Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Cell Mol Life Sci ; 79(1): 45, 2021 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-34913092

RESUMO

PA28γ is a nuclear activator of the 20S proteasome that, unlike the 19S regulatory particle, stimulates hydrolysis of several substrates in an ATP- and ubiquitin-independent manner and whose exact biological functions and molecular mechanism of action still remain elusive. In an effort to shed light on these important issues, we investigated the stimulatory effect of PA28γ on the hydrolysis of different fluorogenic peptides and folded or denatured full-length proteins by the 20S proteasome. Importantly, PA28γ was found to dramatically enhance breakdown rates by 20S proteasomes of several naturally or artificially unstructured proteins, but not of their native, folded counterparts. Furthermore, these data were corroborated by experiments in cell lines with a nucleus-tagged myelin basic protein. Finally, mass spectrometry analysis of the products generated during proteasomal degradation of two proteins demonstrated that PA28γ does not increase, but rather decreases, the variability of peptides that are potentially suitable for MHC class I antigen presentation. These unexpected findings indicate that global stimulation of the degradation of unfolded proteins may represent a more general feature of PA28γ and suggests that this proteasomal activator might play a broader role in the pathway of protein degradation than previously believed.


Assuntos
Autoantígenos/metabolismo , Proteínas Intrinsicamente Desordenadas/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Células HeLa , Humanos , Proteólise , Resposta a Proteínas não Dobradas
2.
Int J Mol Sci ; 23(7)2022 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-35408925

RESUMO

In cattle, phenobarbital (PB) upregulates target drug-metabolizing enzyme (DME) mRNA levels. However, few data about PB's post-transcriptional effects are actually available. This work provides the first, and an almost complete, characterization of PB-dependent changes in DME catalytic activities in bovine liver using common probe substrates and confirmatory immunoblotting investigations. As expected, PB increased the total cytochrome P450 (CYP) content and the extent of metyrapone binding; moreover, an augmentation of protein amounts and related enzyme activities was observed for known PB targets such as CYP2B, 2C, and 3A, but also CYP2E1. However, contradictory results were obtained for CYP1A, while a decreased catalytic activity was observed for flavin-containing monooxygenases 1 and 3. The barbiturate had no effect on the chosen hydrolytic and conjugative DMEs. For the first time, we also measured the 26S proteasome activity, and the increase observed in PB-treated cattle would suggest this post-translational event might contribute to cattle DME regulation. Overall, this study increased the knowledge of cattle hepatic drug metabolism, and further confirmed the presence of species differences in DME expression and activity between cattle, humans, and rodents. This reinforced the need for an extensive characterization and understanding of comparative molecular mechanisms involved in expression, regulation, and function of DMEs.


Assuntos
Fenobarbital , Xenobióticos , Animais , Bovinos , Sistema Enzimático do Citocromo P-450/metabolismo , Indução Enzimática , Fígado/metabolismo , Microssomos Hepáticos/metabolismo , Fenobarbital/farmacologia , Xenobióticos/metabolismo
3.
Mol Cell ; 48(4): 601-11, 2012 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-23041283

RESUMO

Poorly structured domains in proteins enhance their susceptibility to proteasomal degradation. To learn whether the presence of such a domain near either end of a protein determines its direction of entry into the proteasome, directional translocation was enforced on several proteasome substrates. Using archaeal PAN-20S complexes, mammalian 26S proteasomes, and cultured cells, we identified proteins that are degraded exclusively from either the C or N terminus and some showing no directional preference. This property results from interactions of the substrate's termini with the regulatory ATPase and could be predicted based on the calculated relative stabilities of the N and C termini. Surprisingly, the direction of entry into the proteasome affected markedly the spectrum of peptides released and consequently influenced the efficiency of MHC class I presentation. Thus, easily unfolded termini are translocated first, and the direction of translocation influences the peptides generated and presented to the immune system.


Assuntos
Complexo de Endopeptidases do Proteassoma/metabolismo , Desdobramento de Proteína , Proteínas/química , Proteínas/metabolismo , Animais , Calmodulina/química , Calmodulina/imunologia , Calmodulina/metabolismo , Caseínas/química , Caseínas/imunologia , Caseínas/metabolismo , Linhagem Celular Tumoral , Proteínas Ligantes de Maltose/química , Proteínas Ligantes de Maltose/imunologia , Proteínas Ligantes de Maltose/metabolismo , Camundongos , Ovalbumina/química , Ovalbumina/imunologia , Ovalbumina/metabolismo , Complexo de Endopeptidases do Proteassoma/química , Transporte Proteico , Proteínas/imunologia
4.
Blood ; 129(15): 2132-2142, 2017 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-28130214

RESUMO

Systemic light chain (AL) amyloidosis is caused by the clonal production of an unstable immunoglobulin light chain (LC), which affects organ function systemically. Although pathogenic LCs have been characterized biochemically, little is known about the biology of amyloidogenic plasma cells (PCs). Intrigued by the unique response rates of AL amyloidosis patients to the first-in-class proteasome inhibitor (PI) bortezomib, we purified and investigated patient-derived AL PCs, in comparison with primary multiple myeloma (MM) PCs, the prototypical PI-responsive cells. Functional, biochemical, and morphological characterization revealed an unprecedented intrinsic sensitivity of AL PCs to PIs, even higher than that of MM PCs, associated with distinctive organellar features and expression patterns indicative of cellular stress. These consisted of expanded endoplasmic reticulum (ER), perinuclear mitochondria, and a higher abundance of stress-related transcripts, and were consistent with reduced autophagic control of organelle homeostasis. To test whether PI sensitivity stems from AL LC production, we engineered PC lines that can be induced to express amyloidogenic and nonamyloidogenic LCs, and found that AL LC expression alters cell growth and proteostasis and confers PI sensitivity. Our study discloses amyloidogenic LC production as an intrinsic PC stressor, and identifies stress-responsive pathways as novel potential therapeutic targets. Moreover, we contribute a cellular disease model to dissect the biology of AL PCs.


Assuntos
Amiloidose/tratamento farmacológico , Amiloidose/metabolismo , Bortezomib/farmacocinética , Cadeias Leves de Imunoglobulina/biossíntese , Plasmócitos/metabolismo , Inibidores de Proteassoma/farmacocinética , Amiloidose/patologia , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/patologia , Feminino , Humanos , Masculino , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Plasmócitos/patologia
5.
Biochim Biophys Acta ; 1843(9): 1942-7, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24851840

RESUMO

Immunoproteasomes are alternative forms of proteasomes specialized in the generation of MHC class I antigenic peptides and important for efficient cytokine production. We have identified a new biochemical property of 26S immunoproteasomes, namely the ability to hydrolyze basic proteins at greatly increased rates compared to constitutive proteasomes. This enhanced degradative capacity is specific for basic polypeptides, since substrates with a lower content in lysine and arginine residues are hydrolyzed at comparable rates by constitutive and immunoproteasomes. Crucially, selective inhibition of the immunoproteasome tryptic subunit ß2i strongly reduces degradation of basic proteins. Therefore, our data demonstrate the rate limiting function of the proteasomal trypsin-like activity in controlling turnover rates of basic protein substrates and suggest new biological roles for immunoproteasomes in maintaining cellular homeostasis by rapidly removing a potentially harmful excess of free histones that can build up under different pathophysiological conditions.


Assuntos
Complexo de Endopeptidases do Proteassoma/imunologia , Proteólise , Animais , Histonas/metabolismo , Hidrólise/efeitos dos fármacos , Cinética , Leupeptinas/farmacologia , Peso Molecular , Peptídeos/metabolismo , Proteólise/efeitos dos fármacos , Coelhos , Tripsina/metabolismo
6.
Biochimie ; 2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-38631454

RESUMO

PA28γ is a proteasomal interactor whose main and most known function is to stimulate the hydrolytic activity of the 20 S proteasome independently of ubiquitin and ATP. Unlike its two paralogues, PA28α and PA28ß, PA28γ is largely present in the nuclear compartment and plays pivotal functions in important pathways such as cellular division, apoptosis, neoplastic transformation, chromatin structure and organization, fertility, lipid metabolism, and DNA repair mechanisms. Although it is known that a substantial fraction of PA28γ is found in the cell in a free form (i.e. not associated with 20 S), almost all of the studies so far have focused on its ability to modulate proteasomal enzymatic activities. In this respect, the ability of PA28γ to strongly stimulate degradation of proteins, especially if intrinsically disordered and therefore devoid of three-dimensional tightly folded structure, appears to be the main molecular mechanism underlying its multiple biological effects. Initial studies, conducted more than 20 years ago, came to the conclusion that among the many biological functions of PA28γ, the immunological ones were rather limited and circumscribed. In this review, we focus on recent evidence showing that PA28γ fulfills significant functions in cell-mediated acquired immunity, with a particular role in attenuating MHC class I antigen presentation, especially in relation to neoplastic transformation and autoimmune diseases.

7.
Pharmacol Ther ; 241: 108329, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36526014

RESUMO

The immunoproteasome is a specialized form of proteasome equipped with modified catalytic subunits that was initially discovered to play a pivotal role in MHC class I antigen processing and immune system modulation. However, over the last years, this proteolytic complex has been uncovered to serve additional functions unrelated to antigen presentation. Accordingly, it has been proposed that immunoproteasome synergizes with canonical proteasome in different cell types of the nervous system, regulating neurotransmission, metabolic pathways and adaptation of the cells to redox or inflammatory insults. Hence, studying the alterations of immunoproteasome expression and activity is gaining research interest to define the dynamics of neuroinflammation as well as the early and late molecular events that are likely involved in the pathogenesis of a variety of neurological disorders. Furthermore, these novel functions foster the perspective of immunoproteasome as a potential therapeutic target for neurodegeneration. In this review, we provide a brain and retina-wide overview, trying to correlate present knowledge on structure-function relationships of immunoproteasome with the variety of observed neuro-modulatory functions.


Assuntos
Apresentação de Antígeno , Complexo de Endopeptidases do Proteassoma , Humanos , Complexo de Endopeptidases do Proteassoma/metabolismo , Transmissão Sináptica , Encéfalo/metabolismo
8.
Biomolecules ; 12(3)2022 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-35327535

RESUMO

Strictly controlled degradation of the proteome is a key factor in maintaining cellular homeostasis and allows a rapid and effective response to a variety of different stress challenges [...].


Assuntos
Complexo de Endopeptidases do Proteassoma , Proteoma , Homeostase/fisiologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteoma/metabolismo
9.
Biomolecules ; 12(2)2022 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-35204815

RESUMO

Carfilzomib is a last generation proteasome inhibitor (PI) with proven clinical efficacy in the treatment of relapsed/refractory multiple myeloma. This drug is considered to be extremely specific in inhibiting the chymotrypsin-like activity of the 20S proteasome, encoded by the ß5 subunit, overcoming some bortezomib limitations, the first PI approved for multiple myeloma therapy which is however burdened by a significant toxicity profile, due also to its off-target effects. Here, molecular approaches coupled with molecular docking studies have been used to unveil that the Insulin-Degrading Enzyme, a ubiquitous and highly conserved Zn2+ peptidase, often found to associate with proteasome in cell-based models, is targeted by carfilzomib in vitro. The drug behaves as a modulator of IDE activity, displaying an inhibitory effect over 10-fold lower than for the 20S. Notably, the interaction of IDE with the 20S enhances in vitro the inhibitory power of carfilzomib on proteasome, so that the IDE-20S complex is an even better target of carfilzomib than the 20S alone. Furthermore, IDE gene silencing after delivery of antisense oligonucleotides (siRNA) significantly reduced carfilzomib cytotoxicity in rMC1 cells, a validated model of Muller glia, suggesting that, in cells, the inhibitory activity of this drug on cell proliferation is somewhat linked to IDE and, possibly, also to its interaction with proteasome.


Assuntos
Antineoplásicos , Insulisina , Mieloma Múltiplo , Antineoplásicos/farmacologia , Humanos , Insulisina/genética , Insulisina/uso terapêutico , Simulação de Acoplamento Molecular , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/genética , Oligopeptídeos , Preparações Farmacêuticas , Complexo de Endopeptidases do Proteassoma , Inibidores de Proteassoma/farmacologia
10.
Cell Biosci ; 12(1): 108, 2022 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-35842713

RESUMO

BACKGROUND: Diabetic retinopathy (DR) is a microvascular complication of diabetes with a heavy impact on the quality of life of subjects and with a dramatic burden for health and economic systems on a global scale. Although the pathogenesis of DR is largely unknown, several preclinical data have pointed out to a main role of Muller glia (MG), a cell type which spans across the retina layers providing nourishment and support for Retina Ganglion Cells (RGCs), in sensing hyper-glycemia and in acquiring a pro-inflammatory polarization in response to this insult. RESULTS: By using a validated experimental model of DR in vitro, rMC1 cells challenged with high glucose, we uncovered the induction of an early (within minutes) and atypical Nuclear Factor-kB (NF-kB) signalling pathway regulated by a calcium-dependent calmodulin kinase II (CamKII)-proteasome axis. Phosphorylation of proteasome subunit Rpt6 (at Serine 120) by CamKII stimulated the accelerated turnover of IkBα (i.e., the natural inhibitor of p65-50 transcription factor), regardless of the phosphorylation at Serine 32 which labels canonical NF-kB signalling. This event allowed the p65-p50 heterodimer to migrate into the nucleus and to induce transcription of IL-8, Il-1ß and MCP-1. Pharmacological inhibition of CamKII as well as proteasome inhibition stopped this pro-inflammatory program, whereas introduction of a Rpt6 phospho-dead mutant (Rpt6-S120A) stimulated a paradoxical effect on NF-kB probably through the activation of a compensatory mechanism which may involve phosphorylation of 20S α4 subunit. CONCLUSIONS: This study introduces a novel pathway of MG activation by high glucose and casts some light on the biological relevance of proteasome post-translational modifications in modulating pathways regulated through targeted proteolysis.

11.
Blood ; 113(13): 3040-9, 2009 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-19164601

RESUMO

Proteasome inhibitors (PIs) are effective against multiple myeloma (MM), but the mechanisms of action and bases of individual susceptibility remain unclear. Recent work linked PI sensitivity to protein synthesis and proteasome activity, raising the question whether different levels of proteasome expression and workload underlie PI sensitivity in MM cells (MMCs). Exploiting human MM lines characterized by differential PI sensitivity, we report that highly sensitive MMCs express lower proteasome levels and higher proteasomal workload than relatively PI-resistant MMCs, resulting in the accumulation of polyubiquitinated proteins at the expense of free ubiquitin (proteasome stress). Manipulating proteasome expression or workload alters apoptotic sensitivity to PI, demonstrating a cause-effect relationship between proteasome stress and apoptotic responses in MMCs. Intracellular immunostaining in primary, patient-derived MMCs reveals that polyubiquitinated proteins hallmark neoplastic plasma cells, in positive correlation with immunoglobulin (Ig) content, both intra- and interpatient. Moreover, overall proteasome activity of primary MMCs inversely correlates with apoptotic sensitivity to PI. Altogether, our data indicate that the balance between proteasome workload and degradative capacity represents a critical determinant of apoptotic sensitivity of MMCs to PI, potentially providing a framework for identifying indicators of responsiveness and designing novel combination therapies.


Assuntos
Apoptose/efeitos dos fármacos , Inibidores de Cisteína Proteinase/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/fisiologia , Mieloma Múltiplo/tratamento farmacológico , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma , Animais , Linfócitos B/efeitos dos fármacos , Linfócitos B/metabolismo , Linfócitos B/patologia , Diferenciação Celular/efeitos dos fármacos , Inibidores de Cisteína Proteinase/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Células HeLa , Humanos , Leupeptinas/farmacologia , Camundongos , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Complexo de Endopeptidases do Proteassoma/fisiologia , Estresse Fisiológico/efeitos dos fármacos , Estresse Fisiológico/fisiologia , Células Tumorais Cultivadas
12.
Biomolecules ; 11(2)2021 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-33562807

RESUMO

PA28 (also known as 11S, REG or PSME) is a family of proteasome regulators whose members are widely present in many of the eukaryotic supergroups. In jawed vertebrates they are represented by three paralogs, PA28α, PA28ß, and PA28γ, which assemble as heptameric hetero (PA28αß) or homo (PA28γ) rings on one or both extremities of the 20S proteasome cylindrical structure. While they share high sequence and structural similarities, the three isoforms significantly differ in terms of their biochemical and biological properties. In fact, PA28α and PA28ß seem to have appeared more recently and to have evolved very rapidly to perform new functions that are specifically aimed at optimizing the process of MHC class I antigen presentation. In line with this, PA28αß favors release of peptide products by proteasomes and is particularly suited to support adaptive immune responses without, however, affecting hydrolysis rates of protein substrates. On the contrary, PA28γ seems to be a slow-evolving gene that is most similar to the common ancestor of the PA28 activators family, and very likely retains its original functions. Notably, PA28γ has a prevalent nuclear localization and is involved in the regulation of several essential cellular processes including cell growth and proliferation, apoptosis, chromatin structure and organization, and response to DNA damage. In striking contrast with the activity of PA28αß, most of these diverse biological functions of PA28γ seem to depend on its ability to markedly enhance degradation rates of regulatory protein by 20S proteasome. The present review will focus on the molecular mechanisms and biochemical properties of PA28γ, which are likely to account for its various and complex biological functions and highlight the common features with the PA28αß paralog.


Assuntos
Aterosclerose/genética , Autoantígenos/genética , Neoplasias/genética , Doenças Neurodegenerativas/genética , Complexo de Endopeptidases do Proteassoma/genética , Subunidades Proteicas/genética , Proteostase/genética , Sequência de Aminoácidos , Animais , Aterosclerose/enzimologia , Aterosclerose/patologia , Autoantígenos/química , Autoantígenos/metabolismo , Humanos , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Modelos Moleculares , Neoplasias/enzimologia , Neoplasias/patologia , Doenças Neurodegenerativas/enzimologia , Doenças Neurodegenerativas/patologia , Complexo de Endopeptidases do Proteassoma/química , Complexo de Endopeptidases do Proteassoma/metabolismo , Biossíntese de Proteínas , Conformação Proteica , Multimerização Proteica , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Proteólise , Homologia de Sequência de Aminoácidos , Ubiquitina/genética , Ubiquitina/metabolismo
13.
Cancers (Basel) ; 12(4)2020 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-32295216

RESUMO

Adaptation to import iron for proliferation makes cancer cells potentially sensitive to iron toxicity. Iron loading impairs multiple myeloma (MM) cell proliferation and increases the efficacy of the proteasome inhibitor bortezomib. Here, we defined the mechanisms of iron toxicity in MM.1S, U266, H929, and OPM-2 MM cell lines, and validated this strategy in preclinical studies using Vk*MYC mice as MM model. High-dose ferric ammonium citrate triggered cell death in all cell lines tested, increasing malondialdehyde levels, the by-product of lipid peroxidation and index of ferroptosis. In addition, iron exposure caused dose-dependent accumulation of polyubiquitinated proteins in highly iron-sensitive MM.1S and H929 cells, suggesting that proteasome workload contributes to iron sensitivity. Accordingly, high iron concentrations inhibited the proteasomal chymotrypsin-like activity of 26S particles and of MM cellular extracts in vitro. In all MM cells, bortezomib-iron combination induced persistent lipid damage, exacerbated bortezomib-induced polyubiquitinated proteins accumulation, and triggered cell death more efficiently than individual treatments. In Vk*MYC mice, addition of iron dextran or ferric carboxymaltose to the bortezomib-melphalan-prednisone (VMP) regimen increased the therapeutic response and prolonged remission without causing evident toxicity. We conclude that iron loading interferes both with redox and protein homeostasis, a property that can be exploited to design novel combination strategies including iron supplementation, to increase the efficacy of current MM therapies.

14.
Oncoimmunology ; 9(1): 1761205, 2020 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-32923122

RESUMO

The success of CD8+ T cell-based cancer immunotherapy emphasizes the importance of understanding the mechanisms of generation of MHC-I peptide ligands and the possible pathways of tumor cell escape from immunosurveillance. Recently, we showed that peptides generated in the nucleus during a pioneer round of mRNA translation (pioneer translation products, or PTPs) are an important source of tumor specific peptides which correlates with the aberrant splicing and transcription events associated with oncogenesis. Here we show that up-regulation of PSME3 proteasome activator in cancer cells results in increased destruction of PTP-derived peptides in the nucleus thus enabling cancer cell to subvert immunosurveillance. These findings unveil a previously unexpected role for PSME3 in antigen processing and identify PSME3 as a druggable target to improve the efficacy of cancer immunotherapy.


Assuntos
Apresentação de Antígeno , Complexo de Endopeptidases do Proteassoma , Antígenos de Histocompatibilidade Classe I , Monitorização Imunológica , Complexo de Endopeptidases do Proteassoma/genética , Evasão Tumoral
15.
Stem Cells ; 26(8): 2142-52, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18483423

RESUMO

The isolation of human embryonic and somatic stem cells of different types has made it possible to design novel gene and cell replacement therapies. Vectors derived from retro/lentiviruses are used to stably introduce genes into stem cells and their progeny. However, the permissivity to retroviral infection varies among cell types. We previously showed that hematopoietic stem cells are poorly permissive to human immunodeficiency virus (HIV)-derived vectors and that pharmacological inhibition of the proteasome strongly enhances gene transfer. Here we report that the proteasome limits lentiviral gene transfer in all stem cell types tested, including embryonic, mesenchymal, and neural, of both human and mouse origin. Remarkably, this inhibitory activity was sharply reduced upon differentiation of the stem cells, suggesting that it represents a novel feature of the stem cell/immature progenitor phenotype. Proteasome-mediated inhibition was specific for lentiviral vectors and occurred at a postentry infection step. It was not mediated by activation of nuclear factor-kappaB, a major signaling pathway modulated by the proteasome, and did not correlate with high proteasome activity. Interaction of the virion core with cyclophilin A was required to maximize the effect of proteasome inhibitor on the infection pathway. These findings are relevant to uncover new mediators of HIV gene transfer and help in designing more effective protocols for the genetic modification of stem cells. Disclosure of potential conflicts of interest is found at the end of this article.


Assuntos
Técnicas de Transferência de Genes , Lentivirus/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Células-Tronco/citologia , Animais , Células Cultivadas , Vetores Genéticos , HIV/genética , Humanos , Interferons/metabolismo , Camundongos , NF-kappa B/metabolismo , Fenótipo , Transdução de Sinais
16.
Sci Rep ; 7(1): 17626, 2017 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-29247244

RESUMO

Based on promising results in preclinical models, clinical trials have been performed to evaluate the efficacy of the first-in-class proteasome inhibitor bortezomib towards malignant pleural mesothelioma (MPM), an aggressive cancer arising from the mesothelium of the serous cavities following exposure to asbestos. Unexpectedly, only minimal therapeutic benefits were observed, thus implicating that MPM harbors inherent resistance mechanisms. Identifying the molecular bases of this primary resistance is crucial to develop novel pharmacologic strategies aimed at increasing the vulnerability of MPM to bortezomib. Therefore, we assessed a panel of four human MPM lines with different sensitivity to bortezomib, for functional proteasome activity and levels of free and polymerized ubiquitin. We found that highly sensitive MPM lines display lower proteasome activity than more bortezomib-resistant clones, suggesting that reduced proteasomal capacity might contribute to the intrinsic susceptibility of mesothelioma cells to proteasome inhibitors-induced apoptosis. Moreover, MPM equipped with fewer active proteasomes accumulated polyubiquitinated proteins, at the expense of free ubiquitin, a condition known as proteasome stress, which lowers the cellular apoptotic threshold and sensitizes mesothelioma cells to bortezomib-induced toxicity as shown herein. Taken together, our data suggest that an unfavorable load-versus-capacity balance represents a critical determinant of primary apoptotic sensitivity to bortezomib in MPM.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Bortezomib/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Mesotelioma/tratamento farmacológico , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma/farmacologia , Linhagem Celular Tumoral , Epitélio/patologia , Humanos , Mesotelioma Maligno , Proteínas Ubiquitinadas/metabolismo
17.
Vet Microbiol ; 192: 152-162, 2016 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-27527777

RESUMO

Small ruminant lentiviruses (SRLV) globally affect welfare and production of sheep and goats and are mainly controlled through elimination of infected animals, independently of the viral kinetics within the single animal. Control programs are based on highly sensitive serological tests, however the existence of low antibody responders leads to the permanent presence of seronegative infected animals in the flock, thus perpetuating the infection. On the other hand, long-term non-progressors show a detectable antibody response not indicative of a shedding animal, suggesting immune contention of infection. In this study, we analyse two goat populations within the same herd, harbouring low or high proviral SRLV loads respectively, both showing a robust antibody response. In vivo findings were confirmed in vitro since fibroblastic cell lines obtained from one high and one low proviral load representative goats, showed respectively a high and a faint production of virus upon infection with reference and field circulating SRLV strains. Differences in virus production were relieved when strain CAEV-Co was used for experimental infection. We analysed LTR promoter activity, proviral load, entry step and production of virus and viral proteins. Intriguingly, proteasomal activity was higher in fibroblasts from low proviral load animals and proteasome inhibition increased viral production in both cell lines, suggesting the implication of active proteasome-dependent restriction factors. Among them, we analysed relative expression and sequences of TRIM5α, APOBEC3 (Z1, Z2, Z3 and Z2-Z3) and BST-2 (Tetherin) and found a global antiviral status in low proviral carriers that may confer protection against viral shedding and disease onset.


Assuntos
Doenças das Cabras/virologia , Infecções por Lentivirus/veterinária , Lentivirus/classificação , Desaminases APOBEC/genética , Desaminases APOBEC/metabolismo , Animais , Biomarcadores , Linhagem Celular , DNA Viral/genética , Ensaio de Imunoadsorção Enzimática/veterinária , Fibroblastos/metabolismo , Fibroblastos/virologia , Regulação da Expressão Gênica , Predisposição Genética para Doença , Cabras , Humanos , Lentivirus/genética , Infecções por Lentivirus/virologia , Provírus , Purinas , Proteínas Virais/genética , Proteínas Virais/metabolismo , Internalização do Vírus , Eliminação de Partículas Virais
18.
Methods Enzymol ; 398: 336-52, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16275341

RESUMO

PA28 (also named REG or 11S) is a ring-shaped (180-kDa) interferon-gamma-induced complex that associates with the 20S proteasome and dramatically stimulates the breakdown of short peptides. Immunoprecipitation studies indicate that in vivo PA28 also exists in larger complexes that also contain the 19S particle, which is required for the ATP-ubiquitin-dependent degradation of proteins. However, because of its lability (e.g., it does not withstand exposure to high ionic strength buffers), this larger complex cannot be purified by standard biochemical protocols. Therefore, we developed a method to reconstitute in vitro such hybrid proteasomes (i.e., PA28-20S-19S) from highly purified components. This chapter describes conditions that allow the association of PA28 with "singly capped" 26S (i.e., 19S-20S) particles. In addition assays are described to measure absolute rates of degradation of several non-ubiquitinated proteins by 26S and 20S proteasomes and methods to analyze the pattern and size distribution of peptides generated during the degradation of these proteins.


Assuntos
Fragmentos de Peptídeos/análise , Fragmentos de Peptídeos/metabolismo , Complexo de Endopeptidases do Proteassoma/análise , Complexo de Endopeptidases do Proteassoma/isolamento & purificação , Animais , Apresentação de Antígeno/fisiologia , Cromatografia em Gel/métodos , Fluorescamina/metabolismo , Antígenos de Histocompatibilidade Classe I/fisiologia , Camundongos , Fragmentos de Peptídeos/ultraestrutura , Peptídeo Hidrolases/metabolismo , Complexo de Endopeptidases do Proteassoma/genética , Complexo de Endopeptidases do Proteassoma/ultraestrutura , Proteínas/genética , Proteínas/metabolismo , Coelhos , Coloração e Rotulagem
19.
Vet J ; 170(1): 84-90, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15993791

RESUMO

Sixty-nine cats were treated for injection-site sarcomas at the Veterinary Teaching Hospital of Grugliasco, Turin (Italy). The animals were divided into two subgroups: those subjected to four doxorubicin cycles combined with radical surgical excision 10 days after the second chemotherapy cycle (group A, 49 cats) or those treated with surgery alone (group B, 20 cats). Each cat was monitored for lung metastasis and local recurrence. In group A, 28 cats were alive at the end of the follow-up period. In this group, the recurrence rate was 40.8% while lung metastasis occurred in 12% of cats. In group B, eight animals were alive at the end of the follow-up period, while the rates of recurrence and metastasis were 35% and 10%. Neither the median disease-free interval nor the median overall survival was reached in either group. Moreover, there were no statistically significant differences between the two groups.


Assuntos
Antibióticos Antineoplásicos/uso terapêutico , Doenças do Gato/mortalidade , Doxorrubicina/uso terapêutico , Recidiva Local de Neoplasia/veterinária , Sarcoma/veterinária , Neoplasias de Tecidos Moles/veterinária , Animais , Antibióticos Antineoplásicos/administração & dosagem , Doenças do Gato/tratamento farmacológico , Doenças do Gato/patologia , Doenças do Gato/cirurgia , Gatos , Quimioterapia Adjuvante/veterinária , Intervalo Livre de Doença , Doxorrubicina/administração & dosagem , Esquema de Medicação , Feminino , Itália/epidemiologia , Masculino , Terapia Neoadjuvante/veterinária , Metástase Neoplásica , Recidiva Local de Neoplasia/mortalidade , Sarcoma/mortalidade , Neoplasias de Tecidos Moles/mortalidade , Análise de Sobrevida , Vacinação/efeitos adversos , Vacinação/veterinária
20.
Mol Immunol ; 39(3-4): 147-64, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12200047

RESUMO

Three different proteolytic processes have been shown to be important in the generation of antigenic peptides displayed on MHC-class I molecules. The great majority of these peoptides are derived from oligopeptides produced during the degradation of intracellular proteins by the ubiquitin-proteasome pathway. Novel methods were developed to follow this process in vitro. When pure 26S proteasomes degrade the model substrate, ovalbumin, they produce the immunodominant peptide, SIINFEKL, occasionally, but more often an N-extended form of SIINFEKL. Interferon-gamma stimulates antigen presentation in part by inducing new forms of the proteasome that are more efficient in antigen presentation, and in vitro these immunoproteasomes specifically produce more of the N-extended versions of SIINFEKL. In addition, gamma-interferon induces a novel 26S complex containing the 19S and 20S particles and the proteasome activator, PA28, which we show cleaves proteins in distinct ways. In vivo studies established that proteasomal cleavages produce the C-termini of antigenic peptides, but not their N-termini, which can be formed efficiently by aminopeptidases that trim longer proteasomal products to the presented epitopes. gamma-interferon stimulates this trimming process by inducing in the cytosol leucine aminopeptidase and a novel aminopeptidase in the ER. Peptides released by proteasomes, including antigenic peptides, are labile in cytosolic extracts, and most of the longer proteasome products are rapidly cleaved by the cytosolic enzyme, thymet oligopeptidase (TOP). If cells express large amounts of TOP, class I presentation decreases, and if TOP is inhibited, presentation increases. Thus, peptide degradation in the cytosol appears to limit the efficiency of antigen presentation.


Assuntos
Apresentação de Antígeno , Cisteína Endopeptidases/fisiologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Complexos Multienzimáticos/fisiologia , Peptídeos/metabolismo , Aminoácidos/metabolismo , Animais , Citosol/metabolismo , Humanos , Interferon gama/farmacologia , Complexo de Endopeptidases do Proteassoma , Ubiquitina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA