Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Nature ; 628(8006): 122-129, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38448590

RESUMO

Genomic imprinting-the non-equivalence of maternal and paternal genomes-is a critical process that has evolved independently in many plant and mammalian species1,2. According to kinship theory, imprinting is the inevitable consequence of conflictive selective forces acting on differentially expressed parental alleles3,4. Yet, how these epigenetic differences evolve in the first place is poorly understood3,5,6. Here we report the identification and molecular dissection of a parent-of-origin effect on gene expression that might help to clarify this fundamental question. Toxin-antidote elements (TAs) are selfish elements that spread in populations by poisoning non-carrier individuals7-9. In reciprocal crosses between two Caenorhabditis tropicalis wild isolates, we found that the slow-1/grow-1 TA is specifically inactive when paternally inherited. This parent-of-origin effect stems from transcriptional repression of the slow-1 toxin by the PIWI-interacting RNA (piRNA) host defence pathway. The repression requires PIWI Argonaute and SET-32 histone methyltransferase activities and is transgenerationally inherited via small RNAs. Remarkably, when slow-1/grow-1 is maternally inherited, slow-1 repression is halted by a translation-independent role of its maternal mRNA. That is, slow-1 transcripts loaded into eggs-but not SLOW-1 protein-are necessary and sufficient to counteract piRNA-mediated repression. Our findings show that parent-of-origin effects can evolve by co-option of the piRNA pathway and hinder the spread of selfish genes that require sex for their propagation.


Assuntos
Caenorhabditis , Impressão Genômica , RNA de Interação com Piwi , Sequências Repetitivas de Ácido Nucleico , Animais , Feminino , Masculino , Alelos , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Caenorhabditis/genética , Caenorhabditis/metabolismo , Cruzamentos Genéticos , Pai , Genoma/genética , Impressão Genômica/genética , Organismos Hermafroditas/genética , Histona Metiltransferases/genética , Histona Metiltransferases/metabolismo , Mães , Oócitos/metabolismo , RNA de Interação com Piwi/genética , Biossíntese de Proteínas , Sequências Repetitivas de Ácido Nucleico/genética , RNA Mensageiro/genética , Toxinas Biológicas/genética , Transcrição Gênica
3.
J Cell Sci ; 134(16)2021 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-34291805

RESUMO

Plasmodium falciparum, the parasite responsible for the deadliest form of human malaria, replicates within the erythrocytes of its host, where it encounters numerous pressures that cause extensive DNA damage, which must be repaired efficiently to ensure parasite survival. Malaria parasites, which have lost the non-homologous end joining (NHEJ) pathway for repairing DNA double-strand breaks, have evolved unique mechanisms that enable them to robustly maintain genome integrity under such harsh conditions. However, the nature of these adaptations is unknown. We show that a highly conserved RNA splicing factor, P. falciparum (Pf)SR1, plays an unexpected and crucial role in DNA repair in malaria parasites. Using an inducible and reversible system to manipulate PfSR1 expression, we demonstrate that this protein is recruited to foci of DNA damage. Although loss of PfSR1 does not impair parasite viability, the protein is essential for their recovery from DNA-damaging agents or exposure to artemisinin, the first-line antimalarial drug, demonstrating its necessity for DNA repair. These findings provide key insights into the evolution of DNA repair pathways in malaria parasites as well as the ability of the parasite to recover from antimalarial treatment.


Assuntos
Malária Falciparum , Malária , Parasitos , Animais , Reparo do DNA/genética , Humanos , Malária Falciparum/tratamento farmacológico , Malária Falciparum/genética , Plasmodium falciparum/genética , Proteínas de Protozoários/genética
4.
J Clin Invest ; 124(5): 2071-5, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24905461

RESUMO

The transcription factor steroidogenic factor 1 (SF-1; also known as NR5A1) is a crucial mediator of both steroidogenic and nonsteroidogenic tissue differentiation. Mutations within SF1 underlie different disorders of sexual development (DSD), including sex reversal, spermatogenic failure, ovarian insufficiency, and adrenocortical deficiency. Here, we identified a recessive mutation within SF1 that resulted in a substitution of arginine to glutamine at codon 103 (R103Q) in a child with both severe 46,XY-DSD and asplenia. The R103Q mutation decreased SF-1 transactivation of TLX1, a transcription factor that has been shown to be essential for murine spleen development. Additionally, the SF1 R103Q mutation impaired activation of steroidogenic genes, without affecting synergistic SF-1 and sex-determining region Y (SRY) coactivation of the testis development gene SOX9. Together, our data provide evidence that SF-1 is required for spleen development in humans via transactivation of TLX1 and that mutations that only impair steroidogenesis, without altering the SF1/SRY transactivation of SOX9, can lead to 46,XY-DSD.


Assuntos
Proteínas de Homeodomínio/biossíntese , Proteínas Proto-Oncogênicas/biossíntese , Baço/crescimento & desenvolvimento , Fator Esteroidogênico 1/metabolismo , Ativação Transcricional/fisiologia , Substituição de Aminoácidos , Animais , Células CHO , Células COS , Chlorocebus aethiops , Códon/genética , Códon/metabolismo , Cricetinae , Cricetulus , Células HEK293 , Síndrome de Heterotaxia/genética , Síndrome de Heterotaxia/metabolismo , Síndrome de Heterotaxia/patologia , Proteínas de Homeodomínio/genética , Humanos , Masculino , Camundongos , Mutação de Sentido Incorreto , Proteínas Proto-Oncogênicas/genética , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/metabolismo , Proteína da Região Y Determinante do Sexo/genética , Proteína da Região Y Determinante do Sexo/metabolismo , Baço/metabolismo , Fator Esteroidogênico 1/genética
5.
Proc Natl Acad Sci U S A ; 109(19): 7403-8, 2012 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-22529396

RESUMO

The Prader-Willi syndrome/Angelman syndrome (PWS/AS) imprinted domain is regulated by a bipartite imprinting control center (IC) composed of a sequence around the SNRPN promoter (PWS-IC) and a 880-bp sequence located 35 kb upstream (AS-IC). The AS-IC imprint is established during gametogenesis and confers repression upon PWS-IC on the maternal allele. Mutation at PWS-IC on the paternal allele leads to gene silencing across the entire PWS/AS domain. This silencing implies that PWS-IC functions on the paternal allele as a bidirectional activator. Here we examine the mechanism by which PWS-IC activates the paternally expressed genes (PEGs) using transgenes that include the PWS-IC sequence in the presence or absence of AS-IC and NDN, an upstream PEG, as an experimental model. We demonstrate that PWS-IC is in fact an activator of NDN. This activation requires an unmethylated PWS-IC in the gametes and during early embryogenesis. PWS-IC is dispensable later in development. Interestingly, a similar activation of a nonimprinted gene (APOA1) was observed, implying that PWS-IC is a universal activator. To decipher the mechanism by which PWS-IC confers activation of remote genes, we performed methylated DNA immunoprecipitation (MeDIP) array analysis on lymphoblast cell lines that revealed dispersed, rather than continued differential methylation. However, chromatin conformation capture (3c) experiments revealed a physical interaction between PWS-IC and the PEGs, suggesting that activation of PEGs may require their proximity to PWS-IC.


Assuntos
Síndrome de Angelman/genética , Metilação de DNA , Impressão Genômica , Síndrome de Prader-Willi/genética , Animais , Apolipoproteína A-I/genética , Sequência de Bases , Sítios de Ligação/genética , Southern Blotting , Encéfalo/metabolismo , Pai , Feminino , Fibroblastos/metabolismo , Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Modelos Genéticos , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Regiões Promotoras Genéticas/genética , Espermatozoides/metabolismo , Proteínas Centrais de snRNP/genética
6.
Am J Hum Genet ; 89(4): 572-9, 2011 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-21963259

RESUMO

XX female gonadal dysgenesis (XX-GD) is a rare, genetically heterogeneous disorder characterized by lack of spontaneous pubertal development, primary amenorrhea, uterine hypoplasia, and hypergonadotropic hypogonadism as a result of streak gonads. Most cases are unexplained but thought to be autosomal recessive. We elucidated the genetic basis of XX-GD in a highly consanguineous Palestinian family by using homozygosity mapping and candidate-gene and whole-exome sequencing. Affected females were homozygous for a 3 bp deletion (NM_016556.2, c.600_602del) in the PSMC3IP gene, leading to deletion of a glutamic acid residue (p.Glu201del) in the highly conserved C-terminal acidic domain. Proteasome 26S subunit, ATPase, 3-Interacting Protein (PSMC3IP)/Tat Binding Protein Interacting Protein (TBPIP) is a nuclear, tissue-specific protein with multiple functions. It is critical for meiotic recombination as indicated by the known role of its yeast ortholog, Hop2. Through the C terminus (not present in yeast), PSMC3IP also coactivates ligand-driven transcription mediated by estrogen, androgen, glucocorticoid, progesterone, and thyroid nuclear receptors. In cell lines, the p.Glu201del mutation abolished PSMC3IP activation of estrogen-driven transcription. Impaired estrogenic signaling can lead to ovarian dysgenesis both by affecting the size of the follicular pool created during fetal development and by failing to counteract follicular atresia during puberty. PSMC3IP joins previous genes known to be mutated in XX-GD, the FSH receptor, and BMP15, highlighting the importance of hormonal signaling in ovarian development and maintenance and suggesting a common pathway perturbed in isolated XX-GD. By analogy to other XX-GD genes, PSMC3IP is also a candidate gene for premature ovarian failure, and its role in folliculogenesis should be further investigated.


Assuntos
Cromossomos Humanos X , Estrogênios/metabolismo , Disgenesia Gonadal/genética , Proteínas Nucleares/genética , Transativadores/genética , Consanguinidade , Feminino , Deleção de Genes , Marcadores Genéticos , Genótipo , Disgenesia Gonadal 46 XX/genética , Haplótipos , Perda Auditiva Neurossensorial/genética , Homozigoto , Humanos , Masculino , Linhagem , Complexo de Endopeptidases do Proteassoma/metabolismo , Transcrição Gênica
7.
Proc Natl Acad Sci U S A ; 106(25): 10242-7, 2009 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-19506242

RESUMO

Imprinting of the PWS/AS 2.4 Mb domain in the human is controlled by a paternally active imprinting center (PWS-IC). PWS-IC on the maternal allele is methylated and inactivated by an 880-bp sequence (AS-IC) located 30 kb upstream. In this communication, we report the identification of 7 cis acting elements within AS-IC. The elements: DMR, DNS, 2 OCTA sequences, SOX, E1, and E2 bind specific proteins that form at least 2 protein complexes. Using variants of an imprinted transgene, mutated at the elements each at a time, we show that (i) all 7 elements are involved in the methylation and inactivation of the maternal PWS-IC; (ii) the OCTA and SOX elements that bind a protein complex, and the E1 and E2 elements, function in establishing the primary imprint that constitutes an active and unmethylated AS-IC in the oocyte; (iii) DNS and DMR bind a multiprotein complex that may facilitate interaction between AS-IC and PWS-IC, mediating the inactivation in cis of PWS-IC; and (iv) all 7 elements participate in maintaining an unmethylated PWS-IC in the oocyte, which is essential for its maternal methylation later in development. Altogether, the above observations imply that the cis acting elements on AS-IC display diverse functions in establishing the imprints at both AS-IC and PWS-IC in the oocyte. A postulated epigenetic mark imprints the PWS-IC in the oocyte and maintains its inactive status during development before it is translated into maternal methylation.


Assuntos
Síndrome de Angelman/genética , Impressão Genômica , Complexos Multiproteicos/metabolismo , Oócitos/metabolismo , Síndrome de Prader-Willi/genética , Animais , Sequência de Bases , Metilação de DNA/genética , Ensaio de Desvio de Mobilidade Eletroforética , Feminino , Humanos , Camundongos , Camundongos Transgênicos , Ligação Proteica/genética
8.
Epigenetics ; 2(4): 214-22, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18075316

RESUMO

Disruptions in the expression of the BDNF gene that encodes a neurotrophic factor involved in neuronal survival, differentiation and synaptic plasticity has been proposed to contribute to the molecular pathogenesis of Rett syndrome. Rett syndrome (RTT) is a neurodevelopmental disorder, caused by mutations in the X-linked methyl CpG binding protein 2 gene (MeCP2). MeCP2 deficiency in the brain has been shown to decrease overall expression of BDNF in spite of an observed increase in the activity of promoter III that appears to be controlled directly by MeCP2. Therefore, how MeCP2 deficiency causes an overall downregulation of BDNF expression was an enigma. Here we report that MeCP2 deficiency in human and mouse brain causes an increase in expression of two neuronal gene transcriptional repressors REST (RE1 silencing transcription factor), and CoREST. MeCP2 binds to and is involved in repression of Rest and CoRest promoters despite their unmethylated state. MeCP2 depletion is associated with a change in the histone modification profile to a more active conformation. The elevated levels of REST and CoREST in the brain of RTT patients and MeCP2 deficient mice result in downregulation of BDNF, apparently by their binding to the RE1 (element) located between the first two promoters of the BDNF gene. Interestingly, the NTRK2 gene that encodes the BDNF receptor, TRKB, was overexpressed in MeCP2 deficient human and mouse brains either directly or as an attempt to compensate for BDNF deficiency.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Encéfalo/metabolismo , Proteínas de Ligação a DNA/fisiologia , Proteína 2 de Ligação a Metil-CpG/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Receptor trkB/biossíntese , Proteínas Repressoras/fisiologia , Animais , Sequência de Bases , Fator Neurotrófico Derivado do Encéfalo/genética , Imunoprecipitação da Cromatina , Proteínas Correpressoras , Metilação de DNA , Primers do DNA , Feminino , Humanos , Masculino , Proteína 2 de Ligação a Metil-CpG/genética , Camundongos , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas , Reação em Cadeia da Polimerase Via Transcriptase Reversa
9.
Blood ; 107(8): 3288-94, 2006 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-16368880

RESUMO

The reduced folate carrier (RFC) is the dominant route for the uptake of various antifolates including PT523, a potent dihydrofolate reductase inhibitor (Ki = 0.35 pM) and an excellent transport substrate of the RFC (Kt = 0.7 microM). Here, we describe the multiple mechanisms of RFC inactivation in human leukemia PT523-resistant cells originally harboring 3 RFC alleles. Cellular exposure to gradually increasing PT523 concentrations resulted in sublines displaying up to 3500-fold resistance to various hydrophilic antifolates that rely on RFC for their cellular uptake. Antifolate-resistant cells lost RFC gene expression (65%-99% loss) due to impaired promoter binding of various transcription factors that regulate RFC gene expression. Additionally, DNA sequencing revealed that PT523-resistant cells contained a cluster of 4 nearly consecutive mutations residing on a single RFC allele including L143P, A147V, R148G, and Q150Stop. Southern blot analysis established the loss of an RFC allele in PT523-resistant cells. These alterations resulted in markedly decreased RFC protein levels (approximately 80%-99% loss) and consequently impaired [3H]methotrexate transport (87%-99% loss). This study provides the first evidence that acquisition of PT523 resistance in human leukemia cells harboring 3 RFC alleles is due to multiple coexisting alterations including transcriptional silencing, inactivating mutations, and RFC allele loss.


Assuntos
Alelos , Resistencia a Medicamentos Antineoplásicos/genética , Inativação Gênica , Leucemia/genética , Perda de Heterozigosidade/genética , Proteínas de Membrana Transportadoras/genética , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/genética , Linhagem Celular Tumoral , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Regulação Leucêmica da Expressão Gênica/genética , Inativação Gênica/efeitos dos fármacos , Humanos , Leucemia/tratamento farmacológico , Leucemia/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Metotrexato/farmacologia , Ornitina/análogos & derivados , Ornitina/farmacologia , Ornitina/uso terapêutico , Pterinas/farmacologia , Pterinas/uso terapêutico , Proteína Carregadora de Folato Reduzido , Elementos de Resposta/efeitos dos fármacos , Elementos de Resposta/genética , Tetra-Hidrofolato Desidrogenase/genética , Tetra-Hidrofolato Desidrogenase/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
10.
Hum Mol Genet ; 14(8): 1049-58, 2005 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15757975

RESUMO

Rett syndrome (RS) is a severe and progressive neurodevelopmental disorder caused by heterozygous mutations in the X-linked methyl CpG binding protein 2 (MeCP2) gene. MeCP2 is a nuclear protein that binds specifically to methylated DNA and functions as a general transcription repressor in the context of chromatin remodeling complexes. RS shares clinical features with those of Angelman syndrome (AS), an imprinting neurodevelopmental disorder. In AS patients, the maternally expressed copy of UBE3A that codes for the ubiquitin protein ligase 3A (E6-AP) is repressed. The similar phenotype of these two syndromes led us to hypothesize that part of the RS phenotype is due to MeCP2-associated silencing of UBE3A. Indeed, UBE3A mRNA and protein are shown here to be significantly reduced in human and mouse MECP2 deficient brains. This reduced UBE3A level was associated with biallelic production of the UBE3A antisense RNA. In addition, MeCP2 deficiency resulted in elevated histone H3 acetylation and H3(K4) methylation and reduced H3(K9) methylation at the PWS/AS imprinting center, with no effect on DNA methylation or SNRPN expression. We conclude, therefore, that MeCP2 deficiency causes epigenetic aberrations at the PWS imprinting center. These changes in histone modifications result in loss of imprinting of the UBE3A antisense gene in the brain, increase in UBE3A antisense RNA level and, consequently reduction in UBE3A production.


Assuntos
Proteínas Cromossômicas não Histona/deficiência , Proteínas de Ligação a DNA/deficiência , Epigênese Genética , Impressão Genômica , Síndrome de Rett/metabolismo , Ubiquitina-Proteína Ligases/genética , Síndrome de Angelman/genética , Síndrome de Angelman/metabolismo , Animais , Encéfalo/metabolismo , Proteínas Cromossômicas não Histona/genética , Proteínas de Ligação a DNA/genética , Humanos , Proteína 2 de Ligação a Metil-CpG , Camundongos , Síndrome de Prader-Willi/genética , Síndrome de Prader-Willi/metabolismo , Regiões Promotoras Genéticas , Proteínas Repressoras/genética , Síndrome de Rett/genética , Ubiquitina-Proteína Ligases/biossíntese
11.
Hum Mol Genet ; 13(22): 2767-79, 2004 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-15367489

RESUMO

The Prader-Willi/Angelman imprinted domain on human chromosome 15q11-q13 is regulated by an imprinting control center (IC) composed of a sequence around the SNRPN promoter (PWS-SRO) and a sequence located 35 kb upstream (AS-SRO). We have previously hypothesized that the primary imprint is established on AS-SRO, which then confers imprinting on PWS-SRO. Here we examine this hypothesis using a transgene that includes both AS-SRO and PWS-SRO sequences and carries out the entire imprinting process. The epigenetic features of this transgene resemble those previously observed on the endogenous locus, thus allowing analyses in the gametes and early embryo. We demonstrate that the primary imprint is in fact established in the gametes, creating a differentially methylated CpG cluster (DMR) on AS-SRO, presumably by an adjacent de novo signal (DNS). The DMR and DNS bind specific proteins: an allele-discrimination protein (ADP) and a de novo methylation protein, respectively. ADP, being a maternal protein, is involved in both the establishment of DMR in the gametes and in its maintenance through implantation when methylation of PWS-SRO on the maternal allele takes place. Importantly, while the AS-SRO is required in the gametes to confer methylation on PWS-SRO, it is dispensable later in development.


Assuntos
Síndrome de Angelman/genética , Embrião de Mamíferos/metabolismo , Epigênese Genética , Impressão Genômica , Células Germinativas/metabolismo , Síndrome de Prader-Willi/genética , Regiões Promotoras Genéticas , Animais , Sequência de Bases , Blastocisto/metabolismo , Imunoprecipitação da Cromatina , Ilhas de CpG , Metilação de DNA , Desenvolvimento Embrionário , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
12.
Cancer ; 100(4): 773-82, 2004 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-14770434

RESUMO

BACKGROUND: Although the majority of children with acute lymphoblastic leukemia (ALL) are cured with combination chemotherapy containing methotrexate (MTX), drug resistance contributes to treatment failure for a substantial fraction of patients. The primary transporter for folates and MTX is the reduced folate carrier (RFC). Impaired drug transport is a documented mechanism of MTX resistance in patients with ALL; however, to the authors' knowledge it is not known whether inactivating RFC mutations are a contributing factor. METHODS: The authors devised a genomic polymerase chain reaction-single strand conformational polymorphism assay followed by sequencing and screened the entire RFC coding region for sequence alterations in DNA from 246 leukemia specimens from patients with diverse ethnic variation, 24 at the time of recurrence and the rest at the time of diagnosis. This cohort was comprised of 203 B-precursor ALL specimens (82.5%), 32 T-lineage ALL specimens (13%), and 11 acute myeloblastic leukemia specimens (4.5%). RESULTS: Of 246 DNA samples, only 3 diagnosis B-precursor ALL specimens (1.2%) were found to harbor alterations in the RFC gene, including heterozygous single nucleotide changes resulting in D56H and D522N substitutions in the first extracellular loop and the C-terminus of this transporter, respectively. The third sample had a sequence alteration in exon 3 that could not be identified because of the lack of availability of DNA. CONCLUSIONS: Whereas inactivating RFC mutations are a frequent mechanism of MTX resistance in human leukemia cell lines and in patients with osteosarcoma, they are not common and do not appear to play any significant role in intrinsic or acquired resistance to MTX in childhood leukemia. This is the first study of RFC mutations in multiple pediatric leukemia specimens.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Proteínas de Transporte/genética , Proteínas de Transporte/farmacologia , Proteínas de Membrana Transportadoras , Metotrexato/farmacologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Northern Blotting , Criança , Análise Mutacional de DNA , Resistencia a Medicamentos Antineoplásicos , Humanos , Osteossarcoma/tratamento farmacológico , Osteossarcoma/genética , Reação em Cadeia da Polimerase , Polimorfismo Genético , Polimorfismo Conformacional de Fita Simples , Proteína Carregadora de Folato Reduzido , Células Tumorais Cultivadas
13.
J Biol Chem ; 279(1): 374-84, 2004 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-14551190

RESUMO

The human reduced folate carrier (hRFC) is the major uptake route for antifolates used in cancer chemotherapy. Here we explored the molecular basis for the decrease or loss of hRFC gene expression in seventeen tumor cell lines with resistance to multiple antifolates due to impaired antifolate transport. We studied the role of various cis-acting elements including CRE/AP-1-like element and GC-box in hRFC promoters A and B, respectively, as well as AP-2, Mzf-1 and E-box that are contained within or near four tandemly repeated sequences upstream of promoter A. Decreased or abolished binding either to [32P]GC-box, Mzf-1, AP-1, E-box, or CRE oligonucleotides was detected in approximately 50-80% of antifolate-resistant cell lines. Strikingly, approximately 80% of the cell lines displayed a simultaneously decreased binding to three or more of these hRFC promoter elements, whereas normal AP-2 binding was retained. The possible contribution of promoter methylation to hRFC gene silencing was also explored. None of the antifolate-resistant cell lines, except for MDA-MB-231 cells, showed hRFC promoter methylation; consistently, MDA-MB-231 was the only cell line that retained binding to all six cis-acting elements. Western blot analysis demonstrated decreased expression of transcriptional activators (pCREB-1, pATF-1, USF-1, c-Fos, c-Jun, Sp1, and Sp3) and/or increased expression of repressors (short Sp3 isoforms), whereas normal AP2alpha levels were retained. Transient expression of the relevant transcription factors restored, at least partially, both promoter binding and hRFC gene expression. This is the first report that transcriptional silencing of the hRFC gene in multiple tumor cell lines with resistance to various novel antifolates is a result of a simultaneous loss of function of multiple transcription factors but not promoter methylation.


Assuntos
Proteínas de Transporte/genética , Metilação de DNA , Inativação Gênica , Proteínas de Membrana Transportadoras , Mutação , Regiões Promotoras Genéticas , Receptores de Superfície Celular , Fatores de Transcrição/metabolismo , Transcrição Gênica , Substituição de Aminoácidos , Sequência de Bases , Linhagem Celular , Códon de Terminação , Fosfatos de Dinucleosídeos/genética , Receptores de Folato com Âncoras de GPI , Humanos , Leucemia de Células T , Mutação de Sentido Incorreto , Oligodesoxirribonucleotídeos , Proteína Carregadora de Folato Reduzido , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas
14.
Biochem J ; 367(Pt 3): 741-50, 2002 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-12139489

RESUMO

We have studied the molecular basis of resistance of multiple human leukaemia CCRF-CEM sublines to the novel antifolates ZD9331, GW1843, AG2034, PT523 and edatrexate, which use the reduced folate carrier (RFC) as their main cellular uptake route and that target different folate-dependent enzymes. Antifolate-resistant sublines established by stepwise and single-step selections displayed up to 2135-fold resistance to the selection drug, and up to 2323-fold cross-resistance to various hydrophilic antifolates. In contrast, these sublines were up to 17- and 20-fold hypersensitive to the lipophilic antifolates AG377 and trimetrexate, respectively. The total reduced folate pool of these antifolate-resistant sublines shrunk by 87-96%, resulting in up to 42-fold increased folic acid growth requirement. These sublines lost 92-97% of parental [(3)H]methotrexate influx rates. Genomic PCR single-strand conformational polymorphism analysis and sequencing revealed that most of these drug-resistant sublines harboured RFC mutations that surprisingly clustered in two confined regions in exons 2 and 3. The majority of these mutations resulted in frame-shift and/or premature translation termination and lack of RFC protein expression. The remaining mutations involved single amino acid substitutions predominantly residing in the first transmembrane domain (TMD1). Some RFC-inactivating mutations emerged during the early stages of antifolate selection and were stably retained during further drug selection. Furthermore, some sublines displayed a markedly decreased or abolished RFC mRNA and/or protein expression. This constitutes the first demonstration of clustering of multiple human RFC mutations in TMD1, thereby suggesting that it plays a functional role in folate/antifolate binding and/or translocation. This is the first molecular characterization of human RFC-associated modalities of resistance to various novel antifolates in multiple leukaemia sublines.


Assuntos
Proteínas de Transporte/genética , Resistencia a Medicamentos Antineoplásicos/genética , Antagonistas do Ácido Fólico/farmacologia , Leucemia/metabolismo , Proteínas de Membrana Transportadoras , Mutação , Sequência de Bases , Western Blotting , Primers do DNA , Antagonistas do Ácido Fólico/farmacocinética , Humanos , Leucemia/patologia , Metotrexato/farmacocinética , Metotrexato/farmacologia , Polimorfismo Conformacional de Fita Simples , Proteína Carregadora de Folato Reduzido , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA