T3-E1 osteoblast cells and Osterix (Osx)-knockdown stable cells were cultured for 14 days to investigate osteoblast differentiation and nodule formation in the presence of the recombinant GST-UCMA protein. The differentiated cells were assessed by alizarin red S staining and quantitative PCR of the osteoblast differentiation marker osteocalcin. In addition, cell viability in the presence of the recombinant GST-UCMA protein was determined by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and cell adhesion assay. RESULTS: The isolation of both purified recombinant GST-only and GST-UCMA proteins were confirmed at 26 kDa and 34 kDa, respectively, by Coomassie staining and western blot analysis. Neither dose-dependent nor time-dependent presence of recombinant GST-UCMA affected MC3T3-E1 cell viability. However, MC3T3-E1 cell adhesion to the recombinant GST-UCMA protein increased dose-dependently. Osteoblast differentiation and nodule formation were promoted in both MC3T3-E1 osteoblast cells and Osxknockdown stable cells when cultured in the presence of recombinant GST-UCMA protein. CONCLUSION: A recombinant GST-UCMA protein induces osteogenic differentiation and mineralization, suggesting its potential use as an anabolic drug to increase low bone mass in osteoporotic patients.


Assuntos
Osteoblastos , Osteogênese , Cartilagem/metabolismo , Diferenciação Celular , Humanos , Proteínas Matrilinas/metabolismo , Proteínas Matrilinas/farmacologia , Osteocalcina/metabolismo , Osteocalcina/farmacologia
5.
J Immunother Cancer ; 9(7)2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34330763

RESUMO

BACKGROUND: Statins preferentially promote tumor-specific apoptosis by depleting isoprenoid such as farnesyl pyrophosphate and geranylgeranyl pyrophosphate. However, statins have not yet been approved for clinical cancer treatment due, in part, to poor understanding of molecular determinants on statin sensitivity. Here, we investigated the potential of statins to elicit enhanced immunogenicity of KRAS-mutant (KRASmut) tumors. METHODS: The immunogenicity of treated cancer cells was determined by western blot, flow cytometry and confocal microscopy. The immunotherapeutic efficacy of mono or combination therapy using statin was assessed in KRASmut tumor models, including syngeneic colorectal cancer and genetically engineered lung and pancreatic tumors. Using NanoString analysis, we analyzed how statin influenced the gene signatures associated with the antigen presentation of dendritic cells in vivo and evaluated whether statin could induce CD8+ T-cell immunity. Multiplex immunohistochemistry was performed to better understand the complicated tumor-immune microenvironment. RESULTS: Statin-mediated inhibition of KRAS prenylation provoked severe endoplasmic reticulum (ER) stress by attenuating the anti-ER stress effect of KRAS mutation, thereby resulting in the immunogenic cell death (ICD) of KRASmut cancer cells. Moreover, statin-mediated ICD enhanced the cross-priming ability of dendritic cells, thereby provoking CD8+ T-cell immune responses against KRASmut tumors. Combination therapy using statin and oxaliplatin, an ICD inducer, significantly enhanced the immunogenicity of KRASmut tumors and promoted tumor-specific immunity in syngeneic and genetically engineered KRASmut tumor models. Along with immune-checkpoint inhibitors, the abovementioned combination therapy overcame resistance to PD-1 blockade therapies, improving the survival rate of KRASmut tumor models. CONCLUSIONS: Our findings suggest that KRAS mutation could be a molecular target for statins to elicit potent tumor-specific immunity.


Assuntos
Estresse do Retículo Endoplasmático/genética , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Proteínas Proto-Oncogênicas p21(ras)/efeitos dos fármacos , Animais , Humanos , Masculino , Camundongos , Mutação , Transfecção
6.
Biochem Biophys Res Commun ; 533(4): 710-716, 2020 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-33187645

RESUMO

CFL2, a skeletal muscle-specific member of the actin depolymerizing factor/cofilin protein family, is known to be involved in the regulation of actin filament dynamics. Although the impact of CFL2 has been studied in human myopathy, its functional contribution to myogenic differentiation, in terms of its effects on cell proliferation, cell cycle, and myogenic factor modulation, remains largely unknown. Here, we report that CFL2 is required for the myogenic differentiation of C2C12 myoblasts by regulating proliferation and myogenic transcription factors expressions. CFL2 expression was induced during myogenic progression, and its knockdown by siRNA in myoblasts enhanced phalloidin staining, indicating increased filamentous actin formation. Interestingly, CFL2 depletion stimulated cell proliferation and induced a cell cycle shift from G0/G1 to G2/M phases, which are known to inhibit progenitor cell differentiation. CFL2 knockdown markedly downregulated the protein expressions of myogenic transcription factors (MyoD, MyoG, and MEF2C) and thereby impaired the differentiation and myotube formation of C2C12 myoblasts. Collectively, this study highlights the roles played by CFL2 on cell cycle progression and proliferation and suggests a novel regulatory mechanism of myogenic differentiation mediated by CFL2.


Assuntos
Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Cofilina 2/metabolismo , Desenvolvimento Muscular/genética , Fibras Musculares Esqueléticas/metabolismo , Mioblastos/citologia , Animais , Proliferação de Células/genética , Regulação para Baixo , Pontos de Checagem da Fase G2 do Ciclo Celular/genética , Regulação da Expressão Gênica/genética , Técnicas de Silenciamento de Genes , Inativação Gênica , Fatores de Transcrição MEF2/metabolismo , Camundongos , Proteína MyoD/metabolismo , Miogenina/metabolismo , RNA Interferente Pequeno , Regulação para Cima
7.
Int J Mol Sci ; 21(19)2020 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-33008099

RESUMO

The placenta undergoes reconstruction at different times during fetal development to supply oxygen and nutrients required throughout pregnancy. To accommodate the rapid growth of the fetus, small spiral arteries undergo remodeling in the placenta. This remodeling includes apoptosis of endothelial cells that line spiral arteries, which are replaced by trophoblasts of fetal origin. Removal of dead cells is critical during this process. Stabilin-1 (Stab1) and stabilin-2 (Stab2) are important receptors expressed on scavenger cells that absorb and degrade apoptotic cells, and Stab1 is expressed in specific cells of the placenta. However, the role of Stab1 and Stab2 in placental development and maintenance remain unclear. In this study, we assessed Stab1 and Stab2 expression in the placenta and examined the reproductive capacity and placental development using a double-knockout mouse strain lacking both Stab1 and Stab2 (Stab1/2 dKO mice). Most pregnant Stab1/2 dKO female mice did not produce offspring and exhibited placental defects, including decidual hemorrhage and necrosis. Findings of this study offer the first description of the phenotypic characteristics of placentas and embryos of Stab1/2 dKO females during pregnancy, suggesting that Stab1 and Stab2 are involved in placental development and maintenance.


Assuntos
Moléculas de Adesão Celular Neuronais/genética , Placenta/metabolismo , Placentação/genética , Animais , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Feminino , Desenvolvimento Fetal/genética , Humanos , Camundongos , Camundongos Knockout , Oxigênio/metabolismo , Placenta/patologia , Gravidez , Reprodução/genética , Trofoblastos/metabolismo , Trofoblastos/patologia , Remodelação Vascular/genética
8.
Biomolecules ; 9(8)2019 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-31434355

RESUMO

Phosphatidylserine is a membrane phospholipid that is localized to the inner leaflet of the plasma membrane. Phosphatidylserine externalization to the outer leaflet of the plasma membrane is an important signal for various physiological processes, including apoptosis, platelet activation, cell fusion, lymphocyte activation, and regenerative axonal fusion. Stabilin-1 and stabilin-2 are membrane receptors that recognize phosphatidylserine on the cell surface. Here, we discuss the functions of Stabilin-1 and stabilin-2 as phosphatidylserine receptors in apoptotic cell clearance (efferocytosis) and cell fusion, and their ligand-recognition and signaling pathways.


