880 resultados para PROTEASOME INHIBITORS
Resumo:
Peptides derived from cytosolic, mitochondrial, and nuclear proteins have been detected in extracts of animal tissues and cell lines. To test whether the proteasome is involved in their formation, HEK293T cells were treated with epoxomicin (0.2 or 2 mu M) for 1 h and quantitative peptidomics analysis was performed. Altogether, 147 unique peptides were identified by mass spectrometry sequence analysis. Epoxomicin treatment decreased the levels of the majority of intracellular peptides, consistent with inhibition of the proteasome beta-2 and beta-5 subunits. Treatment with the higher concentration of epoxomicin elevated the levels of some peptides. Most of the elevated peptides resulted from cleavages at acidic residues, suggesting that epoxomicin increased the processing of proteins through the beta-1 subunit. Interestingly, some of the peptides that were elevated by the epoxomicin treatment had hydrophobic residues in P1 cleavage sites. Taken together, these findings suggest that, while the proteasome is the major source of intracellular peptides, other peptide-generating mechanisms exist. Because intracellular peptides are likely to perform intracellular functions, studies using proteasome inhibitors need to be interpreted with caution, as it is possible that the effects of these inhibitors are due to a change in the peptide levels rather than inhibition of protein degradation.
Resumo:
The use of proteasome inhibitors in cancer has received much attention with the recent FDA approval of bortezomib (Velcade/PS-341). However, in the chronic lymphocytic leukemia (CLL) clinical trial, bortezomib was not as effective as it was in vitro. Accordingly, results in prostate cancer were not remarkable, although regression of lymphadenopathy was observed. This response was also seen in CLL. ^ The proteasome degrades ∼80% of intracellular proteins. Although specific pathways affected by proteasome inhibitors are known, there are still unidentified mechanisms by which they induce apoptosis. The efficacy and mechanism of action of the reversible proteasome inhibitor bortezomib were compared to the novel irreversible inhibitor NPI-0052 in this study, and their mechanisms of action in CLL and prostate cancer were examined. ^ NPI-0052 inhibited proteasome activity and induced apoptosis with more rapid kinetics than bortezomib in CLL. Inhibition of proteasome activity with NPI-0052 was also more durable. Interestingly, bortezomib is cleared from the serum within 15min, which is insufficient time for bortezomib to effectively inhibit the proteasome. However, only 5min exposure was needed for NPI-0052 to produce maximal proteasome inhibition. The data suggest that bortezomib's slow kinetics and reversible nature limit its potential in vivo and the use of NPI-0052 should be considered. ^ In examining the mechanism(s) by which bortezomib and NPI-0052 induce apoptosis in CLL, both were found to elicit the ER stress pathway. A stromal cell co-culture system prevented apoptosis induced by both proteasome inhibitors, suggesting that if such factors in vivo were responsible for reducing bortezomib's efficacy, NPI-0052 would not prove useful either. Finally, Lyn, a Src family kinase (SFK), was decreased in response to bortezomib and NPI-0052 and correlated with apoptosis induction in CLL and prostate cancer. Both proteasome inhibitors specifically targeted Lyn rather than SFKs in general. ^ SFKs are overexpressed in cancer and involved in cell signaling, survival, and metastasis. In prostate cancer cells, both proteasome inhibition and Lyn-silencing significantly inhibited migration. Preliminary evidence also suggested that Lyn downregulation decreases invasion potential. Together, these data suggest that proteasome inhibitors are potential candidates for anti-metastasic therapy and further investigation is warranted for the use of Lyn-targeted therapy to treat metastases. ^
Resumo:
The proteasome degrades approximately 80% of intracellular proteins to maintain homeostasis. Proteasome inhibition is a validated therapeutic strategy, and currently, proteasome inhibitor bortezomib is FDA approved for the treatment of MM and MCL. Specific pathways affected by proteasome inhibition have been identified, but mechanisms of the anti-tumor effects of proteasome inhibition are not fully characterized and cancer cells display marked heterogeneity in terms of their sensitivity to proteasome inhibitor induced cell death. ^ The antitumor effects of proteasome inhibition involve suppression of tumor angiogenesis and vascular endothelial growth factor (VEGF) expression, but the mechanisms involved have not been clarified. In this dissertation I investigated the mechanisms underlying the effects of two proteasome inhibitors, bortezomib and NPI-0052, on VEGF expression in human prostate cancer cells. I found that proteasome inhibitors selectively downregulated hypoxia inducible factor 1alpha (HIF-1α) protein and its transcriptional activity to inhibit VEGF expression. Mechanistic studies demonstrated that proteasome inhibitors mediate the induction of the unfolded protein response (UPR) and that downregulation of HIF-1α is caused by eukaryotic translation initiation factor 2α (eIF2α) phosphorylation and translation repression. Importantly, I showed that proteasome inhibitors activated the UPR in some cells but not in others. My observation may have implications for the design of combination regimens that are based on exploiting proteasome inhibitor-induced ER stress.^ Although proteasome inhibitors have shown modest activity on prostate cancer, there is general consensus that no single agent is likely to have significant activity in prostate cancer. In the second part of this dissertation I attempted to exploit the effects of proteasome inhibition on the UPR to design a combination therapy that would enhance cancer cell death. Autophagy is a lysosome dependent degradation pathway that functions to eliminate long-lived protein and subcellular structures. Targeting autophagy has been shown to inhibit tumors in preclinical studies. I found that inhibition of autophagy with chloroquine or 3-methyladenine enhanced proteasome inhibitor induced cell death and the effects were associated with increased intracellular stress as marked by aggresome formation. Multiple cancers appear to be resistant to proteasome inhibition treatment alone. The implications of synergy for the combined inhibition of autophagy and the proteasome would likely apply to other cancers aside from prostate cancer. ^
Resumo:
In eukaryotic cells, the ubiquitin–proteasome pathway is the major mechanism for the targeted degradation of proteins with short half-lives. The covalent attachment of ubiquitin to lysine residues of targeted proteins is a signal for the recognition and rapid degradation by the proteasome, a large multi-subunit protease. In this report, we demonstrate that the human estrogen receptor (ER) protein is rapidly degraded in mammalian cells in an estradiol-dependent manner. The treatment of mammalian cells with the proteasome inhibitor MG132 inhibits activity of the proteasome and blocks ER degradation, suggesting that ER protein is turned over through the ubiquitin–proteasome pathway. In addition, we show that in vitro ER degradation depends on ubiquitin-activating E1 enzyme (UBA) and ubiquitin-conjugating E2 enzymes (UBCs), and the proteasome inhibitors MG132 and lactacystin block ER protein degradation in vitro. Furthermore, the UBA/UBCs and proteasome inhibitors promote the accumulation of higher molecular weight forms of ER. The UBA and UBCs, which promote ER degradation in vitro, have no significant effect on human progesterone receptor and human thyroid hormone receptor β proteins.
Resumo:
Retrovirus assembly and maturation involve folding and transport of viral proteins to the virus assembly site followed by subsequent proteolytic cleavage of the Gag polyprotein within the nascent virion. We report that inhibiting proteasomes severely decreases the budding, maturation, and infectivity of HIV. Although processing of the Env glycoproteins is not changed, proteasome inhibitors inhibit processing of Gag polyprotein by the viral protease without affecting the activity of the HIV-1 viral protease itself, as demonstrated by in vitro processing of HIV-1 Gag polyprotein Pr55. Furthermore, this effect occurs independently of the virus release function of the HIV-1 accessory protein Vpu and is not limited to HIV-1, as proteasome inhibitors also reduce virus release and Gag processing of HIV-2. Electron microscopy analysis revealed ultrastructural changes in budding virions similar to mutants in the late assembly domain of p6gag, a C-terminal domain of Pr55 required for efficient virus maturation and release. Proteasome inhibition reduced the level of free ubiquitin in HIV-1-infected cells and prevented monoubiquitination of p6gag. Consistent with this, viruses with mutations in PR or p6gag were resistant to detrimental effects mediated by proteasome inhibitors. These results indicate the requirement for an active proteasome/ubiquitin system in release and maturation of infectious HIV particles and provide a potential pharmaceutical strategy for interfering with retrovirus replication.
Resumo:
A library of 426 FDA-approved drugs was screened for in vitro activity against E. multilocularis metacestodes employing the phosphoglucose isomerase (PGI) assay. Initial screening at 20 µM revealed that 7 drugs induced considerable metacestode damage, and further dose-response studies revealed that bortezomib (BTZ), a proteasome inhibitor developed for the chemotherapy of myeloma, displayed high anti-metacestodal activity with an EC50 of 0.6 µM. BTZ treatment of E. multilocularis metacestodes led to an accumulation of ubiquinated proteins and unequivocally parasite death. In-gel zymography assays using E. multilocularis extracts demonstrated BTZ-mediated inhibition of protease activity in a band of approximately 23 kDa, the same size at which the proteasome subunit beta 5 of E. multilocularis could be detected by Western blot. Balb/c mice experimentally infected with E. multilocularis metacestodes were used to assess BTZ treatment, starting at 6 weeks post-infection by intraperitoneal injection of BTZ. This treatment led to reduced parasite weight, but to a degree that was not statistically significant, and it induced adverse effects such as diarrhea and neurological symptoms. In conclusion, the proteasome was identified as a drug target in E. multilocularis metacestodes that can be efficiently inhibited by BTZ in vitro. However, translation of these findings into in vivo efficacy requires further adjustments of treatment regimens using BTZ, or possibly other proteasome inhibitors.
Resumo:
Protein degradation is regulated during the cell cycle of all eukaryotic cells and is mediated by the ubiquitin-proteasome pathway. Potent and specific peptide-derived inhibitors of the 20S proteasome have been developed recently as anti-cancer agents, based on their ability to induce apoptosis in rapidly dividing cells. Here, we tested a novel small molecule dipeptidyl boronic acid proteasome inhibitor, named MLN-273 on blood and liver stages of Plasmodium species, both of which undergo active replication, probably requiring extensive proteasome activity. The inhibitor blocked Plasmodium falciparum erythrocytic development at an early ring stage as well as P. berghei exoerythrocytic progression to schizonts. Importantly, neither uninfected erythrocytes nor hepatocytes were affected by the drug. MLN-273 caused an overall reduction in protein degradation in P. falciparum, as demonstrated by immunoblots using anti-ubiquitin antibodies to label ubiquitin-tagged protein conjugates. This led us to conclude that the target of the drug was the parasite proteasome. The fact that proteasome inhibitors are presently used as anti-cancer drugs in humans forms a solid basis for further development and makes them potentially attractive drugs also for malaria chemotherapy.
Resumo:
The ability of angiotensin I (Ang I) and II (Ang II) to induce directly protein degradation in skeletal muscle has been studied in murine myotubes. Angiotensin I stimulated protein degradation with a parabolic dose-response curve and with a maximal effect between 0.05 and 0.1 μM. The effect was attenuated by coincubation with the angiotensin-converting enzyme (ACE) inhibitor imidaprilat, suggesting that angiotensin I stimulated protein degradation through conversion to Ang II. Angiotensin II also stimulated protein breakdown with a similar dose-response curve, and with a maximal effect between 1 and 2.5 μM. Total protein degradation, induced by both Ang I and Ang II, was attenuated by the proteasome inhibitors lactacystin (5 μM) and MG132 (10 μM), suggesting that the effect was mediated through upregulation of the ubiquitin-proteasome proteolytic pathway. Both Ang I and Ang II stimulated an increased proteasome 'chymotrypsin-like' enzyme activity as well as an increase in protein expression of 20S proteasome α-subunits, the 19S subunits MSSI and p42, at the same concentrations as those inducing protein degradation. The effect of Ang I was attenuated by imidaprilat, confirming that it arose from conversion to Ang II. These results suggest that Ang II stimulates protein degradation in myotubes through induction of the ubiquitin-proteasome pathway. Protein degradation induced by Ang II was inhibited by insulin-like growth factor and by the polyunsaturated fatty acid, eicosapentaenoic acid. These results suggest that Ang II has the potential to cause muscle atrophy through an increase in protein degradation. The highly lipophilic ACE inhibitor imidapril (Vitor™) (30 mg kg-1) attenuated the development of weight loss in mice bearing the MAC16 tumour, suggesting that Ang II may play a role in the development of cachexia in this model. © 2005 Cancer Research.
Resumo:
9 p.
Resumo:
Chronic myeloid leukemia (CML) is treated effectively with tyrosine kinase inhibitors (TKIs); however, 2 key problems remain-the insensitivity of CML stem and progenitor cells to TKIs and the emergence of TKI-resistant BCR-ABL mutations. BCR-ABL activity is associated with increased proteasome activity and proteasome inhibitors (PIs) are cytotoxic against CML cell lines. We demonstrate that bortezomib is antiproliferative and induces apoptosis in chronic phase (CP) CD34(+) CML cells at clinically achievable concentrations. We also show that bortezomib targets primitive CML cells, with effects on CD34(+)38(-), long-term culture-initiating (LTC-IC) and nonobese diabetic/severe combined immunodeficient (NOD/SCID) repopulating cells. Bortezomib is not selective for CML cells and induces apoptosis in normal CD34(+)38(-) cells. The effects against CML cells are seen when bortezomib is used alone and in combination with dasatinib. Bortezomib causes proteasome but not BCR-ABL inhibition and is also effective in inhibiting proteasome activity and inducing apoptosis in cell lines expressing BCR-ABL mutations, including T315I. By targeting both TKI-insensitive stem and progenitor cells and TKI-resistant BCR-ABL mutations, we believe that bortezomib offers a potential therapeutic option in CML. Because of known toxicities, including myelosuppression, the likely initial clinical application of bortezomib in CML would be in resistant and advanced disease. (Blood. 2010;115:2241-2250)
Resumo:
Background BRCA1 and cyclin D1 are both essential for normal breast development and mutation or aberration of their expression is associated with breast cancer [1,2]. Cyclin D1 is best known as a G1 cyclin where it regulates the G1 to S phase transition by acting as a rate-limiting subunit of CDK4/6 kinase activity. More recently, however, Stacey has demonstrated that cyclin D1 levels in G2/M determine whether a cell continues to proliferate or exits the cell cycle [3]. The majority of BRCA1 in the cell is bound to BARD1 through their N-terminal RING domains. Heterodimerization is essential for the stability and correct localization of the complex and confers ubiquitin ligase activity to BRCA1. The importance of the ligase activity of BRCA1 to breast cancer development is inferred from the fact that N-terminal diseaseassociated mutations are proposed to reduce ligase activity [4]. Methods Protein–protein interactions were demonstrated using yeast-two-hybrid and coimmunoprecipitation. Protein levels were altered through overexpression, siRNA and antisense technology. The effect of proteasome inhibitors and cycloheximide treatment was also examined. Results We initially identified cyclin D1 as a binding partner of BARD1 in a yeast-two-hybrid screen and defined the minimal binding region as the N-terminus of BARD1. This interaction was confirmed in vivo by coimmunoprecipitation. The N-terminus of BARD1 also binds BRCA1 and imparts ubiquitin ligase activity to the complex. Covalent modification of proteins with ubiquitin is a common regulatory mechanism in eukaryotic cells. Traditionally polyubiquitin chains linked through lysine 48 target proteins for degradation by the 26 S proteasome. We have demonstrated that cyclin D1 protein levels are inversely related to BRCA1 and BARD1 levels in several model systems. Furthermore, regulation of cyclin D1 levels occurs through a post-transcriptional mechanism and requires the ligase activity of BRCA1. Interestingly, this phenomenon is cell-cycle regulated, occurring in G2/M. Conclusion We propose that cyclin D1 is a potential substrate for BRCA1 ubiquitination and that this targets cyclin D1 for proteasomal-mediated degradation. Future work will focus on ascertaining the functional consequence of cyclin D1 regulation by the BRCA1–BARD1 complex; in particular, the impact of BRCA1, mediated through regulation of cyclin D1, on the proliferation versus differentiation decision.
Resumo:
HHV-6 is a ubiquitous human herpesvirus. Most individuals become infected at the age of 2 years. Primary infection by the virus causes a self-limiting febrile illness called exanthem subitum or roseola. In adults, primary infection may cause mononucleosis-like illnesses. The infection usually remains latent in healthy individuals, but often reactivates in immunocompromised individuals, for example, transplant patients and AIDS patients. The virus has also been associated with cancers and lymphoproliferative disorders. The virus encodes two proteins that interact with p53. However, little is known concerning the impact of the virus on cell cycle progression in human cells. The investigations reported in the thesis were focused on this issue. We show here that that HHV-6 infection delays the cell cycle progression in human T cell line HSB-2, as well as in primary human T cells and causes their accumulation in S and G2/M phase. By degrading the viral DNA in the virus-infected cells, we show that the infected cells accumulate in the G2/M and not in the S phase. We observed an increase in the kinase activity of cdc2 in virus-infected cells despite lower levels of its catalytic partners, cyclin A and cyclin B. We show here that the viral early antigen p41 associates with, and increases the kinase activity of, CDK1. Our studies have shown that there is a drastic reduction of p21 protein, despite the virus-induced stabilization and activation of p53 suggesting that p53 may be transcriptionally inactivated in the virus-infected cells. This decrease of p21 in infected cells was partially restored by proteasome inhibitors. These results suggest that HHV-6 causes perturbations in the normal progression of cell cycle in human T cells. Autophagy is a physiological cell process during which old cellular constituents and long-lived proteins in cells are degraded. This process is regulated in a cell cycle-dependent manner. We show here that infection with HHV-6 induces autophagy in HSB-2 cells. This was shown by the induction of LC-3 II as well as by the appearance of autophagic vacuoles in the virus-infected cells. However, we found that the virus inhibits fusion between autophagic vacuoles and lysosomes formed in infected cells, thus evading the autophagic response of infected host cells. Finally we tried to investigate replication of the virus in human cells in the absence of P53; a tumor suppressor gene which is also known as "the guardian of the genome ". During these investigations, we found that that inhibition of p53 gene expression mediated by siRNA as well as its inhibition by pharmacological inhibitors leads to massive cell death in human T cell line HSB-2 that carries a wild-type p53. We show that this death also occurs in another cell line CEM, which carries a transcriptionally mutated p53. Interestingly, the cell death could be prevented by pharmacological inhibitors of autophagy and necroptosis. Taken together, our results provide important novel insights concerning the impact of HHV-6 on cell cycle regulation and autophagy as well as of basal level p53 in cell survival.
Resumo:
Das Zytoskelett eukaryotischer Zellen besteht aus drei verschiedenen Protein-Netzwerken: den Aktinfilamenten, Mikrotubuli und Intermediärfilamenten. Intermediärfilamente wurden ursprünglich als statische Strukturen angesehen, die die mechanische Stabilisierung der Zellen übernehmen. In den letzten Jahren hat sich dieses Bild jedoch geändert: Intermediärfilament-Netzwerke sind hochdynamisch und unterliegen kontinuierlichen Veränderungen, welche durch Phosphorylierungen reguliert werden. Sie interagieren mit anderen Zytoskelett-Proteinen und greifen in die Regulation von Schlüsselsignalwegen, die Zellwachstum und Zellteilung sowie Apoptose und Stressantwort bestimmen, ein. Die Mechanismen der Filamentplastizität konnten bisher jedoch nicht vollständig aufgeklärt werden. So ist beispielsweise unklar, wo Auf- und Abbau der Filamente stattfindet und welche Faktoren an der Netzwerkmodulation beteiligt sind. Ziel meiner Arbeit war es, einen Beitrag zur Aufklärung dieser Mechanismen am Beispiel der epithelialen Keratin-Intermediärfilamente zu leisten. Mit Hilfe von mikroskopischen Zeitrafferaufnahmen von fluoreszenzmarkierten Zellklonen wurden Nukleationszentren in der Zellperipherie identifiziert, in denen Keratinfilamentvorläufer gebildet werden. Es handelt sich dabei um fokale Adhäsionskomplexe, die als Anheftungsstellen zwischen der extrazellulären Matrix und dem intrazellulären Aktinfilament-System dienen. Es konnte gezeigt werden, dass diese Filamentvorläufer-Entstehung für alle untersuchten Keratinisoformen gültig ist und in epitelialen als auch nicht-epithelialen Zelltypen abläuft. Knock-Down der Adhäsionskomponente Talin verhinderte die Keratinfilamentbildung. Modulation der fokalen Adhäsionskinase, die den Auf- und Abbau der Adhäsionskomplexe koordiniert, beeinflusste ebenso die Bildung der Keratinfilamentnetzwerke. Es konnte weiterhin beobachtet werden, dass die N-terminalen Isoformen IE und IF des Zytolinkers Plectin in fokalen Adhäsionen lokalisieren und damit möglicherweise an der Vernetzung von Keratinfilamentvorläufern, Zelladhäsionen und Aktinfilamenten beteiligt sind. Letztlich stellte sich heraus, dass die Bildung der Keratinfilamentvorläufer unabhängig von Proteintranslation ist. In den mikroskopischen Zeitrafferaufnahmen wurde im Anschluss an die Keratinfilamentbildung ein kontinuierlicher zentripetaler Transport der wachsenden Vorläuferpartikel beobachtet. An Hand von pharmakologischen Experimenten konnte gezeigt werden, dass dieser Transport Aktinfilament-abhängig ist. Zeitgleich kommt es zu Partikelfusion und Integration in das periphere Netzwerk, das sich weiterhin in Richtung auf das Zellzentrum bewegt. Mit Hilfe von Photoaktivierungsversuchen und Zellfusionsexperimenten konnte die Hypothese bestätigt werden, dass der Abbau der einwandernden Keratinfilamente in lösliche, rasch diffusible Zwischenstufen den kontinuierlichen peripheren Neuaufbau ermöglicht. Aus den Beobachtungen und bereits bekannten Ergebnissen wurde ein Modell des Keratin-Zyklus entwickelt, das die folgenden Stadien umfasst: Nukleation von Keratinfilamentvorläufern an fokalen Adhäsionen in der Zellperipherie, Elongation und Fusion der Keratinfilamentvorläufer bei zeitgleichem Aktinfilament-abhängigem zentripetalen Transport, Integration der Keratinfilamentvorläufer in das periphere Netzwerk, Bündelung der Filamente, Filamentabbau in lösliche Untereinheiten und Neubeginn des Zyklus in der Zellperipherie. Eine Störung dieses Zyklus liegt bei mutierten Keratinen vor, welche die Ursache von Blasen-bildenden Hauterkrankungen sind. In der vorliegenden Arbeit wurde am Beispiel von Keratin 6a-Mutanten, welche die Hauterkrankung Pachyonychia congenita verursachen, gezeigt, dass bei diesen Keratinen die Nukleation zwar im Bereich der Adhäsionskomplexe regelrecht abläuft, die anschließende Elongation und Netzwerkbildung aber gestört ist, so dass statt dessen kurzlebige, hyperphosphorylierte Granula entstehen. Der resultierende frustrane Keratin-Zyklus in der Zellperipherie ist stark beschleunigt und kann durch p38-Inhibierung gestoppt werden. Bei Proteasomeninhibierung wird der Zyklus in Richtung der Granulabildung verschoben. In dieser Arbeit wird erstmals das Keratin-Tretmühlen-Modell vorgestellt, das den regulierbaren Auf- und Abbau-Zyklus des Keratinnetzwerks beschreibt. Damit liegen testbare Hypothesen für die Aufklärung der Keratinfilament-Plastizität in physiologischen und pathologischen Situationen vor, die nach unseren ersten Ergebnissen auch von Relevanz für andere Intermediärfilamenttypen sind.
Resumo:
The existence of Multiple Myeloma Stem cells (MMSCs)is supposed to be one of the major causes of MM drug-resistance. However, very little is known about the molecular characteristics of MMSCs, even if some studies suggested that these cells resembles the memory B cells. In order to molecularly characterize MMSCs, we isolated the 138+138- population. For each cell fraction we performed a VDJ rearrangement analysis. The complete set of aberrations were performed by SNP Array 6.0 and HG-U133 Plus 2.0 microarray analyses (Affymetrix). The VDJ rearrangement analyses confirmed the clonal relationship between the 138+ clone and the immature clone. Both BM and PBL 138+ clones showed exactly the same genomic macroalterations. In the BM and PBL 138-19+27+ cell fractions several micro-alterations (range: 1-350 Kb) unique of the memory B cells clone were highlighted. Any micro-alterations detected were located out of any genomic variants region and are presumably associated to the MM pathogenesis, as confirmed by the presence of KRAS, WWOX and XIAP genes among the amplified regions. To get insight into the biology of the clonotypic B cell population, we compared the gene expression profile of 8 MM B cells samples 5 donor B cells vs, thus showing a differential expression of 11480 probes (p-value: <0,05). Among the self-renewal mechanisms, we observed the down-regulation of Hedgehog pathway and the iperactivation of Notch and Wnt signaling. Moreover, these immature cells showed a particular phenotype correlated to resistance to proteasome inhibitors (IRE1α-XBP1: -18.0; -19.96. P<0,05). Data suggested that the MM 138+ clone might resume the end of the complex process of myelomagenesis, whereas the memory B cells have some intriguing micro-alterations and a specific transcriptional program, supporting the idea that these post germinal center cells might be involved in the transforming event that originate and sustain the neoplastic clone.
Resumo:
Ribosome-inactivating proteins (RIPs) are a family of plant toxic enzymes that permanently damage ribosomes and possibly other cellular substrates, thus causing cell death involving different and still not completely understood pathways. The high cytotoxic activity showed by many RIPs makes them ideal candidates for the production of immunotoxins (ITs), chimeric proteins designed for the selective elimination of unwanted or malignant cells. Saporin-S6, a type 1 RIP extracted from Saponaria officinalis L. seeds, has been extensively employed to construct anticancer conjugates because of its high enzymatic activity, stability and resistance to conjugation procedures, resulting in the efficient killing of target cells. Here we investigated the anticancer properties of two saporin-based ITs, anti-CD20 RTX/S6 and anti-CD22 OM124/S6, designed for the experimental treatment of B-cell NHLs. Both ITs showed high cytotoxicity towards CD20-positive B-cells, and their antitumor efficacy was enhanced synergistically by a combined treatment with proteasome inhibitors or fludarabine. Furthermore, the two ITs showed differencies in potency and ability to activate effector caspases, and a different behavior in the presence of the ROS scavenger catalase. Taken together, these results suggest that the different carriers employed to target saporin might influence saporin intracellular routing and saporin-induced cell death mechanisms. We also investigated the early cellular response to stenodactylin, a recently discovered highly toxic type 2 RIP representing an interesting candidate for the design and production of a new IT for the experimental treatment of cancer.