Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Adv Sci (Weinh) ; 10(35): e2305550, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37828611

RESUMO

Effective treatment for metastasis, a leading cause of cancer-associated death, is still lacking. To seed on a distal organ, disseminated cancer cells (DCCs) must adapt to the local tissue microenvironment. However, it remains elusive how DCCs respond the pro-metastatic niche signals. Here, systemic motif-enrichment identified myocyte enhancer factor 2D (MEF2D) as a critical sensor of niche signals to regulate DCCs adhesion and colonization, leading to intrahepatic metastasis and recurrence of liver cancer. In this context, MEF2D transactivates Itgb1 (coding ß1-integrin) and Itgb4 (coding ß4-integrin) to execute temporally unique functions, where ITGB1 recognizes extracellular matrix for early seeding, and ITGB4 acts as a novel sensor of neutrophil extracellular traps-DNA (NETs-DNA) for subsequent chemotaxis and colonization. In turn, an integrin-FAK circuit promotes a phosphorylation-dependent USP14-orchastrated deubiquitination switch to stabilize MEF2D via circumventing degradation by the E3-ubiquitin-ligase MDM2. Clinically, the USP14(pS432)-MEF2D-ITGB1/4 feedback loop is often hyper-active and indicative of inferior outcomes in human malignancies, while its blockade abrogated intrahepatic metastasis of DCCs. Together, DCCs exploit a deubiquitination-dependent switch on MEF2D to integrate niche signals in the liver mesenchyme, thereby amplifying the pro-metastatic integrin-FAK signaling. Disruption of this feedback loop is clinically applicable with fast-track potential to block microenvironmental cues driving metastasis.


Assuntos
Neoplasias Hepáticas , Ubiquitina , Humanos , Fatores de Transcrição MEF2/genética , Fatores de Transcrição MEF2/metabolismo , Integrinas , DNA , Microambiente Tumoral , Ubiquitina Tiolesterase
2.
J Hematol Oncol ; 16(1): 59, 2023 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-37277776

RESUMO

Amino acids are basic nutrients for immune cells during organ development, tissue homeostasis, and the immune response. Regarding metabolic reprogramming in the tumor microenvironment, dysregulation of amino acid consumption in immune cells is an important underlying mechanism leading to impaired anti-tumor immunity. Emerging studies have revealed that altered amino acid metabolism is tightly linked to tumor outgrowth, metastasis, and therapeutic resistance through governing the fate of various immune cells. During these processes, the concentration of free amino acids, their membrane bound transporters, key metabolic enzymes, and sensors such as mTOR and GCN2 play critical roles in controlling immune cell differentiation and function. As such, anti-cancer immune responses could be enhanced by supplement of specific essential amino acids, or targeting the metabolic enzymes or their sensors, thereby developing novel adjuvant immune therapeutic modalities. To further dissect metabolic regulation of anti-tumor immunity, this review summarizes the regulatory mechanisms governing reprogramming of amino acid metabolism and their effects on the phenotypes and functions of tumor-infiltrating immune cells to propose novel approaches that could be exploited to rewire amino acid metabolism and enhance cancer immunotherapy.


Assuntos
Neoplasias , Humanos , Neoplasias/tratamento farmacológico , Metabolismo Energético , Imunidade , Imunoterapia , Aminoácidos/metabolismo , Microambiente Tumoral
3.
Cell Oncol (Dordr) ; 46(4): 867-883, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37010700

RESUMO

The epithelial lining of the stomach undergoes rapid turnover to preserve its structural and functional integrity, a process driven by long-lived stem cells residing in the antral and corpus glands. Several subpopulations of gastric stem cells have been identified and their phenotypic and functional diversities linked to spatiotemporal specification of stem cells niches. Here, we review the biological features of gastric stem cells at various locations of the stomach under homeostatic conditions, as demonstrated by reporter mice, lineage tracing, and single cell sequencing. We also review the role of gastric stem cells in epithelial regeneration in response to injury. Moreover, we discuss emerging evidence demonstrating that accumulation of oncogenic drivers or alteration of stemness signaling pathways in gastric stem cells promotes gastric cancer. Given a fundamental role of the microenvironment, this review highlights the role reprogramming of niche components and signaling pathways under pathological conditions in dictating stem cell fate. Several outstanding issues are raised, such as the relevance of stem cell heterogeneity and plasticity, and epigenetic regulatory mechanisms, to Helicobacter pylori infection-initiated metaplasia-carcinogenesis cascades. With the development of spatiotemporal genomics, transcriptomics, and proteomics, as well as multiplexed screening and tracing approaches, we anticipate that more precise definition and characterization of gastric stem cells, and the crosstalk with their niche will be delineated in the near future. Rational exploitation and proper translation of these findings may bring forward novel modalities for epithelial rejuvenation and cancer therapeutics.


Assuntos
Infecções por Helicobacter , Helicobacter pylori , Neoplasias Gástricas , Camundongos , Animais , Mucosa Gástrica/metabolismo , Helicobacter pylori/fisiologia , Estômago/patologia , Células-Tronco/metabolismo , Carcinogênese/patologia , Transformação Celular Neoplásica/metabolismo , Neoplasias Gástricas/patologia , Homeostase , Células Epiteliais/patologia , Microambiente Tumoral
4.
Front Oncol ; 12: 1047907, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36439438

RESUMO

Mesenchymal stem cells (MSCs) have been used to treat various diseases including Alzheimer's disease and cancer. In particular, the immunomodulatory function of MSCs plays a major role in cancer therapy using stem cells. However, MSCs exert promotive and inhibitory effects on cancer. The immunomodulatory effects of MSCs in the tumor microenvironment (TME) are ambiguous, which is the primary reason for the different outcomes of MSCs therapies for tumors. This review discusses the use of MSCs in cancer immunotherapy and their immunomodulatory mechanisms in cancers.

5.
J Thorac Dis ; 14(4): 1193-1202, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35572907

RESUMO

Background: A number of studies have demonstrated that trichosanthin (TCS) can induce apoptosis in numerous types of tumor cell lines. However, whether TCS can induce pyroptosis has not yet been reported. This study aimed to investigate the role of TCS and its inhibitory effect on tumor growth by modulating pyroptosis in non-small cell lung cancer (NSCLC). Methods: Effects of different concentrations of TCS on the cell viability, proliferation, migration and invasion of NSCLC were detected by Cell Counting Kit-8 (CCK-8), colony formation, migration, and invasion assays. Immunofluorescence was used to detect the effect of TCS on the expression of pyroptosis marker protein gasdermin-D (GSDMD)-N in A549 cells. A tumor xenograft animal model was established by injecting A549 cells into nude mice. Results: In the present study, we found that TCS significantly inhibited the proliferation, migration, and invasion of A549 cells in a concentration-dependent manner. In addition, TCS at a high concentration (40 µg/mL) significantly promoted the expression of pyroptosis-related proteins [GSDMD-N, NLR family pyrin domain containing 3 (NLRP3), apoptosis-associated speck-like protein containing a CARD (ASC), caspase-1, and GSDMD], which showed an inhibitory effect on the pyroptosis of A549 cells. Additionally, we found that necrosulfonamide (NSA) significantly reversed the inhibitory effect of high concentrations of TCS on the pyroptosis of A549 cells. The in vivo experiments showed that TCS effectively reduced the tumor volume and inhibited the expression of Ki-67, whereas it increased the expression of GSDMD-N. Conclusions: Taken together, these results indicated that TCS could inhibit the progression of NSCLC by promoting pyroptosis. These findings provide further information on the possible underlying mechanism of TCS in the treatment of NSCLC.

6.
RSC Adv ; 11(25): 14891-14898, 2021 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-35424028

RESUMO

Spinel LiMn2O4 (LMO) with a three-dimensional structure has become one of the cathode materials that has gained the most interest due to its safety, low price and abundant resources. However, the lithium ion transmission is limited by large particle size and particle agglomeration of LMO. Thus, reducing the particle size and agglomeration of LMO can effectively improve its lithium ion transmission. Here, we synthesized a LMO cathode material with a nanoscale crystal size using the flexible expanded graphite (EG) soft template and Pichini method. EG-controlled particle size and particle agglomeration of LMO is conducive to charge transfer and diffusion of lithium ions between LMO and the electrolyte, meanwhile, there are more redox sites on the nanosized LMO particles, which makes the redox reaction of LMO more thorough during the charge and discharge process, resulting in high capacitance performance. In order to obtain the considerably required lithium-ion capacitors (LICs) with high energy density and power density, we assembled aqueous LMO//activated carbon (AC) LICs with 5 M LiNO3 as the aqueous electrolytes, which are environmentally friendly, safe, low cost and have higher electrical conductivity than organic electrolytes. The optimal LIC has an energy density of 32.63 W h kg-1 at a power density of 500 W kg-1 and an energy density of 8.06 W h kg-1 at a power density of 10 000 W kg-1, which is higher than most of the LMO-based LICs in previous reports. After 2000 cycles, the specific capacitance retention rate was 75.9% at a current density of 3 A g-1. Therefore, our aqueous LMO//AC LICs synthesized by the soft template/Pichini method have wide prospects and are suitable for low-cost, high-safety and high-power applications.

7.
Gastroenterology ; 158(3): 664-678.e24, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31678303

RESUMO

BACKGROUND & AIMS: Immune checkpoint inhibitors have some efficacy in the treatment of hepatocellular carcinoma (HCC). Programmed cell death 1 ligand 1 (PD-L1), expressed on some cancer cells, binds to the receptor programmed cell death 1 (PDCD1, also called PD1) on T cells to prevent their proliferation and reduce the antigen-tumor immune response. Immune cells that infiltrate some types of HCCs secrete interferon gamma (IFNG). Some HCC cells express myocyte enhancer factor 2D (MEF2D), which has been associated with shorter survival times of patients. We studied whether HCC cell expression of MEF2D regulates expression of PD-L1 in response to IFNG. METHODS: We analyzed immune cells from 20 fresh HCC tissues by flow cytometry. We analyzed 225 fixed HCC tissues (from 2 cohorts) from patients in China by immunohistochemistry and obtained survival data. We created mice with liver-specific knockout of MEF2D (MEF2DLPC-KO mice). We knocked out or knocked down MEF2D, E1A binding protein p300 (p300), or sirtuin 7 (SIRT7) in SMMC-7721, Huh7, H22, and Hepa1-6 HCC cell lines, some incubated with IFNG. We analyzed liver tissues from mice and cell lines by RNA sequencing, immunoblot, dual luciferase reporter, and chromatin precipitation assays. MEF2D protein acetylation and proteins that interact with MEF2D were identified by coimmunoprecipitation and pull-down assays. H22 cells, with MEF2D knockout or without (controls), were transplanted into BALB/c mice, and some mice were given antibodies to deplete T cells. Mice bearing orthotopic tumors grown from HCC cells, with or without knockout of SIRT7, were given injections of an antibody against PD1. Growth of tumors was measured, and tumors were analyzed by immunohistochemistry and flow cytometry. RESULTS: In human HCC specimens, we found an inverse correlation between level of MEF2D and numbers of CD4+ and CD8+ T cells; level of MEF2D correlated with percentages of PD1-positive or TIM3-positive CD8+ T cells. Knockout of MEF2D from H22 cells reduced their growth as allograft tumors in immune-competent mice but not in immune-deficient mice or mice with depletion of CD8+ T cells. When MEF2D-knockout cells were injected into immune-competent mice, they formed smaller tumors that had increased infiltration and activation of T cells compared with control HCC cells. In human and mouse HCC cells, MEF2D knockdown or knockout reduced expression of PD-L1. MEF2D bound the promoter region of the CD274 gene (encodes PD-L1) and activated its transcription. Overexpression of p300 in HCC cells, or knockout of SIRT7, promoted acetylation of MEF2D and increased its binding, along with acetylated histones, to the promoter region of CD274. Exposure of HCC cells to IFNG induced expression of p300 and its binding MEF2D, which reduced the interaction between MEF2D and SIRT7. MEF2D-induced expression of PD-L1 upon IFNG exposure was independent of interferon-regulatory factors 1 or 9. In HCC cells not exposed to IFNG, SIRT7 formed a complex with MEF2D that attenuated expression of PD-L1. Knockout of SIRT7 reduced proliferation of HCC cells and growth of tumors in immune-deficient mice. Compared with allograft tumors grown from control HCC cells, in immune-competent mice, tumors grown from SIRT7-knockout HCC cells expressed higher levels of PD-L1 and had reduced infiltration and activation of T cells. In immune-competent mice given antibodies to PD1, allograft tumors grew more slowly from SIRT7-knockout HCC cells than from control HCC cells. CONCLUSIONS: Expression of MEF2D by HCC cells increases their expression of PD-L1, which prevents CD8+ T-cell-mediated antitumor immunity. When HCC cells are exposed to IFNG, p300 acetylates MEF2D, causing it to bind the CD274 gene promoter and up-regulate PD-L1 expression. In addition to promoting HCC cell proliferation, SIRT7 reduced acetylation of MEF2D and expression of PD-L1 in HCC cells not exposed to IFNG. Strategies to manipulate this pathway might increase the efficacy of immune therapies for HCC.


Assuntos
Antígeno B7-H1/genética , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , Sirtuínas/genética , Adolescente , Adulto , Idoso , Animais , Contagem de Linfócito CD4 , Linfócitos T CD8-Positivos/metabolismo , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Proteína p300 Associada a E1A/genética , Proteína p300 Associada a E1A/metabolismo , Feminino , Técnicas de Inativação de Genes , Receptor Celular 2 do Vírus da Hepatite A/metabolismo , Humanos , Imunocompetência , Interferon gama/farmacologia , Neoplasias Hepáticas/patologia , Fatores de Transcrição MEF2/genética , Fatores de Transcrição MEF2/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Pessoa de Meia-Idade , Transplante de Neoplasias , Receptor de Morte Celular Programada 1/metabolismo , Sirtuínas/metabolismo , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/genética , Adulto Jovem
8.
RSC Adv ; 9(25): 14407-14416, 2019 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-35519317

RESUMO

It is reported that olivine-type lithium iron phosphate (LFP) for Li-ion batteries is one of the most widely utilized cathode materials, but its high-power applications are limited due to its intrinsically poor ion transfer rate and conductivity. Therefore, it is highly desired to fabricate LFP Li-ion capacitors (LICs) with high power performance and excellent cyclic reversibility, especially in safe, low cost and environmentally friendly aqueous electrolytes. Here, we fabricate LFP/expanded graphite (EG) nanocomposites by a one-step process, in which polyethylene glycol (PEG) is used as the particle growth inhibitor combined with vacuum infiltration of the LFP precursor into EG as a conductive sub-phase, and further investigate their high-power performance in aqueous LICs. Embedding spherical LFP nanoparticles with well-controlled size and agglomeration into the pores of EG and wrapping LFP nanoparticles by EG films contribute to the rapid electron and ion diffusion in LFP/EG composites, resulting in excellent cyclic reversibility and rate performance of LFP/EG composites. The aqueous LFP/EG//active carbon (AC) LICs were assembled in LiNO3 electrolytes with LFP/EG composites and AC as the positive and negative electrodes, respectively. The optimal LIC shows a power density of 2367.9 W kg-1 at an energy density of 6.5 W h kg-1, dramatically favorable rate characteristics and excellent cycle life with 82.1% capacitance retention of its primary capacitance at 2 A g-1 after 6000 cycles, markedly higher than those of the commercial LFP LIC. The presented aqueous LFP/EG//AC LICs with excellent electrochemical performance are expected to have broad high-power appliances that are cost-sensitive and highly secure.

9.
Sheng Wu Gong Cheng Xue Bao ; 34(5): 752-760, 2018 May 25.
Artigo em Chinês | MEDLINE | ID: mdl-29893083

RESUMO

This study was aimed to investigate the effect of Forkhead Box G1 (FOXG1) on the epithelial-mesenchymal transition (EMT) of colorectal cancer (CRC) cells and the underlying mechanism. For this purpose, FOXG1 lentiviral interference (shRNA) plasmid and expression plasmid were constructed. Western blotting was used to analyze the expression of FOXG1 protein in five CRC cells, namely RKO, SW480, SW620, LoVo and DLD-1. The shRNA fragment of FOXG1 (shFOXG1) was designed and synthesized. Recombinant plasmids were obtained with the aid of DNA recombination technique. Double digestion and sequencing were used to identify the recombinant plasmids, and then lentivirus packaging, purification and stable transfection were carried out. Additionally, stable CRC cell lines were screened out. The changes of FOXG1 knockdown and overexpression efficiency, E-cadherin, Vimentin, Fibronectin, Snail, Twist mRNA and protein were investigated respectively by Western blotting and qRT-PCR analysis. Furthermore, the changes of cell morphology after knockdown and cell migration ability were evaluated respectively with optical microscopy, scratch test and Transwell assay. FOXG1 had the highest protein expression in RKO and the lowest in DLD-1 among the five CRC cells. Compared with those of the control group, the cell morphology in FOXG1 knockdown RKO group was changed from spindle into round or polygonal shape, cell polarization was enhanced and tight junction assembly was acclerated while cell migration distance was noticeably decreased. Moreover, the number of cells invaded and migrated through chambers was significantly reduced. Among these key factors of EMT, the expression of E-cadherin was increased while the expressions of Vimentin, Fibronectin, Snail and Twist were decreased. The opposite was the case in the overexpressed FOXG1 group. The overexpression of FOXG1 in CRC promoted the invasion and metastasis of CRC cells and played a crucial role in regulating the EMT. Thus, FOXG1 might be a novel therapeutic target in CRC treatment.


Assuntos
Neoplasias Colorretais/patologia , Transição Epitelial-Mesenquimal , Fatores de Transcrição Forkhead/metabolismo , Invasividade Neoplásica , Metástase Neoplásica , Proteínas do Tecido Nervoso/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Neoplasias Colorretais/metabolismo , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos
10.
Mol Med Rep ; 16(3): 2740-2746, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28677757

RESUMO

Chondrocyte hypertrophy is a physiological process in endochondral ossification. However, the hypertrophic­like alterations of chondrocytes at the articular surface may result in osteoarthritis (OA). In addition, the generation of fibrocartilage with a decreased biological function in tissue engineered cartilage, has been attributed to chondrocyte hypertrophy. Therefore, suppressing chondrocyte hypertrophy in OA and the associated regeneration of non­active cartilage is of primary concern. The present study examined the effects of xanthotoxin (XAT), which is classified as a furanocoumarin, on chondrocyte hypertrophic differentiation of mesenchymal stem cells. Following XAT treatment, the expression levels of genes associated with chondrocyte hypertrophy were detected via immunohistochemistry, western blotting and reverse transcription­quantitative polymerase chain reaction. The results revealed that XAT inhibited the expression of various chondrocyte hypertrophic markers, including runt related transcription factor 2 (Runx2), matrix metalloproteinase 13 and collagen type X α1 chain. Further exploration indicated that XAT reduced the activation of p38­mitogen activated protein kinase and then increased the expression of histone deacetylase 4 to suppress Runx2. The findings indicated that XAT maintained the chondrocyte phenotype in regenerated cartilage and therefore may exhibit promise as a potential drug for the treatment of OA in the future.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Condrogênese/efeitos dos fármacos , Histona Desacetilases/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Metoxaleno/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Linhagem Celular , Condrócitos/citologia , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Condrócitos/patologia , Reagentes de Ligações Cruzadas/farmacologia , Hipertrofia/metabolismo , Hipertrofia/patologia , Hipertrofia/prevenção & controle , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Camundongos , Transdução de Sinais/efeitos dos fármacos
11.
Cancer Lett ; 400: 117-126, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28478181

RESUMO

Myocyte enhancer factor 2D (MEF2D) is involved in many aspects of cancer progression, including cell proliferation, invasion, and migration. However, little is known about the role of MEF2D in tumor angiogenesis. Using clinical specimens, colorectal cancer (CRC) cell lines and a mouse model in the present study, we found that MEF2D expression was positively correlated with CD31-positive microvascular density in CRC tissues. MEF2D promoted tumor angiogenesis in vitro and in vivo and induced the expression of proangiogenic cytokines in CRC cells. MEF2D was found to be a downstream effector of hypoxia-inducible factor (HIF)-1α in the induction of tumor angiogenesis. HIF-1α transactivates MEF2D expression by binding to the MEF2D gene promoter. These results demonstrate that the HIF-1α/MEF2D axis can serve as a therapeutic target for the treatment of CRC.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias Colorretais/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Neovascularização Patológica , Animais , Sítios de Ligação , Biomarcadores Tumorais/genética , Células CACO-2 , Neoplasias Colorretais/irrigação sanguínea , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Meios de Cultivo Condicionados/metabolismo , Citocinas/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Células HCT116 , Células HT29 , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Fatores de Transcrição MEF2/genética , Fatores de Transcrição MEF2/metabolismo , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Microvasos/metabolismo , Microvasos/patologia , Pessoa de Meia-Idade , Comunicação Parácrina , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Prognóstico , Regiões Promotoras Genéticas , Interferência de RNA , Transdução de Sinais , Fatores de Tempo , Ativação Transcricional , Transfecção , Carga Tumoral , Hipóxia Tumoral , Microambiente Tumoral
12.
BMB Rep ; 49(10): 548-553, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27439604

RESUMO

Mesenchymal stem cells (MSCs) are widely used in cartilage tissue engineering to repair articular cartilage defects. However, hypertrophy of chondrocytes derived from MSCs might hinder the stabilization of hyaline cartilage. Thus, it is very important to find a suitable way to maintain the chondrogenic phenotype of chondrocytes. It has been reported that cordycepin has anti-inflammatory and anti-tumor functions. However, the role of cordycepin in chondrocyte hypertrophy remains unclear. Therefore, the objective of this study was to determine the effect of cordycepin on chondrogenesis and chondrocyte hypertrophy in MSCs and ATDC5 cells. Cordycepin upregulated chondrogenic markers including Sox9 and collagen type II while down-regulated hypertrophic markers including Runx2 and collagen type X. Further exploration showed that cordycepin promoted chondrogenesis through inhibiting Nrf2 while activating BMP signaling. Besides, cordycepin suppressed chondrocyte hypertrophy through PI3K/Bapx1 pathway and Notch signaling. Our results indicated cordycepin had the potential to maintain chondrocyte phenotype and reconstruct engineered cartilage. [BMB Reports 2016; 49(10): 548-553].


Assuntos
Antineoplásicos/farmacologia , Diferenciação Celular/efeitos dos fármacos , Desoxiadenosinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Antineoplásicos/química , Células Cultivadas , Condrócitos/citologia , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Condrogênese/efeitos dos fármacos , Colágeno Tipo II/metabolismo , Colágeno Tipo X/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Desoxiadenosinas/química , Regulação para Baixo/efeitos dos fármacos , Proteínas de Homeodomínio/metabolismo , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Receptores Notch/genética , Receptores Notch/metabolismo , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/metabolismo , Fatores de Transcrição/metabolismo , Regulação para Cima/efeitos dos fármacos
13.
Sci Rep ; 6: 21499, 2016 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-26856880

RESUMO

Bone is a dynamic organ continuously undergoing shaping, repairing and remodeling. The homeostasis of bone is maintained by the balance between osteoblastic bone formation and osteoclastic bone resorption. Osteoclasts (OCs) are specialized multinucleated cells derived from hematopoietic stem cells (HSCs) or monocytes/macrophage progenitor cells. There are different stages during osteoclastogenesis, and one of the most important steps to form functional osteoclasts is realized by cell-cell fusion. In our study, microarray was performed to detect the expression profiles of lncRNA, mRNA, circRNA and miRNA at different stages during osteoclastogenesis of RAW264.7 cells. Often changed RNAs were selected and clustered among the four groups with Venn analysis. The results revealed that expressions of 518 lncRNAs, 207 mRNAs, 24 circRNAs and 37 miRNAs were often altered at each stage during OC differentiation. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) biological pathway analysis were performed to predict the functions of differentially expressed lncRNAs and co-expressed potential targeting genes. Co-expression networks of lncRNA-mRNA and circRNA-miRNA were constructed based on the correlation analysis between the differentially expressed RNAs. The present study provided a systematic perspective on the potential function of non-coding RNAs (ncRNAs) during osteoclastogenesis.


Assuntos
Diferenciação Celular/fisiologia , Regulação da Expressão Gênica/fisiologia , MicroRNAs , Osteoclastos/metabolismo , RNA Longo não Codificante , RNA Mensageiro , Animais , Linhagem Celular , Perfilação da Expressão Gênica , Camundongos , MicroRNAs/biossíntese , MicroRNAs/genética , Análise de Sequência com Séries de Oligonucleotídeos , Osteoclastos/citologia , RNA Longo não Codificante/biossíntese , RNA Longo não Codificante/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética
14.
ACS Appl Mater Interfaces ; 8(7): 4489-99, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26824825

RESUMO

Insufficient blood perfusion is one of the critical problems that hamper the clinical application of tissue engineering bone (TEB). Current methods for improving blood vessel distribution in TEB mainly rely on delivering exogenous angiogenic factors to promote the proliferation, migration, differentiation, and vessel formation of endothelial cells (ECs) and/or endothelial progenitor cells (EPCs). However, obstacles including limited activity preservation, difficulty in controlled release, and high cost obstructed the practical application of this strategy. In this study, TEB scaffold were modified with cerium oxide nanoparticles (CNPs) and the effects of CNPs existed at the scaffold surface on the growth and paracrine behavior of mesenchymal stem cells (MSCs) were investigated. The CNPs could improve the proliferation and inhibit the apoptosis of MSCs. Meanwhile, the interaction between the cell membrane and the nanoparticle surface could activate the calcium channel of MSCs leading to the rise of intracellular free Ca(2+) level, which subsequently augments the stability of HIF-1α. These chain reactions finally resulted in high expression of angiogenic factor VEGF. The improved paracrine of VEGF could thereby promote the proliferation, differentiation, and tube formation ability of EPCs. Most importantly, in vivo ectopic bone formation experiment demonstrated this method could significantly improve the blood vessel distribution inside of TEB.


Assuntos
Regeneração Óssea/efeitos dos fármacos , Transplante Ósseo/métodos , Células-Tronco Mesenquimais/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos , Animais , Canais de Cálcio/genética , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Cério/administração & dosagem , Cério/química , Células Endoteliais/efeitos dos fármacos , Glicosaminoglicanos , Humanos , Camundongos , Nanopartículas/administração & dosagem , Nanopartículas/química , Engenharia Tecidual , Alicerces Teciduais/química
15.
J Cell Physiol ; 231(3): 558-67, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25545964

RESUMO

Bone homeostasis is maintained by the balance between osteoblastic bone formation and osteoclastic bone resorption. Osteoclasts are multinucleated cells derived from hematopoietic stem cells (HSCs) or monocyte/macrophage progenitor cells and formed by osteoclasts precursors (OCPs) fusion. Cyanidin is an anthocyanin widely distributed in food diet with novel antioxidant activity. However, the effect of cyanidin on osteoclasts is still unknown. We investigated the effect of cyanidin on RANKL-induced osteoclasts differentiation and cell fusion. The results showed that cyanidin had a dual effect on RANKL-induced osteoclastogenesis. Lower dosage of cyanidin (< 1 µg/ml) has a promoting effect on osteoclastogenesis while higher dosage of cyanidin (> 10 µg/ml) has an inhibitory effect. Fusogenic genes like CD9, ATP6v0d2, DC-STAMP, OC-STAMP, and osteoclasts related genes like NFATc1, mitf, and c-fos were all regulated by cyanidin consistent to its dual effect. Further exploration showed that low concentration of cyanidin could increase osteoclasts fusion whereas higher dosage of cyanidin lead to the increase of LXR-ß expression and activation which is suppressive to osteoclasts differentiaton. All these results showed that cyanidin exhibits therapeutic potential in prevention of osteoclasts related bone disorders.


Assuntos
Antocianinas/farmacologia , Osso e Ossos/metabolismo , Diferenciação Celular/efeitos dos fármacos , Osteoclastos/efeitos dos fármacos , Ligante RANK/efeitos dos fármacos , Animais , Antocianinas/metabolismo , Células da Medula Óssea/citologia , Reabsorção Óssea , Fusão Celular , Células Cultivadas , Camundongos , Osteoclastos/citologia , Osteoclastos/metabolismo , Osteogênese/efeitos dos fármacos , Ligante RANK/metabolismo , Transdução de Sinais
16.
Biochim Biophys Acta ; 1839(11): 1084-96, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25123438

RESUMO

DC-STAMP is a key regulating molecule of osteoclastogenesis and osteoclast precursor (OCP) fusion. Emerging lines of evidence showed that microRNAs play crucial roles in bone metabolism and osteoclast differentiation, but no microRNA has yet been reported to be directly related to OCPs fusion. Through a microarray, we found that the expression of miR-7b in RAW264.7 cells was significantly decreased after induction with M-CSF and RANKL. The overexpression of miR-7b in RAW264.7 cells attenuated the number of TRAP-positive cells number and the formation of multinucleated cells, whereas the inhibition of miR-7b enhanced osteoclastogenesis. Through a dual luciferase reporter assay, we confirmed that miR-7b directly targets DC-STAMP. Other fusogenic molecules, such as CD47, ATP6v0d2, and OC-STAMP, were detected to be down-regulated in accordance with the inhibition of DC-STAMP. Because DC-STAMP also participates in osteoclast differentiation through the ITAM-ITIM network, multiple osteoclast-specific genes in the ITAM-ITIM network were detected to identify how DC-STAMP is involved in this process. The results showed that molecules associated with the ITAM-ITIM network, such as NFATc1 and OSCAR, which are crucial in osteoclastogenesis, were consistently altered due to DC-STAMP inhibition. These findings suggest that miR-7b inhibits osteoclastogenesis and cell-cell fusion by directly targeting DC-STAMP. In addition, the inhibition of DC-STAMP and its downstream signals changed the expression of other fusogenic genes and key regulating genes, such as Nfatc1, c-fos, Akt, Irf8, Mapk1, and Traf6. In conclusion, our findings indicate that miR-7b may be a potential therapeutic target for the treatment of osteoclast-related bone disorders.


Assuntos
Diferenciação Celular/genética , Genes fos , Proteínas de Membrana/genética , MicroRNAs/fisiologia , Fatores de Transcrição NFATC/genética , Proteínas do Tecido Nervoso/genética , Osteoclastos/fisiologia , Animais , Fusão Celular , Células Cultivadas , Regulação para Baixo/genética , Genes fos/genética , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Transcrição NFATC/metabolismo , Interferência de RNA/fisiologia , Transdução de Sinais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...