Assuntos
Moléculas de Adesão Celular Neuronais/metabolismo , Animais , Humanos
9.
Biochim Biophys Acta Mol Cell Res ; 1866(10): 1595-1607, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31301364

RESUMO

The rapid and precise clearance of apoptotic cells (efferocytosis) involves a series of phagocytic processes through which apoptotic cells are recognized, engulfed, and degraded within phagocytes. The Rho-family GTPases critically rearrange the cytoskeleton for these phagocytic processes, but we know little about the mechanisms by which regulatory proteins control the spatiotemporal activities of the Rho-family GTPases. Here, we identify ArhGAP12 as a functional GTPase-activating protein (GAP) of Rac1 during Stabilin-2 mediated efferocytosis. ArhGAP12 constitutively forms a complex with the phosphatidylserine receptor, Stabilin-2, via direct interaction with the downstream protein, GULP, but is released from the complex when Stabilin-2 interacts with apoptotic cells. When the phagocytic cup is closed and the apoptotic cell is surrounded by the phagosomal membrane, ArhGAP12 localizes to the phagocytic cup via a specific interaction with phosphatidylinositol-4,5-bisphosphate, which is transiently biosynthesized in the phagocytic cup. Down-regulation of ArhGAP12 results in sustained Rac1 activity, arrangement of F-actin, and delayed phagosome-lysosome fusion. Our results collectively suggest that ArhGAP12 carries dual roles in Stabilin-2 mediated efferocytosis: it binds to GULP/Stabilin-2 and switches off Rac1 basal activity and switches on the Rac1 by releasing itself from the complex. In addition, the spatiotemporal membrane targeting of ArhGAP12 inactivates Rac1 in a time-specific and spatially coordinated manner to orchestrate phagosome maturation. This may shed light on how other RhoGAPs spatiotemporally inactivate Rac or Cdc42 during phagocytosis by various cells, in different circumstances.


Assuntos
Moléculas de Adesão Celular Neuronais/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Fagocitose/fisiologia , Fagossomos/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Actinas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Moléculas de Adesão Celular Neuronais/farmacologia , Linhagem Celular , Regulação para Baixo , Proteínas Ativadoras de GTPase/genética , Técnicas de Silenciamento de Genes , Humanos , Lisossomos/metabolismo , Camundongos , Fagócitos , Fagocitose/efeitos dos fármacos , Fosfatidilinositol 4,5-Difosfato/metabolismo
10.
Calcif Tissue Int ; 105(2): 205-214, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31025051

RESUMO

Stabilin-1 is a transmembrane receptor that regulates molecule recycling and cell homeostasis by controlling the intracellular trafficking and participates in cell-cell adhesion and transmigration. Stabilin-1 expression is observed in various organs, including bones; however, its function and regulatory mechanisms in the bone remain unclear. In this study, we evaluated the physiological function of stabilin-1 in bone cells and tissue using a stabilin-1 knockout (Stab1 KO) mouse model. In wild-type (WT) mice, stabilin-1 was expressed in osteoblasts and osteoclasts, and its expression was maintained during osteoblast differentiation but significantly decreased after osteoclast differentiation. There was no difference in osteoblast differentiation and function, or the expression of osteoblast differentiation markers between mesenchymal stem cells isolated from Stab1 KO and WT mice. However, osteoclast differentiation marker levels demonstrated a non-significant increase and bone-resorbing activity was significantly increased in vitro in RANKL-induced osteoclasts from Stab1-deficient bone marrow macrophages (BMMs) compared with those of WT BMMs. Microcomputed tomography showed a negligible difference between WT and Stab1 KO mice in bone volume and trabecular thickness and number. Moreover, no in vivo functional defect in bone formation by osteoblasts was observed in the Stab1 KO mice. The osteoclast surface and number showed an increased tendency in Stab1 KO mice compared to WT mice in vivo, but this difference was not statistically significant. Overall, these results indicate that Stab1 does not play an essential role in in vivo bone development and bone cell function, but it does affect in vitro osteoclast maturation and function for bone resorption.


Assuntos
Moléculas de Adesão Celular Neuronais/genética , Moléculas de Adesão Celular Neuronais/metabolismo , Osteoclastos/citologia , Animais , Células da Medula Óssea/citologia , Reabsorção Óssea , Osso e Ossos , Adesão Celular , Diferenciação Celular , Linhagem Celular , Movimento Celular , Feminino , Genótipo , Macrófagos/citologia , Masculino , Camundongos , Camundongos Knockout , Osteoblastos/citologia , Osteócitos/citologia , Osteogênese , Microtomografia por Raio-X
11.
J Korean Med Sci ; 34(12): e100, 2019 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-30940996

RESUMO

BACKGROUND: Increased expression of MDR1 gene is one of the major mechanisms responsible for multidrug resistance in cancer cells. Two alternative promoters, upstream and downstream, are responsible for transcription of MDR1 gene in the human. However, the molecular mechanism regarding the transactivation of MDR1 upstream promoter (USP) has not been determined. METHODS: Dual-luciferase reporter gene assays were used to assess the effect of Nkx-2.5 on MDR1 USP activity using reporter plasmids for human MDR1 USP and its mutants. MDR1 mRNA level was examined by quantitative real-time PCR. The direct binding of Nkx-2.5 to the USP of MDR1 was evaluated by promoter enzyme immunoassays and chromatin immunoprecipitation assays. RESULTS: Nkx-2.5 significantly stimulates the transactivation of MDR1 USP and increases MDR1 mRNA expression in MCF7 breast cancer cells. Reporter gene assays with deleted MDR1 USPs showed that the Nkx-2.5-binding site is located between positions -71 and +12. Mutation of the Nkx-2.5-binding site at nucleotide +4 to +10 markedly reduced the Nkx-2.5-mediated activation of MDR1 USP activity. A promoter binding immunoassay and a chromatin immunoprecipitation assay revealed that Nkx-2.5 binds directly to the region +4/+10 of human MDR1 USP. CONCLUSION: The results in the present study show Nkx-2.5 is a positive regulator for the transactivation of MDR1 USP in MCF7 breast cancer cells. Our findings will help elucidate the regulatory mechanism responsible for the multidrug resistant cancer phenotype.


Assuntos
Proteína Homeobox Nkx-2.5/metabolismo , Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Regulação Neoplásica da Expressão Gênica , Genes Reporter , Proteína Homeobox Nkx-2.5/antagonistas & inibidores , Proteína Homeobox Nkx-2.5/genética , Humanos , Células MCF-7 , Regiões Promotoras Genéticas , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Ativação Transcricional
12.
Cancer Lett ; 452: 51-58, 2019 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-30910590

RESUMO

Myeloid lineage immune cells, such as macrophages and dendritic cells, play important roles in the induction of antitumor immunity during the initial stage of the cancer-immunity cycle, eliciting antitumor adaptive immunity by phagocytosing cancer cells and processing cancer-specific antigens, and then presenting these antigens to T cells. During this process, cancer cell phagocytosis can be prevented by inhibitory signals, and the signaling cascades that elicit immune responses against cancer antigens can be inhibited by immunosuppressive myeloid cells in the tumor microenvironment. A number of therapeutic strategies for enhancing cancer cell phagocytosis and promoting antitumor immunity by targeting myeloid lineage cells have recently been developed. Here, we discuss recent advances in cancer immunotherapy that involve the targeting of myeloid lineage immune cells to induce effective antitumor immunity.


Assuntos
Antineoplásicos Imunológicos/uso terapêutico , Linhagem da Célula , Imunoterapia Adotiva , Imunoterapia/métodos , Células Mieloides/efeitos dos fármacos , Neoplasias/terapia , Animais , Antineoplásicos Imunológicos/efeitos adversos , Humanos , Imunoterapia/efeitos adversos , Imunoterapia Adotiva/efeitos adversos , Terapia de Alvo Molecular , Células Mieloides/imunologia , Células Mieloides/metabolismo , Células Mieloides/patologia , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/patologia , Fagocitose/efeitos dos fármacos , Transdução de Sinais , Resultado do Tratamento , Evasão Tumoral , Microambiente Tumoral
13.
Biochem Biophys Res Commun ; 511(2): 221-227, 2019 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-30791984

RESUMO

Unique cartilage matrix-associated protein (UCMA) is a secretory γ-carboxyglutamate (Gla) containing protein that is mainly expressed in the cartilage. Ucma, a downstream gene of both Runx2 and Osterix, has recently been described to promote osteoblast differentiation and matrix mineralization. However, till date, no studies have focused on the role of downstream target genes of Ucma in osteogenesis. Here, by Affymetrix GeneChip microarray analysis, we determined 45 differentially expressed genes in response to Ucma stable overexpression or knockdown in osteoblast cells, which provided insight into molecular mechanisms underlying osteoblast differentiation. In particular, we showed that fibrillin-2 (FBN2) expression was proportional to Ucma expression in osteoblasts as validated by quantitative PCR. We also showed that even though Gla-containing UCMA and calcium-binding EGF-like domain-containing FBN2 are known to have a high affinity for calcium, FBN2 whose expression was regulated by UCMA directly interacted with the UCMA protein, independent of calcium.


Assuntos
Cálcio/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Fibrilina-2/metabolismo , Osteoblastos/metabolismo , Animais , Linhagem Celular , Proteínas da Matriz Extracelular/genética , Fibrilina-2/genética , Regulação da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Osteoblastos/citologia , Osteogênese , Mapas de Interação de Proteínas
14.
Genes Genomics ; 41(4): 397-406, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30474828

RESUMO

BACKGROUND: C1q and TNF related protein 1 (C1QTNF1) is known to be associated with coronary artery diseases. However, the molecular function of C1QTNF1 on the vascular smooth muscles remains to be investigated. OBJECTIVE: This study was therefore undertaken to investigate the effect of C1QTNF1 on gene expression of human smooth muscle cells and to reveal potential molecular mechanisms mediated by C1QTNF1. METHODS: Vascular smooth muscle cells were incubated with recombinant C1QTNF1 for 16 h, followed by determining any change in mRNA expressions by Affymetrix genechip. Gene ontology (GO), KEGG pathway, and protein-protein interaction (PPI) network were analyzed in differentially expressed genes. In addition, validation of microarray data was performed using quantitative real-time PCR. RESULTS: The mRNA expressions of annotated 74 genes were significantly altered after incubation with recombinant C1QTNF1; 41 genes were up-regulated and 33 down-regulated. The differentially expressed genes were enriched in biological processes and KEGG pathways associated with inflammatory responses. In the PPI network analysis, IL-6, CCL2, and ICAM1 were identified as potential key genes with relatively high degree. The cluster analysis in the PPI network identified a significant module composed of upregulated genes, such as IL-6, CCL2, NFKBIA, SOD2, and ICAM1. The quantitative real-time PCR results of potential key genes were consistent with microarray data. CONCLUSION: The results in the present study provide insights on the effects of C1QTNF1 on gene expression of smooth muscle cells. We believe our findings will help to elucidate the molecular mechanisms regarding the functions of C1QTNF1 on smooth muscle cells in inflammatory diseases.


Assuntos
Aorta/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Proteínas/farmacologia , Transcriptoma , Células Cultivadas , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Perfilação da Expressão Gênica , Humanos , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Miócitos de Músculo Liso/metabolismo , Inibidor de NF-kappaB alfa/genética , Inibidor de NF-kappaB alfa/metabolismo , Mapas de Interação de Proteínas , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo
15.
Thromb Haemost ; 118(10): 1776-1789, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30235477

RESUMO

Sepsis develops because of overwhelming inflammatory responses to bacterial infection, and disrupts vascular integrity. Stabilin-1 (STAB-1) is a phagocytic receptor, which mediates efferocytosis in a phosphatidylserine (PS)-dependent manner. STAB-1 is expected to play important roles in efferocytosis during sepsis. Here, we determined the role of STAB-1 in maintaining and restoring vascular integrity. Macrophages and vascular endothelial cells were used to assess the effect of STAB-1 on survival rate, phagocytic activity, vascular permeability and transendothelial migration (TEM). Additionally, we investigated whether the high-mobility group box 1 (HMGB1)-receptor for advanced glycated end products complex interfered with the binding of Stab1 to PS. Mortality rate was higher in the Stab1-knockout mice than in the wild-type mice, and STAB-1 deficiency was related to reduced macrophage-mediated efferocytosis and the disruption of vascular integrity, which increased vascular permeability, and enhanced TEM. STAB-1 deficiency promoted lung injury, and elevated the expression of sepsis markers. The exogenous application of the anti-HMGB1 neutralizing antibody improved efferocytosis, vascular integrity and survival rate in sepsis. Collectively, our findings indicated that STAB-1 regulated and maintained vascular integrity through the clearance of infected apoptotic endothelial cells. Moreover, our results suggested that interventions targeting vascular integrity by STAB-1 signalling are promising therapeutic approaches to sepsis.


Assuntos
Moléculas de Adesão Celular Neuronais/metabolismo , Endotélio Vascular/fisiologia , Inflamação/imunologia , Macrófagos/fisiologia , Sepse/imunologia , Animais , Permeabilidade Capilar , Moléculas de Adesão Celular Neuronais/genética , Feminino , Proteína HMGB1/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fagocitose , Fosfatidilserinas/metabolismo , Migração Transendotelial e Transepitelial
16.
Nat Commun ; 9(1): 2165, 2018 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-29867097

RESUMO

Activation of T cell immune response is critical for the therapeutic efficacy of cancer immunotherapy. Current immunotherapies have shown remarkable clinical success against several cancers; however, significant responses remain restricted to a minority of patients. Here, we show a therapeutic strategy that combines enhancing the phagocytic activity of antigen-presenting cells with immunogenic cell death to trigger efficient antitumour immunity. Rho-kinase (ROCK) blockade increases cancer cell phagocytosis and induces antitumour immunity through enhancement of T cell priming by dendritic cells (DCs), leading to suppression of tumour growth in syngeneic tumour models. Combining ROCK blockade with immunogenic chemotherapy leads to increased DC maturation and synergistic CD8+ cytotoxic T cell priming and infiltration into tumours. This therapeutic strategy effectively suppresses tumour growth and improves overall survival in a genetic mouse mammary tumour virus/Neu tumour model. Collectively, these results suggest that boosting intrinsic cancer immunity using immunogenic killing and enhanced phagocytosis is a promising therapeutic strategy for cancer immunotherapy.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Imunidade/efeitos dos fármacos , Neoplasias Experimentais/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Quinases Associadas a rho/antagonistas & inibidores , Amidas/administração & dosagem , Amidas/farmacologia , Animais , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Morte Celular/efeitos dos fármacos , Morte Celular/imunologia , Linhagem Celular Tumoral , Células Cultivadas , Cisplatino/administração & dosagem , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Doxorrubicina/administração & dosagem , Humanos , Imunidade/imunologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/patologia , Inibidores de Proteínas Quinases/administração & dosagem , Piridinas/administração & dosagem , Piridinas/farmacologia , Linfócitos T Citotóxicos/efeitos dos fármacos , Linfócitos T Citotóxicos/imunologia , Quinases Associadas a rho/imunologia , Quinases Associadas a rho/metabolismo
17.
Adv Mater ; 30(10)2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29333661

RESUMO

A growing appreciation of the relationship between the immune system and the tumorigenesis has led to the development of strategies aimed at "re-editing" the immune system to kill tumors. Here, a novel tactic is reported for overcoming the activation-energy threshold of the immunosuppressive tumor microenvironment and mediating the delivery and presentation of tumor neoantigens to the host's immune system. This nature-derived nanocage not only efficiently presents ligands that enhance cancer cell phagocytosis, but also delivers drugs that induce immunogenic cancer cell death. The designed nanocage-therapeutics induce the release of neoantigens and danger signals in dying tumor cells, and leads to enhancement of tumor cell phagocytosis and cross-priming of tumor specific T cells by neoantigen peptide-loaded antigen-presenting cells. Potent inhibition of tumor growth and complete eradication of tumors is observed through systemic tumor-specific T cell responses in tumor draining lymph nodes and the spleen and further, infiltration of CD8+ T cells into the tumor site. Remarkably, after removal of the primary tumor, all mice treated with this nanocage-therapeutics are protected against subsequent challenge with the same tumor cells, suggesting development of lasting, tumor-specific responses. This designed nanocage-therapeutics "awakens" the host's immune system and provokes a durable systemic immune response against cancer.


Assuntos
Fagocitose , Animais , Linfócitos T CD8-Positivos , Morte Celular , Células Dendríticas , Camundongos , Camundongos Endogâmicos C57BL
18.
FEBS J ; 284(21): 3575-3588, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28881496

RESUMO

Xk-related protein 8 (Xkr8) is a scramblase and responsible for phosphatidylserine (PS) exposure on the cell surface in a caspase-dependent manner. Although PS exposure is found to be important for myotube formation during myoblast differentiation, the role of Xkr8 during myogenesis has not been elucidated. Here we show that Xkr8 contributes to myoblast differentiation. Xkr8 overexpression induced the formation of large myotubes during early differentiation, but this phenotype was not related to caspase-dependent cleavage of Xkr8. Furthermore, forced Xkr8 expression accelerated myoblast differentiation and conferred cell-death resistance after the induction of differentiation. Consistent with these results, Xkr8-knocked-down myoblasts exhibited impaired differentiation and more apoptotic cells during differentiation, implying the involvements of Xkr8 in the survival and proliferation of myoblasts. Taken together, the study shows Xkr8 influences myogenesis by acting as a positive regulator of terminal differentiation and myoblast survival.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Proteínas de Membrana/metabolismo , Mioblastos/citologia , Mioblastos/metabolismo , Animais , Proteínas Reguladoras de Apoptose/genética , Diferenciação Celular , Sobrevivência Celular , Humanos , Proteínas de Membrana/genética , Camundongos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
19.
Exp Mol Med ; 49(5): e331, 2017 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-28496201

RESUMO

The clearance of apoptotic cells is an essential process for tissue homeostasis. To this end, cells undergoing apoptosis must display engulfment signals, such as 'find-me' and 'eat-me' signals. Engulfment signals are recognized by multiple types of phagocytic machinery in phagocytes, leading to prompt clearance of apoptotic cells. In addition, apoptotic cells and phagocytes release tolerogenic signals to reduce immune responses against apoptotic cell-derived self-antigens. Here we discuss recent advances in our knowledge of engulfment signals, the phagocytic machinery and the signal transduction pathways for apoptotic cell engulfment.


Assuntos
Apoptose , Fagócitos/metabolismo , Fagocitose , Transdução de Sinais , Animais , Humanos
20.
PLoS One ; 12(4): e0174603, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28376111

RESUMO

The removal of unwanted or damaged cells by phagocytes is achieved via a finely regulated cleaning process called efferocytosis. To characterize the mechanisms through which phagocytes control the intake of apoptotic cells, we investigated how the phagocyte's appetite for engulfed cells may be coordinated by RhoA and Rac1 in the phagocytic cup. We used FRET biosensors to visualize the spatiotemporal dynamics of Rho-family GTPases, and found that RhoA, which is known to be downregulated during phagocytosis, was transiently upregulated at the phagocytic cup immediately prior to ingestion. Conversely, Rac1 was upregulated during the engulfment process and then downregulated prior to phagosomal maturation. Moreover, disturbance of the dynamic activities of RhoA led to uncontrolled engulfment, such as fast and undiscerning eating. Our results reveal that the temporal activity of RhoA GTPase alters the Rac1/RhoA balance at the phagocytic cup prior to ingestion, and that this plays a distinct role in orchestrating efferocytosis, with RhoA modulating the rate of engulfment to ensure that the phagocyte engulfs an appropriate amount of the correct material.


Assuntos
Fagócitos/fisiologia , Fagocitose/fisiologia , Proteínas rac1 de Ligação ao GTP/fisiologia , Proteína rhoA de Ligação ao GTP/fisiologia , Animais , Apoptose/fisiologia , Técnicas Biossensoriais , Transferência Ressonante de Energia de Fluorescência , Humanos , Células L , Macrófagos Peritoneais/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Modelos Biológicos , Imagem com Lapso de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA