Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cancers (Basel) ; 14(6)2022 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-35326710

RESUMO

There are limited and discrepant data on prostate cancer (PCa) and vitamin D. We investigated changes in three vitamin D3 metabolites in PCa patients after prostatectomy with zoledronic acid (ZA) treatment regarding their metastasis statuses over four years. In 32 patients from the ZEUS trial, 25(OH)D3, 24,25(OH)2D3, and 1,25(OH)2D3 were measured with liquid chromatography coupled with tandem mass spectrometry at four time points. All the patients received daily calcium and vitamin D3. Bone metastases were detected in 7 of the 17 ZA-treated patients and in 5 of the 15 controls (without ZA), without differences between the groups (p = 0.725). While 25(OH)D3 and 24,25(OH)2D3 increased significantly after the study's start, with following constant values, the 1,25(OH)2D3 concentrations remained unchanged. ZA treatment did not change the levels of the three metabolites. 25(OH)D3 and 24,25(OH)2D3 were not associated with the development of bone metastases. In contrast, 1,25(OH)2D3 was also higher in patients with bone metastasis before the study's start. Thus, in high-risk PCa patients after prostatectomy, 25(OH)D3, 24,25(OH)2D3, and 1,25(OH)2D3 were not affected by supportive ZA treatment or by the development of metastasis over four years, with the exception of 1,25(OH)2D3, which was constantly higher in metastatic patients. There might be potential prognostic value if the results can be confirmed.

2.
J Cell Biochem ; 123(1): 77-90, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34180073

RESUMO

By regulating several hallmarks of cancer, BAG3 exerts oncogenic functions in a wide variety of malignant diseases including glioblastoma (GBM) and triple-negative breast cancer (TNBC). Here we performed global proteomic/phosphoproteomic analyses of CRISPR/Cas9-mediated isogenic BAG3 knockouts of the two GBM lines U343 and U251 in comparison to parental controls. Depletion of BAG3 evoked major effects on proteins involved in ciliogenesis/ciliary function and the activity of the related kinases aurora-kinase A and CDK1. Cilia formation was significantly enhanced in BAG3 KO cells, a finding that could be confirmed in BAG3-deficient versus -proficient BT-549 TNBC cells, thus identifying a completely novel function of BAG3 as a negative regulator of ciliogenesis. Furthermore, we demonstrate that enhanced ciliogenesis and reduced expression of SNAI1 and ZEB1, two key transcription factors regulating epithelial to mesenchymal transition (EMT) are correlated to decreased cell migration, both in the GBM and TNBC BAG3 knockout cells. Our data obtained in two different tumor entities identify suppression of EMT and ciliogenesis as putative synergizing mechanisms of BAG3-driven tumor aggressiveness in therapy-resistant cancers.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Neoplasias Encefálicas/metabolismo , Cílios/enzimologia , Glioblastoma/metabolismo , Transdução de Sinais/genética , Neoplasias de Mama Triplo Negativas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Reguladoras de Apoptose/genética , Aurora Quinase A/metabolismo , Neoplasias Encefálicas/patologia , Proteína Quinase CDC2/metabolismo , Sistemas CRISPR-Cas , Linhagem Celular Tumoral , Movimento Celular/genética , Resistencia a Medicamentos Antineoplásicos/genética , Transição Epitelial-Mesenquimal/genética , Técnicas de Inativação de Genes/métodos , Glioblastoma/patologia , Humanos , Proteômica/métodos , Fatores de Transcrição da Família Snail/metabolismo , Neoplasias de Mama Triplo Negativas/patologia , Homeobox 1 de Ligação a E-box em Dedo de Zinco/metabolismo
3.
Front Cell Dev Biol ; 8: 208, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32296703

RESUMO

Short linear motifs (SLiMs) located in disordered regions of multidomain proteins are important for the organization of protein-protein interaction networks. By dynamic association with their binding partners, SLiMs enable assembly of multiprotein complexes, pivotal for the regulation of various aspects of cell biology in higher organisms. Despite their importance, there is a paucity of molecular tools to study SLiMs of endogenous proteins in live cells. LC3 interacting regions (LIRs), being quintessential for orchestrating diverse stages of autophagy, are a prominent example of SLiMs and mediate binding to the ubiquitin-like LC3/GABARAP family of proteins. The role of LIRs ranges from the posttranslational processing of their binding partners at early stages of autophagy to the binding of selective autophagy receptors (SARs) to the autophagosome. In order to generate tools to study LIRs in cells, we engineered high affinity binders of LIR motifs of three archetypical SARs: OPTN, p62, and NDP52. In an array of in vitro and cellular assays, the engineered binders were shown to have greatly improved affinity and specificity when compared with the endogenous LC3/GABARAP family of proteins, thus providing a unique possibility for modulating LIR interactions in living systems. We exploited these novel tools to study the impact of LIR inhibition on the fitness and the responsiveness to cytarabine treatment of THP-1 cells - a model for studying acute myeloid leukemia (AML). Our results demonstrate that inhibition of LIR of a single autophagy receptor is insufficient to sensitize the cells to cytarabine, while simultaneous inhibition of three LIR motifs in three distinct SARs reduces the IC50 of the chemotherapeutic.

4.
Mol Cell ; 77(1): 164-179.e6, 2020 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-31732457

RESUMO

The family of bacterial SidE enzymes catalyzes non-canonical phosphoribosyl-linked (PR) serine ubiquitination and promotes infectivity of Legionella pneumophila. Here, we describe identification of two bacterial effectors that reverse PR ubiquitination and are thus named deubiquitinases for PR ubiquitination (DUPs; DupA and DupB). Structural analyses revealed that DupA and SidE ubiquitin ligases harbor a highly homologous catalytic phosphodiesterase (PDE) domain. However, unlike SidE ubiquitin ligases, DupA displays increased affinity to PR-ubiquitinated substrates, which allows DupA to cleave PR ubiquitin from substrates. Interfering with DupA-ubiquitin binding switches its activity toward SidE-type ligase. Given the high affinity of DupA to PR-ubiquitinated substrates, we exploited a catalytically inactive DupA mutant to trap and identify more than 180 PR-ubiquitinated host proteins in Legionella-infected cells. Proteins involved in endoplasmic reticulum (ER) fragmentation and membrane recruitment to Legionella-containing vacuoles (LCV) emerged as major SidE targets. The global map of PR-ubiquitinated substrates provides critical insights into host-pathogen interactions during Legionella infection.


Assuntos
Enzimas Desubiquitinantes/metabolismo , Serina/metabolismo , Ubiquitina/metabolismo , Ubiquitinação/fisiologia , Células A549 , Proteínas de Bactérias/metabolismo , Domínio Catalítico/fisiologia , Linhagem Celular , Linhagem Celular Tumoral , Retículo Endoplasmático/metabolismo , Células HEK293 , Células HeLa , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Legionella pneumophila/patogenicidade , Doença dos Legionários/metabolismo , Vacúolos/metabolismo
5.
Cancers (Basel) ; 11(3)2019 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-30871073

RESUMO

Glioblastoma is one of the deadliest malignancies and is virtually incurable. Accumulating evidence indicates that a small population of cells with a stem-like phenotype is the major culprit of tumor recurrence. Enhanced DNA repair capacity and expression of stemness marker genes are the main characteristics of these cells. Elimination of this population might delay or prevent tumor recurrence following radiochemotherapy. The aim of this study was to analyze whether interference with the Hedgehog signaling (Hh) pathway or combined Hh/Notch blockade using small-molecule inhibitors can efficiently target these cancer stem cells and sensitize them to therapy. Using tumor sphere lines and primary patient-derived glioma cultures we demonstrate that the Hh pathway inhibitor GANT61 (GANT) and the arsenic trioxide (ATO)-mediated Hh/Notch inhibition are capable to synergistically induce cell death in combination with the natural anticancer agent (-)-Gossypol (Gos). Only ATO in combination with Gos also strongly decreased stemness marker expression and prevented sphere formation and recovery. These synergistic effects were associated with distinct proteomic changes indicating diminished DNA repair and markedly reduced stemness. Finally, using an organotypic brain slice transplantation model, we show that combined ATO/Gos treatment elicits strong growth inhibition or even complete elimination of tumors. Collectively, our data show for the first time that ATO and Gos, two drugs that can be used in the clinic, represent a promising targeted therapy approach for the synergistic elimination of glioma stem-like cells.

6.
Nature ; 557(7707): 734-738, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29795347

RESUMO

Conventional ubiquitination regulates key cellular processes by catalysing the ATP-dependent formation of an isopeptide bond between ubiquitin (Ub) and primary amines in substrate proteins 1 . Recently, the SidE family of bacterial effector proteins (SdeA, SdeB, SdeC and SidE) from pathogenic Legionella pneumophila were shown to use NAD+ to mediate phosphoribosyl-linked ubiquitination of serine residues in host proteins2, 3. However, the molecular architecture of the catalytic platform that enables this complex multistep process remains unknown. Here we describe the structure of the catalytic core of SdeA, comprising mono-ADP-ribosyltransferase (mART) and phosphodiesterase (PDE) domains, and shed light on the activity of two distinct catalytic sites for serine ubiquitination. The mART catalytic site is composed of an α-helical lobe (AHL) that, together with the mART core, creates a chamber for NAD+ binding and ADP-ribosylation of ubiquitin. The catalytic site in the PDE domain cleaves ADP-ribosylated ubiquitin to phosphoribosyl ubiquitin (PR-Ub) and mediates a two-step PR-Ub transfer reaction: first to a catalytic histidine 277 (forming a transient SdeA H277-PR-Ub intermediate) and subsequently to a serine residue in host proteins. Structural analysis revealed a substrate binding cleft in the PDE domain, juxtaposed with the catalytic site, that is essential for positioning serines for ubiquitination. Using degenerate substrate peptides and newly identified ubiquitination sites in RTN4B, we show that disordered polypeptides with hydrophobic residues surrounding the target serine residues are preferred substrates for SdeA ubiquitination. Infection studies with L. pneumophila expressing substrate-binding mutants of SdeA revealed that substrate ubiquitination, rather than modification of the cellular ubiquitin pool, determines the pathophysiological effect of SdeA during acute bacterial infection.


Assuntos
Biocatálise , Legionella pneumophila/enzimologia , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Serina/metabolismo , Ubiquitinação , ADP Ribose Transferases/química , ADP Ribose Transferases/metabolismo , Difosfato de Adenosina/metabolismo , Sequência de Aminoácidos , Proteínas de Bactérias , Sítios de Ligação , Domínio Catalítico , Cristalografia por Raios X , Legionella pneumophila/genética , Legionella pneumophila/patogenicidade , Doença dos Legionários/microbiologia , Proteínas de Membrana/genética , Modelos Moleculares , Diester Fosfórico Hidrolases/química , Diester Fosfórico Hidrolases/metabolismo , Estrutura Secundária de Proteína , Especificidade por Substrato , Ubiquitina/metabolismo
7.
Neoplasia ; 20(3): 263-279, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29462756

RESUMO

Target-specific treatment modalities are currently not available for triple-negative breast cancer (TNBC), and acquired chemotherapy resistance is a primary obstacle for the treatment of these tumors. Here we employed derivatives of BT-549 and MDA-MB-468 TNBC cell lines that were adapted to grow in the presence of either 5-Fluorouracil, Doxorubicin or Docetaxel in an aim to identify molecular pathways involved in the adaptation to drug-induced cell killing. All six drug-adapted BT-549 and MDA-MB-468 cell lines displayed cross resistance to chemotherapy and decreased apoptosis sensitivity. Expression of the anti-apoptotic co-chaperone BAG3 was notably enhanced in two thirds (4/6) of the six resistant lines simultaneously with higher expression of HSP70 in comparison to parental controls. Doxorubicin-resistant BT-549 (BT-549rDOX20) and 5-Fluorouracil-resistant MDA-MB-468 (MDA-MB-468r5-FU2000) cells were chosen for further analysis with the autophagy inhibitor Bafilomycin A1 and lentiviral depletion of ATG5, indicating that enhanced cytoprotective autophagy partially contributes to increased drug resistance and cell survival. Stable lentiviral BAG3 depletion was associated with a robust down-regulation of Mcl-1, Bcl-2 and Bcl-xL, restoration of drug-induced apoptosis and reduced cell adhesion in these cells, and these death-sensitizing effects could be mimicked with the BAG3/Hsp70 interaction inhibitor YM-1 and by KRIBB11, a selective transcriptional inhibitor of HSF-1. Furthermore, BAG3 depletion was able to revert the EMT-like transcriptional changes observed in BT-549rDOX20 and MDA-MB-468r5-FU2000 cells. In summary, genetic and pharmacological interference with BAG3 is capable to resensitize TNBC cells to treatment, underscoring its relevance for cell death resistance and as a target to overcome therapy resistance of breast cancer.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Reguladoras de Apoptose/genética , Apoptose/genética , Autofagia/genética , Resistencia a Medicamentos Antineoplásicos/genética , Neoplasias de Mama Triplo Negativas/genética , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Linhagem Celular Tumoral , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Proteínas Proto-Oncogênicas c-bcl-2/genética , Neoplasias de Mama Triplo Negativas/tratamento farmacológico
8.
Ther Apher Dial ; 22(2): 189-195, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29316258

RESUMO

Technical problems during clinical lipid apheresis interfere with fractionator performance. Therefore, a large animal model was established to characterize a new plasma fractionation membrane. Four sheep were randomized, controlled, and crossover subjected to double ofiltration lipoprotein apheresis with three specimens of FractioPESR having slightly different HDL sieving coefficients (SK ) (FPESa, 0.30, FPESb, 0.26, and FPESc, 0.22) versus a control fractionator (EVAL). SK and reduction ratios were determined for LDL, HDL, fibrinogen, IgG, and albumin. Compared to EVAL (0.42 ± 0.04 to 0.74 ± 0.08) and FPESa (0.36 ± 0.06 to 0.64 ± 0.04), SK for HDL were lower (P < 0.05) with FPESc (0.30 ± 0.04 to 0.49 ± 0.10). Fibrinogen SK were higher (P < 0.05) with EVAL (0.02 ± 0.01 to 0.40 ± 0.08) compared to FPESb (0.05 ± 0.02 to 0.26 ± 0.34) and FPESc (0.01 ± 0.01 to 0.21 ± 0.16). No further differences were determined. The animal model distinguished between minor differences in fractionation membrane permeability, demonstrating equivalent sieving of FPESa and EVAL and slightly inferior permeability of FPESb and FPESc.


Assuntos
Remoção de Componentes Sanguíneos/instrumentação , Remoção de Componentes Sanguíneos/métodos , Filtração/instrumentação , Filtração/métodos , Lipoproteínas , Animais , Feminino , Modelos Animais , Polímeros , Povidona , Ovinos , Sulfonas
9.
Cell ; 167(6): 1636-1649.e13, 2016 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-27912065

RESUMO

Conventional ubiquitination involves the ATP-dependent formation of amide bonds between the ubiquitin C terminus and primary amines in substrate proteins. Recently, SdeA, an effector protein of pathogenic Legionella pneumophila, was shown to mediate NAD-dependent and ATP-independent ubiquitin transfer to host proteins. Here, we identify a phosphodiesterase domain in SdeA that efficiently catalyzes phosphoribosylation of ubiquitin on a specific arginine via an ADP-ribose-ubiquitin intermediate. SdeA also catalyzes a chemically and structurally distinct type of substrate ubiquitination by conjugating phosphoribosylated ubiquitin to serine residues of protein substrates via a phosphodiester bond. Furthermore, phosphoribosylation of ubiquitin prevents activation of E1 and E2 enzymes of the conventional ubiquitination cascade, thereby impairing numerous cellular processes including mitophagy, TNF signaling, and proteasomal degradation. We propose that phosphoribosylation of ubiquitin potently modulates ubiquitin functions in mammalian cells.


Assuntos
Legionella pneumophila/fisiologia , Doença dos Legionários/microbiologia , Ubiquitinação , Trifosfato de Adenosina/análogos & derivados , Trifosfato de Adenosina/metabolismo , Proteínas de Bactérias , Enzimas Reparadoras do DNA , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Doença dos Legionários/metabolismo , Proteínas de Membrana/metabolismo , Diester Fosfórico Hidrolases/metabolismo , Monoéster Fosfórico Hidrolases , Complexo de Endopeptidases do Proteassoma/metabolismo , Pirofosfatases/metabolismo , Saccharomyces cerevisiae , Serina/metabolismo , Ubiquitina/metabolismo
10.
PLoS Genet ; 7(10): e1002325, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22022284

RESUMO

We report an early onset spastic ataxia-neuropathy syndrome in two brothers of a consanguineous family characterized clinically by lower extremity spasticity, peripheral neuropathy, ptosis, oculomotor apraxia, dystonia, cerebellar atrophy, and progressive myoclonic epilepsy. Whole-exome sequencing identified a homozygous missense mutation (c.1847G>A; p.Y616C) in AFG3L2, encoding a subunit of an m-AAA protease. m-AAA proteases reside in the mitochondrial inner membrane and are responsible for removal of damaged or misfolded proteins and proteolytic activation of essential mitochondrial proteins. AFG3L2 forms either a homo-oligomeric isoenzyme or a hetero-oligomeric complex with paraplegin, a homologous protein mutated in hereditary spastic paraplegia type 7 (SPG7). Heterozygous loss-of-function mutations in AFG3L2 cause autosomal-dominant spinocerebellar ataxia type 28 (SCA28), a disorder whose phenotype is strikingly different from that of our patients. As defined in yeast complementation assays, the AFG3L2(Y616C) gene product is a hypomorphic variant that exhibited oligomerization defects in yeast as well as in patient fibroblasts. Specifically, the formation of AFG3L2(Y616C) complexes was impaired, both with itself and to a greater extent with paraplegin. This produced an early-onset clinical syndrome that combines the severe phenotypes of SPG7 and SCA28, in additional to other "mitochondrial" features such as oculomotor apraxia, extrapyramidal dysfunction, and myoclonic epilepsy. These findings expand the phenotype associated with AFG3L2 mutations and suggest that AFG3L2-related disease should be considered in the differential diagnosis of spastic ataxias.


Assuntos
Proteases Dependentes de ATP/genética , Encéfalo/anormalidades , Metaloendopeptidases/genética , Mitocôndrias/enzimologia , Paraplegia Espástica Hereditária/genética , Degenerações Espinocerebelares/genética , ATPases Associadas a Diversas Atividades Celulares , Adolescente , Sequência de Aminoácidos , Animais , Encéfalo/patologia , Criança , Diagnóstico Diferencial , Exoma/genética , Genótipo , Células HeLa , Homozigoto , Humanos , Masculino , Metaloendopeptidases/metabolismo , Camundongos , Dados de Sequência Molecular , Mutação de Sentido Incorreto , Paraplegia , Dobramento de Proteína , Irmãos , Paraplegia Espástica Hereditária/patologia , Ataxias Espinocerebelares/congênito , Degenerações Espinocerebelares/patologia , Leveduras/genética
11.
EMBO J ; 30(13): 2545-56, 2011 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-21610694

RESUMO

m-AAA proteases exert dual functions in the mitochondrial inner membrane: they mediate the processing of specific regulatory proteins and ensure protein quality control degrading misfolded polypeptides to peptides. Loss of these activities leads to neuronal cell death in several neurodegenerative disorders. However, it is unclear how the m-AAA protease chooses between specific processing and complete degradation. A central and conserved function of the m-AAA protease is the processing of the ribosomal subunit MrpL32, which regulates ribosome biogenesis and the formation of respiratory complexes. Here, we demonstrate that the formation of a tightly folded domain harbouring a conserved CxxC-X(9)-CxxC sequence motif halts degradation initiated from the N-terminus and triggers the release of mature MrpL32. Oxidative stress impairs folding of MrpL32, resulting in its degradation by the m-AAA protease and decreased mitochondrial translation. Surprisingly, MrpL32 folding depends on its mitochondrial targeting sequence. Presequence-assisted folding of MrpL32 requires the complete import of the MrpL32 precursor before maturation occurs and therefore explains the need for post-translocational processing by the m-AAA protease rather than co-translocational cleavage by the general mitochondrial processing peptidase.


Assuntos
Sequência de Aminoácidos/fisiologia , Metaloendopeptidases/metabolismo , Mitocôndrias/metabolismo , Dobramento de Proteína , Proteínas Ribossômicas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Domínio Catalítico/genética , Deinococcus/enzimologia , Deinococcus/metabolismo , Metaloendopeptidases/genética , Metaloendopeptidases/fisiologia , Mitocôndrias/fisiologia , Modelos Biológicos , Modelos Moleculares , Dados de Sequência Molecular , Organismos Geneticamente Modificados , Ligação Proteica/fisiologia , Processamento de Proteína Pós-Traducional , Transporte Proteico/genética , Transporte Proteico/fisiologia , Proteínas Ribossômicas/química , Proteínas Ribossômicas/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Análise de Sequência de Proteína , Homologia de Sequência de Aminoácidos
12.
Nat Genet ; 42(4): 313-21, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20208537

RESUMO

Autosomal dominant spinocerebellar ataxias (SCAs) are genetically heterogeneous neurological disorders characterized by cerebellar dysfunction mostly due to Purkinje cell degeneration. Here we show that AFG3L2 mutations cause SCA type 28. Along with paraplegin, which causes recessive spastic paraplegia, AFG3L2 is a component of the conserved m-AAA metalloprotease complex involved in the maintenance of the mitochondrial proteome. We identified heterozygous missense mutations in five unrelated SCA families and found that AFG3L2 is highly and selectively expressed in human cerebellar Purkinje cells. m-AAA-deficient yeast cells expressing human mutated AFG3L2 homocomplex show respiratory deficiency, proteolytic impairment and deficiency of respiratory chain complex IV. Structure homology modeling indicates that the mutations may affect AFG3L2 substrate handling. This work identifies AFG3L2 as a novel cause of dominant neurodegenerative disease and indicates a previously unknown role for this component of the mitochondrial protein quality control machinery in protecting the human cerebellum against neurodegeneration.


Assuntos
Adenosina Trifosfatases/genética , Mutação de Sentido Incorreto , Degenerações Espinocerebelares/genética , Proteases Dependentes de ATP , ATPases Associadas a Diversas Atividades Celulares , Adenosina Trifosfatases/metabolismo , Sequência de Bases , Respiração Celular , Cerebelo/metabolismo , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Teste de Complementação Genética , Humanos , Dados de Sequência Molecular , Células de Purkinje/metabolismo , Saccharomyces cerevisiae/genética
13.
Hum Mutat ; 31(5): 617-21, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20186691

RESUMO

An autosomal recessive form of hereditary spastic paraplegia (AR-HSP) is primarily caused by mutations in the SPG7 gene, which codes for paraplegin, a subunit of the hetero-oligomeric m-AAA protease in mitochondria. In the current study, sequencing of the SPG7 gene in the genomic DNA of 25 unrelated HSP individuals/families led to the identification of two HSP patients with compound heterozygous mutations (p.G349S/p.W583C and p.A510V/p.N739KfsX741) in the coding sequence of the SPG7 gene. We used a yeast complementation assay to evaluate the functional consequence of novel SPG7 sequence variants detected in the HSP patients. We assessed the proteolytic activity of hetero-oligomeric m-AAA proteases composed of paraplegin variant(s) and proteolytically inactive forms of AFG3L2 (AFG3L2(E575Q) or AFG3L2(K354A)) upon expression in m-AAA protease-deficient yeast cells. We demonstrate that the newly identified paraplegin variants perturb the proteolytic function of hetero-oligomeric m-AAA protease. Moreover, commonly occurring silent polymorphisms such as p.T503A and p.R688Q could be distinguished from mutations (p.G349S, p.W583C, p.A510V, and p.N739KfsX741) in our HSP cohort. The yeast complementation assay thus can serve as a reliable system to distinguish a pathogenic mutation from a silent polymorphism for any novel SPG7 sequence variant, which will facilitate the interpretation of genetic data for SPG7.


Assuntos
Análise Mutacional de DNA/métodos , Metaloendopeptidases/genética , Saccharomyces cerevisiae/metabolismo , Paraplegia Espástica Hereditária/genética , Proteases Dependentes de ATP , ATPases Associadas a Diversas Atividades Celulares , Adenosina Trifosfatases/genética , Humanos , Polimorfismo Genético , Saccharomyces cerevisiae/genética , Paraplegia Espástica Hereditária/diagnóstico , Paraplegia Espástica Hereditária/metabolismo
14.
Mol Biol Cell ; 20(19): 4216-24, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19656850

RESUMO

m-AAA proteases are ATP-dependent proteolytic machines in the inner membrane of mitochondria which are crucial for the maintenance of mitochondrial activities. Conserved nuclear-encoded subunits, termed paraplegin, Afg3l1, and Afg3l2, form various isoenzymes differing in their subunit composition in mammalian mitochondria. Mutations in different m-AAA protease subunits are associated with distinct neuronal disorders in human. However, the biogenesis of m-AAA protease complexes or of individual subunits is only poorly understood. Here, we have examined the processing of nuclear-encoded m-AAA protease subunits upon import into mitochondria and demonstrate autocatalytic processing of Afg3l1 and Afg3l2. The mitochondrial processing peptidase MPP generates an intermediate form of Afg3l2 that is matured autocatalytically. Afg3l1 or Afg3l2 are also required for maturation of newly imported paraplegin subunits after their cleavage by MPP. Our results establish that mammalian m-AAA proteases can act as processing enzymes in vivo and reveal overlapping activities of Afg3l1 and Afg3l2. These findings might be of relevance for the pathogenesis of neurodegenerative disorders associated with mutations in different m-AAA protease subunits.


Assuntos
Metaloendopeptidases/metabolismo , Mitocôndrias/enzimologia , Proteínas Mitocondriais/metabolismo , Proteases Dependentes de ATP , ATPases Associadas a Diversas Atividades Celulares , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Animais , Catálise , Núcleo Celular/enzimologia , Células Cultivadas , Eletroforese em Gel de Poliacrilamida , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Immunoblotting , Masculino , Metaloendopeptidases/genética , Camundongos , Camundongos Knockout , Proteínas Mitocondriais/genética , Mutação , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Transporte Proteico , Transfecção , Peptidase de Processamento Mitocondrial
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA