Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
J Immunol ; 212(1): 154-164, 2024 01 01.
Article in English | MEDLINE | ID: mdl-37966267

ABSTRACT

The proteasome inhibitor bortezomib (BTZ) is proposed to deplete activated B cells and plasma cells. However, a complete picture of the mechanisms underlying BTZ-induced apoptosis in B lineage cells remains to be established. In this study, using a direct in vitro approach, we show that deletion of the tumor suppressor and cell cycle regulator p53 rescues recently activated mouse B cells from BTZ-induced apoptosis. Furthermore, BTZ treatment elevated intracellular p53 levels, and p53 deletion constrained apoptosis, as recently stimulated cells first transitioned from the G1 to S phase of the cell cycle. Moreover, combined inhibition of the p53-associated cell cycle regulators and E3 ligases MDM2 and anaphase-promoting complex/cyclosome induced cell death in postdivision B cells. Our results reveal that efficient cell cycle progression of activated B cells requires proteasome-driven inhibition of p53. Consequently, BTZ-mediated interference of proteostasis unleashes a p53-dependent cell cycle-associated death mechanism in recently activated B cells.


Subject(s)
Antineoplastic Agents , Proteasome Inhibitors , Animals , Mice , Bortezomib/pharmacology , Bortezomib/metabolism , Proteasome Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Tumor Suppressor Protein p53/metabolism , Cell Line, Tumor , Proteasome Endopeptidase Complex/metabolism , Apoptosis
2.
Curr Drug Targets ; 24(10): 781-789, 2023.
Article in English | MEDLINE | ID: mdl-37469152

ABSTRACT

Some diseases caused by trypanosomatid parasites, like Leishmaniasis, Chagas Disease, and Human African Trypanosomiasis (HTA), are challenging to manage, mainly concerning pharmacological therapy because they are associated with vulnerable populations. Unfortunately, there is a lack of significant investments in the search for new drugs. Therefore, one of the strategies to aid the discovery of new drugs is to identify and inhibit molecular targets essential to the parasite's survival, such as the proteasome, which degrades most proteins in the parasite cells. Our study has presented several proteasome inhibitors with various pharmacophoric cores, and two of them, 5, and 13, have stood out in the clinical phase of treatment for leishmaniasis.


Subject(s)
Chagas Disease , Leishmaniasis , Trypanosomiasis, African , Animals , Humans , Proteasome Endopeptidase Complex , Trypanosomiasis, African/drug therapy , Chagas Disease/drug therapy , Leishmaniasis/drug therapy , Proteasome Inhibitors/pharmacology , Proteasome Inhibitors/therapeutic use
3.
Cells ; 10(12)2021 12 11.
Article in English | MEDLINE | ID: mdl-34944009

ABSTRACT

The proteasome increases its activity at the onset of sperm capacitation due to the action of the SACY/PRKACA pathway; this increase is required for capacitation to progress. PRKA activity also increases and remains high during capacitation. However, intracellular levels of cAMP decrease in this process. Our goal was to evaluate the role of the proteasome in regulating PRKA activity once capacitation has started. Viable human sperm were incubated in the presence and absence of epoxomicin or with 0.1% DMSO. The activity of PRKA; the phosphorylation pattern of PRKA substrates (pPRKAs); and the expression of PRKAR1, PRKAR2, and AKAP3 were evaluated by Western blot. The localization of pPRKAs, PRKAR1, PRKAR2, and AKAP3 was evaluated by immunofluorescence. Treatment with epoxomicin changed the localization and phosphorylation pattern and decreased the percentage of pPRKAs-positive sperm. PRKA activity significantly increased at 1 min of capacitation and remained high throughout the incubation. However, epoxomicin treatment significantly decreased PRKA activity after 30 min. In addition, PRKAR1 and AKAP3 were degraded by the proteasome but with a different temporal kinetic. Our results suggest that PRKAR1 is the target of PRKA regulation by the proteasome.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Proteasome Endopeptidase Complex/metabolism , Sperm Capacitation/physiology , A Kinase Anchor Proteins/metabolism , Adult , Humans , Phosphorylation/drug effects , Proteasome Inhibitors/pharmacology , Proteolysis/drug effects , Signal Transduction/drug effects , Sperm Capacitation/drug effects , Subcellular Fractions/metabolism , Substrate Specificity/drug effects , Young Adult
4.
Inorg Chem ; 59(20): 15004-15018, 2020 Oct 19.
Article in English | MEDLINE | ID: mdl-32997499

ABSTRACT

In this paper, a series of new ruthenium complexes of the general formula [Ru(NS)(dpphpy)(dppb)]PF6 (Ru1-Ru3), where dpphpy = diphenyl-2-pyridylphosphine, NS ligands = 2-thiazoline-2-thiol (tzdt, Ru1), 2-mercaptopyrimidine (pySm, Ru2), and 4,6-diamino-2-mercaptopyrimidine (damp, Ru3), and dppb = 1,4-bis(diphenylphosphino)butane, were synthesized and characterized by elemental analysis, spectroscopic techniques (IR, UV/visible, and 1D and 2D NMR), and X-ray diffraction. In the characterization, the correlation between the phosphorus atoms and their respective aromatic hydrogen atoms of the compounds in the assignment stands outs, by 1H-31P HMBC experiments. The compounds show anticancer activities against A549 (lung) and MDA-MB-231 (breast) cancer cell lines, higher than the clinical drug cisplatin. All of the complexes are more cytotoxic against the cancer cell lines than against the MRC-5 (lung) and MCF-10A (breast) nontumorigenic human cell lines. For A549 tumor cells, cell cycle analysis upon treatment with Ru2 showed that it inhibits the mitotic phase because arrest was observed in the Sub-G1 phase. Additionally, the compound induces cell death by an apoptotic pathway in a dose-dependent manner, according to annexin V-PE assay. The multitargeted character of the compounds was investigated, and the biomolecules were DNA, topoisomerase IB, and proteasome, as well as the fundamental biomolecule in the pharmacokinetics of drugs, human serum albumin. The experimental results indicate that the complexes do not target DNA in the cells. At low concentrations, the compounds showed the ability to partially inhibit the catalytic activity of topoisomerase IB in the process of relaxation of the DNA plasmid. Among the complexes assayed in cultured cells, complex Ru3 was able to diminish the proteasomal chymotrypsin-like activity to a greater extent.


Subject(s)
Antineoplastic Agents/pharmacology , Coordination Complexes/pharmacology , DNA Topoisomerases, Type I/metabolism , Proteasome Inhibitors/pharmacology , Topoisomerase I Inhibitors/pharmacology , Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Coordination Complexes/chemical synthesis , Drug Screening Assays, Antitumor , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Phosphines/chemical synthesis , Phosphines/pharmacology , Proteasome Inhibitors/chemical synthesis , Ruthenium/chemistry , Sulfhydryl Compounds/chemical synthesis , Sulfhydryl Compounds/pharmacology , Topoisomerase I Inhibitors/chemical synthesis
5.
Parasitol Res ; 119(7): 2159-2176, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32424554

ABSTRACT

The proteasome is the key player in the cellular protein degradation machinery and is pivotal for protein homeostasis and Schistosoma mansoni (S. mansoni) survival. Our group study provides insights into proteasome inhibitors and reveals that selective schistosomiasis agents represent an interesting branch of proteasome research linked to the development of new drugs for this neglected disease. Here, we explored the phenotypic response of S. mansoni to b-AP15, a bis-benzylidine piperidone that inhibits 26S proteasome deubiquitinases (DUBs), ubiquitin-specific protease 14 (USP14), and ubiquitin carboxyl-terminal hydrolase 5 (UCHL5). b-AP15 induces a modest decrease in egg production in vitro and reduces viability, leading to the death of parasite couples. This inhibitor also induces a twofold increase in the accumulation of polyubiquitinated proteins in S. mansoni adult worms and causes tegument changes such as disintegration, wrinkling, and bubble formation, both throughout the length of the parasite and in the oral sucker. b-AP15 alters the cell organelles of adult S. mansoni worms, and we specifically observed mitochondrial alterations, which are suggestive of proteotoxic stress leading to autophagy. Taken together, these results indicate that the deubiquitinase function of the proteasome is essential for the parasite and support the hypothesis that the proteasome constitutes an interesting drug target for the treatment of schistosomiasis.


Subject(s)
Deubiquitinating Enzymes/antagonists & inhibitors , Oviposition/drug effects , Proteasome Inhibitors/pharmacology , Schistosoma mansoni/drug effects , Animals , Female , Helminth Proteins/metabolism , Piperidones/pharmacology , Proteasome Endopeptidase Complex/metabolism , Schistosoma mansoni/metabolism , Schistosoma mansoni/physiology , Ubiquitination/drug effects
6.
PLoS One ; 15(5): e0233591, 2020.
Article in English | MEDLINE | ID: mdl-32453773

ABSTRACT

The heart is critically dependent on mitochondrial respiration for energy supply. Ischemia decreases oxygen availability, with catastrophic consequences for cellular energy systems. After a few minutes of ischemia, the mitochondrial respiratory chain halts, ATP levels drop and ion gradients across cell membranes collapse. Activation of cellular proteases and generation of reactive oxygen species by mitochondria during ischemia alter mitochondrial membrane permeability, causing mitochondrial swelling and fragmentation and eventually cell death. The mitochondria, therefore, are important targets of cardioprotection against ischemic injury. We have previously shown that ixazomib (IXA), a proteasome inhibitor used for treating multiple myeloma, effectively reduced the size of the infarct produced by global ischemia in isolated rat hearts and prevented degradation of the sarcoplasmic reticulum calcium release channel RyR2. The aim of this work was to further characterize the protective effect of IXA by determining its effect on mitochondrial morphology and function after ischemia. We also quantified the effect of IXA on levels of mitofusin-2, a protein involved in maintaining mitochondrial morphology and mitochondria-SR communication. We found that mitochondria were significantly preserved and functional parameters such as oxygen consumption, the ability to generate a membrane potential, and glutathione content were improved in mitochondria isolated from hearts perfused with IXA prior to ischemia. IXA also blocked the release of cytochrome c observed in ischemia and significantly preserved mitofusin-2 integrity. These beneficial effects resulted in a significant decrease in the left ventricular end diastolic pressure upon reperfusion and a smaller infarct in isolated hearts.


