Inhibition of Interleukin-6 Signaling with CNTO328 Sensitizes Multiple Myeloma Cells to Bortezomib and Attenuates Proteasome Inhibitor-Mediated Induction of the Anti-Apoptotic Heat Shock Protein Response.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1574-1574
Author(s):  
Peter M. Voorhees ◽  
Deborah J. Kuhn ◽  
George W. Small ◽  
John S. Strader ◽  
Robert Corringham ◽  
...  

Abstract The proteasome inhibitor bortezomib represents a significant advance in the treatment of multiple myeloma, but its efficacy is limited by a number of resistance mechanisms. One of the most important is the heat shock protein (HSP) and stress response pathways which, through members such as HSP-70 and mitogen-activated protein kinase (MAPK) phosphatase (MKP)-1, oppose the pro-apoptotic activities of bortezomib. Because interleukin (IL)-6 signaling augments the heat shock response through signal transducer and activator of transcription (STAT)-1 and heat shock transcription factor (HSF)-1, we hypothesized that downregulation of IL-6 signaling would attenuate HSP induction by bortezomib, thereby enhancing its anti-myeloma activity. Treatment of the IL-6-dependent multiple myeloma cell lines KAS-6 and ANBL-6 with the combination of bortezomib and CNTO328, a chimeric monoclonal IL-6 neutralizing antibody, resulted in greater reduction of cell viability than with either drug alone in a time- and concentration-dependent manner. This was associated with an enhanced induction of apoptosis which, under some conditions, was greater than the sum of the two individual agents alone, suggesting a synergistic interaction. Similar findings were not seen when using isotype control antibodies, and in studies of the IL-6-independent RPMI 8226 myeloma cell line. Increased activity was seen when cells were pre-treated with CNTO328 followed by bortezomib, or when they were treated with both agents concurrently, compared to treatment with bortezomib followed by CNTO328. Treatment with CNTO328 potently inhibited IL-6-mediated downstream signaling pathways, as demonstrated by marked blockade of STAT-3 and p44/42 MAPK phosphorylation. Enhanced activity of the combination regimen correlated with attenuated induction by bortezomib of the heat shock and stress response proteins HSP-70 and MKP-1 by up to 45% and 90%, respectively. Notably, CNTO328 markedly reduced levels of transcriptionally active phospho-STAT-1 and hyperphosphorylated HSF-1. Other strategies to suppress the heat shock response, including the use of the pharmacologic inhibitor KNK437, also yielded evidence for a synergistic anti-myeloma effect in combination with bortezomib. The synergistic activity of KNK437 and bortezomib was reproduced in normal mouse embryo fibroblasts (MEFs), but blunted in HSF-1 knockout MEFs. Taken together, the above data demonstrate that inhibition of IL-6 signaling enhances the anti-myeloma activity of bortezomib. They also support the hypothesis that this occurs, at least in part, by attenuating proteasome inhibitor-mediated induction of the heat shock response through downregulation of transcriptionally active STAT-1 and HSF-1. These findings provide a strong rationale for future translation of the CNTO328/bortezomib combination into the clinic.

2015 ◽  
Vol 5 (1) ◽  
Author(s):  
Masayu Azizah ◽  
Rostika Flora ◽  
Theodoru Theodoru

AbstrakHeat Shock Protein (HSP) merupakan suatu protein yang dihasilkan karena adanya Heat Shock Response (HSR). HSR diperlukan sebagai tanggapan sel terhadap berbagai macam gangguan, baik yang bersifat fisiologis maupun yang berasal dari lingkungan. Peningkatan kadar HSP70 otot jantung ini dikarenakan adanya aktivitas fisik anaerobik. Aktivitas fisik ini mengakibatkan tubuh mengalami peningkatan suhu tubuh, stres panas dan latihan.Tujuan penelitian ini adalah untuk menganalisa pengaruh aktivitas fisik anaerobik yang dilakukan setiap hari terhadap kadar Heat Shock Protein (HSP)70 otot jantung tikus wistar. Berdasarkan hasil penelitian, kelompok anaerobik menunjukkan peningkatan kadar HSP otot jantung dibandingkan kelompok pembanding.Kata kunci : aktivitas fisik anaerobik tiap hari, HSP70, treadmill, tikus rattus novergicus wistar.AbstractHeat Shock Protein (HSP) is a protein that is produced because of the Heat Shock Response (HSR). HSR is required as a response of cells to a variety of disorders, both physiological as well as from the environment. Increased levels of HSP70 heart muscle is due to anaerobic physical activity. Physical activity causes the body to increase body temperature, heat stress and latihan.Tujuan this study is to analyze the influence of anaerobic physical activity performed every day on levels of Heat Shock Protein (HSP) 70 wistar rat cardiac muscle. Based on the results of the study, the group showed increased levels of HSP anaerobic heart muscle than the comparison group.Keywords : anaerobic physical activity every day , HSP70 , body temperature, rattus novergicus wistar rat.


RSC Advances ◽  
2015 ◽  
Vol 5 (73) ◽  
pp. 59003-59013 ◽  
Author(s):  
Yen Chin Koay ◽  
Jeanette R. McConnell ◽  
Yao Wang ◽  
Shelli R. McAlpine

C-terminal inhibitors of heat shock protein 90 (hsp90) modulate the C-terminus and do not elicit a heat shock response.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 3364-3364
Author(s):  
Christopher Maisel ◽  
Nizar Bahlis ◽  
Yanling Miao ◽  
Lili Liu ◽  
Stanton Gerson

Abstract While proteasome inhibitors are effective therapy for multiple myeloma (MM), their efficacy could be improved by synergistic targeting of apoptosis. The intracellular serine/threonine kinase Akt has been demonstrated to have a central role in MM cell growth, survival, and drug resistance. Akt is activated by extracellular cytokines such as IGF-1 and IL-6, and contributes to MM resistance by ameliorating the apoptotic effects of proteasome inhibition. Akt requires chaperone proteins for proper stability and function, including the 90 kD molecule Heat Shock Protein 90 (HSP-90). HSP-90 function is abrogated by geldanamycin and its derivative, 17-allylamino-17-demethoxygeldanamycin (17-AAG). In the U266 MM cell line, the IC50 of the proteasome inhibitor MG-132 and 17-AAG was 100 nM and 800 nM, respectively. Following exposure of U266 MM cells to either drug alone, or the combination at a fixed-ratio of their IC50s (1:8), apoptosis was determined by Annexin V staining and FACScan analysis. Synergy analysis was performed using Calcusyn (Biosoft, Cambridge, UK). We found that the combination index (CI) was synergistic (CI<1) throughout the dose range, with a CI = 0.449 ± 0.298 at the combination IC50 (highly significant). For example, the apoptotic effect of 50 nM MG-132 and 400 nM 17-AAG was 6 ±2 % and 23 ±3 %, respectively, whereas the 50:400 nM combination produced apoptosis in 68 ± 2 % of the cells. To analyze effects on Akt and its substrates, we incubated U266 MM cells with MG-132 (50 nM), 17-AAG (400 nM), or the combination. We harvested lysates after zero, two, six, and 24 hours incubation, and Western blot analysis was performed. Co-incubation with MG-132 and 17-AAG, but not either alone, depleted Akt by 24 hours post-therapy. Co-treatment also produced significantly greater upregulation of HSP-90 and HSP-70 than 17-AAG alone, thus demonstrating greater functional inhibition of Akt. The combination also demonstrated the greatest abrogation of Akt-mediated effects on mitochondrial apoptosis: Co-treatment produced the greatest expression of BAD, decreased BCL-XL expression, reduced phosphorylation of GSK-3, and produced the greatest activation of caspase 3. Monotherapy with 17-AAG upregulated HSP-90 and HSP-70, reduced BCL-XL expression, and activated caspase 9. MG-132 monotherapy produced none of these effects. These findings demonstrate that synergy between proteasome inhibitors and 17-AAG is mediated by Akt depletion and abrogation of Akt signaling, predominantly by MG-132 augmentation of 17-AAG-mediated decay of Akt. Down-regulation of Akt-mediated resistance allows dual-apoptotic signaling and synergistic effect of combination therapy. These findings demonstrate a mechanistic rationale for utilizing heat shock protein inhibitors in combination with proteasome inhibitors as therapy for MM.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2953-2953
Author(s):  
Shardule P Shah ◽  
Sagar Lonial ◽  
Lawrence H. Boise

Abstract Abstract 2953 Multiple myeloma is a plasma cell disorder with an average incidence of 21,000 new cases per year in the United States. Recent advances in therapeutic approaches such as the use of proteasome inhibitors have resulted in a significant increase in the overall survival of myeloma patients. Myeloma cells maintain many of the characteristics of normal plasma cells, including constitutive immunoglobulin production and secretion, therefore management of ER stress plays a role in myeloma cell sensitivity to proteasome inhibition. However, myeloma cells also upregulate protective genes in response to the proteotoxic stress that can limit the therapeutic response. Previous groups have published on the importance of the heat shock response and the heat shock protein (HSP) family, supporting preclinical and clinical exploration of HSP inhibition in myeloma. Our group had interest in regulation of the HSP response and has evaluated the master regulator HSF1 as a potential therapeutic target. We found that siRNA-mediated silencing of HSF1 enhances bortezomib-induced apoptosis in a myeloma cell line. To define the effectors of the heat shock response important in regulating bortezomib response, we determined which heat shock response genes are induced by bortezomib in an HSF1-dependent manner. From a realtime PCR array of 84 HSP family genes, we found 21 genes that were induced greater than 2-fold by bortezomib. Of these 21 genes, 10 genes showed >50% reduction in HSF1-silenced cells. 7/10 genes were confirmed by independent qRT-PCR and western blot analysis. These genes include: CRYAB (alpha-crystallin B chain), DNAJB1 (HSP40 subfamily B), HSPA1A (HSP70-1A), HSPA1B (HSP70-1B), HSPB1 (HSP27), HSPH1 (HSP105/110), and HSP90AB1 (HSP90b1). To begin to determine which of these genes was important for the HSF1-dependent protective response we silenced the 7 genes individually and subsequently treated the cells with bortezomib. Surprisingly only 1 of the 7 genes silenced individually, DNAJB1, had an observable effect on bortezomib-induced death. However DNAJB1 silencing does not account for all the HSF1 activity as the increase in cell death due to bortezomib is only 48% of that observed with HSF1 silencing. Thus targeting HSF1 is more effective at sensitizing multiple myeloma cells to bortezomib-induced apoptosis than targeting individual HSPs. Moreover these data suggest that HSP90 inhibitors are functioning by inhibiting at least two members of this family to be effective as single agents. Therefore, while clinical trials for individual HSP and HSP in combination with bortezomib are being conducted, a more effective strategy for apoptosis induction is achieved through inhibition of HSP regulators such as HSF1 in combination with bortezomib. These results provide support for investigating HSP regulation in response to PI to increase the efficacy of myeloma therapy. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1929-1929
Author(s):  
Shardule P. Shah ◽  
Vikas A. Gupta ◽  
Shannon M. Matulis ◽  
Ajay K. Nooka ◽  
Sagar Lonial ◽  
...  

Abstract Multiple myeloma (MM) is a plasma cell malignancy with an estimated 22,350 new cases and 10,710 deaths in the United States in 2013. Novel treatments including autologous stem cell transplant, immunomodulatory drugs (IMiDs), and the proteasome inhibitor, bortezomib, have led to an increase in patient life span and long-term survival. Bortezomib is a highly selective and reversible 26S proteasome inhibitor. Proteasome inhibition can affect multiple signaling cascades and lead to a toxic buildup of misfolded proteins and eventually, cell death. As part of the response to this protein buildup following proteasome inhibition, myeloma cells activate the cytoprotective heat shock response. This includes upregulation of heat shock proteins (HSPs) such as HSP40, HSP70, and HSP90. Previous attempts at using HSP-specific inhibitors in combination with bortezomib have been disappointing. These results underscore the need to disrupt broad scale activation of the entire heat shock response. This can be achieved by inhibition of the master regulator, Heat Shock Factor 1 (HSF1). Here we show that in four human MM cell lines, MM1.s, KMS11, KMS18, and U266, HSF1 inhibition leads to downregulation of the bortezomib-induced heat shock response and ultimately, increased cell death. While HSF1 is activated by proteasome inhibition, the mechanism of activation has yet to be determined. HSF1 is regulated through a complex series of post-translational modifications. Here we show that bortezomib induces HSF1 phosphorylation in MM1.s and KMS18, and in freshly isolated patient samples. To determine which kinase pathways are responsible for HSF1 phosphorylation, we treated MM1.s and KMS18 with a non-lethal dose (10 μM) of PI3K, MEK, JNK, and p38 inhibitors in combination with bortezomib. Bortezomib-induced HSF1 phosphorylation was inhibited by the p38 inhibitor SB 203580, while inhibitors to PI3K, MEK, and JNK had no effect on bortezomib-induced HSF1 phosphorylation. To determine the consequence of p38 inhibition on HSF1 function, we performed RT-qPCR to probe for the expression of HSF1-dependent gene targets (HSPB1 [HSP27], HSP40B, HSPA1A [HSP70/72], HSPA1B [HSP70/72], HSP90AA1, HSP90A1B) following treatment with bortezomib with or without SB 203580. Surprisingly, gene expression for each of the targets increased when proteasome inhibition was combined with p38 inhibition compared to proteasome inhibition alone. The observed change ranged from 31% (HSPA1A) to 99% (HSP90AA1). Our results show a previously undescribed link between proteasome inhibition and HSF1 regulation; bortezomib-induced p38-dependent phosphorylation. This is consistent with studies in HeLa cells showing that the p38 effector MK2 negatively regulates HSF1 via phosphorylation of S121. Together these findings underscore the complexity of the cellular response to proteasome inhibition, and that understanding both the positive and negative regulatory events during HSF1 activation could lead to the development of novel partners for use with proteasome inhibitors. Disclosures: Lonial: Millennium: Consultancy; Celgene: Consultancy; Novartis: Consultancy; BMS: Consultancy; Sanofi: Consultancy; Onyx: Consultancy. Boise:Onyx Pharmaceuticals: Consultancy.


Viruses ◽  
2020 ◽  
Vol 12 (2) ◽  
pp. 245 ◽  
Author(s):  
Alexander J. McMenamin ◽  
Katie F. Daughenbaugh ◽  
Michelle L. Flenniken

Honey bees (Apis mellifera) are an agriculturally important pollinator species that live in easily managed social groups (i.e., colonies). Unfortunately, annual losses of honey bee colonies in many parts of the world have reached unsustainable levels. Multiple abiotic and biotic stressors, including viruses, are associated with individual honey bee and colony mortality. Honey bees have evolved several antiviral defense mechanisms including conserved immune pathways (e.g., Toll, Imd, JAK/STAT) and dsRNA-triggered responses including RNA interference and a non-sequence specific dsRNA-mediated response. In addition, transcriptome analyses of virus-infected honey bees implicate an antiviral role of stress response pathways, including the heat shock response. Herein, we demonstrate that the heat shock response is antiviral in honey bees. Specifically, heat-shocked honey bees (i.e., 42 °C for 4 h) had reduced levels of the model virus, Sindbis-GFP, compared with bees maintained at a constant temperature. Virus-infection and/or heat shock resulted in differential expression of six heat shock protein encoding genes and three immune genes, many of which are positively correlated. The heat shock protein encoding and immune gene transcriptional responses observed in virus-infected bees were not completely recapitulated by administration of double stranded RNA (dsRNA), a virus-associated molecular pattern, indicating that additional virus–host interactions are involved in triggering antiviral stress response pathways.


2017 ◽  
Vol 1 (21) ◽  
pp. 1848-1853
Author(s):  
Shardule P. Shah ◽  
Ajay K. Nooka ◽  
Sagar Lonial ◽  
Lawrence H. Boise

Key Points Proteasome inhibition activates multiple kinases in myeloma cells resulting in the phosphorylation of p53, HSP27, c-JUN, and HSF1. TG02 inhibits proteasome inhibitor (PI)–induced HSF1 pS326, representing a novel mechanism for a TG02 and PI combination.


2015 ◽  
Vol 13 (16) ◽  
pp. 4627-4631 ◽  
Author(s):  
Y. Wang ◽  
S. R. McAlpine

The cellular protection mechanism, the heat shock response, is only activated by classical heat shock 90 inhibitors (Hsp90) that “target” the N-terminus of the protein, but not by those that modulate the C-terminus.


Sign in / Sign up

Export Citation Format

Share Document