Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Oncol ; 2022: 4181730, 2022.
Article in English | MEDLINE | ID: mdl-35310912

ABSTRACT

Objective: To investigate the effects of LncRNA SNHG1 on the proliferation, migration, and epithelial-mesenchymal transition (EMT) of colorectal cancer cells (CRCs). Methods: 4 pairs of CRC tissue samples and their corresponding adjacent samples were analyzed by the human LncRNA microarray chip. The expression of LncSNHG1 in CRC cell lines was verified by qRT-PCR. Colony formation assays and CCK8 assays were applied to study the changes in cell proliferation. The transwell assay and wound healing experiments were used to verify the cell invasion and migration. EMT progression was confirmed finally. Results: LncSNHG1 was overexpressed both in CRC tissues and cell lines, while the miR-181b-5p expression was decreased in CRC cell lines. After knock-down of LncSNHG1, the proliferation, invasion, and migration of HT29 and SW620 cells were all decreased. Meanwhile, LncSNHG1 enhanced EMT progress through regulation of the miR-181b-5p/SMAD2 axis. Conclusion: LncSNHG1 promotes colorectal cancer cell proliferation and invasion through the miR-181b-5p/SMAD2 axis.

2.
J Control Release ; 341: 828-843, 2022 01.
Article in English | MEDLINE | ID: mdl-34942304

ABSTRACT

The development of new reagents combining with nanotechnology has become an efficient strategy for improving the immune escaping ability and increasing local drug concentration for natural compounds with low therapy efficiency. In this study, we prepared biomimetic membrane-coated Prussian blue nanoparticles (PB NPs) for the treatment of atherosclerosis, using the function of Artemisinin (ART) and Procyanidins (PC) on the lipid influx and cholesterol efflux of macrophages, two logical steps involved in the plaque progression. In vitro results indicated that the prepared nanocomplexes have significant scavenging effect on ROS and NO, followed by inhibiting NF-κB/NLRP3 pathway, leading to the suppression of lipid influx. Meanwhile, they can notably reduce the uptake and internalization of oxLDL through significantly enhancing AMPK/mTOR/autophagy pathway, accompanied by promoting cholesterol efflux. In vivo study showed that the improved biocompatibility and immune-escape ability of nanocomplexes allowed less drug clearance during the circulation and high drug accumulation in the atherosclerotic plaque of ApoE-/- mice model. More importantly, the ART and PC co-loaded nanocomplexes showed the high efficacy against atherosclerosis of ApoE-/- mice model with both 8-week low dosage treatment or 1-week high dosage treatment. These findings indicated that ART and PC co-loaded nanocomplexes was promising for the targeted treatment of atherosclerosis.


Subject(s)
Artemisinins , Atherosclerosis , Plaque, Atherosclerotic , Proanthocyanidins , Animals , Artemisinins/therapeutic use , Atherosclerosis/drug therapy , Atherosclerosis/metabolism , Cholesterol/metabolism , Mice , Plaque, Atherosclerotic/drug therapy , Proanthocyanidins/therapeutic use
3.
J Control Release ; 339: 259-273, 2021 11 10.
Article in English | MEDLINE | ID: mdl-34597747

ABSTRACT

A pro-nanodrug combinational strategy for efficient cervical cancer therapy with intrinsic tumor microenvironment (TME)-responsive elements and low side effects is highly desired. Here, a pro-nanodrug complexes with GSH and NIR responsive manner is reported to boost gamabufotalin induced chemo-photothermal therapy with the assistance of reprogrammed TME by indomethacin. In addition, hybrid cell membrane was used to endow nanocomplexes with the prolonging circulation time and high accumulation of drug at tumor tissue. Indomethacin activated by the high level GSH can attenuate tumor inflammation microenvironment triggered by PTT and sensitize tumor cells to gamabufotalin through inhibiting PGE2 secretion. The released low-dose gamabufotalin with low side effects can efficiently kill tumor cells by ROS production and COX-2 low expression. In vitro and in vivo assays demonstrated that strong anti-tumor activity of nanocomplexes in tumor-bearing mice through chemo-photothermal therapy, which was reflected by the eradication of cervical tumor and significant extension of survival time of mice.


Subject(s)
Hyperthermia, Induced , Nanoparticles , Uterine Cervical Neoplasms , Animals , Biomimetics , Doxorubicin , Female , Humans , Indomethacin , Mice , Phototherapy , Photothermal Therapy , Tumor Microenvironment , Uterine Cervical Neoplasms/therapy
4.
Acta Biomater ; 113: 554-569, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32569637

ABSTRACT

As a breast cancer subtype with high mortality in women, the efficient treatment of Triple-negative breast cancer (TNBC) is still a challenge due to its unique metastatic mode and poor prognosis. In this study, we developed a biomimetic nanodelivery system (denoted as GTDC@M-R NPs) based on erythrocyte membrane (M)-camouflaged graphene oxide quantum dots (GOQDs, G) for TNBC therapy. The TAT (T) and RGD (R) peptides were used to endow targeting accumulation ability of Gamabufotalin (CS-6, C) and doxorubicin (DOX, D) in tumor tissue. In vitro assay indicated good biocompatibility, prolonged blood circulation time (3-fold longer than GT NPs), and effectively enhanced cell and nucleus targeting capability of this nanosystem. Fluorescence activated cell sorter (FACS) analysis indicated that the combination of DOX and CS-6 induced TNBC cell apoptosis more than 89 % under the ratio of 10:1. In vivo assay indicated that the accumulation of GTDC@M-R NPs in tumor sites increased about 2-fold compared to naked GTDC NPs, which was accompanied by high tumor apoptosis rates through blocking chemotherapy-activated cyclooxygenase-2 (COX-2) and enhancing DOX's anti-tumor activity of chemical drugs (85%). Moreover, comparing with the control, the average number of lung metastatic nodules in tumor-bearing mice reduced 84%, the molecular mechanism of which is related to the down expression of COX-2, matrix metalloproteinase 9 (MMP9) and vascular endothelial growth factor (VEGF). Taken together, our results proved that the developed GTDC@M-R NPs can inhibit the growth and suppress metastasis of TNBC, which broaden our insights into the application of combinational strategy for efficient TNBC therapy. STATEMENT OF SIGNIFICANCE: In this study, we developed a biomimetic nanodelivery system (denoted as GTDC@M-R NPs) based on erythrocyte membrane (M)-camouflaged graphene oxide quantum dots (GOQDs, G) for TNBC therapy. The TAT (T) and RGD (R) peptides were used to endow targeting accumulation ability of Gamabufotalin (CS-6, C) and doxorubicin (DOX, D) in tumor tissue. These GTDC@M-R NPs indicated synergistic chemotherapy against TNBC cells through the precise cell and nuclear targeting, immune escape, and improved DOX sensitivity. A effective inhibition of tumor growth and metastasis was achieved by inhibiting Bcl-2/BAX, COX-2 and VEGF related signal pathways. Our finding suggests that the developed GTDC@M-R NPs present great treating effects in the preclinical models of TNBC, which broaden our insights into the application of combinational strategy for efficient TNBC therapy.


Subject(s)
Lung Neoplasms , Nanoparticles , Triple Negative Breast Neoplasms , Animals , Biomimetics , Cell Line, Tumor , Doxorubicin/pharmacology , Humans , Lung Neoplasms/drug therapy , Mice , Triple Negative Breast Neoplasms/drug therapy , Vascular Endothelial Growth Factor A
5.
Exp Cell Res ; 376(2): 114-123, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30763586

ABSTRACT

Relapse of minimal residual disease (MRD) is a major problem after conventional chemotherapy in patients with acute myeloid leukemia (AML). The bone marrow stroma can protect AML cells from insults of chemotherapy, partly contributing to AML relapse. Arsenic trioxide (ATO) is the main component of arsenical traditional Chinese medicines and has been widely used for the treatment of hematologic malignancies particularly acute promyelocytic leukemia over the past three decades. ATO acts through a direct arsenic binding to cysteine residues in zinc fingers located in promyelocytic leukemia protein (PML), thus killing the leukemia stem cells (LSCs). Our prior study has demonstrated that adhesion to stroma cells could render AML cells resistant to ATO but the detailed mechanism remains to be explored. Here, we report that the adhesion-induced resistance to ATO is related to the up-regulation of myeloid cell leukemia-1 (Mcl-1). Homoharringtonine (HHT) can potentiate the anti-leukemia effects of ATO on adhered AML cells by suppressing Mcl-1 through glycogen synthase kinase-3ß (GSK3ß). Furthermore, a potentiating effect of HHT on ATO was also observed in primary AML cells and AML xenografted tumors. Thus, these data indicate that HHT could enhance ATO anti-leukemia activity both in vitro and in vivo.


Subject(s)
Antineoplastic Agents/therapeutic use , Arsenic Trioxide/therapeutic use , Homoharringtonine/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Adult , Aged , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Adhesion , Cell Line, Tumor , Coculture Techniques , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Drug Synergism , Female , Humans , Male , Mice, SCID , Middle Aged , Xenograft Model Antitumor Assays , Young Adult
6.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 34(4): 346-353, 2018 Apr.
Article in Chinese | MEDLINE | ID: mdl-29973326

ABSTRACT

Objective To investigate the molecular mechanisms underlying the effect of homoharringtonine (HHT) on the proliferation and apoptosis in HT29 human colorectal tumor cells. Methods HT29 cells were treated by 0, 0.007812, 0.015625, 0.03125, 0.0625, 0.125, 0.25, 0.5, 1, 2, 4, 8 µg/mL HHT for 24, 48 and 72 hours. CCK-8 assay was used to assess the cell viability. Colony formation assay was performed to detect the cell proliferation ability. Flow cytometry was used to analyze cell apoptosis. Hoechst33258 fluorescent staining was used to observe the morphology of the cell nuclei. The real-time quantitative PCR was used to detect the mRNA expressions of BAX, Bcl2, caspase-3, caspase-9, mammalian target of rapamycin (mTOR), PI3K, pyruvate dehydrogenase kinase-1 (PDK1), protein kinase B (AKT), raptor, rictor. The protein levels of Bax, Bcl2, pro-caspase-3, cleaved caspase 3 (c-caspase-3), pro-caspase-9, cleaved caspase-9 (c-caspase- 9), poly(ADP-ribose)polymerase (PARP), cleaved PARP (c-PARP), mTOR, raptor, rictor, PI3K, PDK1, AKT, p-AKT were detected by Western blotting. Results Compared with the control group, the proliferation of HT29 cells was inhibited when treated with HHT. Meanwhile, the nuclear fragmentation, chromatin condensation, and apoptotic body of the cells could be observed. Treatment of HHT could increase the mRNA expressions of BAX/Bcl2, caspase-3, caspase-9 and raptor, and decrease PI3K, AKT and rictor in the HT29 cells. The protein levels of pro-caspase-3, pro-caspase-9, PARP, PI3K, PDK1, AKT, mTOR, and rictor were down-regulated, and the c-caspase-3, c-caspase-9, c-PARP, BAX and raptor were up-regulated. Conclusion HHT has the function of inhibiting the HT29 cell proliferation and inducing its apoptosis by blockage of mTOR signaling pathway.


Subject(s)
Apoptosis , Cell Proliferation , Colorectal Neoplasms , Homoharringtonine/pharmacology , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , HT29 Cells , Humans , Phosphatidylinositol 3-Kinases , Sirolimus
7.
Leuk Res ; 57: 119-126, 2017 06.
Article in English | MEDLINE | ID: mdl-28463768

ABSTRACT

Despite the great progress in the treatment, multiple myeloma (MM) still remains incurable. Bortezomib (BTZ), a reversible inhibitor of the 26S proteasome, is very effective against MM but unable to eradicate the MM cells in bone marrow niche eventually causing the disease relapse. Homoharringtonine (HHT) is a known anti-leukemia drug that inhibits MM both in vitro and in vivo. This study aimed to investigate whether HHT could potentiate the anti-tumor activity of BTZ in MM cells cocultured with bone marrow stromal cells and in vivo xenograft models. We found that coculture of myeloma cells with a human stroma cell line significantly decreased the sensitivity of myeloma cells to BTZ treatment. HHT inhibited the proliferation of MM cells and potentiated the anti-myeloma effects of BTZ by inhibition of both canonical and noncanonical NF-κB pathways. HHT also enhanced the anti-myeloma effect of BTZ in vivo xenograft models. Taken together, our data suggest that HHT can enhance the anti-myeloma activity of BTZ both in vitro and in vivo, which may represent a new clinical treatment in MM.


Subject(s)
Bortezomib/pharmacology , Harringtonines/pharmacology , Multiple Myeloma/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Bone Marrow , Bortezomib/therapeutic use , Cell Adhesion/drug effects , Coculture Techniques , Drug Synergism , Harringtonines/therapeutic use , Heterografts , Homoharringtonine , Humans , Mice , Mice, SCID , Multiple Myeloma/pathology , Stromal Cells/pathology
8.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 25(1): 50-54, 2017 Feb.
Article in Chinese | MEDLINE | ID: mdl-28245374

ABSTRACT

OBJECTIVE: To detect the expression of MCL-1 in patient with acute myeloid leukemia(AML) with normal karyotype and to investigate the relationship of its expression level with clinical parameters and prognosis. METHODS: The expression of MCL-1 in the bone marrow from 37 newly diagnosed AML patients was measured by real-time fluorescent quantitative PCR(FQ RT-PCR) and Western blot, and the relationship between its expression level and clinical parameters such as the age, sex, WBC count, Hb level, Plt count, blast ratio in BM, and sensitivity to chemotherapeutic drugs in vitro prognosis was analyzed. RESULTS: The expression level of MCL-1 did not correlate with the age, sex, WBC count, Hb level, Plt count and the percentage of blast cells. There was no significant difference of MCL-1 expression in AML patients with or without extramedullary infiltration. The higher level of MCL-1 existed in AML patients who did not achieve complete remission with classical regimen. The resistant rate to chemotherapeutic drugs in vitro was higher in the patients with higher level of MCL-1. CONCLUSION: Overexpression of MCL-1 is closely related with the resistance to chemotherapeutic drugs, and may be used as an early indicator for judging multi-drug resistance and prognosis of AML.


Subject(s)
Leukemia, Myeloid, Acute/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Bone Marrow/metabolism , Drug Resistance, Neoplasm/genetics , Humans , Leukemia, Myeloid, Acute/drug therapy , Prognosis , Remission Induction
9.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 24(6): 1649-1653, 2016 Dec.
Article in Chinese | MEDLINE | ID: mdl-28024471

ABSTRACT

OBJECTIVE: To investigate the effects and mechanisms of the combination of homoharringtonine (HHT) with arsenic trioxide(As2O3) on human myeloid cell line U937 in vitro. METHODS: MTT method was used to determine the antiproliferating effect of different concentrations of HHT, As2O3 and their combination on U937 cells; the flow cytometry with Annexin-V-FITC/PI double staining was used to determine the apoptosis-induced effect of HHT and As2O3 alone or their combination; Western blot method was used to detect the protein expression of P-AktSer473,P-AktThr308,BCL-XL, BID,MCL-1,P-MCL-1 and so on. RESULTS: HHT and As2O3 could significantly inhibit proliferation of U937 cells and induce their apoptosis. The combination of these 2 drugs could significantly enhance the early apoptosis of U937 cells. After combination of these 2 drugs was used, the protein expressions of P-AktSer473,P-AktThr308,MCL-1,P-MCL-1 and BCL-XL were obviously down-regulated, but the expression of BID protein did not change. CONCLUSION: The combination treatment of HHT and As2O3 can synergistically inhibit the growth of U937 cells through inhibition of PI3K/Akt signal way and MCL-1 protein.


Subject(s)
Leukemia, Myeloid, Acute , Antineoplastic Agents , Apoptosis , Arsenic Trioxide , Arsenicals , Harringtonines , Homoharringtonine , Humans , Myeloid Cell Leukemia Sequence 1 Protein , Oxides , Phosphatidylinositol 3-Kinases , U937 Cells
10.
Oncol Res ; 24(4): 215-23, 2016.
Article in English | MEDLINE | ID: mdl-27656831

ABSTRACT

This study aimed to investigate the role of the PI3K/Akt signaling pathway in multidrug resistance of acute myeloid leukemia (AML) cells induced by cocultured stromal cells. Human AML cell lines HL-60 and U937 were adhesion cocultured with human bone marrow stromal cell line HS-5 cells. Such coculturing induced HL-60 and U937 cells resistant to chemotherapeutic drugs including daunorubicin (DNR), homoharringtonine (HHT), and cytosine arabinoside (Ara-C). The coculturing-induced resistance of AML cells to DNR, HHT, and Ara-C can be partially reversed by inhibition of the PI3K/Akt signaling pathway. Clinically, AML patients with a low level of PTEN and a high level of CCND1 had high relapse rates within 1 year, and newly diagnosed AML patients with extramedullary infiltration had a low level of PTEN. This study confirms the involvement of the PI3K/Akt signaling pathway in multidrug resistance in AML cells induced by stroma and suggests that the expression of PTEN and CCND1 may be a prognostic indicator for AML.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Stromal Cells/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Biomarkers , Cell Line, Tumor , Coculture Techniques , Drug Resistance, Multiple/genetics , Drug Resistance, Neoplasm/genetics , Humans , Prognosis
SELECTION OF CITATIONS
SEARCH DETAIL
...