Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 187(7): 1589-1616, 2024 Mar 28.
Article in English | MEDLINE | ID: mdl-38552609

ABSTRACT

The last 50 years have witnessed extraordinary developments in understanding mechanisms of carcinogenesis, synthesized as the hallmarks of cancer. Despite this logical framework, our understanding of the molecular basis of systemic manifestations and the underlying causes of cancer-related death remains incomplete. Looking forward, elucidating how tumors interact with distant organs and how multifaceted environmental and physiological parameters impinge on tumors and their hosts will be crucial for advances in preventing and more effectively treating human cancers. In this perspective, we discuss complexities of cancer as a systemic disease, including tumor initiation and promotion, tumor micro- and immune macro-environments, aging, metabolism and obesity, cancer cachexia, circadian rhythms, nervous system interactions, tumor-related thrombosis, and the microbiome. Model systems incorporating human genetic variation will be essential to decipher the mechanistic basis of these phenomena and unravel gene-environment interactions, providing a modern synthesis of molecular oncology that is primed to prevent cancers and improve patient quality of life and cancer outcomes.


Subject(s)
Neoplasms , Humans , Carcinogenesis , Microbiota , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/therapy , Obesity/complications , Quality of Life
2.
Mol Cell ; 83(14): 2417-2433.e7, 2023 07 20.
Article in English | MEDLINE | ID: mdl-37348497

ABSTRACT

Aged hematopoietic stem cells (HSCs) display diminished self-renewal and a myeloid differentiation bias. However, the drivers and mechanisms that underpin this fundamental switch are not understood. HSCs produce genotoxic formaldehyde that requires protection by the detoxification enzymes ALDH2 and ADH5 and the Fanconi anemia (FA) DNA repair pathway. We find that the HSCs in young Aldh2-/-Fancd2-/- mice harbor a transcriptomic signature equivalent to aged wild-type HSCs, along with increased epigenetic age, telomere attrition, and myeloid-biased differentiation quantified by single HSC transplantation. In addition, the p53 response is vigorously activated in Aldh2-/-Fancd2-/- HSCs, while p53 deletion rescued this aged HSC phenotype. To further define the origins of the myeloid differentiation bias, we use a GFP genetic reporter to find a striking enrichment of Vwf+ myeloid and megakaryocyte-lineage-biased HSCs. These results indicate that metabolism-derived formaldehyde-DNA damage stimulates the p53 response in HSCs to drive accelerated aging.


Subject(s)
Aging , Aldehydes , DNA Damage , Hematopoiesis , Tumor Suppressor Protein p53 , Animals , Mice , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Aldehydes/metabolism , Transcriptome , Single-Cell Gene Expression Analysis , Hematopoietic Stem Cells/cytology , Myeloid Cells/cytology , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology
3.
Mol Cell ; 80(6): 996-1012.e9, 2020 12 17.
Article in English | MEDLINE | ID: mdl-33147438

ABSTRACT

Reactive aldehydes arise as by-products of metabolism and are normally cleared by multiple families of enzymes. We find that mice lacking two aldehyde detoxifying enzymes, mitochondrial ALDH2 and cytoplasmic ADH5, have greatly shortened lifespans and develop leukemia. Hematopoiesis is disrupted profoundly, with a reduction of hematopoietic stem cells and common lymphoid progenitors causing a severely depleted acquired immune system. We show that formaldehyde is a common substrate of ALDH2 and ADH5 and establish methods to quantify elevated blood formaldehyde and formaldehyde-DNA adducts in tissues. Bone-marrow-derived progenitors actively engage DNA repair but also imprint a formaldehyde-driven mutation signature similar to aging-associated human cancer mutation signatures. Furthermore, we identify analogous genetic defects in children causing a previously uncharacterized inherited bone marrow failure and pre-leukemic syndrome. Endogenous formaldehyde clearance alone is therefore critical for hematopoiesis and in limiting mutagenesis in somatic tissues.


Subject(s)
Alcohol Dehydrogenase/genetics , Aldehyde Dehydrogenase, Mitochondrial/genetics , Formaldehyde/blood , Leukemia/genetics , Adolescent , Aldehydes/blood , Animals , Child , Child, Preschool , DNA Adducts/genetics , DNA Damage/drug effects , DNA Repair/drug effects , Female , Formaldehyde/toxicity , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Humans , Infant , Leukemia/blood , Leukemia/pathology , Male , Mice , Mutation/genetics , Substrate Specificity
4.
Nature ; 600(7887): 158-163, 2021 12.
Article in English | MEDLINE | ID: mdl-34819667

ABSTRACT

Endogenous DNA damage can perturb transcription, triggering a multifaceted cellular response that repairs the damage, degrades RNA polymerase II and shuts down global transcription1-4. This response is absent in the human disease Cockayne syndrome, which is caused by loss of the Cockayne syndrome A (CSA) or CSB proteins5-7. However, the source of endogenous DNA damage and how this leads to the prominent degenerative features of this disease remain unknown. Here we find that endogenous formaldehyde impedes transcription, with marked physiological consequences. Mice deficient in formaldehyde clearance (Adh5-/-) and CSB (Csbm/m; Csb is also known as Ercc6) develop cachexia and neurodegeneration, and succumb to kidney failure, features that resemble human Cockayne syndrome. Using single-cell RNA sequencing, we find that formaldehyde-driven transcriptional stress stimulates the expression of the anorexiogenic peptide GDF15 by a subset of kidney proximal tubule cells. Blocking this response with an anti-GDF15 antibody alleviates cachexia in Adh5-/-Csbm/m mice. Therefore, CSB provides protection to the kidney and brain against DNA damage caused by endogenous formaldehyde, while also suppressing an anorexic endocrine signal. The activation of this signal might contribute to the cachexia observed in Cockayne syndrome as well as chemotherapy-induced anorectic weight loss. A plausible evolutionary purpose for such a response is to ensure aversion to genotoxins in food.


Subject(s)
Cockayne Syndrome , DNA Damage , Formaldehyde/adverse effects , Stress, Physiological/drug effects , Transcription, Genetic/drug effects , Alcohol Dehydrogenase/deficiency , Alcohol Dehydrogenase/metabolism , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Cachexia/complications , Cockayne Syndrome/chemically induced , Cockayne Syndrome/complications , Cockayne Syndrome/genetics , Cockayne Syndrome/pathology , DNA Repair Enzymes/deficiency , Disease Models, Animal , Female , Formaldehyde/metabolism , Growth Differentiation Factor 15/antagonists & inhibitors , Growth Differentiation Factor 15/biosynthesis , Growth Differentiation Factor 15/genetics , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Male , Mice , Poly-ADP-Ribose Binding Proteins/deficiency , Renal Insufficiency/complications , Transcription, Genetic/genetics
5.
Nature ; 579(7800): 603-608, 2020 03.
Article in English | MEDLINE | ID: mdl-32132710

ABSTRACT

Acetaldehyde is a highly reactive, DNA-damaging metabolite that is produced upon alcohol consumption1. Impaired detoxification of acetaldehyde is common in the Asian population, and is associated with alcohol-related cancers1,2. Cells are protected against acetaldehyde-induced damage by DNA crosslink repair, which when impaired causes Fanconi anaemia (FA), a disease resulting in failure to produce blood cells and a predisposition to cancer3,4. The combined inactivation of acetaldehyde detoxification and the FA pathway induces mutation, accelerates malignancies and causes the rapid attrition of blood stem cells5-7. However, the nature of the DNA damage induced by acetaldehyde and how this is repaired remains a key question. Here we generate acetaldehyde-induced DNA interstrand crosslinks and determine their repair mechanism in Xenopus egg extracts. We find that two replication-coupled pathways repair these lesions. The first is the FA pathway, which operates using excision-analogous to the mechanism used to repair the interstrand crosslinks caused by the chemotherapeutic agent cisplatin. However, the repair of acetaldehyde-induced crosslinks results in increased mutation frequency and an altered mutational spectrum compared with the repair of cisplatin-induced crosslinks. The second repair mechanism requires replication fork convergence, but does not involve DNA incisions-instead the acetaldehyde crosslink itself is broken. The Y-family DNA polymerase REV1 completes repair of the crosslink, culminating in a distinct mutational spectrum. These results define the repair pathways of DNA interstrand crosslinks caused by an endogenous and alcohol-derived metabolite, and identify an excision-independent mechanism.


Subject(s)
Acetaldehyde/chemistry , Cross-Linking Reagents/chemistry , DNA Damage , DNA Repair , DNA Replication/physiology , DNA/chemistry , Ethanol/chemistry , Fanconi Anemia/metabolism , Animals , Cisplatin/chemistry , Cisplatin/pharmacology , DNA Damage/drug effects , DNA Replication/drug effects , DNA-Directed DNA Polymerase/metabolism , Ethanol/pharmacology , Mutagenesis/drug effects , Nucleotidyltransferases/metabolism , Point Mutation/drug effects , Point Mutation/genetics , Xenopus , Xenopus Proteins/metabolism
6.
Nature ; 575(7781): 234-237, 2019 11.
Article in English | MEDLINE | ID: mdl-31666700

ABSTRACT

The Fanconi anaemia (FA) pathway repairs DNA damage caused by endogenous and chemotherapy-induced DNA crosslinks, and responds to replication stress1,2. Genetic inactivation of this pathway by mutation of genes encoding FA complementation group (FANC) proteins impairs development, prevents blood production and promotes cancer1,3. The key molecular step in the FA pathway is the monoubiquitination of a pseudosymmetric heterodimer of FANCD2-FANCI4,5 by the FA core complex-a megadalton multiprotein E3 ubiquitin ligase6,7. Monoubiquitinated FANCD2 then recruits additional protein factors to remove the DNA crosslink or to stabilize the stalled replication fork. A molecular structure of the FA core complex would explain how it acts to maintain genome stability. Here we reconstituted an active, recombinant FA core complex, and used cryo-electron microscopy and mass spectrometry to determine its structure. The FA core complex comprises two central dimers of the FANCB and FA-associated protein of 100 kDa (FAAP100) subunits, flanked by two copies of the RING finger subunit, FANCL. These two heterotrimers act as a scaffold to assemble the remaining five subunits, resulting in an extended asymmetric structure. Destabilization of the scaffold would disrupt the entire complex, resulting in a non-functional FA pathway. Thus, the structure provides a mechanistic basis for the low numbers of patients with mutations in FANCB, FANCL and FAAP100. Despite a lack of sequence homology, FANCB and FAAP100 adopt similar structures. The two FANCL subunits are in different conformations at opposite ends of the complex, suggesting that each FANCL has a distinct role. This structural and functional asymmetry of dimeric RING finger domains may be a general feature of E3 ligases. The cryo-electron microscopy structure of the FA core complex provides a foundation for a detailed understanding of its E3 ubiquitin ligase activity and DNA interstrand crosslink repair.


Subject(s)
Cryoelectron Microscopy , Fanconi Anemia Complementation Group Proteins/chemistry , Fanconi Anemia Complementation Group Proteins/ultrastructure , Multiprotein Complexes/chemistry , Multiprotein Complexes/ultrastructure , Protein Subunits/chemistry , Animals , Chickens , Fanconi Anemia/enzymology , Fanconi Anemia Complementation Group L Protein/chemistry , Fanconi Anemia Complementation Group L Protein/ultrastructure , Mass Spectrometry , Models, Molecular , Protein Domains , Protein Multimerization , Structure-Activity Relationship , Ubiquitination
7.
Nature ; 567(7747): 267-272, 2019 03.
Article in English | MEDLINE | ID: mdl-30842657

ABSTRACT

Cells often use multiple pathways to repair the same DNA lesion, and the choice of pathway has substantial implications for the fidelity of genome maintenance. DNA interstrand crosslinks covalently link the two strands of DNA, and thereby block replication and transcription; the cytotoxicity of these crosslinks is exploited for chemotherapy. In Xenopus egg extracts, the collision of replication forks with interstrand crosslinks initiates two distinct repair pathways. NEIL3 glycosylase can cleave the crosslink1; however, if this fails, Fanconi anaemia proteins incise the phosphodiester backbone that surrounds the interstrand crosslink, generating a double-strand-break intermediate that is repaired by homologous recombination2. It is not known how the simpler NEIL3 pathway is prioritized over the Fanconi anaemia pathway, which can cause genomic rearrangements. Here we show that the E3 ubiquitin ligase TRAIP is required for both pathways. When two replisomes converge at an interstrand crosslink, TRAIP ubiquitylates the replicative DNA helicase CMG (the complex of CDC45, MCM2-7 and GINS). Short ubiquitin chains recruit NEIL3 through direct binding, whereas longer chains are required for the unloading of CMG by the p97 ATPase, which enables the Fanconi anaemia pathway. Thus, TRAIP controls the choice between the two known pathways of replication-coupled interstrand-crosslink repair. These results, together with our other recent findings3,4 establish TRAIP as a master regulator of CMG unloading and the response of the replisome to obstacles.


Subject(s)
DNA Helicases/chemistry , DNA Helicases/metabolism , DNA Repair , DNA/chemistry , DNA/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , DNA/biosynthesis , DNA Replication , Female , Humans , Minichromosome Maintenance Complex Component 7/metabolism , N-Glycosyl Hydrolases/metabolism , Protein Binding , Ubiquitin/metabolism , Ubiquitination , Xenopus
8.
Nature ; 553(7687): 171-177, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29323295

ABSTRACT

Haematopoietic stem cells renew blood. Accumulation of DNA damage in these cells promotes their decline, while misrepair of this damage initiates malignancies. Here we describe the features and mutational landscape of DNA damage caused by acetaldehyde, an endogenous and alcohol-derived metabolite. This damage results in DNA double-stranded breaks that, despite stimulating recombination repair, also cause chromosome rearrangements. We combined transplantation of single haematopoietic stem cells with whole-genome sequencing to show that this damage occurs in stem cells, leading to deletions and rearrangements that are indicative of microhomology-mediated end-joining repair. Moreover, deletion of p53 completely rescues the survival of aldehyde-stressed and mutated haematopoietic stem cells, but does not change the pattern or the intensity of genome instability within individual stem cells. These findings characterize the mutation of the stem-cell genome by an alcohol-derived and endogenous source of DNA damage. Furthermore, we identify how the choice of DNA-repair pathway and a stringent p53 response limit the transmission of aldehyde-induced mutations in stem cells.


Subject(s)
Acetaldehyde/metabolism , DNA Breaks, Double-Stranded/drug effects , Ethanol/metabolism , Ethanol/pharmacology , Genomic Instability/drug effects , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/pathology , Mutation , Alcohol Dehydrogenase/deficiency , Alcohol Dehydrogenase/genetics , Alcohol Dehydrogenase/metabolism , Animals , Cell Survival/drug effects , DNA End-Joining Repair , Ethanol/administration & dosage , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Fanconi Anemia/pathology , Fanconi Anemia Complementation Group D2 Protein/deficiency , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group D2 Protein/metabolism , Female , Gene Deletion , Genes, p53/genetics , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Ku Autoantigen/metabolism , Male , Mice , Mice, Inbred C57BL , Recombinational DNA Repair/drug effects , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Whole Genome Sequencing
9.
J Am Chem Soc ; 145(2): 953-959, 2023 01 18.
Article in English | MEDLINE | ID: mdl-36584283

ABSTRACT

DNA interstrand cross-links (ICLs) prevent DNA replication and transcription and can lead to potentially lethal events, such as cancer or bone marrow failure. ICLs are typically repaired by proteins within the Fanconi Anemia (FA) pathway, although the details of the pathway are not fully established. Methods to generate DNA containing ICLs are key to furthering the understanding of DNA cross-link repair. A major route to ICL formation in vivo involves reaction of DNA with acetaldehyde, derived from ethanol metabolism. This reaction forms a three-carbon bridged ICL involving the amino groups of adjacent guanines in opposite strands of a duplex resulting in amino and imino functionalities. A stable reduced form of the ICL has applications in understanding the recognition and repair of these types of adducts. Previous routes to creating DNA duplexes containing these adducts have involved lengthy post-DNA synthesis chemistry followed by reduction of the imine. Here, an efficient and high-yielding approach to the reduced ICL using a novel N2-((R)-4-trifluoroacetamidobutan-2-yl)-2'-deoxyguanosine phosphoramidite is described. Following standard automated DNA synthesis and deprotection, the ICL is formed overnight in over 90% yield upon incubation at room temperature with a complementary oligodeoxyribonucleotide containing 2-fluoro-2'-deoxyinosine. The cross-linked duplex displayed a melting transition 25 °C higher than control sequences. Importantly, we show using the Xenopus egg extract system that an ICL synthesized by this method is repaired by the FA pathway. The simplicity and efficiency of this methodology for preparing reduced acetaldehyde ICLs will facilitate access to these DNA architectures for future studies on cross-link repair.


Subject(s)
Acetaldehyde , DNA , Cross-Linking Reagents , DNA/metabolism , DNA Replication , DNA Repair , DNA Damage
10.
Nature ; 548(7669): 549-554, 2017 08 31.
Article in English | MEDLINE | ID: mdl-28813411

ABSTRACT

The folate-driven one-carbon (1C) cycle is a fundamental metabolic hub in cells that enables the synthesis of nucleotides and amino acids and epigenetic modifications. This cycle might also release formaldehyde, a potent protein and DNA crosslinking agent that organisms produce in substantial quantities. Here we show that supplementation with tetrahydrofolate, the essential cofactor of this cycle, and other oxidation-prone folate derivatives kills human, mouse and chicken cells that cannot detoxify formaldehyde or that lack DNA crosslink repair. Notably, formaldehyde is generated from oxidative decomposition of the folate backbone. Furthermore, we find that formaldehyde detoxification in human cells generates formate, and thereby promotes nucleotide synthesis. This supply of 1C units is sufficient to sustain the growth of cells that are unable to use serine, which is the predominant source of 1C units. These findings identify an unexpected source of formaldehyde and, more generally, indicate that the detoxification of this ubiquitous endogenous genotoxin creates a benign 1C unit that can sustain essential metabolism.


Subject(s)
Carbon/metabolism , Folic Acid/chemistry , Folic Acid/metabolism , Formaldehyde/chemistry , Formaldehyde/metabolism , Metabolic Networks and Pathways , Mutagens/chemistry , Mutagens/metabolism , Alcohol Dehydrogenase/metabolism , Animals , Carbon/deficiency , Cell Line , Chickens , Coenzymes/metabolism , Cross-Linking Reagents/metabolism , DNA Damage , DNA Repair , Humans , Inactivation, Metabolic , Mice , Nucleotides/biosynthesis , Oxidation-Reduction , Serine/chemistry , Serine/metabolism , Tetrahydrofolates/metabolism
12.
Mol Cell ; 60(1): 177-88, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26412304

ABSTRACT

Endogenous formaldehyde is produced by numerous biochemical pathways fundamental to life, and it can crosslink both DNA and proteins. However, the consequences of its accumulation are unclear. Here we show that endogenous formaldehyde is removed by the enzyme alcohol dehydrogenase 5 (ADH5/GSNOR), and Adh5(-/-) mice therefore accumulate formaldehyde adducts in DNA. The repair of this damage is mediated by FANCD2, a DNA crosslink repair protein. Adh5(-/-)Fancd2(-/-) mice reveal an essential requirement for these protection mechanisms in hematopoietic stem cells (HSCs), leading to their depletion and precipitating bone marrow failure. More widespread formaldehyde-induced DNA damage also causes karyomegaly and dysfunction of hepatocytes and nephrons. Bone marrow transplantation not only rescued hematopoiesis but, surprisingly, also preserved nephron function. Nevertheless, all of these animals eventually developed fatal malignancies. Formaldehyde is therefore an important source of endogenous DNA damage that is counteracted in mammals by a conserved protection mechanism.


Subject(s)
Alcohol Dehydrogenase/metabolism , Carcinogens/metabolism , Fanconi Anemia Complementation Group D2 Protein/metabolism , Formaldehyde/metabolism , Mutagens/metabolism , Alcohol Dehydrogenase/genetics , Animals , Cells, Cultured , DNA Adducts/metabolism , Fanconi Anemia Complementation Group D2 Protein/genetics , Gene Knockout Techniques , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Kidney/metabolism , Kidney/pathology , Liver/metabolism , Liver/pathology , Mice
13.
Mol Cell ; 57(1): 150-64, 2015 Jan 08.
Article in English | MEDLINE | ID: mdl-25557546

ABSTRACT

We show that central components of the Fanconi anemia (FA) DNA repair pathway, the tumor suppressor proteins FANCI and FANCD2 (the ID complex), are SUMOylated in response to replication fork stalling. The ID complex is SUMOylated in a manner that depends on the ATR kinase, the FA ubiquitin ligase core complex, and the SUMO E3 ligases PIAS1/PIAS4 and is antagonized by the SUMO protease SENP6. SUMOylation of the ID complex drives substrate selectivity by triggering its polyubiquitylation by the SUMO-targeted ubiquitin ligase RNF4 to promote its removal from sites of DNA damage via the DVC1-p97 ubiquitin segregase complex. Deregulation of ID complex SUMOylation compromises cell survival following replication stress. Our results uncover a regulatory role for SUMOylation in the FA pathway, and we propose that ubiquitin-SUMO signaling circuitry is a mechanism that contributes to the balance of activated ID complex dosage at sites of DNA damage.


Subject(s)
Cysteine Endopeptidases/metabolism , Fanconi Anemia Complementation Group D2 Protein/metabolism , Fanconi Anemia Complementation Group Proteins/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Ubiquitin/metabolism , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line, Tumor , Cysteine Endopeptidases/genetics , DNA Damage , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group Proteins/genetics , Gene Expression Regulation , HEK293 Cells , Humans , Hydroxyurea/pharmacology , Nuclear Proteins/genetics , Poly-ADP-Ribose Binding Proteins , Protein Binding , Protein Inhibitors of Activated STAT/genetics , Protein Inhibitors of Activated STAT/metabolism , Protein Interaction Domains and Motifs , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction , Small Ubiquitin-Related Modifier Proteins/genetics , Small Ubiquitin-Related Modifier Proteins/metabolism , Sumoylation , Transcription Factors/genetics , Ubiquitin/genetics , Ubiquitination
14.
Mol Cell ; 55(6): 807-817, 2014 Sep 18.
Article in English | MEDLINE | ID: mdl-25155611

ABSTRACT

Maternal metabolism provides essential nutrients to enable embryonic development. However, both mother and embryo produce reactive metabolites that can damage DNA. Here we discover how the embryo is protected from these genotoxins. Pregnant mice lacking Aldh2, a key enzyme that detoxifies reactive aldehydes, cannot support the development of embryos lacking the Fanconi anemia DNA repair pathway gene Fanca. Remarkably, transferring Aldh2(-/-)Fanca(-/-) embryos into wild-type mothers suppresses developmental defects and rescues embryonic lethality. These rescued neonates have severely depleted hematopoietic stem and progenitor cells, indicating that despite intact maternal aldehyde catabolism, fetal Aldh2 is essential for hematopoiesis. Hence, maternal and fetal aldehyde detoxification protects the developing embryo from DNA damage. Failure of this genome preservation mechanism might explain why birth defects and bone marrow failure occur in Fanconi anemia, and may have implications for fetal well-being in the many women in Southeast Asia that are genetically deficient in ALDH2.


Subject(s)
Acetaldehyde/metabolism , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase/metabolism , Embryo, Mammalian/metabolism , Ethanol/toxicity , Fanconi Anemia Complementation Group A Protein/genetics , Fanconi Anemia/pathology , Acetaldehyde/toxicity , Aldehyde Dehydrogenase 1 Family , Aldehyde Dehydrogenase, Mitochondrial , Animals , Animals, Newborn , DNA Damage , Disease Models, Animal , Embryo, Mammalian/embryology , Female , Genome , Hematopoietic Stem Cells/metabolism , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Mice, Inbred C57BL , Pregnancy , Retinal Dehydrogenase/genetics , Retinal Dehydrogenase/metabolism
15.
Mol Cell ; 54(3): 472-84, 2014 May 08.
Article in English | MEDLINE | ID: mdl-24726326

ABSTRACT

SLX4 binds to three nucleases (XPF-ERCC1, MUS81-EME1, and SLX1), and its deficiency leads to genomic instability, sensitivity to DNA crosslinking agents, and Fanconi anemia. However, it is not understood how SLX4 and its associated nucleases act in DNA crosslink repair. Here, we uncover consequences of mouse Slx4 deficiency and reveal its function in DNA crosslink repair. Slx4-deficient mice develop epithelial cancers and have a contracted hematopoietic stem cell pool. The N-terminal domain of SLX4 (mini-SLX4) that only binds to XPF-ERCC1 is sufficient to confer resistance to DNA crosslinking agents. Recombinant mini-SLX4 enhances XPF-ERCC1 nuclease activity up to 100-fold, directing specificity toward DNA forks. Mini-SLX4-XPF-ERCC1 also vigorously stimulates dual incisions around a DNA crosslink embedded in a synthetic replication fork, an essential step in the repair of this lesion. These observations define vertebrate SLX4 as a tumor suppressor, which activates XPF-ERCC1 nuclease specificity in DNA crosslink repair.


Subject(s)
DNA Repair , DNA-Binding Proteins/metabolism , Endonucleases/metabolism , Recombinases/physiology , Animals , Base Sequence , Bone Marrow Cells/pathology , DNA Adducts/chemistry , DNA Damage , DNA-Binding Proteins/chemistry , Endonucleases/chemistry , Hematopoietic Stem Cells/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/enzymology , Nucleic Acid Conformation , Tumor Suppressor Proteins
16.
Mol Cell ; 54(5): 858-69, 2014 Jun 05.
Article in English | MEDLINE | ID: mdl-24905007

ABSTRACT

Fanconi anaemia (FA) is a cancer predisposition syndrome characterized by cellular sensitivity to DNA interstrand crosslinkers. The molecular defect in FA is an impaired DNA repair pathway. The critical event in activating this pathway is monoubiquitination of FANCD2. In vivo, a multisubunit FA core complex catalyzes this step, but its mechanism is unclear. Here, we report purification of a native avian FA core complex and biochemical reconstitution of FANCD2 monoubiquitination. This demonstrates that the catalytic FANCL E3 ligase subunit must be embedded within the complex for maximal activity and site specificity. We genetically and biochemically define a minimal subcomplex comprising just three proteins (FANCB, FANCL, and FAAP100) that functions as the monoubiquitination module. Residual FANCD2 monoubiquitination activity is retained in cells defective for other FA core complex subunits. This work describes the in vitro reconstitution and characterization of this multisubunit monoubiquitin E3 ligase, providing key insight into the conserved FA DNA repair pathway.


Subject(s)
Avian Proteins/metabolism , Chickens/genetics , Fanconi Anemia Complementation Group D2 Protein/metabolism , Ubiquitination , Animals , Avian Proteins/chemistry , Avian Proteins/genetics , Cell Line , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group D2 Protein/chemistry , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group L Protein/chemistry , Fanconi Anemia Complementation Group L Protein/metabolism , Fanconi Anemia Complementation Group Proteins/chemistry , Fanconi Anemia Complementation Group Proteins/genetics , Fanconi Anemia Complementation Group Proteins/metabolism , Humans , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/metabolism
17.
J Cell Sci ; 129(24): 4449-4454, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27872153

ABSTRACT

As time passes, mutations accumulate in the genomes of all living organisms. These changes promote genetic diversity, but also precipitate ageing and the initiation of cancer. Food is a common source of mutagens, but little is known about how nutritional factors cause lasting genetic changes in the consuming organism. Here, we describe an unusual genetic interaction between DNA repair in the unicellular amoeba Dictyostelium discoideum and its natural bacterial food source. We found that Dictyostelium deficient in the DNA repair nuclease Xpf (xpf-) display a severe and specific growth defect when feeding on bacteria. Despite being proficient in the phagocytosis and digestion of bacteria, over time, xpf- Dictyostelium feeding on bacteria cease to grow and in many instances die. The Xpf nuclease activity is required for sustained growth using a bacterial food source. Furthermore, the ingestion of this food source leads to a striking accumulation of mutations in the genome of xpf- Dictyostelium This work therefore establishes Dictyostelium as a model genetic system to dissect nutritional genotoxicity, providing insight into how phagocytosis can induce mutagenesis and compromise survival fitness.


Subject(s)
Dictyostelium/metabolism , Mutagenesis , Phagocytosis , Protozoan Proteins/metabolism , Amino Acid Sequence , DNA Repair/genetics , Dictyostelium/cytology , Dictyostelium/growth & development , Phagocytosis/genetics , Protozoan Proteins/chemistry , Protozoan Proteins/genetics
18.
Nature ; 489(7417): 571-5, 2012 Sep 27.
Article in English | MEDLINE | ID: mdl-22922648

ABSTRACT

Haematopoietic stem cells (HSCs) regenerate blood cells throughout the lifespan of an organism. With age, the functional quality of HSCs declines, partly owing to the accumulation of damaged DNA. However, the factors that damage DNA and the protective mechanisms that operate in these cells are poorly understood. We have recently shown that the Fanconi anaemia DNA-repair pathway counteracts the genotoxic effects of reactive aldehydes. Mice with combined inactivation of aldehyde catabolism (through Aldh2 knockout) and the Fanconi anaemia DNA-repair pathway (Fancd2 knockout) display developmental defects, a predisposition to leukaemia, and are susceptible to the toxic effects of ethanol-an exogenous source of acetaldehyde. Here we report that aged Aldh2(-/-) Fancd2(-/-) mutant mice that do not develop leukaemia spontaneously develop aplastic anaemia, with the concomitant accumulation of damaged DNA within the haematopoietic stem and progenitor cell (HSPC) pool. Unexpectedly, we find that only HSPCs, and not more mature blood precursors, require Aldh2 for protection against acetaldehyde toxicity. Additionally, the aldehyde-oxidizing activity of HSPCs, as measured by Aldefluor stain, is due to Aldh2 and correlates with this protection. Finally, there is more than a 600-fold reduction in the HSC pool of mice deficient in both Fanconi anaemia pathway-mediated DNA repair and acetaldehyde detoxification. Therefore, the emergence of bone marrow failure in Fanconi anaemia is probably due to aldehyde-mediated genotoxicity restricted to the HSPC pool. These findings identify a new link between endogenous reactive metabolites and DNA damage in HSCs, and define the protective mechanisms that counteract this threat.


Subject(s)
Aldehydes/toxicity , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Mutagens/toxicity , Acetaldehyde/metabolism , Acetaldehyde/toxicity , Aging , Aldehyde Dehydrogenase/deficiency , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase/metabolism , Aldehyde Dehydrogenase, Mitochondrial , Aldehydes/metabolism , Animals , Bone Marrow/pathology , DNA Damage/drug effects , DNA Damage/genetics , DNA Repair , Ethanol/toxicity , Fanconi Anemia/pathology , Fanconi Anemia Complementation Group D2 Protein/deficiency , Fanconi Anemia Complementation Group D2 Protein/genetics , Female , Hematopoietic Stem Cells/enzymology , Hematopoietic Stem Cells/metabolism , Kaplan-Meier Estimate , Leukemia/metabolism , Leukemia/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
19.
J Am Chem Soc ; 139(15): 5338-5350, 2017 04 19.
Article in English | MEDLINE | ID: mdl-28375637

ABSTRACT

Formaldehyde (FA) is a reactive signaling molecule that is continuously produced through a number of central biological pathways spanning epigenetics to one-carbon metabolism. On the other hand, aberrant, elevated levels of FA are implicated in disease states ranging from asthma to neurodegenerative disorders. In this context, fluorescence-based probes for FA imaging are emerging as potentially powerful chemical tools to help disentangle the complexities of FA homeostasis and its physiological and pathological contributions. Currently available FA indicators require direct modification of the fluorophore backbone through complex synthetic considerations to enable FA detection, often limiting the generalization of designs to other fluorophore classes. To address this challenge, we now present the rational, iterative development of a general reaction-based trigger utilizing 2-aza-Cope reactivity for selective and sensitive detection of FA in living systems. Specifically, we developed a homoallylamine functionality that can undergo a subsequent self-immolative ß-elimination, creating a FA-responsive trigger that is capable of masking a phenol on a fluorophore or any other potential chemical scaffold for related imaging and/or therapeutic applications. We demonstrate the utility of this trigger by creating a series of fluorescent probes for FA with excitation and emission wavelengths that span the UV to visible spectral regions through caging of a variety of dye units. In particular, Formaldehyde Probe 573 (FAP573), based on a resorufin scaffold, is the most red-shifted and FA sensitive in this series in terms of signal-to-noise responses and enables identification of alcohol dehydrogenase 5 (ADH5) as an enzyme that regulates FA metabolism in living cells. The results provide a starting point for the broader use of 2-aza-Cope reactivity for probing and manipulating FA biology.


Subject(s)
Aza Compounds/chemistry , Formaldehyde/analysis , Formaldehyde/chemistry , Optical Imaging , Cell Survival , HEK293 Cells , Humans , Molecular Structure
20.
Nature ; 475(7354): 53-8, 2011 Jul 06.
Article in English | MEDLINE | ID: mdl-21734703

ABSTRACT

Reactive aldehydes are common carcinogens. They are also by-products of several metabolic pathways and, without enzymatic catabolism, may accumulate and cause DNA damage. Ethanol, which is metabolised to acetaldehyde, is both carcinogenic and teratogenic in humans. Here we find that the Fanconi anaemia DNA repair pathway counteracts acetaldehyde-induced genotoxicity in mice. Our results show that the acetaldehyde-catabolising enzyme Aldh2 is essential for the development of Fancd2(-/-) embryos. Nevertheless, acetaldehyde-catabolism-competent mothers (Aldh2(+/-)) can support the development of double-mutant (Aldh2(-/-)Fancd2(-/-)) mice. However, these embryos are unusually sensitive to ethanol exposure in utero, and ethanol consumption by postnatal double-deficient mice rapidly precipitates bone marrow failure. Lastly, Aldh2(-/-)Fancd2(-/-) mice spontaneously develop acute leukaemia. Acetaldehyde-mediated DNA damage may critically contribute to the genesis of fetal alcohol syndrome in fetuses, as well as to abnormal development, haematopoietic failure and cancer predisposition in Fanconi anaemia patients.


Subject(s)
Aldehydes/antagonists & inhibitors , Aldehydes/toxicity , Fanconi Anemia Complementation Group D2 Protein/metabolism , Acetaldehyde/metabolism , Acetaldehyde/toxicity , Aldehyde Dehydrogenase/deficiency , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase/metabolism , Aldehyde Dehydrogenase, Mitochondrial , Aldehydes/metabolism , Alleles , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Bone Marrow/drug effects , Bone Marrow/pathology , Bone Marrow/physiopathology , Cell Line , Cell Survival/drug effects , Chickens , Clone Cells/drug effects , DNA Damage/genetics , DNA Repair/genetics , Embryo Loss/chemically induced , Embryo Loss/etiology , Embryo, Mammalian/abnormalities , Embryo, Mammalian/drug effects , Embryo, Mammalian/embryology , Ethanol/metabolism , Ethanol/toxicity , Fanconi Anemia/genetics , Fanconi Anemia/pathology , Fanconi Anemia Complementation Group D2 Protein/deficiency , Fanconi Anemia Complementation Group D2 Protein/genetics , Female , Fetal Alcohol Spectrum Disorders/etiology , Gene Deletion , Genes, Essential , Hematopoiesis/drug effects , Male , Mice , Mice, Inbred C57BL , Precursor Cell Lymphoblastic Leukemia-Lymphoma/chemically induced , Precursor Cell Lymphoblastic Leukemia-Lymphoma/etiology , Pregnancy , Teratogens/metabolism , Teratogens/toxicity , Weaning
SELECTION OF CITATIONS
SEARCH DETAIL