Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Neuron ; 110(7): 1173-1192.e7, 2022 04 06.
Article in English | MEDLINE | ID: mdl-35114102

ABSTRACT

In Huntington's disease (HD), the uninterrupted CAG repeat length, but not the polyglutamine length, predicts disease onset. However, the underlying pathobiology remains unclear. Here, we developed bacterial artificial chromosome (BAC) transgenic mice expressing human mutant huntingtin (mHTT) with uninterrupted, and somatically unstable, CAG repeats that exhibit progressive disease-related phenotypes. Unlike prior mHTT transgenic models with stable, CAA-interrupted, polyglutamine-encoding repeats, BAC-CAG mice show robust striatum-selective nuclear inclusions and transcriptional dysregulation resembling those in murine huntingtin knockin models and HD patients. Importantly, the striatal transcriptionopathy in HD models is significantly correlated with their uninterrupted CAG repeat length but not polyglutamine length. Finally, among the pathogenic entities originating from mHTT genomic transgenes and only present or enriched in the uninterrupted CAG repeat model, somatic CAG repeat instability and nuclear mHTT aggregation are best correlated with early-onset striatum-selective molecular pathogenesis and locomotor and sleep deficits, while repeat RNA-associated pathologies and repeat-associated non-AUG (RAN) translation may play less selective or late pathogenic roles, respectively.


Subject(s)
Huntington Disease , Nerve Tissue Proteins , Animals , Chromosomes, Artificial, Bacterial/genetics , Chromosomes, Artificial, Bacterial/metabolism , Disease Models, Animal , Humans , Huntingtin Protein/genetics , Huntington Disease/genetics , Huntington Disease/pathology , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Neurons/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Trinucleotide Repeat Expansion/genetics
2.
EMBO Mol Med ; 13(11): e14095, 2021 11 08.
Article in English | MEDLINE | ID: mdl-34632710

ABSTRACT

Spinocerebellar ataxia type 8 (SCA8), a dominantly inherited neurodegenerative disorder caused by a CTG•CAG expansion, is unusual because most individuals that carry the mutation do not develop ataxia. To understand the variable penetrance of SCA8, we studied the molecular differences between highly penetrant families and more common sporadic cases (82%) using a large cohort of SCA8 families (n = 77). We show that repeat expansion mutations from individuals with multiple affected family members have CCG•CGG interruptions at a higher frequency than sporadic SCA8 cases and that the number of CCG•CGG interruptions correlates with age at onset. At the molecular level, CCG•CGG interruptions increase RNA hairpin stability, and in cell culture experiments, increase p-eIF2α and polyAla and polySer RAN protein levels. Additionally, CCG•CGG interruptions, which encode arginine interruptions in the polyGln frame, increase toxicity of the resulting proteins. In summary, SCA8 CCG•CGG interruptions increase polyAla and polySer RAN protein levels, polyGln protein toxicity, and disease penetrance and provide novel insight into the molecular differences between SCA8 families with high vs. low disease penetrance.


Subject(s)
Spinocerebellar Degenerations , Trinucleotide Repeat Expansion , Ataxia , Humans , Nerve Tissue Proteins/genetics , Penetrance , Proteins , RNA, Long Noncoding/genetics , Spinocerebellar Degenerations/genetics
3.
Neuron ; 105(4): 645-662.e11, 2020 02 19.
Article in English | MEDLINE | ID: mdl-31831332

ABSTRACT

The intronic C9orf72 G4C2 expansion, the most common genetic cause of ALS and FTD, produces sense- and antisense-expansion RNAs and six dipeptide repeat-associated, non-ATG (RAN) proteins, but their roles in disease are unclear. We generated high-affinity human antibodies targeting GA or GP RAN proteins. These antibodies cross the blood-brain barrier and co-localize with intracellular RAN aggregates in C9-ALS/FTD BAC mice. In cells, α-GA1 interacts with TRIM21, and α-GA1 treatment reduced GA levels, increased GA turnover, and decreased RAN toxicity and co-aggregation of proteasome and autophagy proteins to GA aggregates. In C9-BAC mice, α-GA1 reduced GA as well as GP and GR proteins, improved behavioral deficits, decreased neuroinflammation and neurodegeneration, and increased survival. Glycosylation of the Fc region of α-GA1 is important for cell entry and efficacy. These data demonstrate that RAN proteins drive C9-ALS/FTD in C9-BAC transgenic mice and establish a novel therapeutic approach for C9orf72 ALS/FTD and other RAN-protein diseases.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Antibodies, Monoclonal/genetics , C9orf72 Protein/genetics , Frontotemporal Dementia/genetics , Genetic Therapy/methods , ran GTP-Binding Protein/metabolism , Aged , Amyotrophic Lateral Sclerosis/metabolism , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/metabolism , Brain/metabolism , C9orf72 Protein/metabolism , Cell Line, Tumor , Disease Models, Animal , Female , Frontotemporal Dementia/metabolism , Gene Targeting/methods , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Random Allocation , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , ran GTP-Binding Protein/antagonists & inhibitors
4.
Lab Invest ; 99(7): 929-942, 2019 07.
Article in English | MEDLINE | ID: mdl-30918326

ABSTRACT

More than 40 different neurological diseases are caused by microsatellite repeat expansions. Since the discovery of repeat-associated non-AUG (RAN) translation by Zu et al. in 2011, nine expansion disorders have been identified as RAN-positive diseases. RAN proteins are translated from different types of nucleotide repeat expansions and can be produced from both sense and antisense transcripts. In some diseases, RAN proteins have been shown to accumulate in affected brain regions. Here we review the pathological and molecular aspects associated with RAN protein accumulation for each particular disorder, the correlation between disease pathology and the available in vivo models and the common aspects shared by some of the newly discovered RAN proteins.


Subject(s)
Microsatellite Repeats , Nervous System Diseases/genetics , Proteostasis Deficiencies/etiology , Animals , Gene Expression , Humans , Proteostasis Deficiencies/therapy
5.
EMBO J ; 37(19)2018 10 01.
Article in English | MEDLINE | ID: mdl-30206144

ABSTRACT

Spinocerebellar ataxia type 8 (SCA8) is caused by a bidirectionally transcribed CTG·CAG expansion that results in the in vivo accumulation of CUG RNA foci, an ATG-initiated polyGln and a polyAla protein expressed by repeat-associated non-ATG (RAN) translation. Although RAN proteins have been reported in a growing number of diseases, the mechanisms and role of RAN translation in disease are poorly understood. We report a novel toxic SCA8 polySer protein which accumulates in white matter (WM) regions as aggregates that increase with age and disease severity. WM regions with polySer aggregates show demyelination and axonal degeneration in SCA8 human and mouse brains. Additionally, knockdown of the eukaryotic translation initiation factor eIF3F in cells reduces steady-state levels of SCA8 polySer and other RAN proteins. Taken together, these data show polySer and WM abnormalities contribute to SCA8 and identify eIF3F as a novel modulator of RAN protein accumulation.


Subject(s)
Aging/metabolism , Eukaryotic Initiation Factor-3/metabolism , Nerve Tissue Proteins/metabolism , Spinocerebellar Degenerations/metabolism , White Matter/metabolism , Aging/genetics , Aging/pathology , Animals , Eukaryotic Initiation Factor-3/genetics , HeLa Cells , Humans , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Spinocerebellar Degenerations/genetics , Spinocerebellar Degenerations/pathology , White Matter/pathology
6.
Neuron ; 95(6): 1292-1305.e5, 2017 Sep 13.
Article in English | MEDLINE | ID: mdl-28910618

ABSTRACT

Several microsatellite-expansion diseases are characterized by the accumulation of RNA foci and RAN proteins, raising the possibility of a mechanistic connection. We explored this question using myotonic dystrophy type 2, a multisystemic disease thought to be primarily caused by RNA gain-of-function effects. We demonstrate that the DM2 CCTG⋅CAGG expansion expresses sense and antisense tetrapeptide poly-(LPAC) and poly-(QAGR) RAN proteins, respectively. In DM2 autopsy brains, LPAC is found in neurons, astrocytes, and glia in gray matter, and antisense QAGR proteins accumulate within white matter. LPAC and QAGR proteins are toxic to cells independent of RNA gain of function. RNA foci and nuclear sequestration of CCUG transcripts by MBNL1 is inversely correlated with LPAC expression. These data suggest a model that involves nuclear retention of expansion RNAs by RNA-binding proteins (RBPs) and an acute phase in which expansion RNAs exceed RBP sequestration capacity, are exported to the cytoplasm, and undergo RAN translation. VIDEO ABSTRACT.


Subject(s)
Myotonic Dystrophy/metabolism , Protein Biosynthesis , RNA-Binding Proteins/metabolism , ran GTP-Binding Protein/biosynthesis , Brain/metabolism , Cell Survival , Cells, Cultured , Gene Expression Regulation , Humans , Mutation , RNA/metabolism , RNA-Binding Proteins/genetics , ran GTP-Binding Protein/toxicity
7.
J Clin Invest ; 126(11): 4319-4330, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27721240

ABSTRACT

Huntington's disease (HD) is a polyglutamine disorder caused by a CAG expansion in the Huntingtin (HTT) gene exon 1. This expansion encodes a mutant protein whose abnormal function is traditionally associated with HD pathogenesis; however, recent evidence has also linked HD pathogenesis to RNA stable hairpins formed by the mutant HTT expansion. Here, we have shown that a locked nucleic acid-modified antisense oligonucleotide complementary to the CAG repeat (LNA-CTG) preferentially binds to mutant HTT without affecting HTT mRNA or protein levels. LNA-CTGs produced rapid and sustained improvement of motor deficits in an R6/2 mouse HD model that was paralleled by persistent binding of LNA-CTG to the expanded HTT exon 1 transgene. Motor improvement was accompanied by a pronounced recovery in the levels of several striatal neuronal markers severely impaired in R6/2 mice. Furthermore, in R6/2 mice, LNA-CTG blocked several pathogenic mechanisms caused by expanded CAG RNA, including small RNA toxicity and decreased Rn45s expression levels. These results suggest that LNA-CTGs promote neuroprotection by blocking the detrimental activity of CAG repeats within HTT mRNA. The present data emphasize the relevance of expanded CAG RNA to HD pathogenesis, indicate that inhibition of HTT expression is not required to reverse motor deficits, and further suggest a therapeutic potential for LNA-CTG in polyglutamine disorders.


Subject(s)
Gene Expression Regulation/drug effects , Huntingtin Protein , Huntington Disease , RNA, Antisense , Trinucleotide Repeats , Animals , Cell Line, Tumor , Disease Models, Animal , Humans , Huntingtin Protein/biosynthesis , Huntingtin Protein/genetics , Huntington Disease/genetics , Huntington Disease/metabolism , Huntington Disease/therapy , Male , Mice , Mice, Transgenic , RNA, Antisense/genetics , RNA, Antisense/pharmacology
8.
Neuron ; 88(4): 667-77, 2015 Nov 18.
Article in English | MEDLINE | ID: mdl-26590344

ABSTRACT

Huntington disease (HD) is caused by a CAG ⋅ CTG expansion in the huntingtin (HTT) gene. While most research has focused on the HTT polyGln-expansion protein, we demonstrate that four additional, novel, homopolymeric expansion proteins (polyAla, polySer, polyLeu, and polyCys) accumulate in HD human brains. These sense and antisense repeat-associated non-ATG (RAN) translation proteins accumulate most abundantly in brain regions with neuronal loss, microglial activation and apoptosis, including caudate/putamen, white matter, and, in juvenile-onset cases, also the cerebellum. RAN protein accumulation and aggregation are length dependent, and individual RAN proteins are toxic to neural cells independent of RNA effects. These data suggest RAN proteins contribute to HD and that therapeutic strategies targeting both sense and antisense genes may be required for efficacy in HD patients. This is the first demonstration that RAN proteins are expressed across an expansion located in an open reading frame and suggests RAN translation may also contribute to other polyglutamine diseases.


Subject(s)
Brain/metabolism , Huntington Disease/genetics , Nerve Tissue Proteins/genetics , RNA, Antisense/genetics , Trinucleotide Repeat Expansion/genetics , Adult , Aged, 80 and over , Case-Control Studies , Cerebellum/metabolism , Child , Female , Frontal Lobe/metabolism , HEK293 Cells , Humans , Huntingtin Protein , Huntington Disease/metabolism , Male , Middle Aged , Neostriatum/metabolism , Nerve Tissue Proteins/metabolism , Peptides/metabolism , Protein Aggregation, Pathological/metabolism , Protein Biosynthesis , RNA, Antisense/metabolism , Young Adult
9.
Genome Biol ; 15(4): R57, 2014 Apr 07.
Article in English | MEDLINE | ID: mdl-24708865

ABSTRACT

BACKGROUND: MicroRNAs (miRNAs) are established regulators of development, cell identity and disease. Although nearly two thousand human miRNA genes are known and new ones are continuously discovered, no attempt has been made to gauge the total miRNA content of the human genome. RESULTS: Employing an innovative computational method on massively pooled small RNA sequencing data, we report 2,469 novel human miRNA candidates of which 1,098 are validated by in-house and published experiments. Almost 300 candidates are robustly expressed in a neuronal cell system and are regulated during differentiation or when biogenesis factors Dicer, Drosha, DGCR8 or Ago2 are silenced. To improve expression profiling, we devised a quantitative miRNA capture system. In a kidney cell system, 400 candidates interact with DGCR8 at transcript positions that suggest miRNA hairpin recognition, and 1,000 of the new miRNA candidates interact with Ago1 or Ago2, indicating that they are directly bound by miRNA effector proteins. From kidney cell CLASH experiments, in which miRNA-target pairs are ligated and sequenced, we observe hundreds of interactions between novel miRNAs and mRNA targets. The novel miRNA candidates are specifically but lowly expressed, raising the possibility that not all may be functional. Interestingly, the majority are evolutionarily young and overrepresented in the human brain. CONCLUSIONS: In summary, we present evidence that the complement of human miRNA genes is substantially larger than anticipated, and that more are likely to be discovered in the future as more tissues and experimental conditions are sequenced to greater depth.


Subject(s)
Genome, Human , MicroRNAs/genetics , Argonaute Proteins/metabolism , Eukaryotic Initiation Factors/metabolism , Humans , MCF-7 Cells , MicroRNAs/metabolism , Protein Binding , RNA-Binding Proteins/metabolism
10.
Proc Natl Acad Sci U S A ; 110(51): E4968-77, 2013 Dec 17.
Article in English | MEDLINE | ID: mdl-24248382

ABSTRACT

The finding that a GGGGCC (G4C2) hexanucleotide repeat expansion in the chromosome 9 ORF 72 (C9ORF72) gene is a common cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) links ALS/FTD to a large group of unstable microsatellite diseases. Previously, we showed that microsatellite expansion mutations can be bidirectionally transcribed and that these mutations express unexpected proteins by a unique mechanism, repeat-associated non-ATG (RAN) translation. In this study, we show that C9ORF72 antisense transcripts are elevated in the brains of C9ORF72 expansion-positive [C9(+)] patients, and antisense GGCCCC (G2C4) repeat-expansion RNAs accumulate in nuclear foci in brain. Additionally, sense and antisense foci accumulate in blood and are potential biomarkers of the disease. Furthermore, we show that RAN translation occurs from both sense and antisense expansion transcripts, resulting in the expression of six RAN proteins (antisense: Pro-Arg, Pro-Ala, Gly-Pro; and sense: Gly-Ala, Gly-Arg, Gly-Pro). These proteins accumulate in cytoplasmic aggregates in affected brain regions, including the frontal and motor cortex, hippocampus, and spinal cord neurons, with some brain regions showing dramatic RAN protein accumulation and clustering. The finding that unique antisense G2C4 RNA foci and three unique antisense RAN proteins accumulate in patient tissues indicates that bidirectional transcription of expanded alleles is a fundamental pathologic feature of C9ORF72 ALS/FTD. Additionally, these findings suggest the need to test therapeutic strategies that target both sense and antisense RNAs and RAN proteins in C9ORF72 ALS/FTD, and to more broadly consider the role of antisense expression and RAN translation across microsatellite expansion diseases.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Brain/metabolism , Frontotemporal Dementia/metabolism , Nerve Tissue Proteins/metabolism , Proteins/metabolism , RNA, Antisense/biosynthesis , ran GTP-Binding Protein/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Brain/pathology , C9orf72 Protein , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Nucleus/pathology , Female , Frontotemporal Dementia/genetics , Frontotemporal Dementia/pathology , HEK293 Cells , Humans , Male , Nerve Tissue Proteins/genetics , Proteins/genetics , RNA, Antisense/genetics , Repetitive Sequences, Nucleic Acid , ran GTP-Binding Protein/genetics
11.
BMC Genomics ; 14: 104, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23414127

ABSTRACT

BACKGROUND: MicroRNAs (miRNAs) are short non-coding regulatory RNAs that control gene expression usually producing translational repression and gene silencing. High-throughput sequencing technologies have revealed heterogeneity at length and sequence level for the majority of mature miRNAs (IsomiRs). Most isomiRs can be explained by variability in either Dicer1 or Drosha cleavage during miRNA biogenesis at 5' or 3' of the miRNA (trimming variants). Although isomiRs have been described in different tissues and organisms, their functional validation as modulators of gene expression remains elusive. Here we have characterized the expression and function of a highly abundant miR-101 5'-trimming variant (5'-isomiR-101). RESULTS: The analysis of small RNA sequencing data in several human tissues and cell lines indicates that 5'-isomiR-101 is ubiquitously detected and a highly abundant, especially in the brain. 5'-isomiR-101 was found in Ago-2 immunocomplexes and complementary approaches showed that 5'-isomiR-101 interacted with different members of the silencing (RISC) complex. In addition, 5'-isomiR-101 decreased the expression of five validated miR-101 targets, suggesting that it is a functional variant. Both the binding to RISC members and the degree of silencing were less efficient for 5'-isomiR-101 compared with miR-101. For some targets, both miR-101 and 5'-isomiR-101 significantly decreased protein expression with no changes in the respective mRNA levels. Although a high number of overlapping predicted targets suggest similar targeted biological pathways, a correlation analysis of the expression profiles of miR-101 variants and predicted mRNA targets in human brains at different ages, suggest specific functions for miR-101- and 5'-isomiR-101. CONCLUSIONS: These results suggest that isomiRs are functional variants and further indicate that for a given miRNA, the different isomiRs may contribute to the overall effect as quantitative and qualitative fine-tuners of gene expression.


Subject(s)
Gene Expression Regulation/genetics , Gene Silencing , MicroRNAs/genetics , RNA Isoforms/genetics , Amyloid beta-Protein Precursor/deficiency , Amyloid beta-Protein Precursor/genetics , Argonaute Proteins/metabolism , Cell Line , Cyclooxygenase 2/deficiency , Cyclooxygenase 2/genetics , DEAD-box RNA Helicases/metabolism , Dual Specificity Phosphatase 1/deficiency , Dual Specificity Phosphatase 1/genetics , Enhancer of Zeste Homolog 2 Protein , Humans , MicroRNAs/metabolism , Myeloid Cell Leukemia Sequence 1 Protein , Polycomb Repressive Complex 2/deficiency , Polycomb Repressive Complex 2/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/deficiency , Proto-Oncogene Proteins c-bcl-2/genetics , RNA Isoforms/metabolism
12.
PLoS Genet ; 8(2): e1002481, 2012.
Article in English | MEDLINE | ID: mdl-22383888

ABSTRACT

Huntington's disease (HD) is an autosomal dominantly inherited disorder caused by the expansion of CAG repeats in the Huntingtin (HTT) gene. The abnormally extended polyglutamine in the HTT protein encoded by the CAG repeats has toxic effects. Here, we provide evidence to support that the mutant HTT CAG repeats interfere with cell viability at the RNA level. In human neuronal cells, expanded HTT exon-1 mRNA with CAG repeat lengths above the threshold for complete penetrance (40 or greater) induced cell death and increased levels of small CAG-repeated RNAs (sCAGs), of ≈21 nucleotides in a Dicer-dependent manner. The severity of the toxic effect of HTT mRNA and sCAG generation correlated with CAG expansion length. Small RNAs obtained from cells expressing mutant HTT and from HD human brains significantly decreased neuronal viability, in an Ago2-dependent mechanism. In both cases, the use of anti-miRs specific for sCAGs efficiently blocked the toxic effect, supporting a key role of sCAGs in HTT-mediated toxicity. Luciferase-reporter assays showed that expanded HTT silences the expression of CTG-containing genes that are down-regulated in HD. These results suggest a possible link between HD and sCAG expression with an aberrant activation of the siRNA/miRNA gene silencing machinery, which may trigger a detrimental response. The identification of the specific cellular processes affected by sCAGs may provide insights into the pathogenic mechanisms underlying HD, offering opportunities to develop new therapeutic approaches.


Subject(s)
Huntington Disease/etiology , Nerve Tissue Proteins/genetics , RNA, Messenger , Exons , Gene Silencing , Humans , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/pathology , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Neurons/pathology , Peptides/genetics , Peptides/metabolism , RNA, Messenger/metabolism , Trinucleotide Repeat Expansion
13.
Nucleic Acids Res ; 38(20): 7219-35, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20591823

ABSTRACT

Huntington disease (HD) is a neurodegenerative disorder that predominantly affects neurons of the forebrain. We have applied the Illumina massively parallel sequencing to deeply analyze the small RNA populations of two different forebrain areas, the frontal cortex (FC) and the striatum (ST) of healthy individuals and individuals with HD. More than 80% of the small-RNAs were annotated as microRNAs (miRNAs) in all samples. Deep sequencing revealed length and sequence heterogeneity (IsomiRs) for the vast majority of miRNAs. Around 80-90% of the miRNAs presented modifications in the 3'-terminus mainly in the form of trimming and/or as nucleotide addition variants, while the 5'-terminus of the miRNAs was specially protected from changes. Expression profiling showed strong miRNA and isomiR expression deregulation in HD, most being common to both FC and ST. The analysis of the upstream regulatory regions in co-regulated miRNAs suggests a role for RE1-Silencing Transcription Factor (REST) and P53 in miRNAs downregulation in HD. The putative targets of deregulated miRNAs and seed-region IsomiRs strongly suggest that their altered expression contributes to the aberrant gene expression in HD. Our results show that miRNA variability is a ubiquitous phenomenon in the adult human brain, which may influence gene expression in physiological and pathological conditions.


Subject(s)
Brain/metabolism , Genetic Variation , Huntington Disease/genetics , MicroRNAs/chemistry , Adult , Frontal Lobe/metabolism , Gene Expression Profiling , Gene Expression Regulation , Humans , Huntington Disease/metabolism , MicroRNAs/metabolism , Neostriatum/metabolism , RNA, Small Untranslated/chemistry , Repressor Proteins/metabolism , Sequence Analysis, RNA
14.
Curr Cancer Drug Targets ; 8(8): 709-19, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19075594

ABSTRACT

Choline Kinase (ChoK) comprises a family of cytosolic enzymes involved in the synthesis of phosphatidylcholine (PC), the most abundant phospholipid in eukaryotic cell membranes. One of the ChoK isoforms, Choline Kinase alpha (ChoKalpha), is found over expressed in human tumours. Chemical inhibitors able to interfere with ChoK activity have proven to be effective antitumoral drugs in vitro and in vivo. To validate the use of selective ChoKalpha inhibitors in cancer therapy, we have developed a genetic strategy to interfere specifically with ChoKalpha activity based on the generation of a shRNA against the alpha isoform of ChoK. Here we demonstrate that specific inhibition of ChoKalpha by shRNA has antitumor activity. The specific depletion of ChoKalpha induces apoptosis in several tumor-derived cell lines from breast, bladder, lung and cervix carcinoma tumors, while the viability of normal primary cells is not affected. Furthermore, this selective antiproliferative effect is achieved both under in vitro and in vivo conditions, as demonstrated by an inducible ChoKalpha suppression system in human tumour xenografts. These results demonstrate that ChoKalpha inhibition is a useful antitumoral strategy per se, and provides definitive and non-ambiguous evidence that ChoKalpha can be used as an efficient and selective drug target for cancer therapy.


Subject(s)
Apoptosis/physiology , Choline Kinase/physiology , Base Sequence , Cell Line, Tumor , Choline Kinase/genetics , Choline Kinase/metabolism , DNA Primers , Flow Cytometry , Gene Silencing , Humans , Microscopy, Fluorescence , RNA/genetics , RNA Interference
15.
Cancer Res ; 65(13): 5647-53, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15994937

ABSTRACT

Choline kinase is overexpressed in human breast, lung, colorectal, and prostate tumors, a finding that suggests the involvement of this enzyme in carcinogenesis. Here we show that overexpression of choline kinase induce oncogenic transformation of human embryo kidney fibroblasts and canine epithelial Madin-Darby canine kidney cells. Choline kinase lays downstream of RhoA signaling and is activated through ROCK kinase, one of the best-characterized RhoA effectors. In keeping with this, coexpression of RhoA and choline kinase potentiates both anchorage independent growth and tumorigenesis. Finally, choline kinase-mediated transformation is sensitive to MN58b, a well-characterized specific choline kinase inhibitor. These results provide the definitive evidence that choline kinase has oncogenic properties and that choline kinase inhibition constitutes a novel valid antitumor strategy.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Choline Kinase/genetics , rhoA GTP-Binding Protein/physiology , Animals , Cell Transformation, Neoplastic/genetics , Choline Kinase/antagonists & inhibitors , Choline Kinase/metabolism , Dogs , Enzyme Activation , Humans , Mice , Mice, Nude , NIH 3T3 Cells , Oncogenes , Rats , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism , cdc42 GTP-Binding Protein/physiology , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/physiology , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
16.
J Med Chem ; 48(9): 3354-63, 2005 May 05.
Article in English | MEDLINE | ID: mdl-15857141

ABSTRACT

Studies have been aimed at the establishment of structure-activity relationships that define choline kinase inhibitory and antiproliferative activities of 40 bisquinolinium compounds. These derivatives have electron-releasing groups at position 4 of the quinolinium ring. It is found that the enzymatic inhibition is closely related to the size of the linker, the 3,3'-biphenyl moiety being the most suitable. On the other hand, the antiproliferative activity against the HT-29 cancer cell line is less influenced by the linker type and by substituent R(4). The corresponding QSAR equation was obtained for the whole set of compounds for the antiproliferative activity, the electronic parameter sigma(R) of R(4), the molar refractivity of R(8), and the lipophilic parameters clog P and pi(linker). The most potent antiproliferative agent so far described is 40 for which an IC(50) = 0.45 microM was predicted by the QSAR equation, while its experimental value is IC(50) = 0.20 microM.


Subject(s)
Choline Kinase/antagonists & inhibitors , Quinolines/chemical synthesis , Quinolinium Compounds/chemical synthesis , Biphenyl Compounds/chemistry , HT29 Cells , Humans , Quantitative Structure-Activity Relationship , Quinolines/chemistry , Quinolines/pharmacology , Quinolinium Compounds/chemistry , Quinolinium Compounds/pharmacology
17.
Int J Oncol ; 26(4): 999-1008, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15753995

ABSTRACT

Tumour cells are frequently altered in their phospholipid metabolism. Choline kinase (ChoK, E.C. 2.7.1.32) activity, the first enzyme involved in the synthesis of phosphatidylcholine, is increased in a large number of human tumours and tumour-derived cell lines. We have previously reported that MN58b, a specific inhibitor of ChoK, has anti-tumoral activity. Here we show the high specificity of MN58b as a cytotoxic drug towards tumour cells, and explore further the basis of its mechanism of action in order to provide a rational understanding for its antitumoral activity. A dramatic difference in the response to the treatment of primary, normal and non-tumorigenic human cells when compared to tumour-derived cell lines was observed. In normal cells, blockage of de novo PCho synthesis by MN58b results in a reversible cell cycle arrest at G0/G1 phase. In contrast, ChoK inhibition in tumour cells promotes the induction of apoptosis. This effect depends on the cell cycle phase, being G1 the critical phase. Regarding the mechanism of apoptosis engagement, a loss of mitochondrial potential was observed 10-20 min after cytochrome c release, but caspase 3 activation preceded the loss of mitochondrial potential, indicating that activation of caspase 3 is independent of cytochrome c release. Our results are consistent with a non-intrinsic process as the mechanism underlying the induction of apoptosis by ChoK inhibition in tumoral cells.


Subject(s)
Apoptosis/drug effects , Choline Kinase/antagonists & inhibitors , Choline Kinase/pharmacology , Enzyme Inhibitors/pharmacology , Mitochondria/pathology , Neoplasms/pathology , Tumor Cells, Cultured , Cell Cycle , Female , Humans , Male
18.
J Med Chem ; 47(22): 5433-40, 2004 Oct 21.
Article in English | MEDLINE | ID: mdl-15481981

ABSTRACT

Studies have been aimed to establish the structure-activity relationship that define choline kinase (ChoK) inhibitory potency and antiproliferative activity of a set of 25 bispyridinium compounds with electron-releasing groups at position 4. Here we report that, according to their inhibitory activities against human ChoK, the enzymatic inhibitory potency is closely related to the size of the linker, the 3,3'-biphenyl moiety being the most suitable. The N-methylanilino and its derivatives, 4-chloro-N-methylanilino and 3,5-dichloro-N-methylanilino, render higher ChoK inhibitory and antiproliferative activities against the HT-29 human colon cancer cell line.


Subject(s)
Antineoplastic Agents/chemical synthesis , Choline Kinase/antagonists & inhibitors , Pyridinium Compounds/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Benzene Derivatives/chemical synthesis , Benzene Derivatives/chemistry , Benzene Derivatives/pharmacology , Biphenyl Compounds/chemical synthesis , Biphenyl Compounds/chemistry , Biphenyl Compounds/pharmacology , Cell Line, Tumor , Cyclopropanes/chemical synthesis , Cyclopropanes/chemistry , Cyclopropanes/pharmacology , Drug Screening Assays, Antitumor , Humans , Pyridinium Compounds/chemistry , Pyridinium Compounds/pharmacology , Structure-Activity Relationship
19.
Cancer Res ; 64(18): 6732-9, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15374991

ABSTRACT

Breast cancer is still one of the most important tumors among women in industrialized countries. Improvement in both understanding the molecular events associated with the disease and the development of new additional treatments is still an important goal to be achieved. Choline kinase (ChoK) is increased in human mammary tumors with high incidence, and this activation is associated with clinical variable indicators of greater malignancy. Here, we have investigated the role of ChoK in the development of breast cancer and found that ChoK is both necessary and sufficient for growth factor-induced proliferation in primary human mammary epithelial cells and an absolute requirement for the specific mitogenic response to heregulin in breast tumor-derived cells. These results demonstrate that ChoK plays an essential role in both normal human mammary epithelial cell proliferation and breast tumor progression. Furthermore, inhibition of ChoK shows a strong in vivo antitumor activity against human breast cancer xenografts. Thus, ChoK constitutes a novel bona fide molecular target for the treatment of breast cancer patients.


Subject(s)
Breast Neoplasms/enzymology , Choline Kinase/antagonists & inhibitors , Choline Kinase/metabolism , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Division/drug effects , Cell Division/physiology , Choline Kinase/genetics , Disease Progression , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Female , Humans , Mammary Glands, Human/cytology , Mammary Glands, Human/drug effects , Mammary Glands, Human/enzymology , Mice , Mice, Nude , Neuregulin-1/pharmacology , Phosphorylcholine/metabolism , Transfection , Xenograft Model Antitumor Assays
20.
Int J Oncol ; 25(4): 1097-103, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15375561

ABSTRACT

The compound 6a is a novel bisoxazol derivative with high cytotoxic properties in vitro against different human tumor-derived cell lines and with similar efficiency against epithelial, haematopoietic and mesenchymal tumor cells. Although the molecular mechanism is not yet fully defined, cell cycle analysis revealed that 6a induces efficiently G0/G1 phase arrest in colon adenocarcinoma HT-29 cells in a dose- and time-dependent manner. Induction of cell death is observed, a possible explanation for the antiproliferative profile of the molecule. The compound was well tolerated at doses that allowed to examine its antitumor activity against human xenografts of the HT-29 cell line implanted s.c. in nude mice. Treatment of mice with 4 mg/kg of the compound resulted in a 60% inhibition of tumor growth. These observations support the use of 6a for the generation of more potent derivatives that could be used as new anticancer agents.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Choline Kinase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/toxicity , Cell Division/drug effects , Enzyme Inhibitors/chemical synthesis , HT29 Cells , Humans , Male , Mice , Neoplasm Transplantation , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...