scholarly journals Protein Homeostasis: A Key Target in Myeloma

Blood ◽  
2017 ◽  
Vol 130 (Suppl_1) ◽  
pp. SCI-8-SCI-8
Author(s):  
Lawrence H. Boise

Multiple myeloma is a disease of long-lived plasma cells that remains incurable in the majority of patients. However advances in the treatment have been made in the last 20 years resulting in significant improvement in the survival of patients with this disease. Interestingly almost all of these advances have been made through the targeting of pathways or molecules that are shared between normal and malignant plasma cells and are not due to the precision targeting of myeloma cell dependencies associated with the transformation process. Most notably targeting both specific and bulk protein homeostasis through the action of IMiDs and proteasome inhibitors have become the backbone for the treatment of newly diagnosed myeloma in the United States. We have focused on proteasome function and regulation to better understand why a macromolecular structure that is present and functions in every cell in the body is such an effective target for myeloma therapy. Our initial findings indicated that proteasome inhibition activated an endoplasmic reticulum stress response known as the unfolded protein response (UPR). The UPR is required for normal differentiation and maintenance of plasma cells, although our initial findings suggesting that the PERK arm of the response, which is not required for normal plasma cell maintenance was activated by proteasome inhibitors. We determined that the likely source of unfolded protein was immunoglobulin produced by myeloma cells and that the fate of immunoglobulin was a key determinant of sensitivity to proteasome inhibition. We have now focused on additional factors that determine the response to proteasome inhibition and consistent with previous findings have demonstrated that activation of heat shock response is a early protective event following proteasome inhibition. However we were unable to demonstrate that any single heat shock protein was sufficient to alter the response to proteasome inhibition. Rather it was the activation of the heat shock response itself and that protection was only efficiently blocked by inhibition of the transcriptional regulator of the response, HSF1. HSF1 is activated through post-translational modifications and proteasome inhibition results in phosphorylation of HSF1 at several sites, most notably S326. This can be blocked with the multi-kinase inhibitor TG02 that effectively blocks the proteasome inhibitor-induced heat shock response and sensitizes myeloma cells to proteasome inhibitor-induced cell death. Based on these findings, the combination of TG02 and carfilzomib are being tested clinically. The cellular response to proteasome inhibitors is also controlled through the regulation of proteasome assembly. We have recently demonstrated that 14-3-3ζ an anti-apoptotic phospho-serine binding protein, can limit proteasome assembly and activity through its binding to the PA28α subunit of the 11S regulator. This significantly lowers proteasome capacity and sensitizes cells to proteasome inhibition but not to other therapeutic agents. Finally in addition to regulation of bulk protein homeostasis, targeting of specific protein turnover has proven effective in myeloma treatment. IMiDs can alter the ability of the E3 ligase cereblon to recognize substrates for ubiquitination, resulting in the degradation of key B cell transcription factors IKZF1/3. These can regulate the plasma cell maintenance factor IRF4 as well as play a direct role in the regulation plasma cell superenhancers that control the expression of oncogenes translocated in myeloma. Another key mediator of myeloma cell survival, MCL1 is regulated via protein stability and can targeted in this manner. Direct and indirect targeting of MCL1 will be discussed. Disclosures Boise: Abbvie: Consultancy; Eli Lilly and Company: Research Funding.

2002 ◽  
Vol 282 (4) ◽  
pp. R1016-R1026 ◽  
Author(s):  
Timothy A. Pritts ◽  
Eric S. Hungness ◽  
Dan D. Hershko ◽  
Bruce W. Robb ◽  
Xiaoyan Sun ◽  
...  

In previous studies, the heat shock response, induced by hyperthermia or sodium arsenite, increased interleukin (IL)-6 production in intestinal mucosa and cultured human enterocytes. A novel way to induce the heat shock response, documented in other cell types, is treatment with proteasome inhibitors. It is not known if proteasome inhibition induces heat shock in enterocytes or influences IL-6 production. Here we tested the hypothesis that treatment of cultured Caco-2 cells, a human intestinal epithelial cell line, with proteasome inhibitors induces the heat shock response and stimulates IL-6 production. Treatment of Caco-2 cells with one of the proteasome inhibitors MG-132 or lactacystin activated the transcription factor heat shock factors (HSF)-1 and -2 and upregulated cellular levels of the 72-kDa heat shock protein HSP-72. The same treatment resulted in increased gene and protein expression of IL-6, a response that was blocked by quercetin. Additional experiments revealed that the IL-6 gene promoter contains a HSF-responsive element and that the IL-6 gene may be regulated by the heat shock response. The present results suggest that proteasome inhibition induces heat shock response and IL-6 production in enterocytes and that IL-6 may be a heat shock-responsive gene, at least under certain circumstances. The observations are important considering the multiple biological roles of IL-6, both locally in the gut mucosa and systemically, and considering recent proposals in the literature to use proteasome inhibitors in the clinical setting to induce the heat shock response.


2011 ◽  
Vol 7 (2) ◽  
pp. 340-349 ◽  
Author(s):  
Sandro Santagata ◽  
Ya-ming Xu ◽  
E. M. Kithsiri Wijeratne ◽  
Renee Kontnik ◽  
Christine Rooney ◽  
...  

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.


2010 ◽  
Vol 29 (7) ◽  
pp. 551-554 ◽  
Author(s):  
Suresh IS Rattan ◽  
Taru Deva

The scientific foundations of hormesis are now well established and include various biochemical and molecular criteria for testing the hormetic nature of chemicals and other modulators. In order to claim homeopathy as being hormetic, it is essential that, in addition to the hormetic biphasic dose response, homeopathic remedies should fulfill one or more molecular criteria. Since stress response pathways, such as heat shock response, antioxidative response, autophagic response and unfolded protein response, are integral components of the physiological hormesis, it is important that homeopathic drugs be tested for these pathways if these are to be considered as hormetins and to cause hormesis.


Blood ◽  
2007 ◽  
Vol 110 (7) ◽  
pp. 2641-2649 ◽  
Author(s):  
Emma L. Davenport ◽  
Hannah E. Moore ◽  
Alan S. Dunlop ◽  
Swee Y. Sharp ◽  
Paul Workman ◽  
...  

Plasma cells producing high levels of paraprotein are dependent on the unfolded protein response (UPR) and chaperone proteins to ensure correct protein folding and cell survival. We hypothesized that disrupting client–chaperone interactions using heat shock protein 90 (Hsp90) inhibitors would result in an inability to handle immunoglobulin production with the induction of the UPR and myeloma cell death. To study this, myeloma cells were treated with Hsp90 inhibitors as well as known endoplasmic reticulum stress inducers and proteasome inhibitors. Treatment with thapsigargin and tunicamycin led to the activation of all 3 branches of the UPR, with early splicing of XBP1 indicative of IRE1 activation, upregulation of CHOP consistent with ER resident kinase (PERK) activation, and activating transcription factor 6 (ATF6) splicing. 17-AAG and radicicol also induced splicing of XBP1, with the induction of CHOP and activation of ATF6, whereas bortezomib resulted in the induction of CHOP and activation of ATF6 with minimal effects on XBP1. After treatment with all drugs, expression levels of the molecular chaperones BiP and GRP94 were increased. All drugs inhibited proliferation and induced cell death with activation of JNK and caspase cleavage. In conclusion, Hsp90 inhibitors induce myeloma cell death at least in part via endoplasmic reticulum stress and the UPR death pathway.


Sign in / Sign up

Export Citation Format

Share Document