Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
2.
Mol Cell ; 83(20): 3659-3668.e10, 2023 10 19.
Article in English | MEDLINE | ID: mdl-37832547

ABSTRACT

The integrity of the nuclear envelope (NE) is essential for maintaining the structural stability of the nucleus. Rupture of the NE has been frequently observed in cancer cells, especially in the context of mechanical challenges, such as physical confinement and migration. However, spontaneous NE rupture events, without any obvious physical challenges to the cell, have also been described. The molecular mechanism(s) of these spontaneous NE rupture events remain to be explored. Here, we show that DNA damage and subsequent ATR activation leads to NE rupture. Upon DNA damage, lamin A/C is phosphorylated in an ATR-dependent manner, leading to changes in lamina assembly and, ultimately, NE rupture. In addition, we show that cancer cells with intrinsic DNA repair defects undergo frequent events of DNA-damage-induced NE rupture, which renders them extremely sensitive to further NE perturbations. Exploiting this NE vulnerability could provide a new angle to complement traditional, DNA-damage-based chemotherapy.


Subject(s)
Lamin Type A , Nuclear Envelope , Nuclear Envelope/metabolism , Lamin Type A/genetics , Lamin Type A/metabolism , Phosphorylation , DNA Damage , DNA/metabolism , Cell Nucleus/metabolism
3.
Nature ; 621(7978): 415-422, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37674080

ABSTRACT

DNA double-strand breaks (DSBs) are deleterious lesions that challenge genome integrity. To mitigate this threat, human cells rely on the activity of multiple DNA repair machineries that are tightly regulated throughout the cell cycle1. In interphase, DSBs are mainly repaired by non-homologous end joining and homologous recombination2. However, these pathways are completely inhibited in mitosis3-5, leaving the fate of mitotic DSBs unknown. Here we show that DNA polymerase theta6 (Polθ) repairs mitotic DSBs and thereby maintains genome integrity. In contrast to other DSB repair factors, Polθ function is activated in mitosis upon phosphorylation by Polo-like kinase 1 (PLK1). Phosphorylated Polθ is recruited by a direct interaction with the BRCA1 C-terminal domains of TOPBP1 to mitotic DSBs, where it mediates joining of broken DNA ends. Loss of Polθ leads to defective repair of mitotic DSBs, resulting in a loss of genome integrity. This is further exacerbated in cells that are deficient in homologous recombination, where loss of mitotic DSB repair by Polθ results in cell death. Our results identify mitotic DSB repair as the underlying cause of synthetic lethality between Polθ and homologous recombination. Together, our findings reveal the critical importance of mitotic DSB repair in the maintenance of genome integrity.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , DNA-Directed DNA Polymerase , Mitosis , Protein Serine-Threonine Kinases , Humans , BRCA1 Protein/metabolism , Cell Cycle Proteins/metabolism , Cell Death/genetics , DNA-Directed DNA Polymerase/chemistry , DNA-Directed DNA Polymerase/metabolism , Homologous Recombination/genetics , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Synthetic Lethal Mutations , DNA Polymerase theta , Polo-Like Kinase 1
4.
Nucleic Acids Res ; 51(15): 7988-8004, 2023 08 25.
Article in English | MEDLINE | ID: mdl-37395445

ABSTRACT

Fanconi anemia (FA) is a genetic disorder associated with developmental defects, bone marrow failure and cancer. The FA pathway is crucial for the repair of DNA interstrand crosslinks (ICLs). In this study, we have developed and characterized a new tool to investigate ICL repair: a clickable version of the crosslinking agent melphalan which we name click-melphalan. Our results demonstrate that click-melphalan is as effective as its unmodified counterpart in generating ICLs and associated toxicity. The lesions induced by click-melphalan can be detected in cells by post-labelling with a fluorescent reporter and quantified using flow cytometry. Since click-melphalan induces both ICLs and monoadducts, we generated click-mono-melphalan, which only induces monoadducts, in order to distinguish between the two types of DNA repair. By using both molecules, we show that FANCD2 knock-out cells are deficient in removing click-melphalan-induced lesions. We also found that these cells display a delay in repairing click-mono-melphalan-induced monoadducts. Our data further revealed that the presence of unrepaired ICLs inhibits monoadduct repair. Finally, our study demonstrates that these clickable molecules can differentiate intrinsic DNA repair deficiencies in primary FA patient cells from those in primary xeroderma pigmentosum patient cells. As such, these molecules may have potential for developing diagnostic tests.


Subject(s)
Fanconi Anemia , Melphalan , Humans , Melphalan/pharmacology , Fanconi Anemia/pathology , DNA Repair , DNA Damage , DNA
5.
Nat Cancer ; 2(6): 598-610, 2021 06.
Article in English | MEDLINE | ID: mdl-34179826

ABSTRACT

DNA polymerase theta (POLθ) is synthetic lethal with Homologous Recombination (HR) deficiency and thus a candidate target for HR-deficient cancers. Through high-throughput small molecule screens we identified the antibiotic Novobiocin (NVB) as a specific POLθ inhibitor that selectively kills HR-deficient tumor cells in vitro and in vivo. NVB directly binds to the POLθ ATPase domain, inhibits its ATPase activity, and phenocopies POLθ depletion. NVB kills HR-deficient breast and ovarian tumors in GEMM, xenograft and PDX models. Increased POLθ levels predict NVB sensitivity, and BRCA-deficient tumor cells with acquired resistance to PARP inhibitors (PARPi) are sensitive to NVB in vitro and in vivo. Mechanistically, NVB-mediated cell death in PARPi-resistant cells arises from increased double-strand break end resection, leading to accumulation of single-strand DNA intermediates and non-functional RAD51 foci. Our results demonstrate that NVB may be useful alone or in combination with PARPi in treating HR-deficient tumors, including those with acquired PARPi resistance. (151/150).


Subject(s)
Homologous Recombination , Ovarian Neoplasms , Adenosine Triphosphatases/genetics , Female , Homologous Recombination/genetics , Humans , Ovarian Neoplasms/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology
6.
Mol Cell ; 72(3): 568-582.e6, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30344097

ABSTRACT

Protecting stalled DNA replication forks from degradation by promiscuous nucleases is essential to prevent genomic instability, a major driving force of tumorigenesis. Several proteins commonly associated with the repair of DNA double-strand breaks (DSBs) by homologous recombination (HR) have been implicated in the stabilization of stalled forks. Human CtIP, in conjunction with the MRE11 nuclease complex, plays an important role in HR by promoting DSB resection. Here, we report an unanticipated function for CtIP in protecting reversed forks from degradation. Unlike BRCA proteins, which defend nascent DNA strands from nucleolytic attack by MRE11, we find that CtIP protects perturbed forks from erroneous over-resection by DNA2. Finally, we uncover functionally synergistic effects between CtIP and BRCA1 in mitigating replication-stress-induced genomic instability. Collectively, our findings reveal a DSB-resection- and MRE11-independent role for CtIP in preserving fork integrity that contributes to the survival of BRCA1-deficient cells.


Subject(s)
Carrier Proteins/metabolism , Carrier Proteins/physiology , DNA Replication/physiology , Nuclear Proteins/metabolism , Nuclear Proteins/physiology , BRCA1 Protein , BRCA2 Protein , Cell Line , DNA Breaks, Double-Stranded , DNA Helicases/physiology , DNA Repair , DNA-Binding Proteins , Deoxyribonucleases , Endodeoxyribonucleases , Genomic Instability/physiology , Homologous Recombination/genetics , Humans , MRE11 Homologue Protein/metabolism , Protein Binding
7.
Nat Cell Biol ; 19(11): 1371-1378, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29035360

ABSTRACT

The emergence of resistance to poly-ADP-ribose polymerase inhibitors (PARPi) poses a threat to the treatment of BRCA1 and BRCA2 (BRCA1/2)-deficient tumours. Stabilization of stalled DNA replication forks is a recently identified PARPi-resistance mechanism that promotes genomic stability in BRCA1/2-deficient cancers. Dissecting the molecular pathways controlling genomic stability at stalled forks is critical. Here we show that EZH2 localizes at stalled forks where it methylates Lys27 on histone 3 (H3K27me3), mediating recruitment of the MUS81 nuclease. Low EZH2 levels reduce H3K27 methylation, prevent MUS81 recruitment at stalled forks and cause fork stabilization. As a consequence, loss of function of the EZH2/MUS81 axis promotes PARPi resistance in BRCA2-deficient cells. Accordingly, low EZH2 or MUS81 expression levels predict chemoresistance and poor outcome in patients with BRCA2-mutated tumours. Moreover, inhibition of Ezh2 in a murine Brca2-/- breast tumour model is associated with acquired PARPi resistance. Our findings identify EZH2 as a critical regulator of genomic stability at stalled forks that couples histone modifications to nuclease recruitment. Our data identify EZH2 expression as a biomarker of BRCA2-deficient tumour response to chemotherapy.


Subject(s)
DNA Replication/genetics , DNA-Binding Proteins/metabolism , Endonucleases/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Histones/metabolism , Animals , BRCA1 Protein/metabolism , BRCA2 Protein/metabolism , Biomarkers, Tumor/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line , Cell Line, Tumor , DNA Replication/drug effects , Drug Resistance, Neoplasm/genetics , Female , Genomic Instability/drug effects , Genomic Instability/genetics , HEK293 Cells , HeLa Cells , Humans , Methylation/drug effects , Mice , Mice, Nude , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology
9.
J Clin Invest ; 126(9): 3580-4, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27500492

ABSTRACT

Fanconi anemia (FA) is a recessive genetic disease characterized by congenital abnormalities, chromosome instability, progressive bone marrow failure (BMF), and a strong predisposition to cancer. Twenty FA genes have been identified, and the FANC proteins they encode cooperate in a common pathway that regulates DNA crosslink repair and replication fork stability. We identified a child with severe BMF who harbored biallelic inactivating mutations of the translesion DNA synthesis (TLS) gene REV7 (also known as MAD2L2), which encodes the mutant REV7 protein REV7-V85E. Patient-derived cells demonstrated an extended FA phenotype, which included increased chromosome breaks and G2/M accumulation upon exposure to DNA crosslinking agents, γH2AX and 53BP1 foci accumulation, and enhanced p53/p21 activation relative to cells derived from healthy patients. Expression of WT REV7 restored normal cellular and functional phenotypes in the patient's cells, and CRISPR/Cas9 inactivation of REV7 in a non-FA human cell line produced an FA phenotype. Finally, silencing Rev7 in primary hematopoietic cells impaired progenitor function, suggesting that the DNA repair defect underlies the development of BMF in FA. Taken together, our genetic and functional analyses identified REV7 as a previously undescribed FA gene, which we term FANCV.


Subject(s)
Fanconi Anemia/genetics , Mad2 Proteins/genetics , Mutation , Alleles , Animals , Cell Cycle , Cell Line, Tumor , Child , Chromosomal Instability , Chromosome Breakage , Cohort Studies , Cross-Linking Reagents/chemistry , DNA Damage , DNA Repair , Female , Fibroblasts/metabolism , Gene Silencing , Genetic Complementation Test , Genetic Predisposition to Disease , Genetic Variation , Hematopoietic Stem Cells/cytology , Humans , Lentivirus , Mad2 Proteins/metabolism , Male , Mice , Mice, Knockout , Mitosis , Phenotype
10.
Cell Rep ; 15(11): 2488-99, 2016 06 14.
Article in English | MEDLINE | ID: mdl-27264184

ABSTRACT

BRCA1/2 proteins function in homologous recombination (HR)-mediated DNA repair and cooperate with Fanconi anemia (FA) proteins to maintain genomic integrity through replication fork stabilization. Loss of BRCA1/2 proteins results in DNA repair deficiency and replicative stress, leading to genomic instability and enhanced sensitivity to DNA-damaging agents. Recent studies have shown that BRCA1/2-deficient tumors upregulate Polθ-mediated alternative end-joining (alt-EJ) repair as a survival mechanism. Whether other mechanisms maintain genomic integrity upon loss of BRCA1/2 proteins is currently unknown. Here we show that BRCA1/2-deficient tumors also upregulate FANCD2 activity. FANCD2 is required for fork protection and fork restart in BRCA1/2-deficient tumors. Moreover, FANCD2 promotes Polθ recruitment at sites of damage and alt-EJ repair. Finally, loss of FANCD2 in BRCA1/2-deficient tumors enhances cell death. These results reveal a synthetic lethal relationship between FANCD2 and BRCA1/2, and they identify FANCD2 as a central player orchestrating DNA repair pathway choice at the replication fork.


Subject(s)
BRCA1 Protein/deficiency , BRCA2 Protein/deficiency , DNA End-Joining Repair , DNA Replication , Fanconi Anemia Complementation Group D2 Protein/metabolism , Neoplasms/genetics , Neoplasms/pathology , Animals , BRCA1 Protein/metabolism , BRCA2 Protein/metabolism , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Survival , DNA End-Joining Repair/genetics , DNA Replication/genetics , DNA-Directed DNA Polymerase/metabolism , Endodeoxyribonucleases , Genomic Instability , Humans , Mice, Nude , Mutation/genetics , Nuclear Proteins/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Ubiquitination , Up-Regulation/genetics , DNA Polymerase theta
11.
Nat Rev Mol Cell Biol ; 17(6): 337-49, 2016 06.
Article in English | MEDLINE | ID: mdl-27145721

ABSTRACT

The Fanconi anaemia pathway repairs DNA interstrand crosslinks (ICLs) in the genome. Our understanding of this complex pathway is still evolving, as new components continue to be identified and new biochemical systems are used to elucidate the molecular steps of repair. The Fanconi anaemia pathway uses components of other known DNA repair processes to achieve proper repair of ICLs. Moreover, Fanconi anaemia proteins have functions in genome maintenance beyond their canonical roles of repairing ICLs. Such functions include the stabilization of replication forks and the regulation of cytokinesis. Thus, Fanconi anaemia proteins are emerging as master regulators of genomic integrity that coordinate several repair processes. Here, we summarize our current understanding of the functions of the Fanconi anaemia pathway in ICL repair, together with an overview of its connections with other repair pathways and its emerging roles in genome maintenance.


Subject(s)
DNA Repair , Fanconi Anemia Complementation Group Proteins/physiology , Fanconi Anemia/genetics , Animals , DNA Damage , DNA Replication , Humans
12.
Trends Cell Biol ; 26(1): 52-64, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26437586

ABSTRACT

DNA double-strand breaks (DSBs) are cytotoxic lesions that threaten genomic integrity. Failure to repair a DSB has deleterious consequences, including genomic instability and cell death. Indeed, misrepair of DSBs can lead to inappropriate end-joining events, which commonly underlie oncogenic transformation due to chromosomal translocations. Typically, cells employ two main mechanisms to repair DSBs: homologous recombination (HR) and classical nonhomologous end joining (C-NHEJ). In addition, alternative error-prone DSB repair pathways, namely alternative end joining (alt-EJ) and single-strand annealing (SSA), have been recently shown to operate in many different conditions and to contribute to genome rearrangements and oncogenic transformation. Here, we review the mechanisms regulating DSB repair pathway choice, together with the potential interconnections between HR and the annealing-dependent error-prone DSB repair pathways.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , Animals , Cell Survival , Genomic Instability , Humans , Mutation
13.
Cancer Discov ; 5(11): 1137-54, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26463832

ABSTRACT

UNLABELLED: Approximately 50% of epithelial ovarian cancers (EOC) exhibit defective DNA repair via homologous recombination (HR) due to genetic and epigenetic alterations of HR pathway genes. Defective HR is an important therapeutic target in EOC as exemplified by the efficacy of platinum analogues in this disease, as well as the advent of PARP inhibitors, which exhibit synthetic lethality when applied to HR-deficient cells. Here, we describe the genotypic and phenotypic characteristics of HR-deficient EOCs, discuss current and emerging approaches for targeting these tumors, and present challenges associated with these approaches, focusing on development and overcoming resistance. SIGNIFICANCE: Defective DNA repair via HR is a pivotal vulnerability of EOC, particularly of the high-grade serous histologic subtype. Targeting defective HR offers the unique opportunity of exploiting molecular differences between tumor and normal cells, thereby inducing cancer-specific synthetic lethality; the promise and challenges of these approaches in ovarian cancer are discussed in this review.


Subject(s)
Homologous Recombination , Ovarian Neoplasms/genetics , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers , Carcinoma, Ovarian Epithelial , Clinical Trials as Topic , DNA Repair , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Genetic Variation , Humans , Molecular Targeted Therapy , Neoplasms, Glandular and Epithelial/genetics , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Phenotype , Signal Transduction/drug effects , Treatment Outcome
14.
Nature ; 518(7538): 258-62, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25642963

ABSTRACT

Large-scale genomic studies have shown that half of epithelial ovarian cancers (EOCs) have alterations in genes regulating homologous recombination (HR) repair. Loss of HR accounts for the genomic instability of EOCs and for their cellular hyper-dependence on alternative poly-ADP ribose polymerase (PARP)-mediated DNA repair mechanisms. Previous studies have implicated the DNA polymerase θ (Polθ also known as POLQ, encoded by POLQ) in a pathway required for the repair of DNA double-strand breaks, referred to as the error-prone microhomology-mediated end-joining (MMEJ) pathway. Whether Polθ interacts with canonical DNA repair pathways to prevent genomic instability remains unknown. Here we report an inverse correlation between HR activity and Polθ expression in EOCs. Knockdown of Polθ in HR-proficient cells upregulates HR activity and RAD51 nucleofilament assembly, while knockdown of Polθ in HR-deficient EOCs enhances cell death. Consistent with these results, genetic inactivation of an HR gene (Fancd2) and Polq in mice results in embryonic lethality. Moreover, Polθ contains RAD51 binding motifs and it blocks RAD51-mediated recombination. Our results reveal a synthetic lethal relationship between the HR pathway and Polθ-mediated repair in EOCs, and identify Polθ as a novel druggable target for cancer therapy.


Subject(s)
DNA Breaks, Double-Stranded , DNA End-Joining Repair , DNA-Directed DNA Polymerase/metabolism , Homologous Recombination , Neoplasms, Glandular and Epithelial/genetics , Neoplasms, Glandular and Epithelial/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Amino Acid Motifs , Animals , Carcinoma, Ovarian Epithelial , Cell Cycle , Cell Death , Cell Line, Tumor , DNA End-Joining Repair/genetics , DNA Replication , DNA-Directed DNA Polymerase/deficiency , Embryo Loss , Fanconi Anemia Complementation Group D2 Protein/deficiency , Fanconi Anemia Complementation Group D2 Protein/genetics , Female , Genomic Instability , Homologous Recombination/genetics , Humans , Mice , Molecular Targeted Therapy , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/pathology , Protein Binding , Rad51 Recombinase/antagonists & inhibitors , Rad51 Recombinase/metabolism , Recombinational DNA Repair/genetics , DNA Polymerase theta
15.
Cancer Res ; 75(4): 628-34, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25634215

ABSTRACT

Platinum and PARP inhibitor (PARPi) sensitivity commonly coexist in epithelial ovarian cancer (EOC) due to the high prevalence of alterations in the homologous recombination (HR) DNA repair pathway that confer sensitivity to both drugs. In this report, we describe a unique subset of EOC with alterations in another DNA repair pathway, the nucleotide excision repair (NER) pathway, which may exhibit a discordance in sensitivities to these drugs. Specifically, 8% of high-grade serous EOC from The Cancer Genome Atlas dataset exhibited NER alterations, including nonsynonymous or splice site mutations and homozygous deletions of NER genes. Tumors with NER alterations were associated with improved overall survival (OS) and progression-free survival (PFS), compared with patients without NER alterations or BRCA1/2 mutations. Furthermore, patients with tumors with NER alterations had similar OS and PFS as BRCA1/2-mutated patients, suggesting that NER pathway inactivation in EOC conferred enhanced platinum sensitivity, similar to BRCA1/2-mutated tumors. Moreover, two NER mutations (ERCC6-Q524* and ERCC4-A583T), identified in the two most platinum-sensitive tumors, were functionally associated with platinum sensitivity in vitro. Importantly, neither NER alteration affected HR or conferred sensitivity to PARPi or other double-strand break-inducing agents. Overall, our findings reveal a new mechanism of platinum sensitivity in EOC that, unlike defective HR, may lead to a discordance in sensitivity to platinum and PARPi, with potential implications for previously reported and ongoing PARPi trials in this disease.


Subject(s)
DNA Repair/genetics , Drug Resistance, Neoplasm/genetics , Neoplasms, Glandular and Epithelial/drug therapy , Neoplasms, Glandular and Epithelial/genetics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Poly(ADP-ribose) Polymerases/genetics , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Carboplatin/administration & dosage , Carcinoma, Ovarian Epithelial , Cisplatin/administration & dosage , DNA Breaks, Double-Stranded/drug effects , Disease-Free Survival , Enzyme Inhibitors/administration & dosage , Female , Humans , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/pathology , Poly(ADP-ribose) Polymerase Inhibitors
16.
Mol Cell ; 54(1): 107-118, 2014 Apr 10.
Article in English | MEDLINE | ID: mdl-24657165

ABSTRACT

Timely DNA replication across damaged DNA is critical for maintaining genomic integrity. Translesion DNA synthesis (TLS) allows bypass of DNA lesions using error-prone TLS polymerases. The E3 ligase RAD18 is necessary for proliferating cell nuclear antigen (PCNA) monoubiquitination and TLS polymerase recruitment; however, the regulatory steps upstream of RAD18 activation are less understood. Here, we show that the UBZ4 domain-containing transcriptional repressor ZBTB1 is a critical upstream regulator of TLS. The UBZ4 motif is required for PCNA monoubiquitination and survival after UV damage. ZBTB1 associates with KAP-1, a transcriptional repressor whose phosphorylation relaxes chromatin after DNA damage. ZBTB1 depletion impairs formation of phospho-KAP-1 at UV damage sites and reduces RAD18 recruitment. Furthermore, phosphorylation of KAP-1 is necessary for efficient PCNA modification. We propose that ZBTB1 is required for localizing phospho-KAP-1 to chromatin and enhancing RAD18 accessibility. Collectively, our study implicates a ubiquitin-binding protein in orchestrating chromatin remodeling during DNA repair.


Subject(s)
Cell Nucleus/metabolism , Chromatin Assembly and Disassembly , DNA Damage , DNA Replication , DNA/biosynthesis , Repressor Proteins/metabolism , Animals , Binding Sites , Cell Nucleus/drug effects , Cell Nucleus/radiation effects , Cell Survival , Chickens , DNA-Binding Proteins/metabolism , DNA-Directed DNA Polymerase/metabolism , Enzyme Inhibitors/pharmacology , HEK293 Cells , HeLa Cells , Humans , Nucleic Acid Synthesis Inhibitors , Phosphorylation , Proliferating Cell Nuclear Antigen/metabolism , RNA Interference , Repressor Proteins/genetics , Signal Transduction , Time Factors , Transfection , Tripartite Motif-Containing Protein 28 , Ubiquitin-Protein Ligases , Ubiquitination , Ultraviolet Rays
17.
PLoS One ; 8(7): e69293, 2013.
Article in English | MEDLINE | ID: mdl-23935976

ABSTRACT

Gaucher disease (GD) is an autosomal recessive disorder characterized by lysosomal glucocerebrosidase (GBA) deficiency leading to hematological and skeletal manifestations. Mechanisms underlying these symptoms have not yet been elucidated. In vivo, bone marrow (BM) mesenchymal stem cells (MSCs) have important role in the regulation of bone mass and in the support of hematopoiesis, thus representing potential candidate that could contribute to the disease. GBA deficiency may also directly impair hematopoietic stem/progenitors cells (HSPCs) intrinsic function and induce hematological defect. In order to evaluate the role of BM stem cells in GD pathophysiology, we prospectively analyzed BM-MSCs and HSPCs properties in a series of 10 patients with type 1 GD. GBA activity was decreased in all tested cell subtypes. GD-MSCs had an impaired growth potential, morphological and cell cycle abnormalities, decreased capacities to differentiate into osteoblasts. Moreover, GD-MSCs secreted soluble factors that stimulated osteoclasts resorbing activities. In vitro and in vivo primitive and mature hematopoiesis were similar between patients and controls. However, GD-MSCs had a lower hematopoietic supportive capacity than those from healthy donors. These data suggest that BM microenvironment is altered in GD and that MSCs are key components of the manifestations observed in GD.


Subject(s)
Bone Marrow Cells/pathology , Gaucher Disease/pathology , Glucosylceramidase/deficiency , Hematopoietic Stem Cells/pathology , Mesenchymal Stem Cells/pathology , Osteoblasts/pathology , Osteoclasts/pathology , Adult , Aged , Animals , Bone Marrow Cells/enzymology , Case-Control Studies , Cell Differentiation , Cell Proliferation , Cellular Microenvironment , Female , Gaucher Disease/enzymology , Hematopoietic Stem Cells/enzymology , Humans , Male , Mesenchymal Stem Cells/enzymology , Mice , Middle Aged , Prospective Studies
18.
Cell Stem Cell ; 11(1): 36-49, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22683204

ABSTRACT

Fanconi anemia (FA) is an inherited DNA repair deficiency syndrome. FA patients undergo progressive bone marrow failure (BMF) during childhood, which frequently requires allogeneic hematopoietic stem cell transplantation. The pathogenesis of this BMF has been elusive to date. Here we found that FA patients exhibit a profound defect in hematopoietic stem and progenitor cells (HSPCs) that is present before the onset of clinical BMF. In response to replicative stress and unresolved DNA damage, p53 is hyperactivated in FA cells and triggers a late p21(Cdkn1a)-dependent G0/G1 cell-cycle arrest. Knockdown of p53 rescued the HSPC defects observed in several in vitro and in vivo models, including human FA or FA-like cells. Taken together, our results identify an exacerbated p53/p21 "physiological" response to cellular stress and DNA damage accumulation as a central mechanism for progressive HSPC elimination in FA patients, and have implications for clinical care.


Subject(s)
Bone Marrow/pathology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Damage , Fanconi Anemia/metabolism , Fanconi Anemia/pathology , Hematopoietic Stem Cells/pathology , Tumor Suppressor Protein p53/metabolism , Adolescent , Adult , Adult Stem Cells/metabolism , Adult Stem Cells/pathology , Aging/pathology , Animals , Bone Marrow/metabolism , Child , Child, Preschool , Disease Models, Animal , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/pathology , Fanconi Anemia Complementation Group C Protein/deficiency , Fanconi Anemia Complementation Group C Protein/metabolism , Fanconi Anemia Complementation Group D2 Protein/metabolism , G1 Phase Cell Cycle Checkpoints , G2 Phase Cell Cycle Checkpoints , Gene Knockdown Techniques , Gene Silencing , Hematopoietic Stem Cells/metabolism , Humans , Infant , Mice , Middle Aged , S Phase
19.
J Thorac Cardiovasc Surg ; 144(2): 467-73, 473.e1-2, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22264418

ABSTRACT

OBJECTIVE: Cardiopulmonary bypass remains associated with significant morbidity and mortality, in part caused by a systemic inflammatory response that is unpredictable and variable among patients. Several limited studies have suggested associations of cytokine plasma levels or gene polymorphisms with outcome after cardiopulmonary bypass. The present study was to determine the relationships between several circulating cytokines and their polymorphisms (single nucleotide polymorphisms), and the occurrence of postoperative clinical events in patients who underwent coronary artery bypass grafting under cardiopulmonary bypass. METHODS: Patients were genotyped for single nucleotide polymorphisms of LTA (Cys13Arg, +252A>G), TNF (-308G>A), IL6 (-597G>A, -572G>C, -174G>C), IL10 (-592C>A, c.∗117C>T), and APOE (Cys112Arg, Arg158Cys). Serum samples were collected preoperatively, immediately after cardiopulmonary bypass, and at different postoperative time points to measure cytokine serum levels by enzyme-linked immunosorbent assay. The clinical end point was the composite of postoperative death, low cardiac output syndrome, myocardial infarction, sepsis, and acute renal insufficiency. RESULTS: Single nucleotide polymorphisms IL6-572GC+CC/IL10-592CC were associated with the clinical end point (P=.032 and P=.009, respectively). In addition to preoperative clinical conditions, the other factor associated with the clinical end point was interleukin-10 plasma levels 24 hours after surgery (P=.017). On the basis of these results, a predictive model of postoperative complications after coronary artery bypass grafting was created. CONCLUSIONS: Our data suggest that focused genetic testing of the IL6-572G>C and IL10-592C>A single nucleotide polymorphisms might be a tool for identifying patients at the highest risk of poor tolerance to the inflammatory response to cardiopulmonary bypass and for implementing strategies to mitigate it, provided the generalization of these tests makes them reasonably affordable and thus favorably shifts their cost-to-benefit ratio.


Subject(s)
Cardiopulmonary Bypass/adverse effects , Coronary Artery Bypass , Cytokines/blood , Cytokines/genetics , Polymorphism, Single Nucleotide , Acute Kidney Injury/epidemiology , Acute Kidney Injury/genetics , Aged , Cardiac Output, Low/epidemiology , Cardiac Output, Low/genetics , Enzyme-Linked Immunosorbent Assay , Female , Genotype , Humans , Male , Middle Aged , Myocardial Infarction/epidemiology , Myocardial Infarction/genetics , Postoperative Complications/epidemiology , Postoperative Complications/genetics , Sepsis/epidemiology , Sepsis/genetics
20.
Blood ; 117(15): e161-70, 2011 Apr 14.
Article in English | MEDLINE | ID: mdl-21325596

ABSTRACT

Fanconi anemia (FA) is a genetic condition associated with bone marrow (BM) failure, myelodysplasia (MDS), and acute myeloid leukemia (AML). We studied 57 FA patients with hypoplastic or aplastic anemia (n = 20), MDS (n = 18), AML (n = 11), or no BM abnormality (n = 8). BM samples were analyzed by karyotype, high-density DNA arrays with respect to paired fibroblasts, and by selected oncogene sequencing. A specific pattern of chromosomal abnormalities was found in MDS/AML, which included 1q+ (44.8%), 3q+ (41.4%), -7/7q (17.2%), and 11q- (13.8%). Moreover, cryptic RUNX1/AML1 lesions (translocations, deletions, or mutations) were observed for the first time in FA (20.7%). Rare mutations of NRAS, FLT3-ITD, MLL-PTD, ERG amplification, and ZFP36L2-PRDM16 translocation, but no TP53, TET2, CBL, NPM1, and CEBPα mutations were found. Frequent homozygosity regions were related not to somatic copy-neutral loss of heterozygosity but to consanguinity, suggesting that homologous recombination is not a common progression mechanism in FA. Importantly, the RUNX1 and other chromosomal/genomic lesions were found at the MDS/AML stages, except for 1q+, which was found at all stages. These data have implications for staging and therapeutic managing in FA patients, and also to analyze the mechanisms of clonal evolution and oncogenesis in a background of genomic instability and BM failure.


Subject(s)
Core Binding Factor Alpha 2 Subunit/genetics , Fanconi Anemia/genetics , Genomic Instability/genetics , Leukemia, Myeloid, Acute/genetics , Myelodysplastic Syndromes/genetics , Adolescent , Adult , Bone Marrow/physiology , Child , Child, Preschool , Fanconi Anemia/complications , Female , Gene Dosage/genetics , Gene Expression Profiling , Gene Expression Regulation, Leukemic , Genes, Tumor Suppressor , Homozygote , Humans , Leukemia, Myeloid, Acute/etiology , Male , Middle Aged , Myelodysplastic Syndromes/etiology , Nucleophosmin , Polymorphism, Single Nucleotide , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...