scholarly journals Effects of Histone Deacetylase Inhibitor Panobinostat (LBH589) on Bone Marrow Mononuclear Cells of Relapsed or Refractory Multiple Myeloma Patients and Its Mechanisms

2017 ◽  
Vol 23 ◽  
pp. 5150-5157
Author(s):  
Yanping Ma ◽  
Wenhua Liu ◽  
Ling Zhang ◽  
Gu Jia
Cancer ◽  
2010 ◽  
Vol 117 (2) ◽  
pp. 336-342 ◽  
Author(s):  
Ruben Niesvizky ◽  
Scott Ely ◽  
Tomer Mark ◽  
Sangeeta Aggarwal ◽  
Janice L Gabrilove ◽  
...  

Blood ◽  
2002 ◽  
Vol 99 (6) ◽  
pp. 2248-2251 ◽  
Author(s):  
Masaki Kitazono ◽  
Vemulkonda Koneti Rao ◽  
Rob Robey ◽  
Takashi Aikou ◽  
Susan Bates ◽  
...  

Abstract Adenovirus infection of hematopoietic cells frequently requires high virus concentrations and long incubation times to obtain moderate infection levels because these cells have low levels of Coxsackie and adenovirus receptor (CAR) and αv integrin. The effect of treatment with FR901228 (depsipeptide), a histone deacetylase inhibitor in phase 2 clinical trials, was studied in K562 cells, granulocyte–colony-stimulating factor–mobilized peripheral blood mononuclear cells (PBMCs), and CD34+ peripheral blood stem cells (PBSCs). FR901228 increased CAR and αvintegrin RNA levels and histone H3 acetylation. FR901228 treatment before adenovirus infection was associated with at least a 10-fold increase in transgene expression from a β-galactosidase–expressing adenoviral vector. More than 80% of the PBMCs or CD34+ PBSCs from 7 different donors were β-galactosidase–positive after adenovirus infection with a multiplicity of infection of 10 for 60 minutes. Increased CAR, αv integrin, and acetylated histone H3 levels were observed in PBMCs from a patient treated with FR901228. These studies suggest that FR901228 can increase the efficiency of adenoviral infection in hematopoietic cells.


2015 ◽  
Vol 59 (7) ◽  
pp. 3984-3994 ◽  
Author(s):  
Kasper L. Jønsson ◽  
Martin Tolstrup ◽  
Johan Vad-Nielsen ◽  
Kathrine Kjær ◽  
Anders Laustsen ◽  
...  

ABSTRACTAdjunct therapy with the histone deacetylase inhibitor (HDACi) romidepsin increases plasma viremia in HIV patients on combination antiretroviral therapy (cART). However, a potential concern is that reversing HIV latency with an HDACi may reactivate the virus in anatomical compartments with suboptimal cART concentrations, leading tode novoinfection of susceptible cells in these sites. We tested physiologically relevant romidepsin concentrations known to reactivate latent HIV in order to definitively address this concern. We found that romidepsin significantly inhibited HIV infection in peripheral blood mononuclear cells and CD4+T cells but not in monocyte-derived macrophages. In addition, romidepsin impaired HIV spreading in CD4+T cell cultures. When we evaluated the impact of romidepsin on quantitative viral outgrowth assays with primary resting CD4+T cells, we found that resting CD4+T cells exposed to romidepsin exhibited reduced proliferation and viability. This significantly lowered assay sensitivity when measuring the efficacy of romidepsin as an HIV latency reversal agent. Altogether, our data indicate that romidepsin-based HIV eradication strategies are unlikely to reseed a latent T cell reservoir, even under suboptimal cART conditions, because romidepsin profoundly restrictsde novoHIV infections.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 1965-1965
Author(s):  
Yifeng Sun ◽  
Peng Liu ◽  
Jing Li

Abstract Most patients with multiple myeloma (MM) would finally relapse. Current chemotherapy regimens have limited effect on relapse MM patients. As a new histone deacetylase inhibitor, chidamide has been used in malignancy treatment such as peripheral T-cell lymphoma. However, it is still unknown if chidamide can be used in MM. To determine the target gene of chidamide in MM patients, we performed RNA-Seq analysis using 3 MM patients' bone marrow mononuclear cells. Their BMMCs were cultured with 6μM chidamide or not, and six of the most significantly changed coding genes were selected. Realtime RT-PCR showed that compared with DMSO-treated cells, after adding 6μM chidamide, the expression of SDHA and FCER2 was increased and MRPL30 decreased. The expression of SDHA was upregulated and ITGA7 was downregulated in MM patients. Based on the data above, SDHA was considered as the most valuable target gene of chidamide in MM. Realtime RT PCR also showed that SDHA expression in normal volunteers was the highest and followed by patients with MGUS and initial MM. Patients with relapse MM had the lowest SDHA expression. To assess the effects of chidamide on MM cells, we next performed cell proliferation and invasion assay. Chidamide dramatically inhibited proliferation of myeloma cell lines (H929 and OPM-2). However, when SDHA was knocked down by siRNA, this inhibition effect was not dramatically as before in H929 cells. Similarly, chidamide-treated H929 cells achieved a notably lower percentage of cell invasion than those treated with DMSO. Thus, when SDHA was knocked down, this invasive ability was not significantly changed whether chidamide was added or not. CCK8 assay was used to determine dose-response curves of chemotherapeutic agents and synergistic effect of chidamide combined with other agents. he combination index (CI) of lenalidomide and low concentration of bortezomib yielded many of the data points to the area <1 when combined with chidamide treatment, denoting synergistic interactions in MM cell line. Interestingly, when SDHA was knocked down, cells apoptosis induced by bortezomib and lenalidomide combined with chidamide greatly decreased, especially by lenalidomide, which indicated the synergistic effect between chidamide and other agents was induced by SDHA. When SDHA was knocked down by siRNA, most of CI between chidamide and bortezomib or lenalidomide also raised above 1, which indicated the synergistic effect tended to disappear. In order to gain further mechanism of chidamide-SDHA-MM axis, Western Blot was used. As a result, it showed that when SDHA was knocked down in H929 cells by siRNA, expression of HIFα protein was increased. HIFα decreased after adding 6μM chidamide. However, when SDHA was knocked down, chidamide did not regulate the expression of HIFα any longer. ROS in MM patients were much higher than that in normal people, which caused by higher HIFα expression in large extent. Chidamide inhibited ROS production, and similar to HIFα, the ROS production was not sensitive to chidamide any more when SDHA was knocked down. Our findings clatify the mechanism of chidamide treatment in MM through SDHA and reveal a novel therapeutic strategy of MM treatment. Figure. Figure. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document