Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
PLoS Comput Biol ; 12(2): e1004744, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26844769

ABSTRACT

MicroRNAs are important regulators of gene expression, acting primarily by binding to sequence-specific locations on already transcribed messenger RNAs (mRNA) and typically down-regulating their stability or translation. Recent studies indicate that microRNAs may also play a role in up-regulating mRNA transcription levels, although a definitive mechanism has not been established. Double-helical DNA is capable of forming triple-helical structures through Hoogsteen and reverse Hoogsteen interactions in the major groove of the duplex, and we show physical evidence (i.e., NMR, FRET, SPR) that purine or pyrimidine-rich microRNAs of appropriate length and sequence form triple-helical structures with purine-rich sequences of duplex DNA, and identify microRNA sequences that favor triplex formation. We developed an algorithm (Trident) to search genome-wide for potential triplex-forming sites and show that several mammalian and non-mammalian genomes are enriched for strong microRNA triplex binding sites. We show that those genes containing sequences favoring microRNA triplex formation are markedly enriched (3.3 fold, p<2.2 × 10(-16)) for genes whose expression is positively correlated with expression of microRNAs targeting triplex binding sequences. This work has thus revealed a new mechanism by which microRNAs could interact with gene promoter regions to modify gene transcription.


Subject(s)
DNA/genetics , Gene Expression Regulation/genetics , MicroRNAs/genetics , Algorithms , Base Composition/genetics , Base Sequence , Binding Sites , Computational Biology , DNA/chemistry , Humans , Leukemia/genetics
2.
Cell Mol Life Sci ; 71(11): 2017-32, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24337808

ABSTRACT

The ubiquitous distribution of lysosomes and their heterogeneous protein composition reflects the versatility of these organelles in maintaining cell homeostasis and their importance in tissue differentiation and remodeling. In lysosomes, the degradation of complex, macromolecular substrates requires the synergistic action of multiple hydrolases that usually work in a stepwise fashion. This catalytic machinery explains the existence of lysosomal enzyme complexes that can be dynamically assembled and disassembled to efficiently and quickly adapt to the pool of substrates to be processed or degraded, adding extra tiers to the regulation of the individual protein components. An example of such a complex is the one composed of three hydrolases that are ubiquitously but differentially expressed: the serine carboxypeptidase, protective protein/cathepsin A (PPCA), the sialidase, neuraminidase-1 (NEU1), and the glycosidase ß-galactosidase (ß-GAL). Next to this 'core' complex, the existence of sub-complexes, which may contain additional components, and function at the cell surface or extracellularly, suggests as yet unexplored functions of these enzymes. Here we review how studies of basic biological processes in the mouse models of three lysosomal storage disorders, galactosialidosis, sialidosis, and GM1-gangliosidosis, revealed new and unexpected roles for the three respective affected enzymes, Ppca, Neu1, and ß-Gal, that go beyond their canonical degradative activities. These findings have broadened our perspective on their functions and may pave the way for the development of new therapies for these lysosomal storage disorders.


Subject(s)
Cathepsin A/metabolism , Gangliosidosis, GM1/enzymology , Lysosomal Storage Diseases/enzymology , Lysosomes/enzymology , Mucolipidoses/enzymology , Neuraminidase/metabolism , beta-Galactosidase/metabolism , Animals , Cathepsin A/genetics , Disease Models, Animal , Gangliosidosis, GM1/genetics , Gangliosidosis, GM1/pathology , Gene Expression Regulation , Humans , Lysosomal Storage Diseases/genetics , Lysosomal Storage Diseases/pathology , Lysosomes/genetics , Lysosomes/pathology , Mice , Mice, Knockout , Mucolipidoses/genetics , Mucolipidoses/pathology , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Neuraminidase/genetics , Signal Transduction , beta-Galactosidase/genetics
3.
Biochim Biophys Acta ; 1832(10): 1784-92, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23770387

ABSTRACT

The lysosomal storage disease sialidosis is caused by a primary deficiency of the sialidase N-acetyl-α-neuraminidase-1 (NEU1). Patients with type I sialidosis develop an attenuated, non-neuropathic form of the disease also named cherry red spot myoclonus syndrome, with symptoms arising during juvenile/ adult age. NEU1 requires binding to its chaperone, protective protein/cathepsin A (PPCA), for lysosomal compartmentalization, stability and catalytic activation. We have generated a new mouse model of type I sialidosis that ubiquitously expresses a NEU1 variant carrying a V54M amino acid substitution identified in an adult patient with type I sialidosis. Mutant mice developed signs of lysosomal disease after 1year of age, predominantly in the kidney, albeit low residual NEU1 activity was detected in most organs and cell types. We demonstrate that the activity of the mutant enzyme could be effectively increased in all systemic tissues by chaperone-mediated gene therapy with a liver-tropic recombinant AAV2/8 vector expressing PPCA. This resulted in clear amelioration of the disease phenotype. These results suggest that at least some of the NEU1 mutations associated with type I sialidosis may respond to PPCA-chaperone-mediated gene therapy.


Subject(s)
Dependovirus/genetics , Genetic Therapy , Molecular Chaperones/metabolism , Mucolipidoses/therapy , Recombination, Genetic , Animals , Disease Models, Animal , Mice , Mice, Knockout
4.
Mol Ther ; 20(2): 267-74, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22008912

ABSTRACT

Galactosialidosis (GS) is a lysosomal storage disease linked to deficiency of the protective protein/cathepsin A (PPCA). Similarly to GS patients, Ppca-null mice develop a systemic disease of the reticuloendothelial system, affecting most visceral organs and the nervous system. Symptoms include severe nephropathy, visceromegaly, infertility, progressive ataxia, and shortened life span. Here, we have conducted a preclinical, dose-finding study on a large cohort of GS mice injected intravenously at 1 month of age with increasing doses of a GMP-grade rAAV2/8 vector, expressing PPCA under the control of a liver-specific promoter. Treated mice, monitored for 16 weeks post-treatment, had normal physical appearance and behavior without discernable side effects. Despite the restricted expression of the transgene in the liver, immunohistochemical and biochemical analyses of other systemic organs, serum, and urine showed a dose-dependent, widespread correction of the disease phenotype, suggestive of a protein-mediated mechanism of cross-correction. A notable finding was that rAAV-treated GS mice showed high expression of PPCA in the reproductive organs, which resulted in reversal of their infertility. Together these results support the use of this rAAV-PPCA vector as a viable and safe method of gene delivery for the treatment of systemic disease in non-neuropathic GS patients.


Subject(s)
Dependovirus/physiology , Genetic Therapy , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Liver/metabolism , Lysosomal Storage Diseases/therapy , Viral Tropism , Animals , Cathepsin A/genetics , Cathepsin A/metabolism , Dependovirus/genetics , Disease Models, Animal , Enzyme Activation/genetics , Female , Fertility/genetics , Gene Expression , Genetic Vectors/pharmacokinetics , Humans , Kidney/metabolism , Kidney/pathology , Liver/pathology , Lysosomal Storage Diseases/genetics , Lysosomal Storage Diseases/metabolism , Male , Mice , Mice, Knockout , Neuraminidase/metabolism , Oligosaccharides/urine , Organ Size , Spleen/metabolism , Spleen/pathology
5.
Life Sci Alliance ; 6(3)2023 03.
Article in English | MEDLINE | ID: mdl-36596605

ABSTRACT

PACSIN2 variants are associated with gastrointestinal effects of thiopurines and thiopurine methyltransferase activity through an uncharacterized mechanism that is postulated to involve autophagy. This study aims to clarify the role of PACSIN2 in autophagy and in thiopurine cytotoxicity in leukemic and intestinal models. Higher autophagy and lower PACSIN2 levels were observed in inflamed compared with non-inflamed colon biopsies of inflammatory bowel disease pediatric patients at diagnosis. PACSIN2 was identified as an inhibitor of autophagy, putatively through inhibition of autophagosome formation by a protein-protein interaction with LC3-II, mediated by a LIR motif. Moreover, PACSIN2 resulted a modulator of mercaptopurine-induced cytotoxicity in intestinal cells, suggesting that PACSIN2-regulated autophagy levels might influence thiopurine sensitivity. However, PACSIN2 modulates cellular thiopurine methyltransferase activity via mechanisms distinct from its modulation of autophagy.


Subject(s)
Inflammatory Bowel Diseases , Mercaptopurine , Humans , Child , Mercaptopurine/pharmacology , Inflammatory Bowel Diseases/drug therapy , Intestines , Autophagy , Adaptor Proteins, Signal Transducing/genetics
6.
Sci Rep ; 12(1): 7820, 2022 05 12.
Article in English | MEDLINE | ID: mdl-35551201

ABSTRACT

Ozz, a member of the SOCS-box family of proteins, is the substrate-binding component of CRL5Ozz, a muscle-specific Cullin-RING ubiquitin ligase complex composed of Elongin B/C, Cullin 5 and Rbx1. CRL5Ozz targets for proteasomal degradation selected pools of substrates, including sarcolemma-associated ß-catenin, sarcomeric MyHCemb and Alix/PDCD6IP, which all interact with the actin cytoskeleton. Ubiquitination and degradation of these substrates are required for the remodeling of the contractile sarcomeric apparatus. However, how CRL5Ozz assembles into an active E3 complex and interacts with its substrates remain unexplored. Here, we applied a baculovirus-based expression system to produce large quantities of two subcomplexes, Ozz-EloBC and Cul5-Rbx1. We show that these subcomplexes mixed in a 1:1 ratio reconstitutes a five-components CRL5Ozz monomer and dimer, but that the reconstituted complex interacts with its substrates only as monomer. The in vitro assembled CRL5Ozz complex maintains the capacity to polyubiquitinate each of its substrates, indicating that the protein production method used in these studies is well-suited to generate large amounts of a functional CRL5Ozz. Our findings highlight a mode of assembly of the CRL5Ozz that differs in presence or absence of its cognate substrates and grant further structural studies.


