Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Eng Life Sci ; 22(2): 100-114, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35140557

ABSTRACT

Mammalian cells are commonly used to produce recombinant protein therapeutics, but suffer from a high cost per mg of protein produced. There is therefore great interest in improving protein yields to reduce production cost. We present an entirely novel approach to reach this goal through direct engineering of the cellular translation machinery by introducing the R98S point mutation in the catalytically essential ribosomal protein L10 (RPL10-R98S). Our data support that RPL10-R98S enhances translation levels and fidelity and reduces proteasomal activity in lymphoid Ba/F3 and Jurkat cell models. In HEK293T cells cultured in chemically defined medium, knock-in of RPL10-R98S was associated with a 1.7- to 2.5-fold increased production of four transiently expressed recombinant proteins and 1.7-fold for one out of two stably expressed proteins. In CHO-S cells, eGFP reached a 2-fold increased expression under stable but not transient conditions, but there was no production benefit for monoclonal antibodies. The RPL10-R98S associated production gain thus depends on culture conditions, cell type, and the nature of the expressed protein. Our study demonstrates the potential for using a ribosomal protein mutation for pharmaceutical protein production gains, and further research on how various factors influence RPL10-R98S phenotypes can maximize its exploitability for the mammalian protein production industry.

2.
Nat Commun ; 10(1): 2542, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31186416

ABSTRACT

Somatic ribosomal protein mutations have recently been described in cancer, yet their impact on cellular transcription and translation remains poorly understood. Here, we integrate mRNA sequencing, ribosome footprinting, polysomal RNA sequencing and mass spectrometry datasets from a mouse lymphoid cell model to characterize the T-cell acute lymphoblastic leukemia (T-ALL) associated ribosomal RPL10 R98S mutation. Surprisingly, RPL10 R98S induces changes in protein levels primarily through transcriptional rather than translation efficiency changes. Phosphoserine phosphatase (PSPH), encoding a key serine biosynthesis enzyme, was the only gene with elevated transcription and translation leading to protein overexpression. PSPH upregulation is a general phenomenon in T-ALL patient samples, associated with elevated serine and glycine levels in xenograft mice. Reduction of PSPH expression suppresses proliferation of T-ALL cell lines and their capacity to expand in mice. We identify ribosomal mutation driven induction of serine biosynthesis and provide evidence supporting dependence of T-ALL cells on PSPH.


Subject(s)
Glycine/metabolism , Mutation , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Serine/metabolism , Animals , Cell Line , Gene Expression Profiling , Mice , Phosphoric Monoester Hydrolases , Polyribosomes/genetics , Polyribosomes/metabolism , Protein Biosynthesis , RNA, Messenger/genetics , Ribosomal Protein L10 , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Ribosomes/metabolism , Sequence Analysis, RNA
3.
Leukemia ; 33(4): 1055-1062, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30850735

ABSTRACT

Following the publication of this article, the authors noted that Dr Laura Fancello was not listed among the authors. The corrected author list is given below. Additionally, the following was not included in the author contribution statement: 'L.F. analyzed RNA sequencing data'.

4.
Leukemia ; 33(2): 319-332, 2019 02.
Article in English | MEDLINE | ID: mdl-29930300

ABSTRACT

The R98S mutation in ribosomal protein L10 (RPL10 R98S) affects 8% of pediatric T-cell acute lymphoblastic leukemia (T-ALL) cases, and was previously described to impair cellular proliferation. The current study reveals that RPL10 R98S cells accumulate reactive oxygen species which promotes mitochondrial dysfunction and reduced ATP levels, causing the proliferation defect. RPL10 R98S mutant leukemia cells can survive high oxidative stress levels via a specific increase of IRES-mediated translation of the anti-apoptotic factor B-cell lymphoma 2 (BCL-2), mediating BCL-2 protein overexpression. RPL10 R98S selective sensitivity to the clinically available Bcl-2 inhibitor Venetoclax (ABT-199) was supported by suppression of splenomegaly and the absence of human leukemia cells in the blood of T-ALL xenografted mice. These results shed new light on the oncogenic function of ribosomal mutations in cancer, provide a novel mechanism for BCL-2 upregulation in leukemia, and highlight BCL-2 inhibition as a novel therapeutic opportunity in RPL10 R98S defective T-ALL.


Subject(s)
Internal Ribosome Entry Sites , Mutation , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Biosynthesis , Proto-Oncogene Proteins c-bcl-2/metabolism , Ribosomal Proteins/genetics , Ribosomes/metabolism , Animals , Gene Expression Regulation, Leukemic , Humans , Male , Mice , Mice, Inbred NOD , Oxidative Stress/drug effects , Phosphorylation , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/genetics , Ribosomal Protein L10 , Ribosomal Proteins/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Nat Commun ; 8(1): 293, 2017 08 17.
Article in English | MEDLINE | ID: mdl-28819097

ABSTRACT

The brain cytoplasmic (BC1) RNA is a non-coding RNA (ncRNA) involved in neuronal translational control. Absence of BC1 is associated with altered glutamatergic transmission and maladaptive behavior. Here, we show that pyramidal neurons in the barrel cortex of BC1 knock out (KO) mice display larger excitatory postsynaptic currents and increased spontaneous activity in vivo. Furthermore, BC1 KO mice have enlarged spine heads and postsynaptic densities and increased synaptic levels of glutamate receptors and PSD-95. Of note, BC1 KO mice show aberrant structural plasticity in response to whisker deprivation, impaired texture novel object recognition and altered social behavior. Thus, our study highlights a role for BC1 RNA in experience-dependent plasticity and learning in the mammalian adult neocortex, and provides insight into the function of brain ncRNAs regulating synaptic transmission, plasticity and behavior, with potential relevance in the context of intellectual disabilities and psychiatric disorders.Brain cytoplasmic (BC1) RNA is a non-coding RNA that has been implicated in translational regulation, seizure, and anxiety. Here, the authors show that in the cortex, BC1 RNA is required for sensory deprivation-induced structural plasticity of dendritic spines, as well as for correct sensory learning and social behaviors.


Subject(s)
Learning/physiology , Neocortex/physiology , Neuronal Plasticity/physiology , Pyramidal Cells/physiology , RNA, Small Cytoplasmic/genetics , Animals , Base Sequence , Cells, Cultured , Dendritic Spines/metabolism , Dendritic Spines/physiology , Excitatory Postsynaptic Potentials/genetics , Excitatory Postsynaptic Potentials/physiology , In Situ Hybridization, Fluorescence , Male , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , Neocortex/cytology , Neocortex/metabolism , Neuronal Plasticity/genetics , Pyramidal Cells/metabolism , Pyramidal Cells/ultrastructure , Sensory Deprivation/physiology , Sequence Homology, Nucleic Acid , Social Behavior , Synaptic Transmission/genetics , Synaptic Transmission/physiology , Vibrissae/metabolism , Vibrissae/physiology
6.
Proc Natl Acad Sci U S A ; 114(8): 2006-2011, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28174276

ABSTRACT

The Notch1 gene is a major oncogenic driver and therapeutic target in T-cell acute lymphoblastic leukemia (T-ALL). However, inhibition of NOTCH signaling with γ-secretase inhibitors (GSIs) has shown limited antileukemic activity in clinical trials. Here we performed an expression-based virtual screening to identify highly active antileukemic drugs that synergize with NOTCH1 inhibition in T-ALL. Among these, withaferin A demonstrated the strongest cytotoxic and GSI-synergistic antileukemic effects in vitro and in vivo. Mechanistically, network perturbation analyses showed eIF2A-phosphorylation-mediated inhibition of protein translation as a critical mediator of the antileukemic effects of withaferin A and its interaction with NOTCH1 inhibition. Overall, these results support a role for anti-NOTCH1 therapies and protein translation inhibitor combinations in the treatment of T-ALL.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Drug Resistance, Neoplasm/drug effects , Enzyme Inhibitors/pharmacology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Protein Biosynthesis/drug effects , Receptor, Notch1/antagonists & inhibitors , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Drug Synergism , Enzyme Inhibitors/therapeutic use , Gene Expression Profiling , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Targeted Therapy/methods , Phosphorylation/drug effects , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Signal Transduction/drug effects , Withanolides/pharmacology , Xenograft Model Antitumor Assays , eIF-2 Kinase/metabolism
7.
Blood ; 129(9): 1113-1123, 2017 03 02.
Article in English | MEDLINE | ID: mdl-28115373

ABSTRACT

T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive malignancy caused by the accumulation of genomic lesions that affect the development of T cells. For many years, it has been established that deregulated expression of transcription factors, impairment of the CDKN2A/2B cell-cycle regulators, and hyperactive NOTCH1 signaling play prominent roles in the pathogenesis of this leukemia. In the past decade, systematic screening of T-ALL genomes by high-resolution copy-number arrays and next-generation sequencing technologies has revealed that T-cell progenitors accumulate additional mutations affecting JAK/STAT signaling, protein translation, and epigenetic control, providing novel attractive targets for therapy. In this review, we provide an update on our knowledge of T-ALL pathogenesis, the opportunities for the introduction of targeted therapy, and the challenges that are still ahead.


Subject(s)
Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Humans
9.
Haematologica ; 99(1): 85-93, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23872305

ABSTRACT

The NUP214-ABL1 fusion protein is a constitutively active protein tyrosine kinase that is found in 6% of patients with T-cell acute lymphoblastic leukemia and that promotes proliferation and survival of T-lymphoblasts. Although NUP214-ABL1 is sensitive to ABL1 kinase inhibitors, development of resistance to these compounds is a major clinical problem, underlining the need for additional drug targets in the sparsely studied NUP214-ABL1 signaling network. In this work, we identify and validate the SRC family kinase LCK as a protein whose activity is absolutely required for the proliferation and survival of T-cell acute lymphoblastic leukemia cells that depend on NUP214-ABL1 activity. These findings underscore the potential of SRC kinase inhibitors and of the dual ABL1/SRC kinase inhibitors dasatinib and bosutinib for the treatment of NUP214-ABL1-positive T-cell acute lymphoblastic leukemia. In addition, we used mass spectrometry to identify protein interaction partners of NUP214-ABL1. Our results strongly support that the signaling network of NUP214-ABL1 is distinct from that previously reported for BCR-ABL1. Moreover, we found that three NUP214-ABL1-interacting proteins, MAD2L1, NUP155, and SMC4, are strictly required for the proliferation and survival of NUP214-ABL1-positive T-cell acute lymphoblastic leukemia cells. In conclusion, this work identifies LCK, MAD2L1, NUP155 and SMC4 as four new potential drug targets in NUP214-ABL1-positive T-cell acute lymphoblastic leukemia.


Subject(s)
Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Proliferation , Gene Knockdown Techniques , Humans , Phosphorylation , Protein Binding , Protein Interaction Mapping , RNA Interference , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
10.
Nat Genet ; 45(2): 186-90, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23263491

ABSTRACT

T-cell acute lymphoblastic leukemia (T-ALL) is caused by the cooperation of multiple oncogenic lesions. We used exome sequencing on 67 T-ALLs to gain insight into the mutational spectrum in these leukemias. We detected protein-altering mutations in 508 genes, with an average of 8.2 mutations in pediatric and 21.0 mutations in adult T-ALL. Using stringent filtering, we predict seven new oncogenic driver genes in T-ALL. We identify CNOT3 as a tumor suppressor mutated in 7 of 89 (7.9%) adult T-ALLs, and its knockdown causes tumors in a sensitized Drosophila melanogaster model. In addition, we identify mutations affecting the ribosomal proteins RPL5 and RPL10 in 12 of 122 (9.8%) pediatric T-ALLs, with recurrent alterations of Arg98 in RPL10. Yeast and lymphoid cells expressing the RPL10 Arg98Ser mutant showed a ribosome biogenesis defect. Our data provide insights into the mutational landscape of pediatric versus adult T-ALL and identify the ribosome as a potential oncogenic factor.


Subject(s)
Exome/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Ribosomal Proteins/genetics , Transcription Factors/genetics , Animals , Base Sequence , Drosophila melanogaster , High-Throughput Nucleotide Sequencing , Humans , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation/genetics , Polyribosomes/genetics , RNA Interference , Ribosomal Protein L10 , Saccharomyces cerevisiae , Sequence Alignment
11.
Toxicon ; 56(3): 381-90, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20398688

ABSTRACT

The Macrovipera lebetina venom consists of a complex mixture of proteins belonging to a few main families according to their enzymatic and pharmacological activity. Given the serious pathophysiological effects caused by M. lebetina bites mainly induced by muscle degeneration, we decided to investigate the myotoxic activity of some venom fractions. In the present study we describe the purification and characterization of a 22.600 kDa protein, named in the following Mlp4.2, that shares myotoxic but not haemorrhagic activity in vivo. Herein we report that Mlp4.2 is a metalloproteinase belonging to the PI-SVMPS family able, in vitro, to proteolyse extracellular matrix proteins as laminin and fibronectin. Histological observations of mouse anterior tibialis Mlp4.2-treated muscle, demonstrate that this protein induces a massive degeneration of myofibers but not haemorrhage. The immunofluorescence analysis of protein-treated anterior tibialis, demonstrates that Mlp4.2 is able to disarray the laminin network surrounding muscle fibers. Finally Mlp4.2 did not show any direct cytolytic activity towards the myogenic cell line C2C12 in culture. The data reported herein suggest that the myotoxicity of Mlp4.2 is primarily linked to the disruption of the muscle fibers interaction with extracellular matrix proteins.


Subject(s)
Muscles/drug effects , Viper Venoms/chemistry , Animals , Electrophoresis, Polyacrylamide Gel , Humans , Muscles/pathology , Spectrometry, Mass, Electrospray Ionization , Viper Venoms/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL
...