Subject(s)
Boron Compounds/pharmacology , Glycine/analogs & derivatives , Heart/drug effects , Mitochondria/drug effects , Myocardial Ischemia/drug therapy , Animals , Chymotrypsin/pharmacology , Disease Models, Animal , Glutathione/genetics , Glutathione/metabolism , Glycine/pharmacology , Heart/physiopathology , Humans , Membrane Potentials/drug effects , Mitochondria/genetics , Myocardial Ischemia/genetics , Myocardial Ischemia/physiopathology , Oxygen Consumption/genetics , Proteasome Endopeptidase Complex/drug effects , Proteasome Inhibitors/pharmacology , Rats
7.
PLoS One ; 15(4): e0230981, 2020.
Article in English | MEDLINE | ID: mdl-32251488

ABSTRACT

Autophagy is a mechanism responsible for intracellular degradation and recycling of macromolecules and organelles, essential for cell survival in adverse conditions. More than 40 autophagy-related (ATG) genes have been identified and characterized in fungi, among them ATG4 and ATG8. ATG4 encodes a cysteine protease (Atg4) that plays an important role in autophagy by initially processing Atg8 at its C-terminus region. Atg8 is a ubiquitin-like protein essential for the synthesis of the double-layer membrane that constitutes the autophagosome vesicle, responsible for delivering the cargo from the cytoplasm to the vacuole lumen. The contributions of Atg-related proteins in the pathogenic yeast in the genus Cryptococcus remain to be explored, to elucidate the molecular basis of the autophagy pathway. In this context, we aimed to investigate the role of autophagy-related proteins 4 and 8 (Atg4 and Atg8) during autophagy induction and their contribution with non-autophagic events in C. neoformans. We found that Atg4 and Atg8 are conserved proteins and that they interact physically with each other. ATG gene deletions resulted in cells sensitive to nitrogen starvation. ATG4 gene disruption affects Atg8 degradation and its translocation to the vacuole lumen, after autophagy induction. Both atg4 and atg8 mutants are more resistant to oxidative stress, have an impaired growth in the presence of the cell wall-perturbing agent Congo Red, and are sensitive to the proteasome inhibitor bortezomib (BTZ). By that, we conclude that in C. neoformans the autophagy-related proteins Atg4 and Atg8 play an important role in the autophagy pathway; which are required for autophagy regulation, maintenance of amino acid levels and cell adaptation to stressful conditions.


Subject(s)
Autophagy-Related Protein 8 Family/physiology , Autophagy-Related Proteins/physiology , Cryptococcus neoformans/physiology , Fungal Proteins/physiology , Amino Acids/metabolism , Autophagy/genetics , Autophagy/physiology , Autophagy-Related Protein 8 Family/genetics , Autophagy-Related Proteins/genetics , Bortezomib/pharmacology , Cryptococcosis/drug therapy , Cryptococcosis/microbiology , Cryptococcus neoformans/drug effects , Cryptococcus neoformans/genetics , Drug Resistance, Fungal , Fungal Proteins/genetics , Genes, Fungal , Genetic Complementation Test , Humans , In Vitro Techniques , Mutation , Nitrogen/metabolism , Oxidative Stress/genetics , Proteasome Inhibitors/pharmacology , Protein Processing, Post-Translational , Vacuoles/metabolism
8.
Braz J Microbiol ; 51(3): 989-997, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32333271

ABSTRACT

Fungi in the genus Trichoderma are notorious producers of secondary metabolites with diverse applications, such as antibacterial, antifungal, and plant growth-promoting properties. Peptaibols are linear peptides produced by such fungi, with more than 440 compounds described to date, including tricholongins, longibrachins, trichobrachins, and trichovirins. Peptaibols are synthesized by non-ribosomal peptide synthetases and they have several biological activities. Our research group isolated four peptaibols (6DP2, 6DP3, 6DP4, and 6DP5) with antifungal activity against the plant pathogen Colletotrichum gloeosporioides and the proteasome (a cancer chemotherapy target) from Trichoderma sp. P8BDA1F1, an endophytic fungus from Begonia venosa. The ethyl acetate extract of this endophyte showed activity of 6.01% and 75% against C. gloeosporioides and the proteasome, respectively. The isolated compounds were identified by MS/MS and compared to literature data, suggesting the presence of trilongins BI, BII, BIII, and BIV, which are peptaibols containing 20 amino acid residues. The minimum inhibitory concentration against C. gloeosporioides was 40 µM for trilongin BI, 320 µM for trilongin BII, 160 µM for trilongin BIII, and 310 µM for trilongin BIV. BI-BIV trilongins inhibited proteasome ChTL activity, with IC50 values of 6.5 ± 2.7; 4.7 ± 1.8; 6.3 ± 2.2; and 2.7 ± 0.5 µM, respectively. The compounds were tested ex vivo against the intracellular amastigotes of Leishmania (L.) infantum but showed no selectivity. It is the first report of trilongins BI-BIV with antifungal activity against C. gloeosporioides and the proteasome target.


Subject(s)
Antifungal Agents/pharmacology , Antineoplastic Agents/pharmacology , Begoniaceae/microbiology , Peptaibols/pharmacology , Trichoderma/chemistry , Antifungal Agents/chemistry , Antifungal Agents/isolation & purification , Antineoplastic Agents/chemistry , Antineoplastic Agents/isolation & purification , Colletotrichum/drug effects , Endophytes , Inhibitory Concentration 50 , Microbial Sensitivity Tests , Peptaibols/chemistry , Peptaibols/isolation & purification , Phylogeny , Proteasome Inhibitors/pharmacology , Trichoderma/classification , Trichoderma/genetics , Trichoderma/isolation & purification
9.
Biochim Biophys Acta Mol Basis Dis ; 1866(5): 165659, 2020 05 01.
Article in English | MEDLINE | ID: mdl-31891806

ABSTRACT

Cardiomyocyte loss is the main cause of myocardial dysfunction following an ischemia-reperfusion (IR) injury. Mitochondrial dysfunction and altered mitochondrial network dynamics play central roles in cardiomyocyte death. Proteasome inhibition is cardioprotective in the setting of IR; however, the mechanisms underlying this protection are not well-understood. Several proteins that regulate mitochondrial dynamics and energy metabolism, including Mitofusin-2 (Mfn2), are degraded by the proteasome. The aim of this study was to evaluate whether proteasome inhibition can protect cardiomyocytes from IR damage by maintaining Mfn2 levels and preserving mitochondrial network integrity. Using ex vivo Langendorff-perfused rat hearts and in vitro neonatal rat ventricular myocytes, we showed that the proteasome inhibitor MG132 reduced IR-induced cardiomyocyte death. Moreover, MG132 preserved mitochondrial mass, prevented mitochondrial network fragmentation, and abolished IR-induced reductions in Mfn2 levels in heart tissue and cultured cardiomyocytes. Interestingly, Mfn2 overexpression also prevented cardiomyocyte death. This effect was apparently specific to Mfn2, as overexpression of Miro1, another protein implicated in mitochondrial dynamics, did not confer the same protection. Our results suggest that proteasome inhibition protects cardiomyocytes from IR damage. This effect could be partly mediated by preservation of Mfn2 and therefore mitochondrial integrity.


Subject(s)
GTP Phosphohydrolases/metabolism , Mitochondrial Proteins/metabolism , Myocardial Infarction/drug therapy , Myocardial Reperfusion Injury/prevention & control , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Animals , Animals, Newborn , Apoptosis/drug effects , Cells, Cultured , Disease Models, Animal , Humans , Isolated Heart Preparation , Male , Mitochondria/drug effects , Myocardial Infarction/complications , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Primary Cell Culture , Proteasome Inhibitors/therapeutic use , Rats , rho GTP-Binding Proteins/metabolism
10.
Int J Mol Sci ; 20(14)2019 Jul 10.
Article in English | MEDLINE | ID: mdl-31295808

ABSTRACT

Proteasome inhibitors have been actively tested as potential anticancer drugs and in the treatment of inflammatory and autoimmune diseases. Unfortunately, cells adapt to survive in the presence of proteasome inhibitors activating a variety of cell responses that explain why these therapies have not fulfilled their expected results. In addition, all proteasome inhibitors tested and approved by the FDA have caused a variety of side effects in humans. Here, we describe the different types of proteasome complexes found within cells and the variety of regulators proteins that can modulate their activities, including those that are upregulated in the context of inflammatory processes. We also summarize the adaptive cellular responses activated during proteasome inhibition with special emphasis on the activation of the Autophagic-Lysosomal Pathway (ALP), proteaphagy, p62/SQSTM1 enriched-inclusion bodies, and proteasome biogenesis dependent on Nrf1 and Nrf2 transcription factors. Moreover, we discuss the role of IRE1 and PERK sensors in ALP activation during ER stress and the involvement of two deubiquitinases, Rpn11 and USP14, in these processes. Finally, we discuss the aspects that should be currently considered in the development of novel strategies that use proteasome activity as a therapeutic target for the treatment of human diseases.


Subject(s)
Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Autophagy/drug effects , Endoplasmic Reticulum Stress , Humans , Immunomodulation/drug effects , Lysosomes/metabolism , Neoplasms/drug therapy , Neoplasms/etiology , Neoplasms/metabolism , Neoplasms/pathology , Proteasome Inhibitors/therapeutic use , Protein Processing, Post-Translational/drug effects , Signal Transduction/drug effects , Unfolded Protein Response
11.
Biochem Biophys Res Commun ; 514(3): 574-579, 2019 06 30.
Article in English | MEDLINE | ID: mdl-31056263

ABSTRACT

Kir7.1 is an inwardly rectifying K+ channel present in epithelia where it shares membrane localization with the Na+/K+-pump. In the present communication we report the presence of a novel splice variant of Kir7.1 in mouse tissues including kidney, lung, choroid plexus and retinal pigment epithelium (RPE). The variant named mKir7.1-SV2 lacks most of the C-terminus domain but is predicted to have the two transmembrane domains and permeation pathway unaffected. Similarly truncated predicted proteins, Kir7.1-R166X and Kir7.1-Q219X, would arise from mutations associated with Leber Congenital Amaurosis, a rare recessive hereditary retinal disease that results in vision loss at early age. We found that mKir7.1-SV2 and the pathological variants do not produce any channel activity when expressed alone in HEK-293 cells due to their scarce presence in the plasma membrane. Simultaneous expression with the full length Kir7.1 however leads to a reduction in activity of the wild-type channel that might be due to partial proteasome degradation of WT-mutant channel heteromers.


Subject(s)
Leber Congenital Amaurosis/genetics , Mutation/genetics , Organ Specificity , Potassium Channels, Inwardly Rectifying/genetics , RNA Splicing/genetics , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , HEK293 Cells , Humans , Ion Channel Gating/drug effects , Male , Mice, Inbred C57BL , Mutant Proteins/metabolism , Organ Specificity/drug effects , Peptides/genetics , Potassium/metabolism , Proteasome Inhibitors/pharmacology , RNA Splicing/drug effects
12.
Viruses ; 11(4)2019 04 23.
Article in English | MEDLINE | ID: mdl-31018496

ABSTRACT

Mayaro (MAYV) and Una (UNAV) are emerging arboviruses belonging to the Alphavirus genus of the Togaviridae family. These viruses can produce febrile disease with symptoms such as fever, headache, myalgia, skin rash and incapacitating poly-arthralgia. Serological studies indicate that both viruses are circulating in different countries in Latin America. Viruses need the host cell machinery and resources to replicate effectively. One strategy to find new antivirals consists of identifying key cellular pathways or factors that are essential for virus replication. In this study, we analyzed the role of the ubiquitin-proteasome system (UPS) in MAYV and UNAV replication. Vero-E6 or HeLa cells were treated with the proteasome inhibitors MG132 or Lactacystin, and viral progeny production was quantified using a plaque assay method. In addition, the synthesis of viral proteins was analyzed by Western blot and confocal microscopy. Our results indicate that treatment with proteasome inhibitors decreases MAYV and UNAV protein synthesis, and also causes a significant dose-dependent decrease in MAYV and UNAV replication. Proteasome activity seems to be important at the early stages of MAYV replication. These findings suggest that the ubiquitin-proteasome system is a possible pharmacological target to inhibit these neglected alphaviruses.


Subject(s)
Alphavirus/drug effects , Antiviral Agents/pharmacology , Proteasome Endopeptidase Complex/physiology , Virus Replication , Acetylcysteine/analogs & derivatives , Acetylcysteine/pharmacology , Alphavirus/physiology , Animals , Chlorocebus aethiops , Cysteine Proteinase Inhibitors/pharmacology , Cytoplasm/drug effects , Cytoplasm/virology , HeLa Cells , Humans , Leupeptins/pharmacology , Proteasome Inhibitors/pharmacology , Vero Cells
13.
Curr Mol Med ; 19(2): 112-119, 2019.
Article in English | MEDLINE | ID: mdl-30834832

ABSTRACT

INTRODUCTION: Multiple myeloma (MM) cells accumulate in the bone marrow and produce enormous quantities of immunoglobulins, causing endoplasmatic reticulum stress and activation of protein handling machinery, such as heat shock protein response, autophagy and unfolded protein response (UPR). METHODS: We evaluated cell lines viability after treatment with bortezomib (B) in combination with HSP70 (VER-15508) and autophagy (SBI-0206965) or UPR (STF- 083010) inhibitors. RESULTS: For RPMI-8226, after 72 hours of treatment with B+VER+STF or B+VER+SBI, we observed 15% of viable cells, but treatment with B alone was better (90% of cell death). For U266, treatment with B+VER+STF or with B+VER+SBI for 72 hours resulted in 20% of cell viability and both treatments were better than treatment with B alone (40% of cell death). After both triplet combinations, RPMI-8226 and U266 presented the overexpression of XBP-1 UPR protein, suggesting that it is acting as a compensatory mechanism, in an attempt of the cell to handle the otherwise lethal large amount of immunoglobulin overload. CONCLUSION: Our in vitro results provide additional evidence that combinations of protein homeostasis inhibitors might be explored as treatment options for MM.


Subject(s)
Autophagy , Bortezomib/pharmacology , HSP70 Heat-Shock Proteins/antagonists & inhibitors , Multiple Myeloma/pathology , Proteasome Inhibitors/pharmacology , Proteostasis/drug effects , Unfolded Protein Response/drug effects , Apoptosis/drug effects , Benzamides/pharmacology , Cell Survival/drug effects , Drug Synergism , Humans , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Thiophenes/pharmacology , Tumor Cells, Cultured
14.
Mol Oncol ; 13(2): 290-306, 2019 02.
Article in English | MEDLINE | ID: mdl-30422399

ABSTRACT

In malignant transformation, cellular stress-response pathways are dynamically mobilized to counterbalance oncogenic activity, keeping cancer cells viable. Therapeutic disruption of this vulnerable homeostasis might change the outcome of many human cancers, particularly those for which no effective therapy is available. Here, we report the use of fibroblast growth factor 2 (FGF2) to demonstrate that further mitogenic activation disrupts cellular homeostasis and strongly sensitizes cancer cells to stress-targeted therapeutic inhibitors. We show that FGF2 enhanced replication and proteotoxic stresses in a K-Ras-driven murine cancer cell model, and combinations of FGF2 and proteasome or DNA damage response-checkpoint inhibitors triggered cell death. CRISPR/Cas9-mediated K-Ras depletion suppressed the malignant phenotype and prevented these synergic toxicities in these murine cells. Moreover, in a panel of human Ewing's sarcoma family tumor cells, sublethal concentrations of bortezomib (proteasome inhibitor) or VE-821 (ATR inhibitor) induced cell death when combined with FGF2. Sustained MAPK-ERK1/2 overactivation induced by FGF2 appears to underlie these synthetic lethalities, as late pharmacological inhibition of this pathway restored cell homeostasis and prevented these described synergies. Our results highlight how mitotic signaling pathways which are frequently overridden in malignant transformation might be exploited to disrupt the robustness of cancer cells, ultimately sensitizing them to stress-targeted therapies. This approach provides a new therapeutic rationale for human cancers, with important implications for tumors still lacking effective treatment, and for those that frequently relapse after treatment with available therapies.


Subject(s)
Antineoplastic Agents/pharmacology , Fibroblast Growth Factor 2/pharmacology , Stress, Physiological , Animals , Bortezomib/pharmacology , Cell Cycle/drug effects , Cell Death/drug effects , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/metabolism , Mice , Proteasome Inhibitors/pharmacology , Proto-Oncogene Proteins p21(ras)/metabolism
15.
J Nat Prod ; 81(10): 2296-2300, 2018 10 26.
Article in English | MEDLINE | ID: mdl-30281303

ABSTRACT

The new pyrrole-imidazole and pyrrole-guanidine alkaloids 4-debromooroidin (1), 4-debromougibohlin (2), 5-debromougibohlin (3), and 5-bromopalau'amine (4), along with the known hymenidin (5) and (+)-monobromoisophakellin (6), have been isolated from a Dictyonella sp. marine sponge, collected at the Amazon River mouth. The bromine-substitution pattern observed for compounds 1, 2 and 4 is unusual among bromopyrrole alkaloids isolated from marine sponges. The 20S proteasome inhibitory activities of compounds 1-6 have been recorded, with 5-bromopalau'amine (4) being the most active in this series.


Subject(s)
Porifera/chemistry , Proteasome Inhibitors/chemistry , Proteasome Inhibitors/pharmacology , Pyrroles/chemistry , Pyrroles/pharmacology , Animals , Brazil , Molecular Structure , Proteasome Endopeptidase Complex , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics
16.
Basic Clin Pharmacol Toxicol ; 122(5): 489-500, 2018 May.
Article in English | MEDLINE | ID: mdl-29205851

ABSTRACT

Collapse of the mitochondrial membrane potential (MMP) is often considered the initiation of regulated cell death (RCD). Carbonyl cyanide 3-chlorophenylhydrazone (CCCP) is an uncoupler of the electron transport chain (ETC) that facilitates the translocation of protons into the mitochondrial matrix leading to the collapse of the MMP. Several cell stress responses such as mitophagy, mitochondrial biogenesis and the ubiquitin proteasome system may differentially contribute to restrain the initiation of RCD depending on the extent of mitochondrial damage. We induced graded mitochondrial damage after collapse of MMP with the mitochondrial uncoupler CCCP in Burkitt's lymphoma cells, and we evaluated the effect of several drugs targeting cell stress responses over RCD at 72 hr, using a multiparametric flow cytometry approach. CCCP caused collapse of MMP after 30 min., massive mitochondrial fission, oxidative stress and increased mitophagy within the 5-15 µM low-dose range (LDR) of CCCP. Within the 20-50 µM high-dose range (HDR), CCCP caused lysosomal destabilization and rupture, thus precluding mitophagy and autophagy. Cell death after 72 hr was below 20%, with increased mitochondrial mass (MM). The inhibitors of mitophagy 3-(2,4-dichloro-5-methoxyphenyl)-2,3-dihydro-2-thioxo-4(1H)-quinazolinone (Mdivi-1) and vincristine (VCR) increased cell death from CCCP within the LDR, while valproic acid (an inducer of mitochondrial biogenesis) also increased MM and cell death within the LDR. The proteasome inhibitor, MG132, increased cell death only in the HDR. Doxycycline, an antibiotic that disrupts mitochondrial biogenesis, had no effect on cell survival, while iodoacetamide, an inhibitor of glycolysis, increased cell death at the HDR. We conclude that mitophagy influenced RCD of lymphoma cells after MMP collapse by CCCP only within the LDR, while proteasome activity and glycolysis contributed to survival in the HDR under extensive mitochondria and lysosome damage.


Subject(s)
Burkitt Lymphoma/drug therapy , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , Mitochondria/drug effects , Mitophagy/drug effects , Uncoupling Agents/pharmacology , Autophagosomes/drug effects , Autophagosomes/metabolism , Autophagosomes/pathology , Autophagy/drug effects , Burkitt Lymphoma/metabolism , Burkitt Lymphoma/pathology , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , Iodoacetamide/pharmacology , Leupeptins/pharmacology , Lysosomes/drug effects , Lysosomes/metabolism , Lysosomes/pathology , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Dynamics/drug effects , Oxidative Stress/drug effects , Proteasome Endopeptidase Complex/drug effects , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Quinazolinones/pharmacology , Reactive Oxygen Species/metabolism , Time Factors , Unfolded Protein Response/drug effects , Vincristine/pharmacology
17.
Curr Drug Metab ; 18(12): 1136-1146, 2017.
Article in English | MEDLINE | ID: mdl-29165070

ABSTRACT

BACKGROUND: Despite major advances in transplant medicine, antibody-mediated rejection (AMR) continues to have severe clinical implications and adversely affect graft survival. Therefore, the search for alternative drugs to treat AMR is widely pursued. The first-in-class proteasome inhibitor bortezomib (BZ) is a selective inhibitor of the 26S proteasome, which was initially approved for the treatment of malignant plasma cell disorders. METHODS: This review encompasses how our understanding of inhibiting proteasome pathway created the basis of BZ research and important milestones accomplished in AMR treatment in the transplant setting. It further discusses at length the results of clinical studies, the tolerability profile, drug-drug interactions and the perspectives of BZ use in desensitization protocols. RESULTS: Proteasome inhibition can downregulate NF-κB activity; decrease cell proliferation/differentiation; induce apoptosis via cell cycle arrest, endoplasmic reticulum stress and caspase induction due the accumulation of unfolded or misfolded proteins; and downregulate antigen presentation, cell-cell interaction, and cell migration. Proteasome inhibition is more evident in cells with high rate of protein synthesis and secretion, like plasma cells. These cells play a critical role in the production of antibodies during AMR. CONCLUSIONS: There is accumulating evidence that the proteasome inhibitor BZ may substantially affect the function and integrity of alloantibody-secreting plasma cells in AMR after organ solid transplant, as well as the activation, proliferation and differentiation of T- and B-lymphocytes. Recent clinical studies have provided evidence that BZ has the capability to downregulate circulating antibodies and treat AMR episodes. Additional randomized-controlled studies are required to assess the impact of BZ during short and long follow-ups.


Subject(s)
Antineoplastic Agents/pharmacology , Bortezomib/pharmacology , Graft Survival/drug effects , Kidney Transplantation , Proteasome Inhibitors/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Bortezomib/chemistry , Bortezomib/pharmacokinetics , Humans , Molecular Structure , Proteasome Inhibitors/chemistry , Proteasome Inhibitors/pharmacokinetics
18.
PLoS One ; 12(9): e0184192, 2017.
Article in English | MEDLINE | ID: mdl-28898250

ABSTRACT

Proteasome is a proteolytic complex responsible for intracellular protein turnover in eukaryotes, archaea and in some actinobacteria species. Previous work has demonstrated that in Schistosoma mansoni parasites, the proteasome inhibitor MG-132 affects parasite development. However, the molecular targets affected by MG-132 in S. mansoni are not entirely known. Here, we used expression microarrays to measure the genome-wide changes in gene expression of S. mansoni adult worms exposed in vitro to MG-132, followed by in silico functional analyses of the affected genes using Ingenuity Pathway Analysis (IPA). Scanning electron microscopy was used to document changes in the parasites' tegument. We identified 1,919 genes with a statistically significant (q-value ≤ 0.025) differential expression in parasites treated for 24 h with MG-132, when compared with control. Of these, a total of 1,130 genes were up-regulated and 790 genes were down-regulated. A functional gene interaction network comprised of MG-132 and its target genes, known from the literature to be affected by the compound in humans, was identified here as affected by MG-132. While MG-132 activated the expression of the 26S proteasome genes, it also decreased the expression of 19S chaperones assembly, 20S proteasome maturation, ubiquitin-like NEDD8 and its partner cullin-3 ubiquitin ligase genes. Interestingly, genes that encode proteins related to potassium ion binding, integral membrane component, ATPase and potassium channel activities were significantly down-regulated, whereas genes encoding proteins related to actin binding and microtubule motor activity were significantly up-regulated. MG-132 caused important changes in the worm tegument; peeling, outbreaks and swelling in the tegument tubercles could be observed, which is consistent with interference on the ionic homeostasis in S. mansoni. Finally, we showed the down-regulation of Bax pro-apoptotic gene, as well as up-regulation of two apoptosis inhibitor genes, IAP1 and BRE1, and in contrast, down-regulation of Apaf-1 apoptotic activator, thus suggesting that apoptosis is deregulated in S. mansoni exposed to MG-132. A considerable insight has been gained concerning the potential of MG-132 as a gene expression modulator, and overall the data suggest that the proteasome might be an important molecular target for the design of new drugs against schistosomiasis.


Subject(s)
Leupeptins/pharmacology , Proteasome Inhibitors/pharmacology , Schistosoma mansoni/drug effects , Animals , Cluster Analysis , Gene Expression Profiling , Gene Expression Regulation/drug effects , Gene Regulatory Networks , Reproducibility of Results , Schistosoma mansoni/genetics , Schistosoma mansoni/ultrastructure , Transcriptome
19.
Blood ; 128(25): 2919-2930, 2016 12 22.
Article in English | MEDLINE | ID: mdl-27793878

ABSTRACT

Multiple myeloma (MM) patients with the t(14;16) translocation have a poor prognosis, and unlike other molecular subgroups, their outcome has not improved with the introduction of bortezomib (Bzb). The mechanism underlying innate resistance of MM to Bzb is unknown. In the present study, we have investigated how MAF overexpression impacts resistance to proteasome inhibitor (PI) therapy (Bzb and carfilzomib). High levels of MAF protein were found in t(14;16) cell lines; cell lines from the t(4;14) subgroup had intermediate levels, whereas cell lines from the other subgroups had low levels. High expression of MAF protein in t(14;16) was associated with significantly higher PI half-maximum inhibitory concentration values compared with other molecular subgroups. PI exposure abrogated glycogen synthase kinase 3ß (GSK3ß)-mediated degradation of MAF protein, resulting in increased MAF protein stability and PI resistance. Subsequent studies using loss-of-function and gain-of-function models showed that silencing MAF led to increased sensitivity to PIs, enhanced apoptosis, and activation of caspase-3, -7, -8, -9, poly (ADP-ribose) polymerase, and lamin A/C. In contrast, overexpression of MAF resulted in increased resistance to PIs and reduced apoptosis. These results define the role of MAF and GSK3 in the resistance of t(14;16) MM to PIs and identifies a novel mechanism by which MAF protein levels are regulated by PIs, which in turn confers resistance to PIs.


Subject(s)
Drug Resistance, Neoplasm , Immunity, Innate , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Proteasome Inhibitors/therapeutic use , Proto-Oncogene Proteins c-maf/metabolism , Apoptosis/drug effects , Caspases/metabolism , Cell Line, Tumor , Chromosomes, Human, Pair 14/genetics , Chromosomes, Human, Pair 16/genetics , Drug Resistance, Neoplasm/drug effects , Enzyme Activation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing/drug effects , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Immunity, Innate/drug effects , Lamins/metabolism , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Phosphorylation/drug effects , Poly(ADP-ribose) Polymerases/metabolism , Prognosis , Proteasome Inhibitors/pharmacology , Proteolysis/drug effects , Proto-Oncogene Proteins c-maf/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Translocation, Genetic
20.
Biochim Biophys Acta ; 1864(12): 1775-1786, 2016 12.
Article in English | MEDLINE | ID: mdl-27693249

ABSTRACT

The PR-11 peptide corresponds to the N-terminal and active region of the endogenously synthesized PR-39 molecule, of porcine origin. It is known to possess various biological effects including antimicrobial properties, angiogenic and anti-inflammatory activities. Apart from its reported activity as a proteasome inhibitor, a more comprehensive understanding of its function, at the molecular level, is still lacking. In this study, we used a label-free shotgun strategy to evaluate the proteomic alterations caused by exposure of cultured fibroblasts to the peptide PR-11. This approach revealed that more than half of the identified molecules were related to signalling, transcription and translation. Proteins directly associated to regulation of angiogenesis and interaction with the hypoxia-inducible factor 1-α (HIF-1α) were significantly altered. In addition, at least three differentially expressed molecules of the NF-κB pathway were detected, suggesting an anti-inflammatory property of PR-11. At last, we demonstrated novel potential ligands of PR-11, through its immobilization for affinity chromatography. Among the eluted molecules, gC1qR, a known complement receptor, appeared markedly enriched. This provided preliminary evidence of a PR-11 ligand possibly involved in the internalization of this peptide. Altogether, our findings contributed to a better understanding of the cellular pathways affected by PR-39 derived molecules.


Subject(s)
Antimicrobial Cationic Peptides/pharmacology , Animals , Antimicrobial Cationic Peptides/metabolism , Carrier Proteins/metabolism , Cells, Cultured , Fibroblasts/drug effects , Fibroblasts/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immobilized Proteins/metabolism , Immobilized Proteins/pharmacology , Ligands , Mass Spectrometry , Mitochondrial Proteins/metabolism , NF-kappa B/metabolism , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Proteasome Inhibitors/metabolism , Proteasome Inhibitors/pharmacology , Proteome/drug effects , Proteome/metabolism , Proteomics , Rats , Rats, Wistar , Swine
SELECTION OF CITATIONS
SEARCH DETAIL