Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 110
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Cell Mol Med ; 26(18): 4745-4755, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35922965

RESUMO

Mesenchymal stem cell (MSC) therapy is considered a new treatment for a wide range of diseases and injuries, but challenges remain, such as poor survival, homing and engraftment rates, thus limiting the therapeutic efficacy of the transplanted MSCs. Many strategies have been developed to enhance the therapeutic efficacy of MSCs, such as preconditioning, co-transplantation with graft materials and gene modification. Hepatocyte growth factor (HGF) is secreted by MSCs, which plays an important role in MSC therapy. It has been reported that the modification of the HGF gene is beneficial to the therapeutic efficacy of MSCs, including diseases of the heart, lung, liver, urinary system, bone and skin, lower limb ischaemia and immune-related diseases. This review focused on studies involving HGF/MSCs both in vitro and in vivo. The characteristics of HGF/MSCs were summarized, and the mechanisms of their improved therapeutic efficacy were analysed. Furthermore, some insights are provided for HGF/MSCs' clinical application based on our understanding of the HGF gene and MSC therapy.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Fator de Crescimento de Hepatócito/genética , Fator de Crescimento de Hepatócito/metabolismo , Hepatócitos/metabolismo , Pulmão/metabolismo , Células-Tronco Mesenquimais/metabolismo
2.
Drug Dev Res ; 83(3): 637-645, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-34725841

RESUMO

Epidermal growth factor receptor (EGFR) is a transmembrane glycoprotein, which has been considered as one of the key targets for cancer therapy. However, currently approved therapeutic anti-EGFR antibody may cause the hypersensitivity reaction induced by galactose-α-1,3-galactose (α-Gal) structure, which is inevitable in insect cell expression system. In this study, the Chinese hamster ovary cell line was used to produce a monoclonal antibody containing simplified glycosylation patterns (code: AB01). And cetuximab was used as a control. The two antibodies were highly similar in molecular weight, secondary structure, binding affinity and endocytosis behavior, whereas the glycotypes are extremely distinct. The flow cytometry assay suggested that AB01 induced cell cycle arrest in G1, thus inhibit cell proliferation. Moreover, both cetuximab and AB01 showed similar sensitivity for all tested cell lines in this research. In conclusion, AB01 could be a potential anti-EGFR drug candidate for cancer therapy.


Assuntos
Antineoplásicos , Neoplasias , Animais , Anticorpos Monoclonais Humanizados/farmacologia , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Células CHO , Linhagem Celular Tumoral , Cetuximab/farmacologia , Cricetinae , Cricetulus , Galactose , Neoplasias/tratamento farmacológico
3.
J Thromb Thrombolysis ; 52(3): 880-888, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33826053

RESUMO

In the present study, bifunctional fusion proteins were designed by fusing the kringle 2 and protease domains of tissue-type plasminogen activator (tPA) to the C-terminal fragment of hirudin. The thrombolytic and anticoagulant activities of these recombinant proteins from mammalian cells were investigated using in vitro coagulation models and chromogenic assays. The results showed that all assayed tPA mutants retained catalytic activity. The C-terminal fragment of hirudin may have weak affinity to thrombin and thus was insufficient to suppress thrombin-mediated fibrin agglutination. The strength of the thrombolytic activity only relied on the selected tPA sequences, and the fibrinolytic efficiency of single-chain protein significantly decreased. Our data indicate that truncated tPA combined with a hirudin peptide may provide a framework for the further development of a new antithrombotic agent.


Assuntos
Hirudinas , Ativador de Plasminogênio Tecidual , Animais , Fibrinólise/efeitos dos fármacos , Fibrinolíticos/farmacologia , Hirudinas/farmacologia , Proteínas Recombinantes/farmacologia , Trombina/farmacologia , Ativador de Plasminogênio Tecidual/genética , Ativador de Plasminogênio Tecidual/farmacologia
4.
Int J Mol Sci ; 22(23)2021 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-34884691

RESUMO

Acute liver injury shares a common feature of hepatocytes death, immune system disorders, and cellular stress. Hepassocin (HPS) is a hepatokine that has ability to promote hepatocytes proliferation and to protect rats from D-galactose (D-Gal)- or carbon tetrachloride (CCl4)-induced liver injury by stimulating hepatocytes proliferation and preventing the high mortality rate, hepatocyte death, and hepatic inflammation. In this paper, we generated a pharmaceutical-grade recombinant human HPS using mammalian cells expression system and evaluated the effects of HPS administration on the pathogenesis of acute liver injury in monkey and mice. In the model mice of D-galactosamine (D-GalN) plus lipopolysaccharide (LPS)-induced liver injury, HPS treatment significantly reduced hepatocyte death and inflammation response, and consequently attenuated the development of acute liver failure. In the model monkey of D-GalN-induced liver injury, HPS administration promoted hepatocytes proliferation, prevented hepatocyte apoptosis and oxidation stress, and resulted in amelioration of liver injury. Furthermore, the primary pharmacokinetic study showed natural HPS possesses favorable pharmacokinetics; the acute toxicity study indicated no significant changes in behavioral, clinical, or histopathological parameters of HPS-treated mice, implying the clinical potential of HPS. Our results suggest that exogenous HPS has protective effects on acute liver injury in both mice and monkeys. HPS or HPS analogues and mimetics may provide novel drugs for the treatment of acute liver injury.


Assuntos
Fibrinogênio/uso terapêutico , Falência Hepática Aguda/prevenção & controle , Animais , Células CHO , Cricetulus , Citocinas/sangue , Avaliação Pré-Clínica de Medicamentos , Fibrinogênio/biossíntese , Fibrinogênio/farmacocinética , Fibrinogênio/toxicidade , Galactosamina , Humanos , Lipopolissacarídeos , Macaca fascicularis , Masculino , Camundongos Endogâmicos BALB C , Estresse Oxidativo , Distribuição Aleatória , Ratos Sprague-Dawley , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/uso terapêutico , Proteínas Recombinantes/toxicidade , Testes de Toxicidade Aguda
5.
Biochem Biophys Res Commun ; 526(2): 431-438, 2020 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-32228887

RESUMO

The mRNA precursor 3'-end modification factor NUDT21 is a major regulator of 3'UTR shortening and an important component of pre-mRNA cleavage and polyadenylation. However, its role in pathologic progress of small cell lung cancer (SCLC) remains unclear. In this study, we observed that NUDT21 expression is downregulated in SCLC tissues. Hypoxia-induced down-regulation of NUDT21 through HIF-1α. NUDT21 shRNA transduction promotes proliferation and inhibits apoptosis of A549 cells. NUDT21 inhibition also promotes tumor growth in a mouse xenograft model. Furthermore, we clarified that HIF-1α mediated NUDT21 downregulation which altered the expression patterns of two isoforms of GLS1, GAC and KGA. These results link the hypoxic tumor environments to aberrant glutamine metabolism which is important for cellular energy in SCLC cells. Therefore, NUDT21 could be considered as a potential target for the treatment of SCLC.


Assuntos
Fator de Especificidade de Clivagem e Poliadenilação/metabolismo , Glutaminase/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Splicing de RNA/genética , Carcinoma de Pequenas Células do Pulmão/genética , Carcinoma de Pequenas Células do Pulmão/patologia , Células A549 , Proliferação de Células/genética , Células Cultivadas , Glutaminase/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , MicroRNAs/genética , Poliadenilação , Carcinoma de Pequenas Células do Pulmão/metabolismo
6.
Biochem Biophys Res Commun ; 515(3): 448-454, 2019 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-31160087

RESUMO

Endothelial cell death is linked to vascular diseases such as atherosclerosis and tissue ischemia. miRNA-17-92 (miR-17-92) is a multiple functional oncogenic miRNA cluster which plays vital roles in tumor angiogenesis and tissue development. However, its role in regulation of endothelial cell ferroptosis remains unclear. In this study, we revealed that miR-17-92 protects endothelial HUVEC cells from erastin-induced ferroptosis. miR-17-92 overexpression significantly reduced erastin-induced growth inhibition and ROS generation of HUVEC cells. Furthermore, Zinc lipoprotein A20, a validated target of miR-17-92, was identified as a novel regulator of endothelial cell ferroptosis. Lentivirus mediated A20 overexpression increased ROS generation and enhanced erastin-induced ferroptosis, whereas A20 knockdown inhibited erastin-induced ferroptosis. Mechanistic studies revealed that erastin-induced ferroptosis is associated with GPX4 downregulation and ACSL4 upregulation. miR-17-92 overexpression or A20 inhibition increased the ACSL4 expression in HUVEC cells. A20 was identified to directly with and regulate ACSL4 expression by immunoprecipitation. It suggests that the A20-ACSL4 axis plays important roles in erastin-induced endothelial ferroptosis. In conclusion, this study revealed a novel mechanism through which miR-17-92 protects endothelial cells from erastin-induced ferroptosis by targeting the A20-ACSL4 axis.


Assuntos
Coenzima A Ligases/metabolismo , Citoproteção , Ferroptose/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , MicroRNAs/metabolismo , Piperazinas/farmacologia , Transdução de Sinais , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/metabolismo , Proliferação de Células/efeitos dos fármacos , Citoproteção/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , MicroRNAs/genética , Transdução de Sinais/efeitos dos fármacos
7.
Cell Immunol ; 331: 49-58, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29935762

RESUMO

Identifying targets for chimeric antigen receptor-modulated T lymphocyte (CAR-T) therapy against solid tumors is an urgent problem to solve. In this study, we showed for the first time that the receptor tyrosine kinase, AXL, is overexpressed in various tumor cell lines and patient tumor tissues including triple negative breast cancer (TNBC) cell lines and patient samples, making AXL a potent novel target for cancer therapy, specifically for TNBC treatment. We also engineered T cells with a CAR consisting of a novel single-chain variable fragment against AXL and revealed its antigen-specific cytotoxicity and ability to release cytokines in a TNBC cell line and other AXL-positive tumors in vitro. Furthermore, AXL-CAR-T cells displayed a significant anti-tumor effect and in vivo persistence in a TNBC xenograft model. Taken together, our findings indicate that AXL-CAR-T cells can represent a promising therapeutic strategy against TNBC.


Assuntos
Imunoterapia Adotiva/métodos , Proteínas Proto-Oncogênicas/imunologia , Receptores Proteína Tirosina Quinases/imunologia , Receptores de Antígenos Quiméricos/imunologia , Neoplasias de Mama Triplo Negativas/terapia , Animais , Linhagem Celular Tumoral , Células Cultivadas , Feminino , Humanos , Células MCF-7 , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/metabolismo , Receptores de Antígenos Quiméricos/genética , Receptores de Antígenos Quiméricos/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Neoplasias de Mama Triplo Negativas/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto , Receptor Tirosina Quinase Axl
8.
Cell Commun Signal ; 16(1): 62, 2018 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-30241478

RESUMO

BACKGROUND: TEM8 is a cell membrane protein predominantly expressed in tumor endothelium, which serves as a receptor for the protective antigen (PA) of anthrax toxin. However, the physiological ligands for TEM8 remain unknown. RESULTS: Here we identified uPA as an interacting partner of TEM8. Binding of uPA stimulated the phosphorylation of TEM8 and augmented phosphorylation of EGFR and ERK1/2. Finally, TEM8-Fc, a recombinant fusion protein comprising the extracellular domain of human TEM8 linked to the Fc portion of human IgG1, efficiently abrogated the interaction between uPA and TEM8, blocked uPA-induced migration of HepG2 cells in vitro and inhibited the growth and metastasis of human MCF-7 xenografts in vivo. uPA, TEM8 and EGFR overexpression and ERK1/2 phosphorylation were found co-located on frozen cancer tissue sections. CONCLUSIONS: Taken together, our data provide evidence that TEM8 is a novel receptor for uPA, which may play a significant role in the regulation of tumor growth and metastasis.


Assuntos
Receptores ErbB/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores de Superfície Celular/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Proliferação de Células , Humanos , Cinética , Proteínas dos Microfilamentos , Metástase Neoplásica , Fosforilação , Domínios Proteicos , Receptores de Ativador de Plasminogênio Tipo Uroquinase/química , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/química
9.
J Asian Nat Prod Res ; 20(4): 385-390, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28617053

RESUMO

A novel glucoside bletilloside A (1) was isolated from the tubers of Bletilla striata, together with seven known compounds (2-8). Their structures were determined on the basis of extensive spectroscopic analyses. All compounds were evaluated for the inhibition on NO production effects in RAW 264.7 macrophage cells, while militarine (4) and dactylorhin A (5) exhibited moderate inhibitory effects.


Assuntos
Bibenzilas/isolamento & purificação , Bibenzilas/farmacologia , Glucosídeos/isolamento & purificação , Glucosídeos/farmacologia , Orchidaceae/química , Animais , Bibenzilas/química , Glucosídeos/química , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Camundongos , Estrutura Molecular , Óxido Nítrico/biossíntese , Tubérculos/química
10.
Biochem Biophys Res Commun ; 487(4): 868-874, 2017 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-28461114

RESUMO

miR-17-92 cluster are overexpressed in hematological malignancies including chronic myeloid leukemia (CML). However, their roles and mechanisms that regulate BCR-ABL induced leukemogenesis remain unclear. In this study, we demonstrated that genomic depletion of miR-17-92 inhibited the BCR-ABL induced leukemogenesis by using a mouse model of transplantation of BCR-ABL transduced hematopoietic stem cells. Furthermore, we identified that miR-19b targeted A20 (TNFAIP3). A20 overexpression results in inactivation of NF-κB activity including decrease of phosphorylation of P65 and IκBα, leads to induce apoptosis and inhibit proliferation and cycle in CML CD34 + cells. Thus we proved that miR-17-92 is a critical contributor to CML leukemogenesis via targeting A20 and activation of NF-κB signaling. These findings indicate that miR-17-92 will be important resources for developing novel treatment strategies of CML and better understanding long-term disease control.


Assuntos
Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , MicroRNAs/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/metabolismo , Animais , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias Experimentais/genética , Neoplasias Experimentais/metabolismo , RNA Longo não Codificante
11.
Int J Mol Sci ; 18(4)2017 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-28417919

RESUMO

In advanced prostate cancer, small ubiquitin-like modifier (SUMO)-specific cysteine protease 1 (SENP1) is up-regulated. However, the role of SENP1 in regulating deSUMOylation of TGF-ß/SMADs signaling is unknown. In this study, we developed a lentiviral vector, PLKO.1-shSENP1, to silence SENP1 in prostate cancer cells with high metastatic characteristics (PC3M). Likewise, we also created an adenovirus vector, Ad5/F11p-SENP1 to over-express SENP1 in prostate cancer cells with low metastatic potential (LNCaP). We showed that silencing of SENP1 promoted cellular apoptosis, and inhibited proliferation and migration of PC3M cells. Moreover, SENP1 silencing increased the SMAD4 expression at protein level, up-regulated E-cadherin and down-regulated Vimentin expression, indicating the inhibition of epithelial mesenchymal transition (EMT). Furthermore, SMAD4 interference abolished SENP1-mediated up-regulation of E-cadherin, suggesting that SENP1 regulated E-cadherin expression via SMAD4. SENP1 over-expression in LNCaP cells reduced SMAD4 protein, and promoted EMT via decreasing E-cadherin and increasing Vimentin. Moreover, down-regulation of SMAD4 and E-cadherin were blocked, after transfection with two SUMOylation sites mutated SMAD4, suggesting that SENP1 might reduce SMAD4 levels to regulate E-cadherin expression via deSUMOylation of SMAD4. In conclusion, SENP1 deSUMOylated SMAD4 to promote EMT via up-regulating E-cadherin in prostate cancer cells. Therefore, SENP1 is a potential target for treatment of advanced prostate cancer.


Assuntos
Endopeptidases/metabolismo , Transição Epitelial-Mesenquimal , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Proteína Smad4/metabolismo , Androgênios , Apoptose/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Cisteína Endopeptidases , Transição Epitelial-Mesenquimal/genética , Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Humanos , Masculino , Neoplasias da Próstata/genética , RNA Interferente Pequeno/genética , Transdução de Sinais , Sumoilação , Fator de Crescimento Transformador beta/metabolismo
12.
Tumour Biol ; 37(6): 7741-8, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26695141

RESUMO

The deregulation of HGF/c-Met signaling is implicated in epithelial-mesenchymal transition (EMT) and progress of hepatocellular carcinoma (HCC). However, the epigenetic mechanisms that HGF/c-Met regulates EMT and metastasis of HCC cells are less explored. In this study, we demonstrated that HCC cells express a high level of SUMO/sentrin-specific protease 1 (Senp1) which is induced by HGF/c-Met signals. Lentivirus-mediated small hairpin RNA (shRNA) transduction results in Senp1 silence in HCC cells. Senp1 silence reduces the HGF-induced proliferation and migration of HCC cells. Senp1 inhibition also induces HCC cell apoptosis and growth arrest. Furthermore, Senp1 knockdown inhibits epithelial-to-mesenchymal transition, with increase of E-cadherin and ZO-1 expression, decrease of fibronectin and N-cadherin expression. The EMT-related transcription factor Zeb1 was SUMO-modified and decreased in Senp1-silenced HCC cells. These results delineate that senp1 might play an important role in the regulation of HGF-induced invasion and migration of HCC cells.


Assuntos
Carcinoma Hepatocelular/patologia , Endopeptidases/fisiologia , Transição Epitelial-Mesenquimal/genética , Neoplasias Hepáticas/patologia , Proteínas de Neoplasias/fisiologia , Apoptose , Carcinoma Hepatocelular/genética , Moléculas de Adesão Celular/biossíntese , Moléculas de Adesão Celular/genética , Pontos de Checagem do Ciclo Celular , Divisão Celular , Linhagem Celular Tumoral , Movimento Celular , Cisteína Endopeptidases , Endopeptidases/genética , Fator de Crescimento de Hepatócito/fisiologia , Humanos , Neoplasias Hepáticas/genética , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas c-met/fisiologia , Interferência de RNA , RNA Interferente Pequeno/genética , Transdução de Sinais/genética , Sumoilação , Homeobox 1 de Ligação a E-box em Dedo de Zinco/metabolismo
13.
Biochem Biophys Res Commun ; 460(2): 409-15, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25791478

RESUMO

SUMO/sentrin specific protease 1 (Senp1) is an important regulation protease in the protein sumoylation, which affects the cell cycle, proliferation and differentiation. The role of Senp1 mediated protein desumoylation in pathophysiological progression of multiple myeloma is unknown. In this study, we demonstrated that Senp1 is overexpressed and induced by IL-6 in multiple myeloma cells. Lentivirus-mediated Senp1 knockdown triggers apoptosis and reduces viability, proliferation and colony forming ability of MM cells. The NF-κB family members including P65 and inhibitor protein IkBα play important roles in regulation of MM cell survival and proliferation. We further demonstrated that Senp1 inhibition decreased IL-6-induced P65 and IkBα phosphorylation, leading to inactivation of NF-кB signaling in MM cells. These results delineate a key role for Senp1in IL-6 induced proliferation and survival of MM cells, suggesting it may be a potential new therapeutic target in MM.


Assuntos
Apoptose , Divisão Celular , Endopeptidases/metabolismo , Mieloma Múltiplo/patologia , NF-kappa B/metabolismo , Transdução de Sinais , Linhagem Celular Tumoral , Cisteína Endopeptidases , Endopeptidases/genética , Técnicas de Silenciamento de Genes , Inativação Gênica , Células HEK293 , Humanos , Mieloma Múltiplo/metabolismo
14.
Cytotherapy ; 17(7): 874-84, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25800776

RESUMO

BACKGROUND AIMS: Multipotent mesenchymal stromal cells (MSCs) are promising candidates for innovative cell therapeutic applications. Before their use, however, they usually need to be expanded in vitro with serum-supplemented media. MSCs can undergo replicative senescence during in vitro expansion, but it is not yet clear how serum supplements influence this process. METHODS: In the present study, we compared how media supplemented with fetal bovine serum (FBS) or calf serum (CS) affected morphology, proliferation, differentiation, senescence and other functional characteristics of human umbilical cord-derived MSCs (UC-MSCs). RESULTS: UC-MSCs cultured in both FBS- and CS-containing media were able to differentiate along osteogenic and adipogenic lineages but ultimately reached proliferation arrest. However, senescence-associated characteristics, such as ß-galactosidase activity, reactive oxygen species levels, proliferation rate and gene expression, demonstrate that UC-MSCs grown with FBS have better proliferation potential and differentiation capacity. In contrast, UC-MSCs grown with CS have a higher proportion of apoptotic cells and senescent characteristics. Possible mechanisms for the observed phenotypes include changes in gene expression (Bax, p16, p21 and p53) and cytokine production (interleukin-6 and interleukin-8). CONCLUSIONS: This study demonstrates that FBS-supplemented media provides a better microenvironment for the expansion of UC-MSCs in vitro than CS-supplemented media. This work provides insight into MSCs generation practices for use in basic research and clinical therapies.


Assuntos
Técnicas de Cultura de Células/métodos , Senescência Celular/efeitos dos fármacos , Meios de Cultura/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Bovinos , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Humanos , Interleucina-6/biossíntese , Interleucina-8/biossíntese , Células-Tronco Mesenquimais/citologia , Espécies Reativas de Oxigênio/metabolismo , Soro , Cordão Umbilical/citologia , beta-Galactosidase/metabolismo
15.
Hepatol Res ; 45(13): 1312-22, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25655186

RESUMO

AIM: miRNA-486 (miR-486) was first identified from the human fetal liver cDNA library and considered to be associated with hepatocellular carcinoma (HCC) development. Its roles in regulation of HCC metastasis and chemosensitivity have not been explored yet. METHODS: miR-486 expression in HCC tissues, cell lines and serum was evaluated by real-time polymerase chain reaction. miR-486 overexpression or downregulation in the cell lines SMMC-7721/LM3 was conducted by lentivirus transfection. Cell proliferation, migration and apoptosis were quantitated using commercial assays. Matrix metalloproteinase activity was quantitated by gelatin zymography. The target genes of miR-486 were screened by 3'-untranslated region luciferase report assays and their function was validated by small RNA interference. RESULTS: We show here that miR-486 is frequently down-expressed in HCC tissues and cell lines. Lentivirus-mediated restoration of miR-486 in HCC cell lines resulted in significant reduction in the ability of cell growth, colony formation and migration. However, miR-486 inhibition enhances proliferation and invasion of HCC cells. Two genes, CITRON and CLDN10 which regulate cell proliferation and invasion, respectively, were identified as the direct targets of miR-486 in HCC cells. CITRON and CLDN10 knockdown by siRNA results in similar phenotypes of miR-486 restoration in HCC cell lines. In addition, miR-486 enhances the chemosensitivity of HCC cells to sorafenib. CONCLUSION: Our data indicated that miR-486 may function as a novel tumor suppressor in HCC.

16.
J Pathol ; 230(4): 365-76, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23839903

RESUMO

The hepatic growth factor hepatopoietin Cn (HPPCn) prevents liver injury induced by carbon tetrachloride in rats. Sphingosine 1-phosphate (S1P) is a bioactive sphingolipid produced by sphingosine kinase (SphK). S1P and S1P receptors (S1PRs) are involved in liver fibrogenesis and oxidative injury. This work sought to understand the mechanism by which SphK/S1P/S1PRs are involved in the protective effects of HPPCn on ethanol-induced liver injury and fibrosis. Transgenic mice with liver-specific overexpression of HPPCn (HPPCn(liver) (+/+)) were generated. Two ethanol feeding protocols were used to assess the protective effect of HPPCn on acute and chronic liver injury in mice. Specific inhibitors of S1PR1, S1PR2 and S1PR3 and siRNA were used to examine the roles of S1PRs in hepatic stellate cell (HSC) activation and hepatocyte apoptosis. Increased HPPCn expression in transgenic mice attenuated fibrosis induced by ethanol and carbon tetrachloride (CCl4). Treatment with recombinant human HPPCn prevented human hepatocyte apoptosis and HSC activation. JTE-013 or S1PR2-siRNA attenuated the effect of HPPCn on HSC activation induced by tumour necrosis factor-α (TNF-α). Consistent with the effect of N,N-dimethylsphingosine (DMS), suramin or S1PR3-siRNA treatment blocked HPPCn-induced Erk1/2 phosphorylation in human hepatocytes. This study demonstrated that HPPCn attenuated oxidative injury and fibrosis induced by ethanol feeding and that the SphK1/S1P/S1PRs signalling pathway contributes to the protective effect of HPPCn on hepatocyte apoptosis and HSC activation.


Assuntos
Etanol , Fator de Crescimento de Hepatócito/metabolismo , Cirrose Hepática Alcoólica/prevenção & controle , Fígado/enzimologia , Proteínas Nucleares/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Animais , Apoptose , Células Cultivadas , Modelos Animais de Doenças , Regulação da Expressão Gênica , Células Estreladas do Fígado/enzimologia , Células Estreladas do Fígado/patologia , Fator de Crescimento de Hepatócito/genética , Humanos , Fígado/efeitos dos fármacos , Fígado/patologia , Cirrose Hepática Alcoólica/enzimologia , Cirrose Hepática Alcoólica/etiologia , Cirrose Hepática Alcoólica/genética , Cirrose Hepática Alcoólica/patologia , Lisofosfolipídeos/metabolismo , Camundongos , Camundongos Transgênicos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteínas Nucleares/genética , Estresse Oxidativo , Fosforilação , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Interferência de RNA , RNA Mensageiro/metabolismo , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Receptores de Lisoesfingolipídeo/genética , Transdução de Sinais , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Fatores de Tempo , Transfecção , Fator de Necrose Tumoral alfa/metabolismo
17.
Bioact Mater ; 34: 204-220, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38235309

RESUMO

Skeletal stem cells (SSC) have gained attentions as candidates for the treatment of osteoarthritis due to their osteochondrogenic capacity. However, the immunomodulatory properties of SSC, especially under delivery operations, have been largely ignored. In the study, we found that Pdpn+ and Grem1+ SSC subpopulations owned immunoregulatory potential, and the single-cell RNA sequencing (scRNA-seq) data suggested that the mechanical activation of microgel carriers on SSC induced the generation of Pdpn+Grem1+Ptgs2+ SSC subpopulation, which was potent at suppressing macrophage inflammation. The microgel carriers promoted the YAP nuclear translocation, and the activated YAP protein was necessary for the increased expression of Ptgs2 and PGE2 in microgels-delivered SSC, which further suppressed the expression of TNF-ɑ, IL-1ß and promoted the expression of IL-10 in macrophages. SSC delivered with microgels yielded better preventive effects on articular lesions and macrophage activation in osteoarthritic rats than SSC without microgels. Chemically blocking the YAP and Ptgs2 in microgels-delivered SSC partially abolished the enhanced protection on articular tissues and suppression on osteoarthritic macrophages. Moreover, microgel carriers significantly prolonged SSC retention time in vivo without increasing SSC implanting into osteoarthritic joints. Together, our study demonstrated that microgel carriers enhanced SSC reprogramming towards immunomodulatory phenotype to regulate macrophage phenotype transformation for effectively osteoarthritic therapy by promoting YAP protein translocation into nucleus. The study not only complement and perfect the immunological mechanisms of SSC-based therapy at the single-cell level, but also provide new insight for microgel carriers in stem cell-based therapy.

18.
World J Stem Cells ; 16(5): 575-590, 2024 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-38817328

RESUMO

BACKGROUND: Atherosclerosis (AS), a chronic inflammatory disease of blood vessels, is a major contributor to cardiovascular disease. Dental pulp stem cells (DPSCs) are capable of exerting immunomodulatory and anti-inflammatory effects by secreting cytokines and exosomes and are widely used to treat autoimmune and inflammation-related diseases. Hepatocyte growth factor (HGF) is a pleiotropic cytokine that plays a key role in many inflammatory and autoimmune diseases. AIM: To modify DPSCs with HGF (DPSC-HGF) and evaluate the therapeutic effect of DPSC-HGF on AS using an apolipoprotein E-knockout (ApoE-/-) mouse model and an in vitro cellular model. METHODS: ApoE-/- mice were fed with a high-fat diet (HFD) for 12 wk and injected with DPSC-HGF or Ad-Null modified DPSCs (DPSC-Null) through tail vein at weeks 4, 7, and 11, respectively, and the therapeutic efficacy and mechanisms were analyzed by histopathology, flow cytometry, lipid and glucose measurements, real-time reverse transcription polymerase chain reaction (RT-PCR), and enzyme-linked immunosorbent assay at the different time points of the experiment. An in vitro inflammatory cell model was established by using RAW264.7 cells and human aortic endothelial cells (HAOECs), and indirect co-cultured with supernatant of DPSC-Null (DPSC-Null-CM) or DPSC-HGF-CM, and the effect and mechanisms were analyzed by flow cytometry, RT-PCR and western blot. Nuclear factor-κB (NF-κB) activators and inhibitors were also used to validate the related signaling pathways. RESULTS: DPSC-Null and DPSC-HGF treatments decreased the area of atherosclerotic plaques and reduced the expression of inflammatory factors, and the percentage of macrophages in the aorta, and DPSC-HGF treatment had more pronounced effects. DPSCs treatment had no effect on serum lipoprotein levels. The FACS results showed that DPSCs treatment reduced the percentages of monocytes, neutrophils, and M1 macrophages in the peripheral blood and spleen. DPSC-Null-CM and DPSC-HGF-CM reduced adhesion molecule expression in tumor necrosis factor-α stimulated HAOECs and regulated M1 polarization and inflammatory factor expression in lipopolysaccharide-induced RAW264.7 cells by inhibiting the NF-κB signaling pathway. CONCLUSION: This study suggested that DPSC-HGF could more effectively ameliorate AS in ApoE-/- mice on a HFD, and could be of greater value in stem cell-based treatments for AS.

19.
Biochem Biophys Res Commun ; 432(2): 370-7, 2013 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-23376714

RESUMO

The WL5 antibody is an anti-colorectal cancer antibody secreted by the WL5 hybridoma clone. Flow cytometric analysis showed that WL5 specifically binds to the HT29 and LS180 colorectal cancer cell lines. Immunohistochemical analysis performed on a tissue microarray demonstrated that the WL5 antibody can be used for the specific and sensitive diagnosis of colorectal carcinoma. Furthermore, WL5 mediated antibody dependent cell-mediated cytotoxicity (ADCC) of tumor cells and exhibited similar antitumor activity to adriamycin (ADM) but avoided the cardiomyopathy and decrease in peripheral white blood cell counts associated with prolonged ADM treatment. The glycoprotein, carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1), was identified as the target antigen of WL5 through immunoprecipitation and mass spectrometric analyses, which might provide a potential biomarker and therapeutic target for colorectal cancer.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Anticorpos Antineoplásicos/imunologia , Anticorpos Antineoplásicos/uso terapêutico , Antígenos de Neoplasias/imunologia , Biomarcadores Tumorais/imunologia , Neoplasias Colorretais/diagnóstico , Neoplasias Colorretais/terapia , Animais , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Antígenos CD/imunologia , Moléculas de Adesão Celular/imunologia , Linhagem Celular Tumoral , Feminino , Humanos , Hibridomas , Masculino , Camundongos , Camundongos Endogâmicos BALB C
20.
Biochem Biophys Res Commun ; 430(2): 598-603, 2013 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-23219815

RESUMO

Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) is a member of the immunoglobulin super family and has been observed to have two paradoxical functions: tumor suppression and the promotion of tumor invasion. In the present study, we discovered that CEACAM1 functions as an adhesion inhibitor and a migration promoter. The CEACAM1 transfected cells, either 293-CEACAM1 or LOVO/trans-CEACAM1, was proved to have lower adhesion rate. Furthermore, HT29/siRNA-CEACAM1 cells had a higher adhesion rate than HT29 cells. These results indicated that CEACAM1 was an inhibitor of cell-matrix adhesion. Additionally, 293-CEACAM1 LOVO/trans-CEACAM1 cells exhibited better motility in a trans-well migration assay. N-cadherin expression levels were positively correlated with CEACAM1 in 293-CEACAM1, LOVO/trans-CEACAM1 and HT29/siRNA-CEACAM1 cells. When blocked by a GC-4 antibody, the adhesive capacities of 293-CEACAM1 and LOVO/trans-CEACAM1 were recovered and the motilities of them were suppressed, which suggested that CEACAM1 functioned through N-cadherin.


Assuntos
Antígenos CD/metabolismo , Caderinas/biossíntese , Moléculas de Adesão Celular/metabolismo , Movimento Celular , Metástase Neoplásica/patologia , Antígenos CD/genética , Adesão Celular , Moléculas de Adesão Celular/genética , Células HEK293 , Células HT29 , Humanos , Modelos Biológicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA