Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Am J Hematol ; 96(8): 989-999, 2021 08 01.
Article in English | MEDLINE | ID: mdl-33984160

ABSTRACT

Fanconi anemia (FA) is characterized by chromosome fragility, bone marrow failure (BMF) and predisposition to cancer. As reverse genetic mosaicism has been described as "natural gene therapy" in patients with FA, we sought to evaluate the clinical course of a cohort of FA mosaic patients followed at referral centers in Spain over a 30-year period. This cohort includes patients with a majority of T cells without chromosomal aberrations in the DEB-chromosomal breakage test. Relative to non-mosaic FA patients, we observed a higher proportion of adult patients in the cohort of mosaics, with a later age of hematologic onset and a milder evolution of (BMF). Consequently, the requirement for hematopoietic stem cell transplant (HSCT) was also lower. Additional studies allowed us to identify a sub-cohort of mosaic FA patients in whom the reversion was present in bone marrow (BM) progenitor cells leading to multilineage mosaicism. These multilineage mosaic patients are older, have a lower percentage of aberrant cells, have more stable hematology and none of them developed leukemia or myelodysplastic syndrome when compared to non-mosaics. In conclusion, our data indicate that reverse mosaicism is a good prognostic factor in FA and is associated with more favorable long-term clinical outcomes.


Subject(s)
Fanconi Anemia/therapy , Genetic Therapy/methods , Adolescent , Adult , Child , Fanconi Anemia/genetics , Humans , Male , Mosaicism , Young Adult
2.
Blood ; 130(13): 1535-1542, 2017 09 28.
Article in English | MEDLINE | ID: mdl-28801449

ABSTRACT

Previous Fanconi anemia (FA) gene therapy studies have failed to demonstrate engraftment of gene-corrected hematopoietic stem and progenitor cells (HSPCs) from FA patients, either after autologous transplantation or infusion into immunodeficient mice. In this study, we demonstrate that a validated short transduction protocol of G-CSF plus plerixafor-mobilized CD34+ cells from FA-A patients with a therapeutic FANCA-lentiviral vector corrects the phenotype of in vitro cultured hematopoietic progenitor cells. Transplantation of transduced FA CD34+ cells into immunodeficient mice resulted in reproducible engraftment of myeloid, lymphoid, and CD34+ cells. Importantly, a marked increase in the proportion of phenotypically corrected, patient-derived hematopoietic cells was observed after transplantation with respect to the infused CD34+ graft, indicating the proliferative advantage of corrected FA-A hematopoietic repopulating cells. Our data demonstrate for the first time that optimized protocols of hematopoietic stem cell collection from FA patients, followed by the short and clinically validated transduction of these cells with a therapeutic lentiviral vector, results in the generation of phenotypically corrected HSPCs capable of repopulating and developing proliferation advantage in immunodeficient mice. Our results suggest that clinical approaches for FA gene therapy similar to those used in this study will facilitate hematopoietic repopulation in FA patients with gene corrected HSPCs, opening new prospects for gene therapy of FA patients.


Subject(s)
Fanconi Anemia Complementation Group C Protein/genetics , Fanconi Anemia/therapy , Genetic Therapy/methods , Genetic Vectors , Hematopoietic Stem Cell Transplantation/methods , Transduction, Genetic/methods , Animals , Antigens, CD34/immunology , Child , Child, Preschool , Fanconi Anemia/pathology , Graft Survival , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/pathology , Heterografts , Humans , Lentivirus/genetics , Mice
3.
Am J Hum Genet ; 92(5): 800-6, 2013 May 02.
Article in English | MEDLINE | ID: mdl-23623386

ABSTRACT

Fanconi anemia (FA) is a rare genomic instability disorder characterized by progressive bone marrow failure and predisposition to cancer. FA-associated gene products are involved in the repair of DNA interstrand crosslinks (ICLs). Fifteen FA-associated genes have been identified, but the genetic basis in some individuals still remains unresolved. Here, we used whole-exome and Sanger sequencing on DNA of unclassified FA individuals and discovered biallelic germline mutations in ERCC4 (XPF), a structure-specific nuclease-encoding gene previously connected to xeroderma pigmentosum and segmental XFE progeroid syndrome. Genetic reversion and wild-type ERCC4 cDNA complemented the phenotype of the FA cell lines, providing genetic evidence that mutations in ERCC4 cause this FA subtype. Further biochemical and functional analysis demonstrated that the identified FA-causing ERCC4 mutations strongly disrupt the function of XPF in DNA ICL repair without severely compromising nucleotide excision repair. Our data show that depending on the type of ERCC4 mutation and the resulting balance between both DNA repair activities, individuals present with one of the three clinically distinct disorders, highlighting the multifunctional nature of the XPF endonuclease in genome stability and human disease.


