Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Leukemia ; 31(2): 382-392, 2017 02.
Article in English | MEDLINE | ID: mdl-27479184

ABSTRACT

The notion that plasma cells (PCs) are terminally differentiated has prevented intensive research in multiple myeloma (MM) about their phenotypic plasticity and differentiation. Here, we demonstrated in healthy individuals (n=20) that the CD19-CD81 expression axis identifies three bone marrow (BM)PC subsets with distinct age-prevalence, proliferation, replication-history, immunoglobulin-production, and phenotype, consistent with progressively increased differentiation from CD19+CD81+ into CD19-CD81+ and CD19-CD81- BMPCs. Afterwards, we demonstrated in 225 newly diagnosed MM patients that, comparing to normal BMPC counterparts, 59% had fully differentiated (CD19-CD81-) clones, 38% intermediate-differentiated (CD19-CD81+) and 3% less-differentiated (CD19+CD81+) clones. The latter patients had dismal outcome, and PC differentiation emerged as an independent prognostic marker for progression-free (HR: 1.7; P=0.005) and overall survival (HR: 2.1; P=0.006). Longitudinal comparison of diagnostic vs minimal-residual-disease samples (n=40) unraveled that in 20% of patients, less-differentiated PCs subclones become enriched after therapy-induced pressure. We also revealed that CD81 expression is epigenetically regulated, that less-differentiated clonal PCs retain high expression of genes related to preceding B-cell stages (for example: PAX5), and show distinct mutation profile vs fully differentiated PC clones within individual patients. Together, we shed new light into PC plasticity and demonstrated that MM patients harbouring less-differentiated PCs have dismal survival, which might be related to higher chemoresistant potential plus different molecular and genomic profiles.


Subject(s)
Multiple Myeloma/diagnosis , Multiple Myeloma/metabolism , Plasma Cells/metabolism , Plasma Cells/pathology , Adult , Antigens, CD/metabolism , Biomarkers , Bone Marrow/metabolism , Bone Marrow/pathology , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Case-Control Studies , Cell Cycle , DNA Methylation , Female , Gene Expression Profiling , Genetic Heterogeneity , High-Throughput Nucleotide Sequencing , Humans , Immunophenotyping , In Situ Hybridization, Fluorescence , Male , Middle Aged , Multiple Myeloma/genetics , Multiple Myeloma/mortality , Mutation , Neoplasm Grading , Phenotype , Prognosis , Single-Cell Analysis , Young Adult
3.
Leukemia ; 27(4): 925-31, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23174883

ABSTRACT

Currently, multiple myeloma (MM) patients are broadly grouped into a non-hyperdiploid (nh-MM) group, highly enriched for IgH translocations, or into a hyperdiploid (h-MM) group, which is typically characterized by trisomies of some odd-numbered chromosomes. We compared the micro RNA (miRNA) expression profiles of these two groups and we identified 16 miRNAs that were downregulated in the h-MM group, relative to the nh-MM group. We found that target genes of the most differentially expressed miRNAs are directly involved in the pathogenesis of MM; specifically, the inhibition of hsa-miR-425, hsa-miR-152 and hsa-miR-24, which are all downregulated in h-MM, leads to the overexpression of CCND1, TACC3, MAFB, FGFR3 and MYC, which are the also the oncogenes upregulated by the most frequent IgH chromosomal translocations occurring in nh-MM. Importantly, we showed that the downregulation of these specific miRNAs and the upregulation of their targets also occur simultaneously in primary cases of h-MM. These data provide further evidence on the unifying role of cyclin D pathways deregulation as the key mechanism involved in the development of both groups of MM. Finally, they establish the importance of miRNA deregulation in the context of MM, thereby opening up the potential for future therapeutic approaches based on this molecular mechanism.


Subject(s)
Diploidy , Down-Regulation , Immunoglobulin Heavy Chains/genetics , MicroRNAs/genetics , Multiple Myeloma/genetics , Translocation, Genetic , Base Sequence , Blotting, Western , DNA Methylation , DNA Primers , Humans , Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
4.
Leukemia ; 26(9): 2052-60, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22522792

ABSTRACT

Apart from microRNAs, little is known about the regulation of expression of non-coding RNAs in cancer. We investigated whether small nucleolar RNAs (snoRNAs) accumulation displayed specific signatures in acute myeloblastic and acute lymphoblastic leukemias. Using microarrays and high-throughput quantitative PCR (qPCR), we demonstrate here that snoRNA expression patterns are negatively altered in leukemic cells compared with controls. Interestingly, a specific signature was found in acute promyelocytic leukemia (APL) with ectopic expression of SNORD112-114 snoRNAs located at the DLK1-DIO3 locus. In vitro experiments carried out on APL blasts demonstrate that transcription of these snoRNAs was lost under all-trans retinoic acid-mediated differentiation and induced by enforced expression of the PML-RARalpha fusion protein in negative leukemic cell lines. Further experiments revealed that the SNORD114-1 (14q(II-1)) variant promoted cell growth through cell cycle modulation; its expression was implicated in the G0/G1 to S phase transition mediated by the Rb/p16 pathways. This study thus reports three important observations: (1) snoRNA regulation is different in normal cells compared with cancer cells; (2) a relationship exists between a chromosomal translocation and expression of snoRNA loci; and (3) snoRNA expression can affect Rb/p16 cell cycle regulation. Taken together, these data strongly suggest that snoRNAs have a role in cancer development.


Subject(s)
Biomarkers, Tumor/genetics , Gene Expression Profiling , Leukemia, Myeloid, Acute/genetics , Leukemia, Promyelocytic, Acute/genetics , RNA, Small Nucleolar/genetics , Biomarkers, Tumor/metabolism , Blast Crisis , Blotting, Western , Calcium-Binding Proteins , Cell Cycle , Cell Differentiation , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Leukemia, Myeloid, Acute/metabolism , Leukemia, Promyelocytic, Acute/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , RNA, Messenger/genetics , RNA, Small Nucleolar/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
6.
Leukemia ; 26(7): 1517-26, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22307227

ABSTRACT

Histone deacetylases (HDACs) have been identified as therapeutic targets due to their regulatory function in chromatin structure and organization. Here, we analyzed the therapeutic effect of LBH589, a class I-II HDAC inhibitor, in acute lymphoblastic leukemia (ALL). In vitro, LBH589 induced dose-dependent antiproliferative and apoptotic effects, which were associated with increased H3 and H4 histone acetylation. Intravenous administration of LBH589 in immunodeficient BALB/c-RAG2(-/-)γc(-/-) mice in which human-derived T and B-ALL cell lines were injected induced a significant reduction in tumor growth. Using primary ALL cells, a xenograft model of human leukemia in BALB/c-RAG2(-/-)γc(-/-) mice was established, allowing continuous passages of transplanted cells to several mouse generations. Treatment of mice engrafted with T or B-ALL cells with LBH589 induced an in vivo increase in the acetylation of H3 and H4, which was accompanied with prolonged survival of LBH589-treated mice in comparison with those receiving vincristine and dexamethasone. Notably, the therapeutic efficacy of LBH589 was significantly enhanced in combination with vincristine and dexamethasone. Our results show the therapeutic activity of LBH589 in combination with standard chemotherapy in pre-clinical models of ALL and suggest that this combination may be of clinical value in the treatment of patients with ALL.


Subject(s)
Apoptosis/drug effects , Dexamethasone/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Vincristine/pharmacology , Animals , Antineoplastic Agents, Hormonal/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Combined Chemotherapy Protocols , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Proliferation/drug effects , DNA Methylation , DNA-Binding Proteins/physiology , Drug Synergism , Female , Gene Expression Profiling , Histones/metabolism , Humans , Immunoenzyme Techniques , Indoles , Mice , Mice, Inbred BALB C , Oligonucleotide Array Sequence Analysis , Panobinostat , Polymorphism, Single Nucleotide/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
Leukemia ; 26(3): 395-403, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22143672

ABSTRACT

MicroRNAs (miRNAs) are small non-coding RNA molecules that can negatively regulate gene expression at the post-transcriptional level. miRNA expression patterns are regulated during development and differentiation of the hematopoietic system and have an important role in cell processes such as proliferation, apoptosis, differentiation or even in tumorigenesis of human tumors and in particular of hematological malignancies such as acute leukemias. Various miRNAs and their functions have been intensively studied in acute leukemias but the mechanisms that control their expression are largely unknown for the majority of aberrantly expressed miRNAs. miRNA expression can be regulated by the same genetic mechanism that modulate protein coding genes such as mutation, deletion, amplification, loss of heterozygosity and translocations. In this review we focus on the regulation of miRNAs in acute leukemias mediated by alterations in epigenetic mechanisms such as DNA methylation and histone code, describing the role of these alterations in the pathogenesis, diagnosis and prognosis of acute leukemias and their possible use as new therapeutic targets and biomarkers.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation, Leukemic , Leukemia/genetics , MicroRNAs/genetics , Humans , Leukemia, Lymphoid/genetics , Leukemia, Myeloid, Acute/genetics
8.
J Chromatogr B Analyt Technol Biomed Life Sci ; 879(30): 3490-6, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21983199

ABSTRACT

An ultra high performance liquid chromatography tandem mass spectrometry method (UHPLC-MS/MS) was developed and validated for the quantitation of LBH589, a novel histone deacetylase inhibitor (HDACi), in mouse plasma and tissues (liver, spleen, kidney and lung). Tobramycin was employed as the internal standard. Separation was performed on an Acquity UPLC™ BEH column, with a mobile phase consisting of 10% water (with 0.1% of trifluoroacetic acid) and 90% methanol (with 0.1% trifluoroacetic acid). LBH589 and tobramycin were determined using an electrospray ionization (ESI) interface. Detection was performed on electrospray positive ionization mass spectrometry by multiple reaction monitoring of the transitions of LBH589 at m/z 349.42→157.95 and of tobramycin at 468.2→163. Calibration curves for the UHPLC method (0.0025-1 µg/mL for plasma and tissue homogenates, equivalent to 0.0357-14.2857 µg/g for tissue samples) showed a linear range of detector responses (r>0.998). Intra-batch and inter-batch precision expressed as coefficient of variation (CV) ranged from 0.92 to 8.40%. Accuracy expressed as bias, ranged from -2.41 to 2.62%. The lower limit of quantitation (LLOQ) was 0.0025 µg/mL for both plasma and tissue homogenate samples, equivalent to 0.0357 µg/g tissue. This method was successfully applied to quantify LBH589 in plasma and tissue samples obtained after the intraperitoneal administration of a single dose of 20 mg/kg of LBH589 in BALB/c mice.


Subject(s)
Chromatography, High Pressure Liquid/methods , Hydroxamic Acids/analysis , Spectrometry, Mass, Electrospray Ionization/methods , Tandem Mass Spectrometry/methods , Animals , Drug Stability , Hydroxamic Acids/administration & dosage , Hydroxamic Acids/blood , Hydroxamic Acids/pharmacokinetics , Indoles , Injections, Intraperitoneal , Linear Models , Mice , Mice, Inbred BALB C , Panobinostat , Reproducibility of Results , Sensitivity and Specificity , Tissue Distribution , Tobramycin/analysis
9.
Leukemia ; 25(12): 1882-90, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21778999

ABSTRACT

The anaplastic lymphoma kinase (ALK), tyrosine kinase oncogene is implicated in a wide variety of cancers. In this study we used conditional onco-ALK (NPM-ALK and TPM3-ALK) mouse MEF cell lines (ALK+ fibroblasts) and transgenic models (ALK+ B-lymphoma) to investigate the involvement and regulation of angiogenesis in ALK tumor development. First, we observed that ALK expression leads to downregulation of miR-16 and increased Vascular Endothelial Growth Factor (VEGF) levels. Second, we found that modification of miR-16 levels in TPM3-ALK MEF cells greatly affected VEGF levels. Third, we demonstrated that miR-16 directly interacts with VEGF mRNA at the 3'-untranslated region and that the regulation of VEGF by miR-16 occurs at the translational level. Fourth, we showed that expression of both the ALK oncogene and hypoxia-induced factor 1α (HIF1α) is a prerequisite for miR-16 downregulation. Fifth, in vivo, miR-16 gain resulted in reduced angiogenesis and tumor growth. Finally, we highlighted an inverse correlation between the levels of miR-16 and VEGF in human NPM-ALK+ Anaplastic Large Cell Lymphomas (ALCL). Altogether, our results demonstrate, for the first time, the involvement of angiogenesis in ALK+ ALCL and strongly suggest an important role for hypoxia-miR-16 in regulating VEGF translation.


Subject(s)
Gene Expression Regulation, Neoplastic , Hypoxia/metabolism , Lymphoma, Large-Cell, Anaplastic/genetics , Lymphoma, Large-Cell, Anaplastic/metabolism , MicroRNAs/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Vascular Endothelial Growth Factor A/metabolism , Anaplastic Lymphoma Kinase , Animals , Blotting, Northern , Blotting, Western , Case-Control Studies , Cell Adhesion , Cell Movement , Cells, Cultured , DNA Methylation , Down-Regulation , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Hypoxia/genetics , Hypoxia/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunoenzyme Techniques , Lymphoma, Large-Cell, Anaplastic/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/metabolism , Neovascularization, Pathologic , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptor Protein-Tyrosine Kinases/genetics , Vascular Endothelial Growth Factor A/genetics
11.
Oncogene ; 29(22): 3276-86, 2010 Jun 03.
Article in English | MEDLINE | ID: mdl-20305692

ABSTRACT

The use of c-abl-specific inhibitors such as Imatinib (IM) or Dasatinib has revolutionized the treatment of chronic myeloid leukemia (CML). However, a significant percentage of patients become resistant to IM. In this report, we have analyzed the possibility of using the proteasome as a molecular target in CML. Our results show that cells that express Bcr-Abl1 are more sensitive to the inhibition of the proteasome with Bortezomib (Btz) than control cells. This treatment reduces the proliferation of Bcr-Abl1-expressing cells, by inactivating NF-kappaB2 and decreasing the phosphorylation of Rb, eventually leading to an increase in caspase-dependent apoptosis. Furthermore, we show that Btz also induces cell-cycle arrest and apoptosis in cells expressing Bcr-Abl1 mutants that are resistant to IM. These results unravel a new molecular target of Btz, that is the Rb pathway, and open new possibilities in the treatment of CML especially for patients that become resistant to IM because of the presence of the T315I mutation.


Subject(s)
Apoptosis/drug effects , Boronic Acids/pharmacology , Caspases/metabolism , Fusion Proteins, bcr-abl/biosynthesis , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Piperazines/pharmacology , Pyrazines/pharmacology , Pyrimidines/pharmacology , Retinoblastoma Protein/metabolism , Antineoplastic Agents/pharmacology , Benzamides , Bortezomib , Cell Growth Processes/drug effects , Cell Line, Tumor , Electrophoretic Mobility Shift Assay , Flow Cytometry , Fusion Proteins, bcr-abl/antagonists & inhibitors , Fusion Proteins, bcr-abl/genetics , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Phosphorylation/drug effects
12.
Mol Cancer ; 5: 29, 2006 Jul 19.
Article in English | MEDLINE | ID: mdl-16854228

ABSTRACT

MicroRNAs (miRNAs) are short non-coding RNA molecules playing regulatory roles by repressing translation or cleaving RNA transcripts. Although the number of verified human miRNA is still expanding, only few have been functionally described. However, emerging evidences suggest the potential involvement of altered regulation of miRNA in pathogenesis of cancers and these genes are thought to function as both tumours suppressor and oncogenes. In our study, we examined by Real-Time PCR the expression of 156 mature miRNA in colorectal cancer. The analysis by several bioinformatics algorithms of colorectal tumours and adjacent non-neoplastic tissues from patients and colorectal cancer cell lines allowed identifying a group of 13 miRNA whose expression is significantly altered in this tumor. The most significantly deregulated miRNA being miR-31, miR-96, miR-133b, miR-135b, miR-145, and miR-183. In addition, the expression level of miR-31 was correlated with the stage of CRC tumor. Our results suggest that miRNA expression profile could have relevance to the biological and clinical behavior of colorectal neoplasia.


Subject(s)
Colon/metabolism , Colorectal Neoplasms/metabolism , MicroRNAs/metabolism , Polymerase Chain Reaction/methods , Cluster Analysis , Colon/cytology , Computer Systems , Gene Expression Profiling , Humans , Matched-Pair Analysis , Tumor Cells, Cultured
14.
Oncogene ; 25(13): 1862-70, 2006 Mar 23.
Article in English | MEDLINE | ID: mdl-16314841

ABSTRACT

We have analyzed the regulation and expression of ASPP members, genes implicated in the regulation of the apoptotic function of the TP53 tumor-suppressor gene, in acute lymphoblastic leukemia (ALL). Expression of ASPP1 was significantly reduced in ALL and was dependent on hypermethylation of the ASPP1 gene promoter. Abnormal ASPP1 expression was associated with normal function of the tumor-suppressor gene TP53 in ALL. The analyses of 180 patients with ALL at diagnosis showed that the ASPP1 promoter was hypermethylated in 25% of cases with decreased mRNA expression. Methylation was significantly higher in adult ALL vs childhood ALL (32 vs 17%, P = 0.03) and T-ALL vs B-ALL (50 vs 9%, P = 0.001). Relapse rate (62 vs 44%, P = 0.05) and mortality (59 vs 43%, P = 0.05) were significantly higher in patients with methylated ASPP1. DFS and OS were 32.8 and 33.7% for patients with unmethylated ASPP1 and 6.1 and 9.9% for methylated patients (P < 0.001 y P < 0.02, respectively). On the multivariate analysis, methylation of the ASPP1 gene promoter was an independent poor prognosis factor in ALL patients. Our results demonstrate that decreased expression of ASPP1 in patients with ALL is due to an abnormal methylation of its promoter and is associated with a poor prognosis.


Subject(s)
Carrier Proteins/biosynthesis , Carrier Proteins/genetics , DNA Methylation , Gene Expression Regulation, Neoplastic , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Adaptor Proteins, Signal Transducing , Adolescent , Adult , Aged , Aged, 80 and over , Apoptosis Regulatory Proteins , Child , Child, Preschool , Female , Gene Expression Profiling , Genes, p53 , Humans , Infant , Male , Middle Aged , Precursor Cell Lymphoblastic Leukemia-Lymphoma/physiopathology , Prognosis , Promoter Regions, Genetic , Recurrence , Survival
17.
Br J Cancer ; 91(4): 707-13, 2004 Aug 16.
Article in English | MEDLINE | ID: mdl-15226763

ABSTRACT

DKK-3: is a newly characterised mortalisation-related gene and an antagonist of the Wnt oncogenic signalling pathway whose expression is decreased in a variety of cancer cell lines, suggesting that the Dkk-3 gene, located at chromosome 11p15.1, functions as a tumour suppressor gene. Although 11p15 is a 'hot spot' for methylation in acute lymphoblastic leukaemia (ALL), the role of Dkk-3 abnormalities has never been evaluated in this disease. We analysed CpG island methylation of the Dkk-3 promoter in six ALL cell lines and 183 ALL patients. We observed Dkk-3 hypermethylation in all cell lines and in cells from 33% (60/183) of ALL patients. Moreover, Dkk-3 methylation was associated with decreased Dkk-3 mRNA expression and this expression was restored after exposure to the demethylating agent 5-AzaC. Clinical features did not differ between hypermethylated and unmethylated patients. Estimated disease-free survival (DFS) and overall survival at 10 and 11 years, respectively, were 49.8 and 45.6% for normal patients and 10.5 and 15.1% for hypermethylated patients (P=0.001 and 0.09). Multivariate analysis demonstrated that Dkk-3 methylation was an independent prognostic factor predicting DFS (P=0.0009). Our data suggest that Dkk-3 methylation occurs at an early stage in ALL pathogenesis and probably influences the clinical behaviour of the disease.


Subject(s)
Chromosomes, Human, Pair 11/genetics , DNA Methylation , Gene Silencing , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Protein Biosynthesis , Proteins/genetics , Adaptor Proteins, Signal Transducing , Adolescent , Adult , Aged , Aged, 80 and over , Cell Transformation, Neoplastic , Chemokines , Child , Child, Preschool , CpG Islands , DNA, Neoplasm/metabolism , Female , Humans , Infant , Intercellular Signaling Peptides and Proteins , Male , Middle Aged , Multivariate Analysis , Prognosis , Promoter Regions, Genetic , Survival Analysis , Transcription, Genetic , Tumor Cells, Cultured
19.
Biotechniques ; 32(5): 1064-5, 1068-70, 2002 May.
Article in English | MEDLINE | ID: mdl-12019779

ABSTRACT

For genes that have a substantial number of exons and long intronic sequences, mutation screening by denaturing gradient gel electrophoresis (DGGE) requires the amplification of each exon from genomic DNA by PCR. This results in a high number of fragments to be analyzed by DGGE so that the analysis of large sample sets becomes labor intensive and time consuming. To address this problem, we have developed a new strategy for mutation analysis, lexon-DGGE, which combines the joining of different exons by PCR (also known as lexons) with a highly sensitive technique such as DGGE to screen for mutations. The lexon technique is based on the concatenation of several exons, adjacent or not, from genomic DNA into a single DNA fragment so that this approach could simultaneously be used to check the mutational status of several small genes. To show the feasibility of the approach, we have used the lexon-DGGE technique to analyze all coding exons, intron-exon junctions, noncoding exon 1, and part of the noncoding region of exon 11 of the TP53 gene. The validity and performance of the technique were confirmed by using negative and positive controls for each of the DNAfragments analyzed.


Subject(s)
Exons/genetics , Genetic Testing/methods , Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , DNA Mutational Analysis/methods , DNA, Neoplasm/analysis , Electrophoresis/methods , Humans , Introns/genetics , Mutation/genetics , Nucleic Acid Denaturation
20.
An. sist. sanit. Navar ; 23(3): 451-465, sept. 2000. ilus, tab
Article in Es | IBECS | ID: ibc-32317

ABSTRACT

La mejora de las técnicas de citogenética convencional, el desarrollo de la citogenética molecular y la aplicación de técnicas de biología molecular al análisis genético ha conducido a una verdadera revolución en el conocimiento de los procesos implicados en el desarrollo y progresión de las neoplasias linfoides. De esta manera, se han caracterizado gran parte de las alteraciones presentes en las células malignas estableciendo cuáles son los genes implicados en el proceso transformativo. Esto tiene importantes consecuencias en el manejo clínico de este tipo de enfermedades y permite un diagnóstico más exacto a través de una sistematización de las distintas entidades basada en sus características biológicas. Por otra parte, la introducción de nuevas técnicas de análisis, como la PCR en tiempo real, posibilitará la monitorización cuantitativa de la enfermedad permitiendo valorar la respuesta a los distintos tratamientos y estableciendo valores predictivos de recaídas. En el futuro, todo este conocimiento permitirá el establecimiento de terapias genotipo-específicas y el desarrollo de nuevos fármacos dirigidos a la alteración causante del proceso maligno y con menores efectos colaterales indeseables. (AU)


Subject(s)
Humans , Cytogenetic Analysis , Lymphoma/genetics , Lymphoma/diagnosis , Leukemia/genetics , Leukemia/diagnosis , Leukemia/therapy , Lymphoma/therapy , Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...