Inhibition of the HDM-2 E3 Ligase Induces Types I and II Cell Death in Wild-Type and Mutant p53 Models of Multiple Myeloma, and Acts Synergistically in Both with ABT-737.

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2940-2940
Author(s):  
Dongmin Gu ◽  
Richard Julian Jones ◽  
Hua Wang ◽  
Lance Leopold ◽  
Robert Z. Orlowski

Abstract Abstract 2940 Background: The ubiquitin-proteasome pathway has been validated as a therapeutic target for multiple myeloma by the incorporation of proteasome inhibitors such as bortezomib and carfilzomib into our standards of care against this disease. A minority of patients achieve complete remission with single-agent therapy, however, and proteasome inhibitors may induce on- and off-target anti-apoptotic effects and clinical toxicities that limit their utility. Unlike proteasome inhibitors, which have a broad impact on intracellular proteolysis, E3 ligases are responsible for ubiquitination of only a small subset of proteins. Our hypothesis proposed that inhibition of the Human double minute (HDM)-2 E3 ligase, which in part is responsible for p53 ubiquitination, could be a more rational therapeutic target for myeloma, where p53 mutations are less common than is the case in solid tumors. Methods: We used Nutlin-3a and MI-63, which bind to the p53 binding pocket of HDM-2, as prototypic agents, and studied their activity against both wild-type (wt) and mutant (mut) p53 cell line models of multiple myeloma. These studies were supported by the M. D. Anderson Cancer Center SPORE in Multiple Myeloma. Results: Inhibition of HDM-2 in wt p53 (MM1.S, H929, MOLP-8) cells with Nutlin-3a or MI-63 reduced viability, with a median inhibitory concentration (IC50) in the 0.5–2 μM range. In wt cells, genomic and proteomic studies showed that MI-63 induced transcription of downstream gene targets including p21, PUMA and Bax, and promoted accumulation of p53, p21, p27, and Bax. This was accompanied by cell cycle arrest at G1, and induction of apoptosis as demonstrated by Annexin V staining and caspase activation. Interestingly, these agents also showed activity against mut p53 cells such as RPMI 8226 and U266, though with higher IC50's of 30–40 μM. Knockdown of HDM-2 sensitized mut p53 myeloma cells to MI-63, suggesting that this agent was inducing cell death in an on-target manner. In p53 mutant cells, MI-63 induced both type I cell death and type II, autophagic cell death, as determined by conversion of microtubule-associated protein 1 light chain 3, and activation of transcription of autophagy (ATG) genes ATG3 and 5. Since autophagy can be a mechanism of cell survival or cell death, we tested the impact of the autophagy inhibitors chloroquine and 3-methyladenine, and found that they reduced the impact of MI-63 on viability. Moreover, knockdown of ATG5 helped rescue cells as well, supporting the possibility that autophagic activation was a mechanism of cell death in these myeloma models. Synergistic anti-myeloma effects were seen when MI-63 was combined with the BH3 mimetic ABT-737, with decreased viability and enhanced apoptosis, accompanied by increased Bax activation and caspase cleavage. Notably, ABT-737 also enhanced the effects of MI-63 on mut p53 myeloma models, where increased levels of both type I and II programmed cell death were seen. Conclusions: Together, our findings support the possibility that HDM-2 is a promising therapeutic target for multiple myeloma, and also provide a rationale for translation of HDM-2 inhibitors alone, and in combination with ABT-737, to the clinic. Disclosures: Leopold: Ascenta Therapeutics: Employment, Equity Ownership.

Cancers ◽  
2021 ◽  
Vol 13 (6) ◽  
pp. 1221
Author(s):  
Raquel Lopes ◽  
Bruna Velosa Ferreira ◽  
Joana Caetano ◽  
Filipa Barahona ◽  
Emilie Arnault Carneiro ◽  
...  

Despite the improvement of patient’s outcome obtained by the current use of immunomodulatory drugs, proteasome inhibitors or anti-CD38 monoclonal antibodies, multiple myeloma (MM) remains an incurable disease. More recently, the testing in clinical trials of novel drugs such as anti-BCMA CAR-T cells, antibody–drug conjugates or bispecific antibodies broadened the possibility of improving patients’ survival. However, thus far, these treatment strategies have not been able to steadily eliminate all malignant cells, and the aim has been to induce a long-term complete response with minimal residual disease (MRD)-negative status. In this sense, approaches that target not only myeloma cells but also the surrounding microenvironment are promising strategies to achieve a sustained MRD negativity with prolonged survival. This review provides an overview of current and future strategies used for immunomodulation of MM focusing on the impact on bone marrow (BM) immunome.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Peng Gao ◽  
Xianwei Ma ◽  
Ming Yuan ◽  
Yulan Yi ◽  
Guoke Liu ◽  
...  

AbstractUbiquitination is one of the most prevalent protein posttranslational modifications. Here, we show that E3 ligase Nedd4l positively regulates antiviral immunity by catalyzing K29-linked cysteine ubiquitination of TRAF3. Deficiency of Nedd4l significantly impairs type I interferon and proinflammatory cytokine production induced by virus infection both in vitro and in vivo. Nedd4l deficiency inhibits virus-induced ubiquitination of TRAF3, the binding between TRAF3 and TBK1, and subsequent phosphorylation of TBK1 and IRF3. Nedd4l directly interacts with TRAF3 and catalyzes K29-linked ubiquitination of Cys56 and Cys124, two cysteines that constitute zinc fingers, resulting in enhanced association between TRAF3 and E3 ligases, cIAP1/2 and HECTD3, and also increased K48/K63-linked ubiquitination of TRAF3. Mutation of Cys56 and Cys124 diminishes Nedd4l-catalyzed K29-linked ubiquitination, but enhances association between TRAF3 and the E3 ligases, supporting Nedd4l promotes type I interferon production in response to virus by catalyzing ubiquitination of the cysteines in TRAF3.


PLoS ONE ◽  
2014 ◽  
Vol 9 (9) ◽  
pp. e103015 ◽  
Author(s):  
Dongmin Gu ◽  
Shuhong Wang ◽  
Isere Kuiatse ◽  
Hua Wang ◽  
Jin He ◽  
...  

2018 ◽  
Vol 92 (8) ◽  
Author(s):  
E. Kip ◽  
J. Staal ◽  
L. Verstrepen ◽  
H. G. Tima ◽  
S. Terryn ◽  
...  

ABSTRACTMALT1 is involved in the activation of immune responses, as well as in the proliferation and survival of certain cancer cells. MALT1 acts as a scaffold protein for NF-κB signaling and a cysteine protease that cleaves substrates, further promoting the expression of immunoregulatory genes. Deregulated MALT1 activity has been associated with autoimmunity and cancer, implicating MALT1 as a new therapeutic target. Although MALT1 deficiency has been shown to protect against experimental autoimmune encephalomyelitis, nothing is known about the impact of MALT1 on virus infection in the central nervous system. Here, we studied infection with an attenuated rabies virus, Evelyn-Rotnycki-Abelseth (ERA) virus, and observed increased susceptibility with ERA virus in MALT1−/−mice. Indeed, after intranasal infection with ERA virus, wild-type mice developed mild transient clinical signs with recovery at 35 days postinoculation (dpi). Interestingly, MALT1−/−mice developed severe disease requiring euthanasia at around 17 dpi. A decreased induction of inflammatory gene expression and cell infiltration and activation was observed in MALT1−/−mice at 10 dpi compared to MALT1+/+infected mice. At 17 dpi, however, the level of inflammatory cell activation was comparable to that observed in MALT1+/+mice. Moreover, MALT1−/−mice failed to produce virus-neutralizing antibodies. Similar results were obtained with specific inactivation of MALT1 in T cells. Finally, treatment of wild-type mice with mepazine, a MALT1 protease inhibitor, also led to mortality upon ERA virus infection. These data emphasize the importance of early inflammation and activation of T cells through MALT1 for controlling the virulence of an attenuated rabies virus in the brain.IMPORTANCERabies virus is a neurotropic virus which can infect any mammal. Annually, 59,000 people die from rabies. Effective therapy is lacking and hampered by gaps in the understanding of virus pathogenicity. MALT1 is an intracellular protein involved in innate and adaptive immunity and is an interesting therapeutic target because MALT1-deregulated activity has been associated with autoimmunity and cancers. The role of MALT1 in viral infection is, however, largely unknown. Here, we study the impact of MALT1 on virus infection in the brain, using the attenuated ERA rabies virus in different models of MALT1-deficient mice. We reveal the importance of MALT1-mediated inflammation and T cell activation to control ERA virus, providing new insights in the biology of MALT1 and rabies virus infection.


2001 ◽  
Vol 355 (3) ◽  
pp. 587-595 ◽  
Author(s):  
Paola MATARRESE ◽  
Ugo TESTA ◽  
Roberto CAUDA ◽  
Stefano VELLA ◽  
Lucrezia GAMBARDELLA ◽  
...  

Multidrug resistance caused by P-glycoprotein (P-170) is a phenomenon by which cells exposed to a single drug acquire resistance to other structurally and functionally unrelated drugs. This is a widespread phenomenon described in vivo in the management of infectious as well as non-infectious diseases. Several in vitro models have been developed in order to evaluate physiopathological properties of P-170. Among these are P-170-expressing variants of the human T-lymphoblastoid CEM cell line called VBL100. As a general rule, drug resistance normally results in resistance to apoptosis induction. By contrast, a paradoxical activity is exerted in this cell model by the cytokine tumour necrosis factor-α (TNF-α), which is capable of inducing apoptosis in P-170-expressing variants better than in wild-type (wt) cells. In the present study we partially address the mechanisms underlying this activity. In fact, the susceptibility of VBL100 cells to TNF-α appears to be specifically due to the depolarization of their mitochondrial membrane, a key factor for apoptotic induction. The same was observed with staurosporine, a specific mitochondrion-mediated proapoptotic chemical probe. Conversely, other proapoptotic stimuli, such as Fas/CD95 or the anti-cancer drug etoposide, did induce significant cell death in wild type cells only. Thus, schematically, mitochondrially dependent stimuli appeared to be more effective in VBL100-cell killing, while ‘physiological’ stimuli showed the opposite behaviour. Importantly, under steady-state conditions, VBL100 cells displayed per se a mitochondrial membrane hyperpolarization that appeared strictly related to their high susceptibility to specific apoptotic stimuli. In conclusion, the study of a well-established cell model such as that represented by the wt/VBL CEM lymphoid cell line seems to suggest that the multidrug resistance phenotype can specifically sensitize cells towards ‘unphysiological’, mitochondria-associated cell death cascade or, in the same fashion, it could shift cells from type I (mainly plasma membrane-associated) towards type II (mainly mitochondrial membrane-associated) phenotype.


Blood ◽  
2009 ◽  
Vol 113 (23) ◽  
pp. 5927-5937 ◽  
Author(s):  
Encouse B. Golden ◽  
Philip Y. Lam ◽  
Adel Kardosh ◽  
Kevin J. Gaffney ◽  
Enrique Cadenas ◽  
...  

Abstract The anticancer potency of green tea and its individual components is being intensely investigated, and some cancer patients already self-medicate with this “miracle herb” in hopes of augmenting the anticancer outcome of their chemotherapy. Bortezomib (BZM) is a proteasome inhibitor in clinical use for multiple myeloma. Here, we investigated whether the combination of these compounds would yield increased antitumor efficacy in multiple myeloma and glioblastoma cell lines in vitro and in vivo. Unexpectedly, we discovered that various green tea constituents, in particular (-)-epigallocatechin gallate (EGCG) and other polyphenols with 1,2-benzenediol moieties, effectively prevented tumor cell death induced by BZM in vitro and in vivo. This pronounced antagonistic function of EGCG was evident only with boronic acid–based proteasome inhibitors (BZM, MG-262, PS-IX), but not with several non–boronic acid proteasome inhibitors (MG-132, PS-I, nelfinavir). EGCG directly reacted with BZM and blocked its proteasome inhibitory function; as a consequence, BZM could not trigger endoplasmic reticulum stress or caspase-7 activation, and did not induce tumor cell death. Taken together, our results indicate that green tea polyphenols may have the potential to negate the therapeutic efficacy of BZM and suggest that consumption of green tea products may be contraindicated during cancer therapy with BZM.


2017 ◽  
Vol 35 (15_suppl) ◽  
pp. e18096-e18096
Author(s):  
Anne Roc ◽  
Wendy Turell ◽  
David Dingli

e18096 Background: Newly approved and emerging agents with novel mechanisms of action present unique challenges to treating patients with relapsed/refractory multiple myeloma (RRMM), including keeping abreast of rapid and ever-aggregating data on their safety, tolerability and efficacy. To address these challenges, an education activity focused on new and emerging therapies for RRMM was developed and learner responses were evaluated to determine the impact of education. Methods: A live-online 1-hour video panel discussion with slides, polling, and live questions was produced in 2016 and made available on-demand at OMedLive.com for 6 months. Survey responses (pre-, post-, 8 weeks post-activity), polling responses, and live questions asked were tracked to measure engagement, lessons learned, and additional education gaps. Results: 51 of 288 learners completed all pre/post/follow-up surveys, 67% of which reported the activity positively impacted patient experience or outcome and 61% reported it positively impacted their clinical practice. These learners reported improvements in their ability to: assess the relative safety and efficacy of emerging agents (50%), differentiate available agents and their relative efficacy (50% with proteasome inhibitors, 39% with monoclonal antibodies), see the potential benefit of HDAC inhibitors (35%), use combination regimens (29%), and adjust treatments for patients who’ve experienced disease progression (29%). As the result of education, 95 of 288 learners reported commitments to change in regards to: medical/practice knowledge (95%), care attitudes (89%), practice behavior (76%), and patient clinical outcomes (81%). Learners also demonstrated improved competence via a case scenario which required identifying the optimal next step for a patient with biochemical progression, and improved knowledge on the characteristics of daratumumab, elotuzumab, and selinexor. Conclusions: New and emerging therapies promise to improve the lives of patients with RRMM, but complicates the already difficult task of managing the disease. Ongoing education on RRMM is recommended and can yield immediate and sustained gains in knowledge, competence, and performance.


2021 ◽  
Vol 102 (10) ◽  
Author(s):  
Rheza Gandi Bawono ◽  
Takayuki Abe ◽  
Mengting Qu ◽  
Daisuke Kuroki ◽  
Lin Deng ◽  
...  

Ubiquitin and ubiquitin-like protein modification play important roles in modulating the functions of viral proteins in many viruses. Here we demonstrate that hepatitis B virus (HBV) X protein (HBx) is modified by ISG15, which is a type I IFN-inducible, ubiquitin-like protein; this modification is called ISGylation. Immunoblot analyses revealed that HBx proteins derived from four different HBV genotypes accepted ISGylation in cultured cells. Site-directed mutagenesis revealed that three lysine residues (K91, K95 and K140) on the HBx protein, which are well conserved among all the HBV genotypes, are involved in acceptance of ISGylation. Using expression plasmids encoding three known E3 ligases involved in the ISGylation to different substrates, we found that HERC5 functions as an E3 ligase for HBx-ISGylation. Treatment with type I and type III IFNs resulted in the limited suppression of HBV replication in Hep38.7-Tet cells. When cells were treated with IFN-α, silencing of ISG15 resulted in a marked reduction of HBV replication in Hep38.7-Tet cells, suggesting a role of ISG15 in the resistance to IFN-α. In contrast, the silencing of USP18 (an ISG15 de-conjugating enzyme) increased the HBV replication in Hep38.7-Tet cells. Taken together, these results suggest that the HERC5-mediated ISGylation of HBx protein confers pro-viral functions on HBV replication and participates in the resistance to IFN-α-mediated antiviral activity.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 785-785
Author(s):  
Tricia Nardiello ◽  
Achim A Jungbluth ◽  
Anna Mei ◽  
Maurizio DiLiberto ◽  
Xiangao Huang ◽  
...  

Abstract Abstract 785 The type I Melanoma Antigen GEne (MAGE) MAGE-A3 is commonly present in primary multiple myeloma cells and its expression is correlated with advanced disease and proliferation. MAGE-A3 belongs to the Cancer-Testis antigen (CTAg) family of tumor-associated proteins, which are present in many cancers, but their normal expression is limited to developing germ cells and placental trophoblast. This unique expression pattern fuels speculation on a role for CTAg in oncogenesis; however, very little is known about their function. In gene expression analyses of primary myeloma cells, CTAg were associated with proliferative gene signatures and poor clinical outcome, suggesting they contribute to the pathogenesis or progression of this disease through effects on survival and/or proliferation of myeloma cells. To investigate this, we examined the impact of MAGE-A on disease progression, proliferation, and apoptosis in primary myeloma specimens and human myeloma cell lines (HMCL). MAGE-A3 protein expression was examined by immunohistochemistry in a new, independent set of myeloma bone marrow specimens from two critical clinical milestones, newly diagnosed, untreated patients and patients who relapsed after chemotherapy. MAGE-A3 was detected in a higher percentage of tumor specimens from relapsed patients (77%) compared to those from newly diagnosed patients (36%, p=0.0003). The percentage of proliferating myeloma cells, as measured by staining for the proliferation marker Ki-67, was significantly higher in relapsed specimens (19.0 ± 3.5%) compared to newly diagnosed (6.9 ± 1.3%, p=0.0002), demonstrating a correlation between MAGE-A3, progression of disease and proliferation. The mechanisms for MAGE-A3 activity were investigated by silencing this gene in primary myeloma cells and HMCL by shRNA interference. Targeted lentiviral shRNA transduction efficiently knocked down MAGE-A3 mRNA and protein in MM.1r (p53+/+) and ARP-1 (p53−/−) HMCL and in primary myeloma cells by 48 hours, and this effect was maintained up to 96 hours. Silencing of MAGE-A did not affect cell cycling, as this intervention did not affect the phosphorylation of the Retinoblastoma gene product (Rb) that is required for progression through the G1 cell cycle checkpoints and entry into S phase. In contrast, MAGE-A was required for survival of proliferating myeloma cells. Silencing of MAGE-A led to a precipitous loss of viable cells within 48–72 hrs compared to controls. This was due to activation of intrinsic apoptosis, as demonstrated by increased annexin V staining, loss of mitochondrial membrane polarization, and cleavage/activation of caspase-9. These effects of MAGE-A knock-down were completely reversed by the pan-caspase inhibitor Quinoline-Val-Asp-CH2-OPh. Apoptosis after MAGE-A silencing appeared to be mediated by at least two distinct mechanisms; p53-dependent activation of pro-apoptotic Bax and Bak expression and reduced expression of the Inhibitor of Apoptosis Protein survivin through both p53-dependent and independent mechanisms. These results demonstrate that MAGE-A plays a role in the survival of proliferating multiple myeloma cells through the regulation of two critical apoptotic mechanisms. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3510-3510
Author(s):  
Sneha V. Gupta ◽  
Erin K Hertlein ◽  
Jennifer A. Woyach ◽  
Ellen J. Sass ◽  
Melanie E. Davis ◽  
...  

Abstract Abstract 3510 The reversible proteasome inhibitor bortezomib is effective in the treatment of multiple myeloma and mantle cell lymphoma, but failed to produce objective responses in chronic lymphocytic leukemia (CLL). Carfilzomib (CFZ) is a tetrapeptide ketoepoxide that belongs to a new class of irreversible proteasome inhibitors that specifically target the chymotrypsin-like subunit of the 26S proteasome. Based on preclinical data demonstrating potent cytotoxicity in primary CLL cells, CFZ is currently in a phase I clinical trial at The Ohio State University in patients with relapsed or refractory CLL. However, the mechanism of action of CFZ in CLL is unknown. We have therefore investigated the effects of CFZ on CLL patient cells ex vivo. Here we demonstrate that a short (1 hr) exposure of 100 nM CFZ is sufficient to inhibit the chymotrypsin-like proteasome subunit in CLL cells. This exposure is also rapidly cytotoxic, inducing apoptosis in approximately 50% of cells by 24 hr (annexin+ and/or PI+). Unlike bortezomib, the cytotoxicity of carfilzomib is not diminished in media with human serum compared to fetal bovine serum. Additionally, CFZ is more cytotoxic to normal CD19+ B cells than normal CD3+ T cells at clinically relevant concentrations of 33 to 300 nM, suggesting that this agent could potentially avoid immune-suppressing T-cell depletion that is commonly noted with chemotherapy. CFZ causes CLL cell death ex vivo by a caspase-dependent apoptotic pathway, indicated by PARP cleavage and rescue by the broad caspase inhibitor Boc-D-fmk. Importantly, our studies indicate that CFZ causes cytotoxicity in primary CLL cells irrespective of p53 status. This tumor suppressor, which is functional in most CLL patients at the time of diagnosis, is mutated or deleted in at least one allele in approximately 40% of patients with advanced CLL, and p53 pathway dysfunction is associated with resistance to standard therapies and poor overall survival. Therefore, the ability of CFZ to induce apoptosis irrespective of p53 function is of important clinical significance. Additionally, the pro-apoptotic protein Noxa is increased following CFZ treatment despite a lack of induction in mRNA, suggesting accumulation of protein via inhibition of proteasome-mediated degradation. The NF-kB signaling pathway is broadly implicated in CLL cell survival and resistance to therapy, and proteasome inhibitors have been reported to block this pathway via inhibition of IkB degradation. We therefore investigated the impact of CFZ on NF-kB-mediated transcription in CLL patient cells. Paradoxically, our results indicate that CFZ leads to activation of NF-kB, as evidenced by increased nuclear accumulation of the p50 and p65 subunits of NF-kB, as well as phosphorylated IkBα. This correlates with enhanced binding of the p50/p65 heterodimer to an NF-kB probe in an electrophoretic mobility shift assay. However, despite this apparent NF-kB activation, no transcriptional increases were observed in NF-kB targets genes including Mcl-1, p53, Bcl-2, Bcl2A1 or XIAP. In addition, inhibition of NF-kB activity using a dominant negative (non-phosphorylatable) IkBα did not impair CFZ-induced cytotoxicity. This is the first study suggesting that treatment with a proteasome inhibitor induces a defective NF-κB response in CLL cells. The mechanism and relevance of this effect, as well as the pathway by which CFZ causes CLL cell death, continues to be investigated. Collectively, our data indicate that proteasome inhibition is a relevant therapeutic target in CLL and supports the development of carfilzomib for the treatment of this currently incurable disease. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document