Subject(s)
DNA-Binding Proteins/genetics , Deoxyribonucleases/genetics , Fanconi Anemia/genetics , Genetic Predisposition to Disease/genetics , Phenotype , Apoptosis/genetics , Apoptosis/radiation effects , Base Sequence , Exome/genetics , Fanconi Anemia/pathology , Germ-Line Mutation/genetics , Humans , Immunoblotting , Immunoprecipitation , Molecular Sequence Data , Sequence Analysis, DNA , Ultraviolet Rays
4.
Blood ; 119(13): 3042-9, 2012 Mar 29.
Article in English | MEDLINE | ID: mdl-22310912

ABSTRACT

Fanconi anemia (FA) is an inherited genetic disorder associated with BM failure and cancer predisposition. In the present study, we sought to elucidate the role of microRNAs (miRNAs) in the hematopoietic defects observed in FA patients. Initial studies showed that 3 miRNAs, hsa-miR-133a, hsa-miR-135b, and hsa-miR-181c, were significantly down-regulated in lymphoblastoid cell lines and fresh peripheral blood cells from FA patients. In vitro studies with cells expressing the luciferase reporter fused to the TNFα 3'-untranslated region confirmed in silico predictions suggesting an interaction between hsa-miR-181c and TNFα mRNA. These observations were consistent with the down-regulated expression of TNFα mediated by hsa-miR-181c in cells from healthy donors and cells from FA patients. Because of the relevance of TNFα in the hematopoietic defects of FA patients, in the present study, we transfected BM cells from FA patients with hsa-miR-181c to evaluate the impact of this miRNA on their clonogenic potential. hsa-miR-181c markedly increased the number and size of the myeloid and erythroid colonies generated by BM cells from FA patients. Our results offer new clues toward understanding the biologic basis of BM failure in FA patients and open new possibilities for the treatment of the hematologic dysfunction in FA patients based on miRNA regulation.


Subject(s)
Cell Proliferation , Fanconi Anemia/genetics , Hematopoietic Stem Cells/physiology , MicroRNAs/genetics , Tumor Necrosis Factor-alpha/pharmacology , Blood Cell Count , Bone Marrow Cells/metabolism , Bone Marrow Cells/physiology , Cell Proliferation/drug effects , Cells, Cultured , Down-Regulation/genetics , Fanconi Anemia/metabolism , Gene Expression/drug effects , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Male , MicroRNAs/metabolism , Primary Cell Culture , Transfection
5.
Blood ; 117(14): 3759-69, 2011 Apr 07.
Article in English | MEDLINE | ID: mdl-21273304

ABSTRACT

Fanconi anemia is characterized by congenital abnormalities, bone marrow failure, and cancer predisposition. To investigate the origin, functional role, and clinical impact of FANCA mutations, we determined a FANCA mutational spectrum with 130 pathogenic alleles. Some of these mutations were further characterized for their distribution in populations, mode of emergence, or functional consequences at cellular and clinical level. The world most frequent FANCA mutation is not the result of a mutational "hot-spot" but results from worldwide dissemination of an ancestral Indo-European mutation. We provide molecular evidence that total absence of FANCA in humans does not reduce embryonic viability, as the observed frequency of mutation carriers in the Gypsy population equals the expected by Hardy-Weinberg equilibrium. We also prove that long distance Alu-Alu recombination can cause Fanconi anemia by originating large interstitial deletions involving FANCA and 2 adjacent genes. Finally, we show that all missense mutations studied lead to an altered FANCA protein that is unable to relocate to the nucleus and activate the FA/BRCA pathway. This may explain the observed lack of correlation between type of FANCA mutation and cellular phenotype or clinical severity in terms of age of onset of hematologic disease or number of malformations.


Subject(s)
Fanconi Anemia Complementation Group A Protein/genetics , Fanconi Anemia Complementation Group A Protein/physiology , Fanconi Anemia/genetics , Fanconi Anemia/pathology , Mutation , Adolescent , Age of Onset , Base Sequence , Cell Culture Techniques , Cells, Cultured , Child , Child, Preschool , Chromosome Aberrations , Comparative Genomic Hybridization , DNA Mutational Analysis , Fanconi Anemia/diagnosis , Fanconi Anemia/epidemiology , Fanconi Anemia Complementation Group A Protein/metabolism , Gene Frequency , Humans , Infant , Models, Biological , Molecular Sequence Data , Mutation/physiology , Phenotype , Spain/epidemiology
6.
J Med Genet ; 48(4): 242-50, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21217111

ABSTRACT

BACKGROUND: Fanconi anaemia (FA) is a rare syndrome characterized by bone marrow failure, malformations and cancer predisposition. Chromosome fragility induced by DNA interstrand crosslink (ICL)-inducing agents such as diepoxybutane (DEB) or mitomycin C (MMC) is the 'gold standard' test for the diagnosis of FA. OBJECTIVE: To study the variability, the diagnostic implications and the clinical impact of chromosome fragility in FA. METHODS: Data are presented from 198 DEB-induced chromosome fragility tests in patients with and without FA where information on genetic subtype, cell sensitivity to MMC and clinical data were available. RESULTS: This large series allowed quantification of the variability and the level of overlap in ICL sensitivity among patients with FA and the normal population. A new chromosome fragility index is proposed that provides a cut-off diagnostic level to unambiguously distinguish patients with FA, including mosaics, from non-FA individuals. Spontaneous chromosome fragility and its correlation with DEB-induced fragility was also analysed, indicating that although both variables are correlated, 54% of patients with FA do not have spontaneous fragility. The data reveal a correlation between malformations and sensitivity to ICL-inducing agents. This correlation was also statistically significant when the analysis was restricted to patients from the FA-A complementation group. Finally, chromosome fragility does not correlate with the age of onset of haematological disease. CONCLUSIONS: This study proposes a new chromosome fragility index and suggests that genome instability during embryo development may be related to malformations in FA, while DEB-induced chromosome breaks in T cells have no prognostic value for the haematological disease.


Subject(s)
Chromosome Fragility , Fanconi Anemia/genetics , Cross-Linking Reagents/pharmacology , Epoxy Compounds/pharmacology , Fanconi Anemia/diagnosis , Humans , Mitomycin/pharmacology , Mosaicism , Phenotype
7.
Med Eng Phys ; 110: 103919, 2022 12.
Article in English | MEDLINE | ID: mdl-36564142

ABSTRACT

This paper is aimed at identifying by means of micro-CT the microstructural differences between normal and degenerative mitral marginal chordae tendineae. The control group is composed of 21 normal chords excised from 14 normal mitral valves from heart transplant recipients. The experimental group comprises 22 degenerative fibroelastic chords obtained at surgery from 11 pathological valves after mitral repair or replacement. In the control group the superficial endothelial cells and spongiosa layer remained intact, covering the wavy core collagen. In contrast, in the experimental group the collagen fibers were arranged as straightened thick bundles in a parallel configuration. 100 cross-sections were examined by micro-CT from each chord. Each image was randomized through the K-means machine learning algorithm and then, the global and local Shannon entropies were obtained. The optimum number of clusters, K, was estimated to maximize the differences between normal and degenerative chords in global and local Shannon entropy; the p-value after a nested ANOVA test was chosen as the parameter to be minimized. Optimum results were obtained with global Shannon entropy and 2≤K≤7, providing p < 0.01; for K=3, p = 2.86·10-3. These findings open the door to novel perioperative diagnostic methods in order to avoid or reduce postoperative mitral valve regurgitation recurrences.


Subject(s)
Endothelial Cells , Mitral Valve Insufficiency , Humans , Chordae Tendineae/pathology , Collagen , Mitral Valve/diagnostic imaging , Mitral Valve Insufficiency/diagnostic imaging , Mitral Valve Insufficiency/surgery , X-Ray Microtomography
8.
J Clin Invest ; 132(15)2022 08 01.
Article in English | MEDLINE | ID: mdl-35671096

ABSTRACT

Fanconi anemia (FA) is the most prevalent inherited bone marrow failure (BMF) syndrome. Nevertheless, the pathophysiological mechanisms of BMF in FA have not been fully elucidated. Since FA cells are defective in DNA repair, we hypothesized that FA hematopoietic stem and progenitor cells (HSPCs) might express DNA damage-associated stress molecules such as natural killer group 2 member D ligands (NKG2D-Ls). These ligands could then interact with the activating NKG2D receptor expressed in cytotoxic NK or CD8+ T cells, which may result in progressive HSPC depletion. Our results indeed demonstrated upregulated levels of NKG2D-Ls in cultured FA fibroblasts and T cells, and these levels were further exacerbated by mitomycin C or formaldehyde. Notably, a high proportion of BM CD34+ HSPCs from patients with FA also expressed increased levels of NKG2D-Ls, which correlated inversely with the percentage of CD34+ cells in BM. Remarkably, the reduced clonogenic potential characteristic of FA HSPCs was improved by blocking NKG2D-NKG2D-L interactions. Moreover, the in vivo blockage of these interactions in a BMF FA mouse model ameliorated the anemia in these animals. Our study demonstrates the involvement of NKG2D-NKG2D-L interactions in FA HSPC functionality, suggesting an unexpected role of the immune system in the progressive BMF that is characteristic of FA.


Subject(s)
Fanconi Anemia , Animals , Antigens, CD34 , Fanconi Anemia/genetics , Hematopoietic Stem Cells , Ligands , Mice , NK Cell Lectin-Like Receptor Subfamily K/genetics , Up-Regulation
9.
Mol Ther Methods Clin Dev ; 22: 66-75, 2021 Sep 10.
Article in English | MEDLINE | ID: mdl-34485595

ABSTRACT

Difficulties in the collection of hematopoietic stem and progenitor cells (HSPCs) from Fanconi anemia (FA) patients have limited the gene therapy in this disease. We have investigated (ClinicalTrials.gov, NCT02931071) the safety and efficacy of filgrastim and plerixafor for mobilization of HSPCs and collection by leukapheresis in FA patients. Nine of eleven enrolled patients mobilized beyond the threshold level of 5 CD34+ cells/µL required to initiate apheresis. A median of 21.8 CD34+ cells/µL was reached at the peak of mobilization. Significantly, the oldest patients (15 and 16 years old) were the only ones who did not reach that threshold. A median of 4.27 million CD34+ cells/kg was collected in 2 or 3 aphereses. These numbers were markedly decreased to 1.1 million CD34+ cells/kg after immunoselection, probably because of weak expression of the CD34 antigen. However, these numbers were sufficient to facilitate the engraftment of corrected HSPCs in non-conditioned patients. No procedure-associated serious adverse events were observed. Mobilization of CD34+ cells correlated with younger age, higher leukocyte counts and hemoglobin values, lower mean corpuscular volume, and higher proportion of CD34+ cells in bone marrow (BM). All these values offer crucial information for the enrollment of FA patients for gene therapy protocols.

10.
Mol Cancer Ther ; 7(5): 1309-18, 2008 May.
Article in English | MEDLINE | ID: mdl-18483318

ABSTRACT

Trabectedin (Yondelis; ET-743) is a potent anticancer drug that binds to DNA by forming a covalent bond with a guanine in one strand and one or more hydrogen bonds with the opposite strand. Using a fluorescence-based melting assay, we show that one single trabectedin-DNA adduct increases the thermal stability of the double helix by >20 degrees C. As deduced from the analysis of phosphorylated H2AX and Rad51 foci, we observed that clinically relevant doses of trabectedin induce the formation of DNA double-strand breaks in human cells and activate homologous recombination repair in a manner similar to that evoked by the DNA interstrand cross-linking agent mitomycin C (MMC). Because one important characteristic of this drug is its marked cytotoxicity on cells lacking a functional Fanconi anemia (FA) pathway, we compared the response of different subtypes of FA cells to MMC and trabectedin. Our data clearly show that human cells with mutations in FANCA, FANCC, FANCF, FANCG, or FANCD1 genes are highly sensitive to both MMC and trabectedin. However, in marked contrast to MMC, trabectedin does not induce any significant accumulation of FA cells in G2-M. The critical relevance of FA proteins in the response of human cells to trabectedin reported herein, together with observations showing the role of the FA pathway in cancer suppression, strongly suggest that screening for mutations in FA genes may facilitate the identification of tumors displaying enhanced sensitivity to this novel anticancer drug.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Dioxoles/pharmacology , Fanconi Anemia Complementation Group Proteins/genetics , Tetrahydroisoquinolines/pharmacology , Cell Cycle , DNA/genetics , DNA/metabolism , DNA Breaks, Double-Stranded , DNA Repair , Dose-Response Relationship, Drug , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Fanconi Anemia Complementation Group Proteins/metabolism , Humans , Mitomycin/pharmacology , Time Factors , Trabectedin
11.
Nat Med ; 25(7): 1073-1081, 2019 07.
Article in English | MEDLINE | ID: mdl-31270502

ABSTRACT

Bladder cancer is lethal in its advanced, muscle-invasive phase with very limited therapeutic advances1,2. Recent molecular characterization has defined new (epi)genetic drivers and potential targets for bladder cancer3,4. The immune checkpoint inhibitors have shown remarkable efficacy but only in a limited fraction of bladder cancer patients5-8. Here, we show that high G9a (EHMT2) expression is associated with poor clinical outcome in bladder cancer and that targeting G9a/DNMT methyltransferase activity with a novel inhibitor (CM-272) induces apoptosis and immunogenic cell death. Using an immunocompetent quadruple-knockout (PtenloxP/loxP; Trp53loxP/loxP; Rb1loxP/loxP; Rbl1-/-) transgenic mouse model of aggressive metastatic, muscle-invasive bladder cancer, we demonstrate that CM-272 + cisplatin treatment results in statistically significant regression of established tumors and metastases. The antitumor effect is significantly improved when CM-272 is combined with anti-programmed cell death ligand 1, even in the absence of cisplatin. These effects are associated with an endogenous antitumor immune response and immunogenic cell death with the conversion of a cold immune tumor into a hot tumor. Finally, increased G9a expression was associated with resistance to programmed cell death protein 1 inhibition in a cohort of patients with bladder cancer. In summary, these findings support new and promising opportunities for the treatment of bladder cancer using a combination of epigenetic inhibitors and immune checkpoint blockade.


Subject(s)
Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Urinary Bladder Neoplasms/drug therapy , Animals , Cell Line, Tumor , Cisplatin/therapeutic use , Enhancer of Zeste Homolog 2 Protein/physiology , Female , Histocompatibility Antigens , Humans , Mice , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/pathology
12.
Nat Med ; 25(9): 1396-1401, 2019 09.
Article in English | MEDLINE | ID: mdl-31501599

ABSTRACT

Fanconi anemia (FA) is a DNA repair syndrome generated by mutations in any of the 22 FA genes discovered to date1,2. Mutations in FANCA account for more than 60% of FA cases worldwide3,4. Clinically, FA is associated with congenital abnormalities and cancer predisposition. However, bone marrow failure is the primary pathological feature of FA that becomes evident in 70-80% of patients with FA during the first decade of life5,6. In this clinical study (ClinicalTrials.gov, NCT03157804 ; European Clinical Trials Database, 2011-006100-12), we demonstrate that lentiviral-mediated hematopoietic gene therapy reproducibly confers engraftment and proliferation advantages of gene-corrected hematopoietic stem cells (HSCs) in non-conditioned patients with FA subtype A. Insertion-site analyses revealed the multipotent nature of corrected HSCs and showed that the repopulation advantage of these cells was not due to genotoxic integrations of the therapeutic provirus. Phenotypic correction of blood and bone marrow cells was shown by the acquired resistance of hematopoietic progenitors and T lymphocytes to DNA cross-linking agents. Additionally, an arrest of bone marrow failure progression was observed in patients with the highest levels of gene marking. The progressive engraftment of corrected HSCs in non-conditioned patients with FA supports that gene therapy should constitute an innovative low-toxicity therapeutic option for this life-threatening disorder.


Subject(s)
Fanconi Anemia Complementation Group A Protein/genetics , Fanconi Anemia/therapy , Genetic Therapy , Hematopoietic Stem Cell Transplantation , Adolescent , Adult , Bone Marrow Cells/cytology , Child , Child, Preschool , Fanconi Anemia/genetics , Fanconi Anemia/physiopathology , Female , Genetic Vectors/genetics , Hematopoietic Stem Cells/metabolism , Humans , Infant , Lentivirus/genetics , Male , Mutation/genetics , Spain/epidemiology , Targeted Gene Repair , Transduction, Genetic , Young Adult
13.
J Mech Behav Biomed Mater ; 65: 200-212, 2017 01.
Article in English | MEDLINE | ID: mdl-27591507

ABSTRACT

The influence of the orientation of rat bones on their mechanical response is analyzed in this research. 28 femora obtained from 14 Sprague-Dawley rats were subjected to three-point bending tests, comparing the anteroposterior and posteroanterior orientations. The results show that the whole-bone loading capacity of the femora tested in the posteroanterior orientation clearly exceeds that of the anteroposterior oriented bones. Likewise, the intrinsic (tissue-level) loading capacity of the bones tested in the posteroanterior orientation is manifestly higher than that of the bones tested in the opposite direction. The analysis carried out shows that applying beam theory for symmetric cross-sections leads to underestimating the stress state in the cross-section. In this sense, it is generally recommendable to use the non-symmetric beam theory in order to obtain the normal stresses during bending tests. The geometric, intrinsic and global changes resulting from the orientation of the bones was assessed, finding out that it is the variation in the intrinsic properties which explains the change measured in the whole-bone properties. The experimental scope was increased, including 8 additional femora on which a series of Vickers tests were carried out in the anterior and posterior regions of the cross-section. In all cases the hardness obtained in the anterior region is larger than in the posterior region. This result confirms that the mechanical properties of the bone tissue depend on its position in the cross-section and provides a reliable explanation to understand the response of the bones when subjected to bending tests. These results stress the importance of reporting the orientation of the bones in any scientific paper because, otherwise, it would be impossible to properly assess its impact and relevance.


Subject(s)
Bone and Bones/physiology , Stress, Mechanical , Animals , Biomechanical Phenomena , Rats , Rats, Sprague-Dawley
14.
Radiat Res ; 164(5): 635-41, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16238440

ABSTRACT

Fanconi anemia is a genetically heterogeneous recessive disease characterized mainly by bone marrow failure and cancer predisposition. Although it is accepted that Fanconi cells are highly sensitive to DNA crosslinking agents, their response to ionizing radiation is still unclear. Using pulsed-field gel electrophoresis, we have observed that radiation generates a similar number of DNA double-strand breaks in normal and Fanconi cells from three (FA-A, FA-C and FA-F) of the 11 complementation groups identified. Nonsynchronized as well as nonproliferating Fanconi anemia cells showed an evident defect in rejoining the double-strand breaks generated by ionizing radiation, indicating defective non-homologous end-joining repair. At the cellular level, no difference in the radiosensitivity of normal and FA-A lymphoblast cells was noted, and a modest increase in the radiosensitivity of Fanca-/- hematopoietic progenitor cells was observed compared to Fanca+/+ cells. Finally, when animals were exposed to a fractionated total-body irradiation of 5 Gy, a similar hematopoietic syndrome was observed in wild-type and Fanca-/- mice. Taken together, our observations suggest that Fanconi cells, in particular those having nonfunctional Fanconi proteins upstream of FANCD2, have a defect in the non-homologous end-joining repair of double-strand breaks produced by ionizing radiation, and that compensatory mechanisms of DNA repair and/or stem cell regeneration should limit the impact of this defect in irradiated organisms.


Subject(s)
DNA Repair , Fanconi Anemia/genetics , Hematopoietic Stem Cells/radiation effects , Animals , DNA Damage , Fanconi Anemia Complementation Group D2 Protein/physiology , Hematopoietic Stem Cells/metabolism , Humans , Mice , Mice, Inbred C57BL , Radiation Tolerance
15.
PLoS One ; 10(10): e0139740, 2015.
Article in English | MEDLINE | ID: mdl-26466379

ABSTRACT

Fanconi anaemia (FA) is an inherited disorder characterized by chromosomal instability. The phenotype is variable, which raises the possibility that it may be affected by other factors, such as epigenetic modifications. These play an important role in oncogenesis and may be pharmacologically manipulated. Our aim was to explore whether the epigenetic profiles in FA differ from non-FA individuals and whether these could be manipulated to alter the disease phenotype. We compared expression of epigenetic genes and DNA methylation profile of tumour suppressor genes between FA and normal samples. FA samples exhibited decreased expression levels of genes involved in epigenetic regulation and hypomethylation in the promoter regions of tumour suppressor genes. Treatment of FA cells with histone deacetylase inhibitor Vorinostat increased the expression of DNM3Tß and reduced the levels of CIITA and HDAC9, PAK1, USP16, all involved in different aspects of epigenetic and immune regulation. Given the ability of Vorinostat to modulate epigenetic genes in FA patients, we investigated its functional effects on the FA phenotype. This was assessed by incubating FA cells with Vorinostat and quantifying chromosomal breaks induced by DNA cross-linking agents. Treatment of FA cells with Vorinostat resulted in a significant reduction of aberrant cells (81% on average). Our results suggest that epigenetic mechanisms may play a role in oncogenesis in FA. Epigenetic agents may be helpful in improving the phenotype of FA patients, potentially reducing tumour incidence in this population.


Subject(s)
Epigenesis, Genetic , Fanconi Anemia/genetics , Hydroxamic Acids/chemistry , Neoplasms/prevention & control , Adolescent , Adult , Child , Child, Preschool , Chromatin/chemistry , Chromosomal Instability , Computational Biology , Cross-Linking Reagents/chemistry , DNA/genetics , DNA Methylation , Fanconi Anemia/physiopathology , Female , Gene Expression Profiling , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylases/metabolism , Humans , Immune System , Incidence , Infant , Leukocytes, Mononuclear/cytology , Male , Middle Aged , Neoplasms/genetics , Phenotype , Promoter Regions, Genetic , Real-Time Polymerase Chain Reaction , Vorinostat
16.
Arch Med Res ; 34(6): 589-99, 2003.
Article in English | MEDLINE | ID: mdl-14734099

ABSTRACT

Hematopoietic stem cells constitute a rare population of precursor cells with remarkable properties for being used as targets in gene therapy protocols. The last years have been particularly productive both in the fields of gene therapy and stem cell biology. Results from ongoing clinical trials have shown the first unquestionable clinical benefits of immunodeficient patients transplanted with genetically modified autologous stem cells. On the other hand, severe side effects in a few patients treated with gene therapy have also been reported, indicating the usefulness of further improving the vectors currently used in gene therapy clinical trials. In the field of stem cell biology, evidence showing the plastic potential of adult hematopoietic stem cells and data indicating the multipotency of adult mesenchymal precursor cells have been presented. Also, the generation of embryonic stem cells by means of nuclear transfer techniques has appeared as a new methodology with direct implications in gene therapy.


Subject(s)
Genetic Therapy/methods , Hematologic Diseases/congenital , Hematopoietic Stem Cells/physiology , Animals , Gene Transfer Techniques , Humans , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/therapy
17.
Clin Transl Oncol ; 13(4): 215-21, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21493181

ABSTRACT

The dissection of the molecular pathways participating in genetic instability disorders has rendered invaluable information about the mechanisms of cancer pathogenesis and progression, and is offering a unique opportunity to establish targeted anticancer therapies. Fanconi anaemia (FA) is a paradigm of cancer-prone inherited monogenic disorders. Moreover, accumulated evidence indicates that genetic and epigenetic alterations in FA genes can also play an important role in sporadic cancer in the general population. Here, we summarise current progress in the understanding of the molecular biology of FA and review the principal mechanisms accounting for a disrupted FA pathway in sporadic cancer. Additionally, we discuss the impact of these findings in the development of new anticancer therapies, particularly with DNA interstrand crosslinkers and with new inhibitors of the FA and/or alternative DNA repair pathways.


Subject(s)
Fanconi Anemia/complications , Fanconi Anemia/genetics , Neoplasms/genetics , Precancerous Conditions/complications , Precancerous Conditions/genetics , Cell Transformation, Neoplastic/genetics , Humans , Signal Transduction/physiology
18.
PLoS One ; 5(12): e15525, 2010 Dec 28.
Article in English | MEDLINE | ID: mdl-21203397

ABSTRACT

Chronic myeloid leukemia (CML) is a malignant clonal disorder of the hematopoietic system caused by the expression of the BCR/ABL fusion oncogene. Although it is well known that CML cells are genetically unstable, the mechanisms accounting for this genomic instability are still poorly understood. Because the Fanconi anemia (FA) pathway is believed to control several mechanisms of DNA repair, we investigated whether this pathway was disrupted in CML cells. Our data show that CML cells have a defective capacity to generate FANCD2 nuclear foci, either in dividing cells or after DNA damage. Similarly, human cord blood CD34(+) cells transduced with BCR/ABL retroviral vectors showed impaired FANCD2 foci formation, whereas FANCD2 monoubiquitination in these cells was unaffected. Soon after the transduction of CD34(+) cells with BCR/ABL retroviral vectors a high proportion of cells with supernumerary centrosomes was observed. Similarly, BCR/ABL induced a high proportion of chromosomal abnormalities, while mediated a cell survival advantage after exposure to DNA cross-linking agents. Significantly, both the impaired formation of FANCD2 nuclear foci, and also the predisposition of BCR/ABL cells to develop centrosomal and chromosomal aberrations were reverted by the ectopic expression of BRCA1. Taken together, our data show for the first time a disruption of the FA/BRCA pathway in BCR/ABL cells, suggesting that this defective pathway should play an important role in the genomic instability of CML by the co-occurrence of centrosomal amplification and DNA repair deficiencies.


Subject(s)
Chromosomes/ultrastructure , Fanconi Anemia/metabolism , Fusion Proteins, bcr-abl/metabolism , Genes, BRCA1 , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Antigens, CD34/biosynthesis , Cell Line, Tumor , Centrosome/ultrastructure , Chromosome Aberrations , Cross-Linking Reagents/pharmacology , DNA Damage , Fanconi Anemia Complementation Group D2 Protein/metabolism , Humans , Microscopy, Fluorescence/methods , Retroviridae/genetics
19.
Am J Hum Genet ; 80(5): 895-910, 2007 May.
Article in English | MEDLINE | ID: mdl-17436244

ABSTRACT

FANCD2 is an evolutionarily conserved Fanconi anemia (FA) gene that plays a key role in DNA double-strand-type damage responses. Using complementation assays and immunoblotting, a consortium of American and European groups assigned 29 patients with FA from 23 families and 4 additional unrelated patients to complementation group FA-D2. This amounts to 3%-6% of FA-affected patients registered in various data sets. Malformations are frequent in FA-D2 patients, and hematological manifestations appear earlier and progress more rapidly when compared with all other patients combined (FA-non-D2) in the International Fanconi Anemia Registry. FANCD2 is flanked by two pseudogenes. Mutation analysis revealed the expected total of 66 mutated alleles, 34 of which result in aberrant splicing patterns. Many mutations are recurrent and have ethnic associations and shared allelic haplotypes. There were no biallelic null mutations; residual FANCD2 protein of both isotypes was observed in all available patient cell lines. These analyses suggest that, unlike the knockout mouse model, total absence of FANCD2 does not exist in FA-D2 patients, because of constraints on viable combinations of FANCD2 mutations. Although hypomorphic mutations arie involved, clinically, these patients have a relatively severe form of FA.


Subject(s)
Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia/genetics , Mutation , Alleles , Base Sequence , Cell Line , Child , Child, Preschool , DNA Primers/genetics , Ethnicity/genetics , Exons , Fanconi Anemia/classification , Fanconi Anemia/metabolism , Female , Genetic Complementation Test , Humans , Introns , Male , Mosaicism , Phenotype , Pregnancy , Pseudogenes , RNA Precursors/genetics , RNA Precursors/metabolism , RNA Splicing/genetics
20.
Mol Ther ; 14(4): 525-35, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16859999

ABSTRACT

We have investigated the hematopoietic phenotype of mice with a hypomorphic mutation in the Brca2/Fancd1 gene (Brca2(Delta27/Delta27) mutation). In contrast to observations made in other Fanconi anemia (FA) mouse models, low numbers of hematopoietic colony-forming cells (CFCs) were noted in Brca2(Delta27/Delta27) mice, either young or adult. Additionally, a high incidence of spontaneous chromosomal instability was observed in Brca2(Delta27/Delta27) bone marrow (BM) cells, but not in Brca2(+/Delta27) or Fanca(-/-) BM cells. Although Brca2(Delta27/Delta27) CFCs were not hypersensitive to ionizing radiation, a very severe hematopoietic syndrome was observed in irradiated Brca2(Delta27/Delta27) mice. Conventional BM competition experiments showed a marked repopulation defect in Brca2(Delta27/Delta27) hematopoietic stem cells (HSCs), compared to wild-type HSCs. Moreover, we have observed for the first time in a DNA repair disease model a very significant proliferation defect in Brca2(Delta27/Delta27) HSCs maintained in their natural physiological environment. The progressive repopulation of wild-type HSCs transplanted into unconditioned Brca2(Delta27/Delta27) recipients is reminiscent of the somatic mosaicism phenomenon observed in a number of genetic diseases, including FA. The hematopoietic phenotype associated with the Brca2(Delta27/Delta27) mutation suggests that this FA-D1 mouse model will constitute an important tool for the development of new therapies for FA, including gene therapy.


Subject(s)
Fanconi Anemia/pathology , Hematopoietic System/pathology , Animals , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Proliferation , Chromosome Aberrations/chemically induced , Disease Models, Animal , Fanconi Anemia/classification , Fanconi Anemia/genetics , Fanconi Anemia/therapy , Gene Deletion , Hematopoietic Stem Cell Transplantation , Hematopoietic System/metabolism , Hematopoietic System/radiation effects , Mice , Mice, Inbred BALB C , Mice, Transgenic , Mitomycin/pharmacology , Myeloid Cells/metabolism , Myeloid Cells/pathology , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL