Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Blood ; 143(1): 42-56, 2024 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-37729611

RESUMO

ABSTRACT: The translocation t(11;14) occurs in 20% of patients with multiple myeloma (MM) and results in the upregulation of CCND1. Nearly two-thirds of t(11;14) MM cells are BCL2 primed and highly responsive to the oral BCL2 inhibitor venetoclax. Although it is evident that this unique sensitivity to venetoclax depends on the Bcl-2 homology domain 3- proapoptotic protein priming of BCL2, the biology underlying t(11;14) MM dependency on BCL2 is poorly defined. Importantly, the epigenetic regulation of t(11;14) transcriptomes and its impact on gene regulation and clinical response to venetoclax remain elusive. In this study, by integrating assay for transposase-accessible chromatin by sequencing (ATAC-seq) and RNA-seq at the single-cell level in primary MM samples, we have defined the epigenetic regulome and transcriptome associated with t(11;14) MM. A B-cell-like epigenetic signature was enriched in t(11;14) MM, confirming its phylogeny link to B-cell rather than plasma cell biology. Of note, a loss of a B-cell-like epigenetic signature with a gain of canonical plasma cell transcription factors was observed at the time of resistance to venetoclax. In addition, MCL1 and BCL2L1 copy number gains and structural rearrangements were linked to venetoclax resistance in patients with t(11;14) MM. To date, this is the first study in which both single-cell (sc) ATAC-seq and scRNA-seq analysis are integrated into primary MM cells to obtain a deeper resolution of the epigenetic regulome and transcriptome associated with t(11;14) MM biology and venetoclax resistance.


Assuntos
Antineoplásicos , Mieloma Múltiplo , Humanos , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Mieloma Múltiplo/tratamento farmacológico , Epigênese Genética , Antineoplásicos/uso terapêutico , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Compostos Bicíclicos Heterocíclicos com Pontes/uso terapêutico
2.
Blood Cancer Discov ; 5(1): 56-73, 2024 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-37934799

RESUMO

Immunomodulatory drugs (IMiD) are a backbone therapy for multiple myeloma (MM). Despite their efficacy, most patients develop resistance, and the mechanisms are not fully defined. Here, we show that IMiD responses are directed by IMiD-dependent degradation of IKZF1 and IKZF3 that bind to enhancers necessary to sustain the expression of MYC and other myeloma oncogenes. IMiD treatment universally depleted chromatin-bound IKZF1, but eviction of P300 and BRD4 coactivators only occurred in IMiD-sensitive cells. IKZF1-bound enhancers overlapped other transcription factor binding motifs, including ETV4. Chromatin immunoprecipitation sequencing showed that ETV4 bound to the same enhancers as IKZF1, and ETV4 CRISPR/Cas9-mediated ablation resulted in sensitization of IMiD-resistant MM. ETV4 expression is associated with IMiD resistance in cell lines, poor prognosis in patients, and is upregulated at relapse. These data indicate that ETV4 alleviates IKZF1 and IKZF3 dependency in MM by maintaining oncogenic enhancer activity and identify transcriptional plasticity as a previously unrecognized mechanism of IMiD resistance. SIGNIFICANCE: We show that IKZF1-bound enhancers are critical for IMiD efficacy and that the factor ETV4 can bind the same enhancers and substitute for IKZF1 and mediate IMiD resistance by maintaining MYC and other oncogenes. These data implicate transcription factor redundancy as a previously unrecognized mode of IMiD resistance in MM. See related article by Welsh, Barwick, et al., p. 34. See related commentary by Yun and Cleveland, p. 5. This article is featured in Selected Articles from This Issue, p. 4.


Assuntos
Mieloma Múltiplo , Humanos , Proteínas que Contêm Bromodomínio , Proteínas de Ciclo Celular , Agentes de Imunomodulação , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/genética , Recidiva Local de Neoplasia , Proteínas Nucleares , Proteínas Proto-Oncogênicas c-ets/genética , Fatores de Transcrição/genética , Ubiquitina-Proteína Ligases/fisiologia , Ubiquitina-Proteína Ligases/uso terapêutico
3.
Nat Med ; 29(9): 2295-2306, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37653344

RESUMO

B cell maturation antigen (BCMA) target loss is considered to be a rare event that mediates multiple myeloma (MM) resistance to anti-BCMA chimeric antigen receptor T cell (CAR T) or bispecific T cell engager (TCE) therapies. Emerging data report that downregulation of G-protein-coupled receptor family C group 5 member D (GPRC5D) protein often occurs at relapse after anti-GPRC5D CAR T therapy. To examine the tumor-intrinsic factors that promote MM antigen escape, we performed combined bulk and single-cell whole-genome sequencing and copy number variation analysis of 30 patients treated with anti-BCMA and/or anti-GPRC5D CAR T/TCE therapy. In two cases, MM relapse post-TCE/CAR T therapy was driven by BCMA-negative clones harboring focal biallelic deletions at the TNFRSF17 locus at relapse or by selective expansion of pre-existing subclones with biallelic TNFRSF17 loss. In another five cases of relapse, newly detected, nontruncating, missense mutations or in-frame deletions in the extracellular domain of BCMA negated the efficacies of anti-BCMA TCE therapies, despite detectable surface BCMA protein expression. In the present study, we also report four cases of MM relapse with biallelic mutations of GPRC5D after anti-GPRC5D TCE therapy, including two cases with convergent evolution where multiple subclones lost GPRC5D through somatic events. Immunoselection of BCMA- or GPRC5D-negative or mutant clones is an important tumor-intrinsic driver of relapse post-targeted therapies. Mutational events on BCMA confer distinct sensitivities toward different anti-BCMA therapies, underscoring the importance of considering the tumor antigen landscape for optimal design and selection of targeted immunotherapies in MM.


Assuntos
Mieloma Múltiplo , Receptores de Antígenos Quiméricos , Humanos , Mieloma Múltiplo/genética , Mieloma Múltiplo/terapia , Deriva e Deslocamento Antigênicos , Variações do Número de Cópias de DNA , Recidiva Local de Neoplasia , Imunoterapia , Anticorpos , Proteínas de Membrana
4.
Cancer Cell ; 41(4): 711-725.e6, 2023 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-36898378

RESUMO

Bispecific T cell engagers (TCEs) have shown promise in the treatment of various cancers, but the immunological mechanism and molecular determinants of primary and acquired resistance to TCEs remain poorly understood. Here, we identify conserved behaviors of bone marrow-residing T cells in multiple myeloma patients undergoing BCMAxCD3 TCE therapy. We show that the immune repertoire reacts to TCE therapy with cell state-dependent clonal expansion and find evidence supporting the coupling of tumor recognition via major histocompatibility complex class I (MHC class I), exhaustion, and clinical response. We find the abundance of exhausted-like CD8+ T cell clones to be associated with clinical response failure, and we describe loss of target epitope and MHC class I as tumor-intrinsic adaptations to TCEs. These findings advance our understanding of the in vivo mechanism of TCE treatment in humans and provide the rationale for predictive immune-monitoring and conditioning of the immune repertoire to guide future immunotherapy in hematological malignancies.


Assuntos
Anticorpos Biespecíficos , Mieloma Múltiplo , Humanos , Mieloma Múltiplo/tratamento farmacológico , Linfócitos T CD8-Positivos , Imunoterapia , Células Clonais/patologia , Anticorpos Biespecíficos/uso terapêutico
5.
Multimed Tools Appl ; : 1-17, 2023 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-36820085

RESUMO

Usability is generally considered as a metric to judge the efficacy of any interface. This is also true for the web pages of a website. There are different factors - efficiency, memorability, learnability, errors, and aesthetics play significant roles in order to determine usability. In this work, we proposed a computational model to predict the efficiency with which users can do a particular task on a website. We considered seventeen features of web pages that may affect the efficiency of a task. The statistical significance of these features was tested based on the empirical data collected using twenty websites. For each website, a representative task was identified. Twenty participants completed these tasks using a controlled environment within a group. Task completion times were recorded for feature identification. The one Dimensional ANOVA study reveals sixteen out of the seventeen are statistically significant for efficiency measurement. Using these features, a computational model was developed based on the Support Vector Regression. Experimental results show that our model can predict the efficiency of web pages' tasks with an accuracy of 90.64%.

6.
RSC Med Chem ; 13(6): 726-730, 2022 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-35814928

RESUMO

To develop new degrader molecules from an existing protein ligand a linkage vector must be identified and then joined with a suitable E3 ligase without disrupting binding to the respective targets. This is typically achieved through empirically evaluating the degradation efficacy of a series of synthetic degraders. Our strategy for determining optimal linkage sites utilises biotinylated protein ligands, linked via potential conjugation sites of an inhibitor to confirm whether target protein is maintained after forming a conjugate. This method provides low-cost, qualitative evidence that the addition of a linker moiety at a specific position can be tolerated, guiding further optimisation. We demonstrate the application of this method through the exploration of linkage vectors on A-485, a known ligand of p300/CBP, and found a conjugation site through a urea moiety. Pomalidomide was then conjugated through this site with several different linkers and cell viability and degradation were assessed for this library using a myeloma cell line, MM1.S. Compound 18i, with a PEG4 linker, was found to be the most effective p300 degrader and linker length greater than 10 atoms afforded enhanced degradation.

7.
Front Oncol ; 10: 636, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32432039

RESUMO

Immunotherapy has recently emerged as a promising treatment option for multiple myeloma (MM) patients. Profound immune dysfunction and evasion of immune surveillance are known to characterize MM evolution and disease progression. Along with genomic changes observed in malignant plasma cells, the bone marrow (BM) milieu creates a protective environment sustained by the complex interaction of BM stromal cells (BMSCs) and malignant cells that using bidirectional connections and cytokines released stimulate disease progression, drug resistance and enable immune escape. Local immune suppression and T-cell exhaustion are important mediating factors of clinical outcomes and responses to immune-based approaches. Thus, further characterization of the defects present in the immune system of MM patients is essential to develop novel therapies and to repurpose the existing ones. This review seeks to provide insights into the mechanisms that promote tumor escape, cause inadequate T-cell stimulation and impaired cytotoxicity in MM. Furthermore, it highlights current immunotherapies being used to restore adaptive T-cell immune responses in MM and describes strategies created to escape these multiple immune evasion mechanisms.

8.
J Med Chem ; 62(11): 5522-5540, 2019 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-31117518

RESUMO

Protein-protein interactions (PPIs) have emerged as significant targets for therapeutic development, owing to their critical nature in diverse biological processes. An ideal PPI-based target is the protein myeloid cell leukemia 1 (MCL1), a critical prosurvival factor in cancers such as multiple myeloma where MCL1 levels directly correlate to disease progression. Current strategies for halting the antiapoptotic properties of MCL1 revolve around inhibiting its sequestration of proapoptotic factors. Existing inhibitors disrupt endogenous regulatory proteins; however, this strategy actually leads to an increase of MCL1 protein levels. Here, we show the development of hetero-bifunctional small molecules capable of selectively targeting MCL1 using a proteolysis targeting chimera (PROTAC) methodology leading to successful degradation. We have confirmed the involvement of the E3 ligase CUL4A-DDB1 cereblon ubiquitination pathway, making these PROTACs a first step toward a new class of antiapoptotic B-cell lymphoma 2 family protein degraders.


Assuntos
Proteína de Sequência 1 de Leucemia de Células Mieloides/metabolismo , Proteólise/efeitos dos fármacos , Linhagem Celular , Humanos , Indóis/farmacologia , Modelos Moleculares , Proteína de Sequência 1 de Leucemia de Células Mieloides/química , Complexo de Endopeptidases do Proteassoma/metabolismo , Conformação Proteica , Sulfonamidas/farmacologia , Ubiquitinação/efeitos dos fármacos
9.
Microbiol Mol Biol Rev ; 78(1): 40-73, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24600040

RESUMO

All living organisms are continuously faced with endogenous or exogenous stress conditions affecting genome stability. DNA repair pathways act as a defense mechanism, which is essential to maintain DNA integrity. There is much to learn about the regulation and functions of these mechanisms, not only in human cells but also equally in divergent organisms. In trypanosomatids, DNA repair pathways protect the genome against mutations but also act as an adaptive mechanism to promote drug resistance. In this review, we scrutinize the molecular mechanisms and DNA repair pathways which are conserved in trypanosomatids. The recent advances made by the genome consortiums reveal the complete genomic sequences of several pathogens. Therefore, using bioinformatics and genomic sequences, we analyze the conservation of DNA repair proteins and their key protein motifs in trypanosomatids. We thus present a comprehensive view of DNA repair processes in trypanosomatids at the crossroads of DNA repair and drug resistance.


Assuntos
Reparo do DNA/fisiologia , Resistência a Medicamentos/genética , Trypanosomatina/efeitos dos fármacos , Trypanosomatina/genética , DNA , Humanos , Leishmaniose/tratamento farmacológico , Leishmaniose/parasitologia , Tripanossomíase/tratamento farmacológico , Tripanossomíase/parasitologia
10.
Cell Rep ; 6(3): 553-64, 2014 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-24485656

RESUMO

One envisioned function of homologous recombination (HR) is to find a template for DNA synthesis from the resected 3'-OH molecules that occur during double-strand break (DSB) repair at collapsed replication forks. However, the interplay between DNA synthesis and HR remains poorly understood in higher eukaryotic cells. Here, we reveal functions for the breast cancer proteins BRCA2 and PALB2 at blocked replication forks and show a role for these proteins in stimulating polymerase η (Polη) to initiate DNA synthesis. PALB2, BRCA2, and Polη colocalize at stalled or collapsed replication forks after hydroxyurea treatment. Moreover, PALB2 and BRCA2 interact with Polη and are required to sustain the recruitment of Polη at blocked replication forks. PALB2 and BRCA2 stimulate Polη-dependent DNA synthesis on D loop substrates. We conclude that PALB2 and BRCA2, in addition to their functions in D loop formation, play crucial roles in the initiation of recombination-associated DNA synthesis by Polη-mediated DNA repair.


Assuntos
Proteína BRCA2/metabolismo , Neoplasias da Mama/metabolismo , Replicação do DNA , DNA Polimerase Dirigida por DNA/metabolismo , DNA/biossíntese , Proteínas Nucleares/metabolismo , Recombinação Genética , Proteínas Supressoras de Tumor/metabolismo , Linhagem Celular , Quebras de DNA de Cadeia Dupla , Proteína do Grupo de Complementação N da Anemia de Fanconi , Feminino , Técnicas de Silenciamento de Genes , Humanos , Leishmania infantum/metabolismo , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas
11.
Mol Cell ; 53(1): 7-18, 2014 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-24316220

RESUMO

MRE11 within the MRE11-RAD50-NBS1 (MRN) complex acts in DNA double-strand break repair (DSBR), detection, and signaling; yet, how its endo- and exonuclease activities regulate DSBR by nonhomologous end-joining (NHEJ) versus homologous recombination (HR) remains enigmatic. Here, we employed structure-based design with a focused chemical library to discover specific MRE11 endo- or exonuclease inhibitors. With these inhibitors, we examined repair pathway choice at DSBs generated in G2 following radiation exposure. While nuclease inhibition impairs radiation-induced replication protein A (RPA) chromatin binding, suggesting diminished resection, the inhibitors surprisingly direct different repair outcomes. Endonuclease inhibition promotes NHEJ in lieu of HR, while exonuclease inhibition confers a repair defect. Collectively, the results describe nuclease-specific MRE11 inhibitors, define distinct nuclease roles in DSB repair, and support a mechanism whereby MRE11 endonuclease initiates resection, thereby licensing HR followed by MRE11 exonuclease and EXO1/BLM bidirectional resection toward and away from the DNA end, which commits to HR.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA por Junção de Extremidades , Proteínas de Ligação a DNA/metabolismo , Inibidores Enzimáticos/química , Fase G2 , Reparo de DNA por Recombinação , Linhagem Celular , Cromatina/genética , Cromatina/metabolismo , Enzimas Reparadoras do DNA/genética , Enzimas Reparadoras do DNA/metabolismo , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/genética , Exodesoxirribonucleases/genética , Exodesoxirribonucleases/metabolismo , Raios gama/efeitos adversos , Humanos , Proteína Homóloga a MRE11 , Proteína de Replicação A/genética , Proteína de Replicação A/metabolismo
12.
J Vis Exp ; (80): e50320, 2013 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-24193370

RESUMO

Key assays in enzymology for the biochemical characterization of proteins in vitro necessitate high concentrations of the purified protein of interest. Protein purification protocols should combine efficiency, simplicity and cost effectiveness. Here, we describe the GST-His method as a new small-scale affinity purification system for recombinant proteins, based on a N-terminal Glutathione Sepharose Tag (GST) and a C-terminal 10xHis tag, which are both fused to the protein of interest. The latter construct is used to generate baculoviruses, for infection of Sf9 infected cells for protein expression. GST is a rather long tag (29 kDa) which serves to ensure purification efficiency. However, it might influence physiological properties of the protein. Hence, it is subsequently cleaved off the protein using the PreScission enzyme. In order to ensure maximum purity and to remove the cleaved GST, we added a second affinity purification step based on the comparatively small His-Tag. Importantly, our technique is based on two different tags flanking the two ends of the protein, which is an efficient tool to remove degraded proteins and, therefore, enriches full-length proteins. The method presented here does not require an expensive instrumental setup, such as FPLC. Additionally, we incorporated MgCl2 and ATP washes to remove heat shock protein impurities and nuclease treatment to abolish contaminating nucleic acids. In summary, the combination of two different tags flanking the N- and the C-terminal and the capability to cleave off one of the tags, guaranties the recovery of a highly purified and full-length protein of interest.


Assuntos
Cromatografia de Afinidade/métodos , Glutationa/química , Histidina/química , Proteínas Recombinantes/isolamento & purificação
13.
J Biol Chem ; 288(52): 37010-25, 2013 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-24247247

RESUMO

The mammalian protein arginine methyltransferase 7 (PRMT7) has been implicated in roles of transcriptional regulation, DNA damage repair, RNA splicing, cell differentiation, and metastasis. However, the type of reaction that it catalyzes and its substrate specificity remain controversial. In this study, we purified a recombinant mouse PRMT7 expressed in insect cells that demonstrates a robust methyltransferase activity. Using a variety of substrates, we demonstrate that the enzyme only catalyzes the formation of ω-monomethylarginine residues, and we confirm its activity as the prototype type III protein arginine methyltransferase. This enzyme is active on all recombinant human core histones, but histone H2B is a highly preferred substrate. Analysis of the specific methylation sites within intact histone H2B and within H2B and H4 peptides revealed novel post-translational modification sites and a unique specificity of PRMT7 for methylating arginine residues in lysine- and arginine-rich regions. We demonstrate that a prominent substrate recognition motif consists of a pair of arginine residues separated by one residue (RXR motif). These findings will significantly accelerate substrate profile analysis, biological function study, and inhibitor discovery for PRMT7.


Assuntos
Histonas/química , Processamento de Proteína Pós-Traducional/fisiologia , Proteína-Arginina N-Metiltransferases/química , Motivos de Aminoácidos , Animais , Arginina/química , Arginina/genética , Arginina/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Lisina/química , Lisina/genética , Lisina/metabolismo , Metilação , Camundongos , Proteína-Arginina N-Metiltransferases/genética , Proteína-Arginina N-Metiltransferases/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Células Sf9 , Spodoptera , Especificidade por Substrato
14.
Hum Mol Genet ; 19(23): 4726-34, 2010 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-20858601

RESUMO

Lafora disease (LD) is an autosomal recessive progressive myoclonic epilepsy characterized by the presence of intracellular polyglucosan inclusions commonly known as Lafora bodies in many tissues, including the brain, liver and skin. The disease is caused by mutations in either EPM2A gene, encoding the protein phosphatase, laforin, or EPM2B gene, encoding the ubiquitin ligase, malin. But how mutations in these two genes cause disease pathogenesis is poorly understood. In this study, we show that the Lafora bodies in the axillary skin and brain stain positively for the ubiquitin, the 20S proteasome and the molecular chaperones Hsp70/Hsc70. Interestingly, mutant malins that are misfolded also frequently colocalizes with Lafora bodies in the skin biopsy sample of the respective LD patient. The expression of disease-causing mutations of malin in Cos-7 cells results in the formation of the profuse cytoplasmic aggregates that colocalize with the Hsp70/Hsc70 chaperones and the 20S proteasome. The mutant malin expressing cells also exhibit proteasomal dysfunction and cell death. Overexpression of Hsp70 decreases the frequency of the mutant malin aggregation and protects from mutant malin-induced cell death. These findings suggest that Lafora bodies consist of abnormal proteins, including mutant malin, targeted by the chaperones or the proteasome for their refolding or clearance, and failure of these quality control systems could lead to LD pathogenesis. Our data also indicate that the Hsp70 chaperone could be a potential therapeutic target of LD.


Assuntos
Proteínas de Transporte/genética , Proteínas de Choque Térmico HSC70/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Corpos de Inclusão/metabolismo , Doença de Lafora , Complexo de Endopeptidases do Proteassoma/metabolismo , Animais , Células COS , Morte Celular , Chlorocebus aethiops , Imunofluorescência , Glucanos/genética , Proteínas de Choque Térmico HSC70/genética , Proteínas de Choque Térmico HSP70/genética , Humanos , Corpos de Inclusão/patologia , Doença de Lafora/genética , Doença de Lafora/metabolismo , Doença de Lafora/patologia , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Mutação , Complexo de Endopeptidases do Proteassoma/genética , Ubiquitina/química , Ubiquitina/genética , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases
15.
J Cell Biochem ; 109(5): 933-42, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20069556

RESUMO

Capsaicin is an active component of red pepper having an antiproliferative effect in a variety of cancer cells, which recent evidence suggests due to its ability to induce apoptosis. However, the molecular mechanisms through which capsaicin induces apoptosis are not well understood. Here we demonstrate that capsaicin-induced apoptosis is mediated via the inhibition cellular proteasome function. Treatment of capsaicin to mouse neuro 2a cells results in the inhibition of proteasome activity in a dose- and time-dependent manner that seems to correlate with its effect on cell death. The effect of capsaicin on cellular proteasome function is indirect and probably mediated via the generation of oxidative stress. Exposure of capsaicin also causes increased accumulation of ubiquitinated proteins as wells as various target substrates of proteasome like p53 and Bax and p27. Like many other classical proteasome inhibitors, capsaicin also triggers the intrinsic pathway of apoptosis involving mitochondria and induces neurite outgrowth. Our results strongly support for the use of capsaicin as an anticancer drug.


Assuntos
Apoptose/efeitos dos fármacos , Capsaicina/farmacologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina/metabolismo , Animais , Caspases/metabolismo , Linhagem Celular , Citocromos c/metabolismo , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Proteínas I-kappa B/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Inibidor de NF-kappaB alfa , NF-kappa B/metabolismo , Neuritos/efeitos dos fármacos , Neuritos/metabolismo , Inibidores de Proteassoma , Espécies Reativas de Oxigênio/metabolismo , Especificidade por Substrato/efeitos dos fármacos , Fatores de Tempo
16.
J Biol Chem ; 285(2): 1404-13, 2010 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-19892702

RESUMO

Lafora disease (LD) is an autosomal recessive neurodegenerative disorder caused by mutation in either the dual specificity phosphatase laforin or ubiquitin ligase malin. A pathological hallmark of LD is the accumulation of cytoplasmic polyglucosan inclusions commonly known as Lafora bodies in both neuronal and non-neuronal tissues. How mutations in these two proteins cause disease pathogenesis is not well understood. Malin interacts with laforin and recruits to aggresomes upon proteasome inhibition and was shown to degrade misfolded proteins. Here we report that malin is spontaneously misfolded and tends to be aggregated, degraded by proteasomes, and forms not only aggresomes but also other cytoplasmic and nuclear aggregates in all transfected cells upon proteasomal inhibition. Malin also interacts with Hsp70. Several disease-causing mutants of malin are comparatively more unstable than wild type and form aggregates in most transfected cells even without the inhibition of proteasome function. These cytoplasmic and nuclear aggregates are immunoreactive to ubiquitin and 20 S proteasome. Interestingly, progressive proteasomal dysfunction and cell death is also most frequently observed in the mutant malin-overexpressed cells compared with the wild-type counterpart. Finally, we demonstrate that the co-chaperone carboxyl terminus of the Hsc70-interacting protein (CHIP) stabilizes malin by modulating the activity of Hsp70. All together, our results suggest that malin is unstable, and the aggregate-prone protein and co-chaperone CHIP can modulate its stability.


Assuntos
Proteínas de Transporte/metabolismo , Doença de Lafora/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Dobramento de Proteína , Ubiquitina-Proteína Ligases/metabolismo , Animais , Células COS , Proteínas de Transporte/genética , Chlorocebus aethiops , Glucanos/genética , Glucanos/metabolismo , Proteínas de Choque Térmico HSC70/genética , Proteínas de Choque Térmico HSC70/metabolismo , Humanos , Corpos de Inclusão/genética , Corpos de Inclusão/metabolismo , Doença de Lafora/genética , Mutação , Complexo de Endopeptidases do Proteassoma/genética , Proteínas Tirosina Fosfatases não Receptoras/genética , Proteínas Tirosina Fosfatases não Receptoras/metabolismo , Ubiquitina-Proteína Ligases/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...