Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Arch Biochem Biophys ; 754: 109929, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38367794

ABSTRACT

Diffuse large B-cell lymphoma (DLBCL) is the most common type of non-Hodgkin lymphoma. Although treatment options have improved, a large proportion of patients show low survival rates, highlighting an urgent need for novel therapeutic strategies. The aim of this study was to investigate the efficacy of the new small-molecule compound dihydrocelastrol (DHCE), acquired through the structural modification of celastrol (CE), in the treatment of DLBCL. DHCE showed potent anti-lymphoma efficacy and synergistic effects with doxorubicin. DHCE triggered DLBCL cell apoptosis and G0/G1-phase blockade, thereby hindering angiogenesis. DHCE inhibited B-cell receptor cascade signalling and Jun B and p65 nuclear translocation, thereby suppressing pro-tumourigenic signalling. Finally, DHCE exerted lower toxicity than CE, which showed severe hepatic, renal, and reproductive toxicity in vivo. Our findings support further investigation of the clinical efficacy of DHCE against DLBCL.


Subject(s)
Lymphoma, Large B-Cell, Diffuse , Pentacyclic Triterpenes , Transcription Factor AP-1 , Humans , Transcription Factor AP-1/metabolism , Angiogenesis , Signal Transduction , Apoptosis , Lymphoma, Large B-Cell, Diffuse/metabolism , Cell Line, Tumor , Cell Proliferation
2.
Front Immunol ; 14: 1220558, 2023.
Article in English | MEDLINE | ID: mdl-37600821

ABSTRACT

Background: Despite recent advances in the treatment of aggressive lymphomas, a significant fraction of patients still succumbs to their disease. Thus, novel therapies are urgently needed. As the anti-CD20 antibody rituximab and the CD19-targeting antibody tafasitamab share distinct modes of actions, we investigated if dual-targeting of aggressive lymphoma B-cells by combining rituximab and tafasitamab might increase cytotoxic effects. Methods: Antibody single and combination efficacy was determined investigating different modes of action including direct cytotoxicity, antibody-dependent cell-mediated cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP) in in vitro and in vivo models of aggressive B-cell lymphoma comprising diffuse large B-cell lymphoma (DLBCL) and Burkitt lymphoma (BL). Results: Three different sensitivity profiles to antibody monotherapy or combination treatment were observed in in vitro models: while 1/11 cell lines was primarily sensitive to tafasitamab and 2/11 to rituximab, the combination resulted in enhanced cell death in 8/11 cell lines in at least one mode of action. Treatment with either antibody or the combination resulted in decreased expression of the oncogenic transcription factor MYC and inhibition of AKT signaling, which mirrored the cell line-specific sensitivities to direct cytotoxicity. At last, the combination resulted in a synergistic survival benefit in a PBMC-humanized Ramos NOD/SCID mouse model. Conclusion: This study demonstrates that the combination of tafasitamab and rituximab improves efficacy compared to single-agent treatments in models of aggressive B-cell lymphoma in vitro and in vivo.


Subject(s)
Burkitt Lymphoma , Lymphoma, Large B-Cell, Diffuse , Mice , Animals , Mice, Inbred NOD , Mice, SCID , Rituximab/pharmacology , Rituximab/therapeutic use , Leukocytes, Mononuclear , Antibodies, Monoclonal, Humanized , Burkitt Lymphoma/drug therapy , Lymphoma, Large B-Cell, Diffuse/drug therapy
3.
Acta Biochim Biophys Sin (Shanghai) ; 50(8): 782-792, 2018 Aug 01.
Article in English | MEDLINE | ID: mdl-29961897

ABSTRACT

Mantle cell lymphoma (MCL) is an aggressive and mostly incurable B-cell malignancy with frequent relapses after an initial response to standard chemotherapy. Therefore, novel therapies are urgently required to improve MCL clinical outcomes. In this study, MCL cell lines were treated with pterostilbene (PTE), a non-toxic natural phenolic compound primarily found in blueberries. The antitumor activity of PTE was examined by using the Cell Counting Kit-8, apoptosis assays, cell cycle analysis, JC-1 mitochondrial membrane potential assay, western blot analysis, and tumor xenograft models. PTE treatment induced a dose-dependent inhibition of cell proliferation, including the induction of cell apoptosis and cell cycle arrest at the G0/G1 phase. Moreover, the PI3K/Akt/mTOR pathway was downregulated after PTE treatment, which might account for the anti-MCL effects of PTE. Synergistic cytotoxicity was also observed, both in MCL cells and in xenograft mouse models, when PTE was administered in combination with bortezomib (BTZ). The antitumor effects of PTE shown in our study provide an innovative option for MCL patients with poor responses to standardized therapy. It is noteworthy that the treatment combining PTE with BTZ warrants clinical investigation, which may offer an alternative and effective MCL treatment in the future.


Subject(s)
Lymphoma, Mantle-Cell/drug therapy , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Stilbenes/pharmacology , TOR Serine-Threonine Kinases/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Disease Progression , Female , Humans , Lymphoma, Mantle-Cell/metabolism , Lymphoma, Mantle-Cell/pathology , Mice, Inbred NOD , Mice, SCID , Xenograft Model Antitumor Assays
4.
Acta Biochim Biophys Sin (Shanghai) ; 50(4): 399-407, 2018 Apr 01.
Article in English | MEDLINE | ID: mdl-29534146

ABSTRACT

MCT-1 (multiple copies in T-cell lymphoma-1), a novel oncogene, was originally identified in T-cell lymphoma. A recent study has demonstrated that MCT-1 is highly expressed in 85% of diffuse large B-cell lymphomas (DLBCL). PKC (protein kinase C) plays an essential role in signal transduction for multiple biologically active substances for activating cellular functions and proliferation. In this study, we found that the mRNA and protein expression levels of MCT-1 were visibly decreased after knocking down PKC by siRNA in SUDHL-4 and OCI-LY8 DLBCL cell lines. A selective PKC inhibitor, sotrastaurin, effectively inhibited cell proliferation and induced cell apoptosis in a dose- and time-dependent manner. Meanwhile, we also observed that the cell cycle was arrested in the G1 phase in sotrastaurin-treated cells. In addition, MCT-1 was down-regulated in the sotrastaurin treatment group in vivo. Furthermore, we demonstrated that the PKC inhibitor sotrastaurin induced cell apoptosis and cell cycle arrest in DLBCL cells potentially through regulating the expression of MCT-1. Our data suggest that targeting PKC may be a potential therapeutic approach for lymphomas and related malignancies that exhibit high levels of MCT-1 protein.


Subject(s)
Cell Cycle Proteins/metabolism , Gene Expression Regulation , Lymphoma, Large B-Cell, Diffuse/drug therapy , Oncogene Proteins/metabolism , Protein Kinase C/antagonists & inhibitors , Pyrroles/pharmacology , Quinazolines/pharmacology , Animals , Apoptosis , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Dose-Response Relationship, Drug , Female , Gene Silencing , Humans , Lymphoma/drug therapy , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Signal Transduction
5.
Theranostics ; 7(15): 3690-3699, 2017.
Article in English | MEDLINE | ID: mdl-29109769

ABSTRACT

We synthesized a novel aryl-guanidino compound, DCZ3301, and found that it has potent cytotoxicity against multiple human cancer cell lines. The anticancer activity was most potent against multiple myeloma (MM). DCZ3301 induced cytotoxicity in MM cell lines, as well as patient myeloma cells, in part by decreasing mitochondrial membrane potential to induce apoptosis. In contrast, DCZ3301 had no cytotoxic effect on normal cells. DCZ3301 also inhibited cell cycling and caused a G2/M accumulation that corresponded with downregulation of Cdc25C, CDK1, and Cyclin B1. DCZ3301 retained its activity against MM cells in the presence of exogenous cytokines (IL-6 or VEGF) or bone marrow stromal cells (BMSCs) and reduced activity of multiple signaling pathways (STAT3, NFκB, AKT, ERK1/2) in MM but not normal cells. The STAT3 pathway played an important role in modulating DCZ3301-mediated cytotoxicity. Knockdown of STAT3 using siRNA in MM cells enhanced DCZ3301-induced cytotoxicity, whereas overexpression of STAT3 in MM cells partially protected them from apoptosis. In addition, DCZ3301 inhibited VEGF and IL-6 secretion in a dose-dependent fashion in a co-culture of MM cells and BMSCs. Combining DCZ3301 with bortezomib induced synergistic cytotoxicity in MM cell lines and primary MM cells. Finally, in vivo efficacy of DCZ3301 was confirmed in an MM xenograft mouse model. Together, these results provide a rationale for translation of this small-molecule inhibitor, either alone or in combination, to the clinic against MM.


Subject(s)
Antineoplastic Agents/therapeutic use , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Animals , Apoptosis/drug effects , Blotting, Western , Cell Line , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Humans , Interleukin-6/metabolism , Membrane Potential, Mitochondrial/drug effects , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , STAT3 Transcription Factor/metabolism , Vascular Endothelial Growth Factor A/metabolism
6.
Cell Death Dis ; 8(10): e3111, 2017 10 12.
Article in English | MEDLINE | ID: mdl-29022919

ABSTRACT

Diffuse large B-cell lymphoma (DLBCL) is the most common type of lymphoma in adults, characterized by a rapidly increasing painless mass. A novel compound, DCZ3301, was synthesized that exerted direct cytotoxicity against DLBCL cell lines. The effects of DCZ3301 on DLBCL cells in vitro and in vivo and the associated mechanisms were investigated. DCZ3301 inhibited the viability of DLBCL cell lines, even in the presence of protumorigenesis cytokines. Additionally, the compound induced apoptosis and cell cycle arrest at the G2/M phase by reducing mitochondrial membrane potential. DCZ3301 exerted an antitumor effect through modulation of Akt, extracellular signal-regulated kinases 1/2 (ERK1/2) and janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) signaling pathways. Furthermore, DCZ3301 downregulates STAT3 phosphorylation by inhibiting Lck/Yes-related novel protein tyrosine kinase (Lyn) activation in DLBCL. A synergistic cytotoxic effect on DLBCL cells was observed upon combination of DCZ3301 with panobinostat. In vivo, intraperitoneal injection of xenograft mice with DCZ3301 resulted in reduced tumor volume. Our preliminary results collectively support the utility of the small-molecule inhibitor DCZ3301 as an effective novel therapeutic option for DLBCL that requires further clinical evaluation.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , G2 Phase Cell Cycle Checkpoints/drug effects , Guanidines/pharmacology , Lymphoma, Large B-Cell, Diffuse/drug therapy , Pyridines/pharmacology , STAT3 Transcription Factor/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Janus Kinase 2/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Small Interfering/genetics , STAT3 Transcription Factor/genetics , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
7.
Biomed Res Int ; 2017: 9872073, 2017.
Article in English | MEDLINE | ID: mdl-28785594

ABSTRACT

Pterostilbene is a natural 3,5-dimethoxy analog of trans-resveratrol that has been reported to have antitumor, antioxidant, and anti-inflammatory effects. T-cell leukemia/lymphoma is one of the more aggressive yet uncommon non-Hodgkin lymphomas. Although there has been increasing research into T-cell leukemia/lymphoma, the molecular mechanisms of the antitumor effects of pterostilbene against this malignancy are still largely unknown. The aim of this study is to confirm the effects of pterostilbene in T-cell leukemia/lymphoma. Jurkat and Hut-78 cells treated with pterostilbene were evaluated for cell proliferation using Cell Counting Kit-8, and apoptosis, cell cycle progression, reactive oxygen species generation, and mitochondrial membrane potential were analyzed using flow cytometry. The level of protein expression was detected by western blot. The results demonstrated that pterostilbene significantly inhibited the growth of T-cell leukemia/lymphoma cell lines in vitro and induced apoptosis in a dose- and time-dependent manner. Moreover, pterostilbene treatment markedly induced S-phase cell cycle arrest, which was accompanied by downregulation of cdc25A, cyclin A2, and CDK2. Pterostilbene also induced the generation of reactive oxygen species and the loss of mitochondrial membrane potential and inhibited ERK1/2 phosphorylation. Taken together, our study demonstrated the potential of pterostilbene to be an effective treatment for T-cell leukemia/lymphoma.


Subject(s)
Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Leukemia, T-Cell/enzymology , Leukemia, T-Cell/pathology , MAP Kinase Signaling System/drug effects , Stilbenes/pharmacology , Caspases/metabolism , Cell Proliferation/drug effects , Humans , Indazoles/pharmacology , Jurkat Cells , Membrane Potential, Mitochondrial/drug effects , Phosphorylation/drug effects , Piperazines/pharmacology , Reactive Oxygen Species/metabolism
8.
Oncol Rep ; 38(1): 488-496, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28560392

ABSTRACT

Multiple myeloma (MM) is an incurable hematologic malignancy because of its drug resistance. Pterostilbene (Pter) is found mainly in blueberries and grapes. The effects of Pter and its exact pharmacologic mechanisms on chemoresistant myeloma are not known. Herein, we investigated the anti-myeloma activity of Pter in bortezomib-resistant cell line H929R and explored the related mechanism of action for the first time. We found that Pter inhibited proliferation of H929R cells and promoted apoptosis of the cells through a caspase-dependent pathway, loss of mitochondrial membrane potential, and activation of Akt and p38 mitogen-activated protein kinase (MAPK) signaling pathways. DNA damage and S-phase arrest might be involved in Pter-related toxicity in H929R cells. Pter and the histone deacetylase inhibitors panobinostat or vorinostat inhibited proliferation of H929R cells in a synergistic manner. These data supported that Pter might be a promising natural compound for relapsed/refractory myeloma therapy, especially against myeloma resistant to bortezomib chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Histone Deacetylase Inhibitors/pharmacology , MAP Kinase Signaling System/drug effects , Multiple Myeloma/drug therapy , Neoplasm Recurrence, Local/drug therapy , Stilbenes/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Blueberry Plants/chemistry , Bortezomib/pharmacology , Bortezomib/therapeutic use , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Damage/drug effects , Drug Resistance, Neoplasm , Drug Synergism , Histone Deacetylase Inhibitors/therapeutic use , Humans , Hydroxamic Acids/pharmacology , Hydroxamic Acids/therapeutic use , Indoles/pharmacology , Indoles/therapeutic use , Membrane Potential, Mitochondrial/drug effects , Multiple Myeloma/pathology , Panobinostat , S Phase Cell Cycle Checkpoints/drug effects , Stilbenes/therapeutic use , Vorinostat , p38 Mitogen-Activated Protein Kinases/metabolism
9.
Acta Biochim Biophys Sin (Shanghai) ; 49(5): 420-427, 2017 May 01.
Article in English | MEDLINE | ID: mdl-28338993

ABSTRACT

Multiple myeloma (MM) is the second most frequent malignant hematological disease. Dihydrocelastrol (DHCE) is synthesized by hydrogenated celastrol, a treterpene isolated from Chinese medicinal plant Tripterygium regelii. In this study, we first reported the anti-tumor activity of DHCE on MM cells. We found that DHCE could inhibit cell proliferation and promote apoptosis through caspase-dependent way in vitro. In addition, DHCE could inactivate the expression of interleukin (IL)-6 and downregulate the phosphorylation of extracellular regulated protein kinases (ERK1/2) and the signal transducer and activator of transcription 3 (STAT3) in MM. It also retained its activity against MM cell lines in the presence of IL-6. Furthermore, treatment of MM cells with DHCE resulted in an accumulation of cells in G0/G1 phase of the cell cycle. Notably, DHCE reduced the expression of cyclin D1 and cyclin-dependent kinases 4 and 6 in MM cell lines. Additionally, its efficacy toward the MM cell lines could be enhanced in combination with the histone deacetylase inhibitor panobinostat (LBH589), which implied the possibility of the combination treatment of DHCE and LBH589 as a potential therapeutic strategy in MM. In addition, treatment of NCI-H929 tumor-bearing nude mice with DHCE (10 mg/kg/d, i.p., 1-14 days) resulted in 73% inhibition of the tumor growth in vivo. Taken together, the results of our present study indicated that DHCE could inhibit cellular proliferation and induce cell apoptosis in myeloma cells mediated through different mechanisms, possibly through inhibiting the IL-6/STAT3 and ERK1/2 pathways. And it may provide a new therapeutic option for MM patients.


Subject(s)
Apoptosis/drug effects , Interleukin-6/metabolism , MAP Kinase Signaling System/drug effects , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , STAT3 Transcription Factor/metabolism , Triterpenes/administration & dosage , Animals , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Male , Mice , Mice, Nude , Multiple Myeloma/pathology , Pentacyclic Triterpenes , Treatment Outcome
10.
Sci Rep ; 6: 37417, 2016 11 21.
Article in English | MEDLINE | ID: mdl-27869173

ABSTRACT

Diffuse large B-cell lymphoma (DLBCL) is the most common type of non-Hodgkin lymphoma (NHL). Pterostilbene, a natural dimethylated analog of resveratrol, has been shown to possess diverse pharmacological activities, including anti-inflammatory, antioxidant and anticancer properties. However, to the best of our knowledge, there has been no study of the effects of pterostilbene upon hematological malignancies. Herein, we report the antitumor activity and mechanism of pterostilbene against DLBCL cells both in vitro and in vivo. We found that pterostilbene treatment resulted in a dose-dependent inhibition of cell viability. In addition, pterostilbene exhibited a strong cytotoxic effect, as evidenced not only by reductions of mitochondrial membrane potential (MMP) but also by increases in cellular apoptotic index and reactive oxygen species (ROS) levels, leading to arrest in the S-phase of the cell cycle. Furthermore, pterostilbene treatment directly up-regulated p-p38MAPK and down-regulated p-ERK1/2. In vivo, intravenous administration of pterostilbene inhibited tumor development in xenograft mouse models. Overall, the results suggested that pterostilbene is a potential anti-cancer pharmaceutical against human DLBCL by a mechanism involving the suppression of ERK1/2 and activation of p38MAPK signaling pathways.


Subject(s)
Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Lymphoma, Large B-Cell, Diffuse/pathology , Stilbenes/pharmacology , Animals , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Humans , MAP Kinase Signaling System/drug effects , Membrane Potential, Mitochondrial/drug effects , Mice, Inbred BALB C , Mice, Nude , Reactive Oxygen Species/metabolism , Xenograft Model Antitumor Assays
11.
Int J Mol Sci ; 17(11)2016 Nov 17.
Article in English | MEDLINE | ID: mdl-27869675

ABSTRACT

Multiple myeloma (MM) is the second most common malignancy in the hematologic system, which is characterized by accumulation of plasma cells in bone marrow. Pterostilbene (PTE) is a natural dimethylated analog of resveratrol, which has anti-oxidant, anti-inflammatory and anti-tumor properties. In the present study, we examined the anti-tumor effect of PTE on MM cell lines both in vitro and in vivo using the cell counting kit (CCK)-8, apoptosis assays, cell cycle analysis, reactive oxygen species (ROS) generation, JC-1 mitochondrial membrane potential assay, Western blotting and tumor xenograft models. The results demonstrated that PTE induces apoptosis in the H929 cell line and causes cell cycle arrest at G0/G1 phase by enhancing ROS generation and reducing mitochondrial membrane potential. The anti-tumor effect of PTE may be caused by the activation of the extracellular regulated protein kinases (ERK) 1/2 and c-Jun N-terminal kinase (JNK) signaling pathways. Additionally, mice treated with PTE by intraperitoneal injection demonstrated reduced tumor volume. Taken together, the results of this study indicate that the anti-tumor effect of PTE on MM cells may provide a new therapeutic option for MM patients.


Subject(s)
Antineoplastic Agents/pharmacology , MAP Kinase Signaling System/drug effects , Multiple Myeloma/drug therapy , Stilbenes/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Damage , Female , G1 Phase Cell Cycle Checkpoints , Humans , Membrane Potential, Mitochondrial/drug effects , Mice, Inbred NOD , Mice, SCID , Multiple Myeloma/metabolism , Reactive Oxygen Species/metabolism , Stilbenes/therapeutic use , Tumor Burden , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...