Subject(s)
Cullin Proteins , Ubiquitin-Protein Ligases , Cullin Proteins/genetics , Protein Binding , Sarcomeres/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Ubiquitins/metabolism
7.
J Inflamm Res ; 15: 1183-1194, 2022.
Article in English | MEDLINE | ID: mdl-35221708

ABSTRACT

BACKGROUND: Following our 2015 elucidation of the CASP1/NALP3 inflammasome mechanism of glucocorticoid (GC)-resistance in pediatric acute lymphoblastic leukemia (ALL) patients, we engineered a cell-based CASP1/NALP3 reporter system suitable for high-throughput screening (HTS) of small molecule libraries, with the purpose of identifying compounds capable of inhibiting the CASP1/NALP3 inflammasome and synergizing with GC drugs for the treatment of GC-resistant ALL patients and various autoinflammatory diseases. METHODS: A Dox-controlled system was utilized to induce the expression of the ASC transgene in HEK293 cells while simultaneously overexpressing NLRP3 and CASP1. ASC/CASP1/NALP3 inflammasome complex formation was confirmed by co-immunoprecipitation (co-IP) experiments. Next, a LV fluorescence-based biosensor (CASPorter) was transduced in the HEK293-iASC-NLRP3/CASP1 cell line to monitor the real-time activation of CASP1/NALP3 inflammasome in live cells. The applicability and effectiveness of the CASPorter cell line were tested by co-treatment with Dox and four known CASP1/NLRP3 inhibitors (MCC950, Glyburide, VX-765 and VRT-043198). Inflammasome activation and inhibitions were assessed by Western blotting, fluorescence microscopy and flow cytometry (FC) methods. RESULTS: Dox treatment significantly induced ASC expression and increased levels of cleaved and catalytically active CASP1, co-IPs further demonstrated that CASP1 was pulled-down with NLRP3 in HEK293-iASC-NLRP3/CASP1 cells after induction of ASC by Dox treatment. In HEK293-iASC-NLRP3/CASP1-CASPorter cell system, cleavage of the CASP1 consensus site (YVAD) in the CASPorter protein after Dox treatment causing excitation/emission of green fluorescence and the 71% GFP+ cell population increase quantified by FC (78.1% vs 6.90%). Dox-induced activation of the NLRP3 inflammasome was dose-dependently inhibited by Dox co-treatment with four known CASP1/NLRP3 inhibitors. CONCLUSION: We have established a cell-based CASP1/NLRP3 inflammasome model, utilizing a fluorescence biosensor as readout for qualitatively observing and quantitatively determining the activation of caspase 1 and NLRP3 inflammasomes in living cells and easily define the inhibitory effect of inhibitors with high efficacy.

8.
Cancers (Basel) ; 14(14)2022 Jul 19.
Article in English | MEDLINE | ID: mdl-35884569

ABSTRACT

Vincristine (VCR) is an important component of curative chemotherapy for many childhood cancers. Its main side effect is VCR-induced peripheral neuropathy (VIPN), a dose limiting toxicity. Some children are more susceptible to VIPN, which is at least partially dependent on genetic factors and pharmacokinetics (PK). In this study, we identify and replicate genetic variants associated with VCR PK and VIPN. Patient samples from a randomized clinical trial studying the effect of administration duration of VCR on VIPN in 90 patients were used. PK sampling was conducted on between one and five occasions at multiple time points. A linear two-compartment model with first-order elimination was used, and targeted next-generation DNA sequencing was performed. Genotype-trait associations were analyzed using mixed-effect models or logistic regression analysis for repeated measures, or Poisson regression analysis in which the highest VIPN score per patient was included. Nine single-nucleotide polymorphisms (SNPs) in seven genes (NDRG1, GARS, FIG4, FGD4, SEPTIN9, CEP72, and ETAA1) were associated with VIPN. Furthermore, three SNPs in three genes (MTNR1B, RAB7A and SNU13) were associated with PK of VCR. In conclusion, PK of VCR and VIPN are influenced by SNPs; upfront identification of those that lead to an altered susceptibility to VIPN or VCR exposure could help individualize VCR treatment.

9.
Blood Adv ; 6(11): 3386-3397, 2022 06 14.
Article in English | MEDLINE | ID: mdl-35671062

ABSTRACT

Understanding the genomic and epigenetic mechanisms of drug resistance in pediatric acute lymphoblastic leukemia (ALL) is critical for further improvements in treatment outcomes. The role of transcriptomic response in conferring resistance to l-asparaginase (LASP) is poorly understood beyond asparagine synthetase (ASNS). We defined reproducible LASP response genes in LASP-resistant and LASP-sensitive ALL cell lines as well as primary leukemia samples from newly diagnosed patients. Defining target genes of the amino acid stress response-related transcription factor activating transcription factor 4 (ATF4) in ALL cell lines using chromatin immunoprecipitation sequencing (ChIP-seq) revealed 45% of genes that changed expression after LASP treatment were direct targets of the ATF4 transcription factor, and 34% of these genes harbored LASP-responsive ATF4 promoter binding events. SLC7A11 was found to be a response gene in cell lines and patient samples as well as a direct target of ATF4. SLC7A11 was also one of only 2.4% of LASP response genes with basal level gene expression that also correlated with LASP ex vivo resistance in primary leukemia cells. Experiments using chemical inhibition of SLC7A11 with sulfasalazine, gene overexpression, and partial gene knockout recapitulated LASP resistance or sensitivity in ALL cell lines. These findings show the importance of assessing changes in gene expression following treatment with an antileukemic agent for its association with drug resistance and highlight that many response genes may not differ in their basal expression in drug-resistant leukemia cells.


Subject(s)
Aspartate-Ammonia Ligase , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Activating Transcription Factor 4/genetics , Amino Acids/therapeutic use , Asparaginase/pharmacology , Asparaginase/therapeutic use , Aspartate-Ammonia Ligase/genetics , Aspartate-Ammonia Ligase/metabolism , Cell Line, Tumor , Child , Humans , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics
10.
Biochim Biophys Acta ; 1802(2): 259-68, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19857571

ABSTRACT

The neuraminidase-1 (Neu1) knockout mouse model is a phenocopy of the lysosomal storage disease (LSD) sialidosis, characterized by multisystemic and neuropathic symptoms, including hearing loss. We have characterized the auditory defects in Neu1(-/-) mice and found that hearing loss involves both conductive and sensorineural components. Auditory brainstem response (ABR) thresholds were significantly elevated in Neu1(-/-) mice at P21 (48-55 dB), and hearing loss appeared progressive (53-66 dB at P60). At these ages Neu1(-/-) mice accumulated cerumen in the external ear canal and had a thickened mucosa and inflammation in the middle ear. In cochleae of adult wild-type mice, Neu1 was expressed in several cell types in the stria vascularis, the organ of Corti, and spiral ganglion. Progressive morphological abnormalities such as extensive vacuolization were detected in the Neu1(-/-) cochleae as early as P9. These early morphologic changes in Neu1(-/-) cochleae were associated with oversialylation of several lysosomal associated membrane proteins (Lamps) in the stria vascularis. A marked increase in the expression and apical localization of Lamp-1 in marginal cells of the stria vascularis predicts exacerbation of lysosomal exocytosis into the endolymph. Consequently, the endolymphatic potential in Neu1(-/-) mice was reduced by approximately 20 mV at ages P31-P44, which would cause dysfunction of transduction in sensory hair cells. This study suggests a molecular mechanism that contributes to hearing loss in sialidosis and identifies potential therapeutic targets.


Subject(s)
Cochlea/physiopathology , Hearing Loss/genetics , Neuraminidase/deficiency , Neuraminidase/genetics , Animals , Cochlea/pathology , Disease Models, Animal , Ear, Middle/pathology , Hearing Loss/enzymology , Lysosomal Storage Diseases/enzymology , Lysosomal Storage Diseases/genetics , Mice , Mice, Knockout , Microscopy, Electron, Scanning , Vacuoles/pathology , Vacuoles/ultrastructure
11.
Biochim Biophys Acta ; 1802(7-8): 659-72, 2010.
Article in English | MEDLINE | ID: mdl-20388541

ABSTRACT

Neuraminidase 1 (NEU1) regulates the catabolism of sialoglycoconjugates in lysosomes. Congenital NEU1 deficiency in children is the basis of sialidosis, a severe neurosomatic disorder in which patients experience a broad spectrum of clinical manifestations varying in the age of onset and severity. Osteoskeletal deformities and muscle hypotonia have been described in patients with sialidosis. Here we present the first comprehensive analysis of the skeletal muscle pathology associated with loss of Neu1 function in mice. In this animal model, skeletal muscles showed an expansion of the epimysial and perimysial spaces, associated with proliferation of fibroblast-like cells and abnormal deposition of collagens. Muscle fibers located adjacent to the expanded connective tissue underwent extensive invagination of their sarcolemma, which resulted in the infiltration of the fibers by fibroblast-like cells and extracellular matrix, and in their progressive cytosolic fragmentation. Both the expanded connective tissue and the juxtaposed infiltrated muscle fibers were strongly positive for lysosomal markers and displayed increased proteolytic activity of lysosomal cathepsins and metalloproteinases. These combined features could lead to abnormal remodeling of the extracellular matrix that could be responsible for sarcolemmal invagination and progressive muscle fiber degeneration, ultimately resulting in an overt atrophic phenotype. This unique pattern of muscle damage, which has never been described in any myopathy, might explain the neuromuscular manifestations reported in patients with the type II severe form of sialidosis. More broadly, these findings point to a potential role of NEU1 in cell proliferation and extracellular matrix remodeling.


Subject(s)
Connective Tissue/physiopathology , Muscle Fibers, Skeletal/pathology , Muscular Atrophy/etiology , Muscular Atrophy/pathology , Neuraminidase/genetics , Animals , Apoptosis/genetics , Apoptosis/physiology , Cell Movement/genetics , Cell Proliferation , Connective Tissue/pathology , Extracellular Matrix/pathology , Extracellular Matrix/physiology , Fibroblasts/pathology , Mice , Mice, Knockout , Muscular Atrophy/genetics , Muscular Atrophy/physiopathology , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/pathology , Necrosis/genetics , Necrosis/pathology , Neuraminidase/deficiency , Neuraminidase/physiology , Sarcolemma/pathology
12.
Clin Transl Sci ; 14(3): 964-975, 2021 05.
Article in English | MEDLINE | ID: mdl-33382913

ABSTRACT

To assess whether NLRP3 gene promoter methylation was able to discriminate glucocorticoid (GC)-resistant from GC-sensitive idiopathic nephrotic syndrome (INS), patients with minimal change disease (MCD) or focal segmental glomerulosclerosis (FSGS), we measured the methylation level of NLRP3 promoter in DNA from peripheral blood cells of 10 adult patients with GC-resistant FSGS already in hemodialysis and 18 patients with GC-sensitive INS (13 MCD/5 FSGS) and in 21 pediatric patients with INS with MCD/FSGS before starting any treatment. Association of NLRP3 inflammasome with GC resistance was recapitulated in vitro in monocytic cell lines (THP-1 and U937). In both adults and pediatric patients, NLRP3 promoter methylation was significantly reduced in GC-resistant compared with GC-sensitive patients. Indeed, NLRP3 methylation distinguished GC-resistant and GC-sensitive patients (area under the receiver operating characteristic curve [AUROC] 86.7% in adults, p = 0.00019, and 73.5% in children, p = 0.00097). NLRP3 knock-down augmented sensitivity to GCs in THP-1 cells, whereas NLRP3 inflammasome activation lowered GC receptor concentration, increasing GC resistance in U937 cells. Our results uncovered a new biological mechanism by which patients with INS may acquire GC resistance, that could be used in future as a novel noninvasive diagnostic tool. Study Highlights WHAT IS THE CURRENT KNOWLEDGE ON THE TOPIC? ☑ Approximately 80% of patients with idiopathic nephrotic syndrome (INS) respond to glucocorticoids, with the remaining 20% being steroid-resistant. WHAT QUESTION DID THIS STUDY ADDRESS? ☑ Whether NLRP3 gene promoter methylation was able to discriminate glucocorticoid-resistant from glucocorticoid (GC)-sensitive INS. WHAT DOES THIS STUDY ADD TO OUR KNOWLEDGE? ☑ In both adults and children, NLRP3 promoter methylation was significantly reduced in leukocytes of patients with GC-resistant compared with GC-sensitive INS. NLRP3 inflammasome activation lowered GC receptor concentration and augmented GC resistance, whereas NLRP3 knockdown increased sensitivity to GCs in cell lines representative of monocytes (U937 and THP1). HOW MIGHT THIS CHANGE CLINICAL PHARMACOLOGY OR TRANSLATIONAL SCIENCE? ☑ Our findings uncovered a new biological mechanism whereby patients with INS may develop resistance to GCs that could be used in the future as a novel noninvasive diagnostic tool.


Subject(s)
DNA Methylation , Drug Resistance/genetics , Glucocorticoids/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Nephrotic Syndrome/drug therapy , Adolescent , Adult , Aged , Case-Control Studies , Child , Child, Preschool , Female , Follow-Up Studies , Gene Knockdown Techniques , Glucocorticoids/therapeutic use , Healthy Volunteers , Humans , Inflammasomes/genetics , Inflammasomes/metabolism , Male , Middle Aged , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nephrotic Syndrome/genetics , Promoter Regions, Genetic/genetics , ROC Curve , THP-1 Cells
13.
Clin Transl Sci ; 14(4): 1490-1504, 2021 07.
Article in English | MEDLINE | ID: mdl-33742760

ABSTRACT

Vincristine (VCR) is one of the most widely prescribed medications for treating solid tumors and acute lymphoblastic leukemia (ALL) in children and adults. However, its major dose-limiting toxicity is peripheral neuropathy that can disrupt curative therapy. Peripheral neuropathy can also persist into adulthood, compromising quality of life of childhood cancer survivors. Reducing VCR-induced neurotoxicity without compromising its anticancer effects would be ideal. Here, we show that low expression of NHP2L1 is associated with increased sensitivity of primary leukemia cells to VCR, and that concomitant administration of VCR with inhibitors of NHP2L1 increases VCR cytotoxicity in leukemia cells, prolongs survival of ALL xenograft mice, but decreases VCR effects on human-induced pluripotent stem cell-derived neurons and mitigates neurotoxicity in mice. These findings offer a strategy for increasing VCR's antileukemic effects while reducing peripheral neuropathy in patients treated with this widely prescribed medication.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Peripheral Nervous System Diseases/prevention & control , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Ribonucleoproteins, Small Nuclear/antagonists & inhibitors , Vincristine/adverse effects , Adolescent , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cells, Cultured , Child , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Leukemic , Gene Knockdown Techniques , Humans , Induced Pluripotent Stem Cells , Male , Mice , Neurons , Peripheral Nervous System Diseases/chemically induced , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Primary Cell Culture , Ribonucleoproteins, Small Nuclear/genetics , Ribonucleoproteins, Small Nuclear/metabolism , Vincristine/therapeutic use , Xenograft Model Antitumor Assays , Young Adult
14.
J Biol Chem ; 284(41): 28430-28441, 2009 Oct 09.
Article in English | MEDLINE | ID: mdl-19666471

ABSTRACT

Lysosomal neuraminidase-1 (NEU1) forms a multienzyme complex with beta-galactosidase and protective protein/cathepsin A (PPCA). Because of its association with PPCA, which acts as a molecular chaperone, NEU1 is transported to the lysosomal compartment, catalytically activated, and stabilized. However, the mode(s) of association between these two proteins both en route to the lysosome and in the multienzyme complex has remained elusive. Here, we have analyzed the hydrodynamic properties of PPCA, NEU1, and a complex of the two proteins and identified multiple binding sites on both proteins. One of these sites on NEU1 that is involved in binding to PPCA can also bind to other NEU1 molecules, albeit with lower affinity. Therefore, in the absence of PPCA, as in the lysosomal storage disease galactosialidosis, NEU1 self-associates into chain-like oligomers. Binding of PPCA can reverse self-association of NEU1 by causing the disassembly of NEU1-oligomers and the formation of a PPCA-NEU1 heterodimeric complex. The identification of binding sites between the two proteins allowed us to create innovative structural models of the NEU1 oligomer and the PPCA-NEU1 heterodimeric complex. The proposed mechanism of interaction between NEU1 and its accessory protein PPCA provides a rationale for the secondary deficiency of NEU1 in galactosialidosis.


Subject(s)
Cathepsin A/chemistry , Cathepsin A/metabolism , Molecular Chaperones/chemistry , Molecular Chaperones/metabolism , Neuraminidase/chemistry , Neuraminidase/metabolism , Protein Conformation , Protein Multimerization , Amino Acid Sequence , Animals , Binding Sites , Cathepsin A/genetics , Cells, Cultured , Enzyme Activation , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Knockout , Molecular Chaperones/genetics , Molecular Sequence Data , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Mutagenesis, Site-Directed , Neuraminidase/genetics
15.
Biochim Biophys Acta ; 1790(4): 275-82, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19714866

ABSTRACT

BACKGROUND: Neuraminidase-1 (NEU1) catabolizes the hydrolysis of sialic acids from sialo-glycoconjugates. NEU1 depends on its interaction with the protective protein/cathepsin A (PPCA) for lysosomal compartmentalization and catalytic activation. Murine NEU1 contains 4 N-glycosylation sites, 3 of which are conserved in the human enzyme. The expression of NEU1 gives rise to differentially glycosylated proteins. METHODS: We generated single-point mutations in mouse NEU1 at each of the 4 N-glycosylation sites. Mutant enzymes were expressed in NEU1-deficient cells in the presence and absence of PPCA. RESULTS: All 4 N-glycosylation variants were targeted to the lysosomal/endosomal compartment. All N-glycans, with the exception of the most C-terminal glycan, were important for maintaining stability or catalytic activity. The loss of catalytic activity caused by the deletion of the second N-glycan was rescued by increasing PPCA expression. Similar results were obtained with a human NEU1 N-glycosylation mutant identified in a sialidosis patient. The N-terminal N-glycan of NEU1 is indispensable for its function, whereas the C-terminal N-glycan appears to be non-essential. The omission of the second N-glycan can be compensated for by upregulating the expression of PPCA. GENERAL SIGNIFICANCE: These findings could be relevant for the design of target therapies for patients carrying specific NEU1 mutations.


Subject(s)
Cathepsin A/physiology , Neuraminidase/genetics , Animals , Cathepsin A/genetics , Glycosylation , Humans , Mice , Models, Molecular , Mucolipidoses/genetics , Mucolipidoses/therapy , Mutation, Missense , Point Mutation , Sequence Homology, Amino Acid
16.
Biochem Soc Trans ; 38(6): 1453-7, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21118106

ABSTRACT

The lysosomal system comprises a specialized network of organelles crucial for the sorting, digestion, recycling and secretion of cellular components. With their content of hydrolytic enzymes, lysosomes regulate the degradation of a multitude of substrates that reach these organelles via the biosynthetic or the endocytic route. Gene defects that affect one or more of these hydrolases lead to LSDs (lysosomal storage diseases). This underscores the apparent lack of redundancy of these enzymes and the importance of the lysosomal system in cell and tissue homoeostasis. Some of the lysosomal enzymes may form multiprotein complexes, which usually work synergistically on substrates and, in this configuration, may respond more efficiently to changes in substrate load and composition. A well-characterized lysosomal multienzyme complex is the one comprising the glycosidases ß-gal (ß-galactosidase) and NEU1 (neuramidase-1), and of the serine carboxypeptidase PPCA (protective protein/cathepsin A). Three neurodegenerative LSDs are caused by either single or combined deficiency of these lysosomal enzymes. Sialidosis (NEU1 deficiency) and galactosialidosis (combined NEU1 and ß-gal deficiency, secondary to a primary defect of PPCA) belong to the glycoprotein storage diseases, whereas GM1-gangliosidosis (ß-gal deficiency) is a glycosphingolipid storage disease. Identification of novel molecular pathways that are deregulated because of loss of enzyme activity and/or accumulation of specific metabolites in various cell types has shed light on mechanisms of disease pathogenesis and may pave the way for future development of new therapies for these LSDs.


Subject(s)
Glycoproteins/metabolism , Glycosphingolipids/metabolism , Lysosomal Storage Diseases/metabolism , Lysosomal Storage Diseases/pathology , Animals , Cathepsin A/chemistry , Cathepsin A/genetics , Cathepsin A/metabolism , Disease Models, Animal , Humans , Lysosomal Storage Diseases/genetics , Lysosomes/metabolism , Lysosomes/pathology , Models, Molecular , Multienzyme Complexes/metabolism , Neuraminidase/chemistry , Neuraminidase/genetics , Neuraminidase/metabolism , Protein Conformation , beta-Galactosidase/chemistry , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
17.
J Clin Invest ; 130(12): 6600-6615, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33164984

ABSTRACT

BACKGROUNDInterpatient differences in the accumulation of methotrexate's active polyglutamylated metabolites (MTXPGs) in leukemia cells influence its antileukemic effects.METHODSTo identify genomic and epigenomic and patient variables determining the intracellular accumulation of MTXPGs, we measured intracellular MTXPG levels in acute lymphoblastic leukemia (ALL) cells from 388 newly diagnosed patients after in vivo high-dose methotrexate (HDMTX) (1 g/m2) treatment, defined ALL subtypes, and assessed genomic and epigenomic variants influencing folate pathway genes (mRNA, miRNA, copy number alterations [CNAs], SNPs, single nucleotide variants [SNVs], CpG methylation).RESULTSWe documented greater than 100-fold differences in MTXPG levels, which influenced its antileukemic effects (P = 4 × 10-5). Three ALL subtypes had lower MTXPG levels (T cell ALL [T-ALL] and B cell ALL [B-ALL] with the TCF3-PBX1 or ETV6-RUNX1 fusions), and 2 subtypes had higher MTXPG levels (hyperdiploid and BCR-ABL like). The folate pathway genes SLC19A1, ABCC1, ABCC4, FPGS, and MTHFD1 significantly influenced intracellular MTXPG levels (P = 2.9 × 10-3 to 3.7 × 10-8). A multivariable model including the ALL subtype (P = 1.1 × 10-14), the SLC19A1/(ABCC1 + ABCC4) transporter ratio (P = 3.6 × 10-4), the MTX infusion time (P = 1.5 × 10-3), FPGS mRNA expression (P = 2.1 × 10-3), and MTX systemic clearance (P = 4.4 × 10-2) explained 42% of the variation in MTXPG accumulation (P = 1.1 × 10-38). Model simulations indicated that a longer infusion time (24 h vs. 4 h) was superior in achieving higher intracellular MTXPG levels across all subtypes if ALL.CONCLUSIONSThese findings provide insights into mechanisms underlying interpatient differences in intracellular accumulation of MTXPG in leukemia cells and its antileukemic effectsFUNDINGTHE National Cancer Institute (NCI) and the Institute of General Medical Sciences of the NIH, the Basque Government Programa Posdoctoral de Perfeccionamiento de Personal Investigador doctor, and the American Lebanese Syrian Associated Charities (ALSAC).


Subject(s)
Methotrexate/analogs & derivatives , Neoplasm Proteins , Polyglutamic Acid/analogs & derivatives , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Adolescent , Cell Line, Tumor , Child , Child, Preschool , Female , Humans , Infant , Male , Methotrexate/pharmacokinetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Pharmacogenomic Testing , Polyglutamic Acid/pharmacokinetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology
18.
Nat Cancer ; 1(3): 329-344, 2020 03.
Article in English | MEDLINE | ID: mdl-32885175

ABSTRACT

Identification of genomic and epigenomic determinants of drug resistance provides important insights for improving cancer treatment. Using agnostic genome-wide interrogation of mRNA and miRNA expression, DNA methylation, SNPs, CNAs and SNVs/Indels in primary human acute lymphoblastic leukemia cells, we identified 463 genomic features associated with glucocorticoid resistance. Gene-level aggregation identified 118 overlapping genes, 15 of which were confirmed by genome-wide CRISPR screen. Collectively, this identified 30 of 38 (79%) known glucocorticoid-resistance genes/miRNAs and all 38 known resistance pathways, while revealing 14 genes not previously associated with glucocorticoid-resistance. Single cell RNAseq and network-based transcriptomic modelling corroborated the top previously undiscovered gene, CELSR2. Manipulation of CELSR2 recapitulated glucocorticoid resistance in human leukemia cell lines and revealed a synergistic drug combination (prednisolone and venetoclax) that mitigated resistance in mouse xenograft models. These findings illustrate the power of an integrative genomic strategy for elucidating genes and pathways conferring drug resistance in cancer cells.


Subject(s)
MicroRNAs , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Animals , Drug Resistance, Neoplasm/genetics , Genomics , Glucocorticoids/pharmacology , Humans , Mice , MicroRNAs/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy
19.
Mol Cell Oncol ; 3(1): e1065947, 2016 Jan.
Article in English | MEDLINE | ID: mdl-27308575

ABSTRACT

In primary acute lymphoblastic leukemia cells exhibiting de novo resistance to glucocorticoids, we recently discovered decreased promoter methylation of caspase 1 (CASP1) and NLR family, pyrin domain containing 3 (NLRP3), which resulted in increased transcription, constitutive NALP3 (NACHT, LRR and PYD domains-containing protein 3) inflammasome activation, and caspase 1-mediated cleavage of the glucocorticoid receptor. This revealed a novel mechanism of glucocorticoid resistance that was recapitulated in model systems.

20.
FASEB J ; 18(9): 971-3, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15084520

ABSTRACT

Lysosomal storage diseases (LSDs) are monogenic disorders of metabolism caused by deficiency of hydrolytic enzymes. In several LSDs, cells of the reticuloendothelial (RE) system are the primary targets of the disease. Exogenous administration of recombinant enzymes overproduced in mammalian cells has proved effective for treating the systemic phenotype in nonneuropathic patients with LSDs. However, for the treatment of diseases with primary involvement of the RE system, the production of the therapeutic enzyme in insect cells could be an alternative and advantageous method because glycoproteins expressed in insect cells carry carbohydrates of the pauci-mannose or core-type. These recombinant enzymes are in principle already poised to be internalized by cells that express mannose receptors, including macrophages. Here, we demonstrate that three baculovirus-expressed enzymes, protective protein/cathepsin A (PPCA), neuraminidase (Neu1), and beta-glucosidase, were readily taken up and restored lysosomal function in enzyme-deficient mouse macrophages. The capacity of recombinant PPCA and Neu1 to clear the lysosomal storage in target cells was assessed in PPCA-/- mice, a model of galactosialidosis. Intravenously injected PPCA-/- mice efficiently internalized the corrective enzymes in resident macrophages of many organs. In addition, treated mice showed overall clearance of lysosomal storage in the most affected systemic organs, kidney, liver, and spleen. Our results suggest that ERT with baculovirus-expressed enzymes might be an effective treatment for nonneuropathic patients with galactosialidosis and possibly for others with LSDs that primarily involve the RE system.


Subject(s)
Baculoviridae/genetics , Cathepsin A/therapeutic use , Lysosomal Storage Diseases/drug therapy , Lysosomes/enzymology , Macrophages/enzymology , Neuraminidase/therapeutic use , beta-Glucosidase/therapeutic use , Animals , Catalysis , Cathepsin A/administration & dosage , Cathepsin A/genetics , Cathepsin A/metabolism , Cell Line , Gene Deletion , Humans , Kidney/drug effects , Kidney/pathology , Liver/chemistry , Liver/drug effects , Liver/metabolism , Liver/pathology , Lysosomal Storage Diseases/enzymology , Lysosomal Storage Diseases/pathology , Macrophages/cytology , Macrophages/drug effects , Mice , Neuraminidase/administration & dosage , Neuraminidase/genetics , Neuraminidase/metabolism , Oligosaccharides/chemistry , Oligosaccharides/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology , Spleen/drug effects , Spleen/pathology , Spodoptera/cytology , Spodoptera/virology , Vacuoles/enzymology , Vacuoles/pathology , beta-Glucosidase/administration & dosage , beta-Glucosidase/biosynthesis , beta-Glucosidase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL