Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
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
2.
Biochem Pharmacol ; 194: 114801, 2021 12.
Article in English | MEDLINE | ID: mdl-34678222

ABSTRACT

The regulation of the apoptotic pathway is one of the most studied mechanisms regarding cancer cell resistance. Many mutations have been linked to drug resistance. The DNA fragmentation factor 40 (DFF40) has been gaining interest regarding cancer cell response to chemotherapy and patient outcomes. Glioblastomas and uterine leiomyosarcomas have been shown to have a downregulation of DFF40 expression, conferring a poor patient prognosis. In concordance with these observations, in this study, we showed that DFF40 gene is also downregulated in breast, endocervical, ovarian, lung, pancreas and glioblastomas. DFF40 is the endonuclease responsible of DNA fragmentation during apoptosis. In this study, we sought to determine if a DFF40 deficiency in Jurkat T cells could impact the sensitivity to conventional chemotherapy drugs. CRISPR-cas9 generated DFF40 knockout (DFF40 KO) stable Jurkat cells and wild-type (DFF40 WT) cells were treated with different antimetabolites and topoisomerase II (TOP2) inhibitors, and cell viability was subsequently assessed. DFF40 deficient cells show chemoresistance to antimetabolites (e.g. methotrexate, 6-mercaptopurine and cytarabine) and surprisingly, they are more sensitive to TOP2 inhibitors (e.g. etoposide and teniposide). DFF40 deficient cells exposed to cytarabine present lower phosphatidylserine translocation levels to the outer cell membrane layer. Etoposide exposure in DFF40 deficient cells induces higher mortality levels and downregulation of Bcl-xL cells compared to DFF40 expressing T cells. The abolition of DFF40 expression in Jurkat cells significantly impairs histone H2AX phosphorylation following etoposide and cytarabine treatments. Our findings suggest that DFF40 is a novel key target in cancer cell resistance that potentially regulates genomic stability.


Subject(s)
Apoptosis/physiology , Deoxyribonucleases/deficiency , Drug Resistance, Neoplasm/physiology , Neoplasms/metabolism , Poly-ADP-Ribose Binding Proteins/deficiency , Signal Transduction/physiology , T-Lymphocytes/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Proliferation/physiology , Deoxyribonucleases/genetics , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , Gene Knockout Techniques , HeLa Cells , Humans , Jurkat Cells , Neoplasms/drug therapy , Neoplasms/genetics , Poly-ADP-Ribose Binding Proteins/genetics , Signal Transduction/drug effects , T-Lymphocytes/drug effects
3.
J Immunol ; 206(10): 2453-2467, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33941659

ABSTRACT

The detection of intracellular nucleic acids is a fundamental mechanism of host defense against infections. The dysregulated nucleic acid sensing, however, is a major cause for a number of autoimmune diseases. In this study, we report that GTPase-activating protein SH3 domain-binding protein 1 (G3BP1) is critical for both intracellular DNA- and RNA-induced immune responses. We found that in both human and mouse cells, the deletion of G3BP1 led to the dampened cGAS activation by DNA and the insufficient binding of RNA by RIG-I. We further found that resveratrol (RSVL), a natural compound found in grape skin, suppressed both intracellular DNA- and RNA-induced type I IFN production through inhibiting G3BP1. Importantly, using experimental mouse models for Aicardi-Goutières syndrome, an autoimmune disorder found in humans, we demonstrated that RSVL effectively alleviated intracellular nucleic acid-stimulated autoimmune responses. Thus, our study demonstrated a broader role of G3BP1 in sensing different kinds of intracellular nucleic acids and presented RSVL as a potential treatment for autoimmune conditions caused by dysregulated nucleic acid sensing.


Subject(s)
Autoimmunity/genetics , DNA Helicases/deficiency , DNA Helicases/metabolism , Intracellular Space/metabolism , Nucleic Acids/metabolism , Poly-ADP-Ribose Binding Proteins/deficiency , Poly-ADP-Ribose Binding Proteins/metabolism , RNA Helicases/deficiency , RNA Helicases/metabolism , RNA Recognition Motif Proteins/deficiency , RNA Recognition Motif Proteins/metabolism , Signal Transduction/genetics , A549 Cells , Animals , Autoimmunity/drug effects , Cell Survival/drug effects , DNA Helicases/antagonists & inhibitors , DNA Helicases/genetics , Fibroblasts/metabolism , Gene Knockout Techniques , HEK293 Cells , Humans , Intracellular Space/immunology , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Poly-ADP-Ribose Binding Proteins/antagonists & inhibitors , Poly-ADP-Ribose Binding Proteins/genetics , RNA Helicases/antagonists & inhibitors , RNA Helicases/genetics , RNA Recognition Motif Proteins/antagonists & inhibitors , RNA Recognition Motif Proteins/genetics , Resveratrol/administration & dosage , Signal Transduction/immunology , Transfection
4.
Aging Cell ; 19(12): e13268, 2020 12.
Article in English | MEDLINE | ID: mdl-33166073

ABSTRACT

Cockayne syndrome (CS) is a rare premature aging disease, most commonly caused by mutations of the genes encoding the CSA or CSB proteins. CS patients display cachectic dwarfism and severe neurological manifestations and have an average life expectancy of 12 years. The CS proteins are involved in transcription and DNA repair, with the latter including transcription-coupled nucleotide excision repair (TC-NER). However, there is also evidence for mitochondrial dysfunction in CS, which likely contributes to the severe premature aging phenotype of this disease. While damaged mitochondria and impaired mitophagy were characterized in mice with CSB deficiency, such changes in the CS nematode model and CS patients are not fully known. Our cross-species transcriptomic analysis in CS postmortem brain tissue, CS mouse, and nematode models shows that mitochondrial dysfunction is indeed a common feature in CS. Restoration of mitochondrial dysfunction through NAD+ supplementation significantly improved lifespan and healthspan in the CS nematodes, highlighting mitochondrial dysfunction as a major driver of the aging features of CS. In cerebellar samples from CS patients, we found molecular signatures of dysfunctional mitochondrial dynamics and impaired mitophagy/autophagy. In primary cells depleted for CSA or CSB, this dysfunction can be corrected with supplementation of NAD+ precursors. Our study provides support for the interconnection between major causative aging theories, DNA damage accumulation, mitochondrial dysfunction, and compromised mitophagy/autophagy. Together, these three agents contribute to an accelerated aging program that can be averted by cellular NAD+ restoration.


Subject(s)
Cockayne Syndrome/metabolism , DNA Helicases/metabolism , DNA Repair Enzymes/metabolism , Mitochondria/metabolism , NAD/metabolism , Poly-ADP-Ribose Binding Proteins/metabolism , Transcription Factors/metabolism , AMP-Activated Protein Kinases/metabolism , Aging, Premature/genetics , Aging, Premature/metabolism , Animals , Animals, Genetically Modified , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Cerebellum/metabolism , Cockayne Syndrome/genetics , Cockayne Syndrome/pathology , DNA Helicases/deficiency , DNA Helicases/genetics , DNA Repair Enzymes/deficiency , DNA Repair Enzymes/genetics , Disease Models, Animal , Humans , Longevity/genetics , Longevity/physiology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mitochondria/pathology , Oligonucleotide Array Sequence Analysis , Poly-ADP-Ribose Binding Proteins/deficiency , Poly-ADP-Ribose Binding Proteins/genetics , Signal Transduction , Transcription Factors/deficiency , Transcription Factors/genetics
5.
Nat Commun ; 11(1): 4979, 2020 10 05.
Article in English | MEDLINE | ID: mdl-33020468

ABSTRACT

Cellular senescence is a known driver of carcinogenesis and age-related diseases, yet senescence is required for various physiological processes. However, the mechanisms and factors that control the negative effects of senescence while retaining its benefits are still elusive. Here, we show that the rasGAP SH3-binding protein 1 (G3BP1) is required for the activation of the senescent-associated secretory phenotype (SASP). During senescence, G3BP1 achieves this effect by promoting the association of the cyclic GMP-AMP synthase (cGAS) with cytosolic chromatin fragments. In turn, G3BP1, through cGAS, activates the NF-κB and STAT3 pathways, promoting SASP expression and secretion. G3BP1 depletion or pharmacological inhibition impairs the cGAS-pathway preventing the expression of SASP factors without affecting cell commitment to senescence. These SASPless senescent cells impair senescence-mediated growth of cancer cells in vitro and tumor growth in vivo. Our data reveal that G3BP1 is required for SASP expression and that SASP secretion is a primary mediator of senescence-associated tumor growth.


Subject(s)
Cellular Senescence/physiology , DNA Helicases/metabolism , Neoplasms/pathology , Poly-ADP-Ribose Binding Proteins/metabolism , RNA Helicases/metabolism , RNA Recognition Motif Proteins/metabolism , A549 Cells , Animals , Carcinogenesis , Cell Line , Cell Movement , Cytokines/metabolism , DNA Helicases/antagonists & inhibitors , DNA Helicases/deficiency , Humans , Inflammation , Mice , Neoplasms/metabolism , Nucleotidyltransferases/metabolism , Poly-ADP-Ribose Binding Proteins/antagonists & inhibitors , Poly-ADP-Ribose Binding Proteins/deficiency , RNA Helicases/antagonists & inhibitors , RNA Helicases/deficiency , RNA Recognition Motif Proteins/antagonists & inhibitors , RNA Recognition Motif Proteins/deficiency , STAT3 Transcription Factor/metabolism , Signal Transduction , Transcription Factor RelA/metabolism
6.
Nature ; 581(7807): 209-214, 2020 05.
Article in English | MEDLINE | ID: mdl-32405004

ABSTRACT

Intracellular bodies such as nucleoli, Cajal bodies and various signalling assemblies represent membraneless organelles, or condensates, that form via liquid-liquid phase separation (LLPS)1,2. Biomolecular interactions-particularly homotypic interactions mediated by self-associating intrinsically disordered protein regions-are thought to underlie the thermodynamic driving forces for LLPS, forming condensates that can facilitate the assembly and processing of biochemically active complexes, such as ribosomal subunits within the nucleolus. Simplified model systems3-6 have led to the concept that a single fixed saturation concentration is a defining feature of endogenous LLPS7-9, and has been suggested as a mechanism for intracellular concentration buffering2,7,8,10. However, the assumption of a fixed saturation concentration remains largely untested within living cells, in which the richly multicomponent nature of condensates could complicate this simple picture. Here we show that heterotypic multicomponent interactions dominate endogenous LLPS, and give rise to nucleoli and other condensates that do not exhibit a fixed saturation concentration. As the concentration of individual components is varied, their partition coefficients change in a manner that can be used to determine the thermodynamic free energies that underlie LLPS. We find that heterotypic interactions among protein and RNA components stabilize various archetypal intracellular condensates-including the nucleolus, Cajal bodies, stress granules and P-bodies-implying that the composition of condensates is finely tuned by the thermodynamics of the underlying biomolecular interaction network. In the context of RNA-processing condensates such as the nucleolus, this manifests in the selective exclusion of fully assembled ribonucleoprotein complexes, providing a thermodynamic basis for vectorial ribosomal RNA flux out of the nucleolus. This methodology is conceptually straightforward and readily implemented, and can be broadly used to extract thermodynamic parameters from microscopy images. These approaches pave the way for a deeper understanding of the thermodynamics of multicomponent intracellular phase behaviour and its interplay with the nonequilibrium activity that is characteristic of endogenous condensates.


Subject(s)
Intracellular Space/chemistry , Intracellular Space/metabolism , Organelles/chemistry , Organelles/metabolism , Thermodynamics , Adaptor Proteins, Signal Transducing/deficiency , Cell Nucleolus/chemistry , Cell Nucleolus/metabolism , Coiled Bodies/chemistry , Coiled Bodies/metabolism , Cytoplasmic Granules/chemistry , Cytoplasmic Granules/metabolism , DNA Helicases/deficiency , HeLa Cells , Humans , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Nucleophosmin , Phase Transition , Poly-ADP-Ribose Binding Proteins/deficiency , RNA Helicases/deficiency , RNA Recognition Motif Proteins/deficiency , RNA, Ribosomal/chemistry , RNA, Ribosomal/metabolism , RNA-Binding Proteins , Ribosomes/chemistry , Ribosomes/metabolism
7.
Sci Rep ; 9(1): 12896, 2019 09 09.
Article in English | MEDLINE | ID: mdl-31501480

ABSTRACT

The aberrant accumulation of ubiquitinated protein aggregates in cells plays a critical role in the pathogenesis of several degenerative diseases, including Parkinson disease (PD) and cystic fibrosis (CF). In this study, we found that Ras GTPase-activating protein-binding protein 1 (G3BP1) inhibits ubiquitinated protein aggregations induced by p62 and USP10 in cultured cells. p62 is a ubiquitin receptor, and p62 and its binding partner USP10 have been shown to augment ubiquitinated protein aggregation. G3BP1 interacted with p62 and USP10 and inhibited p62/USP10-induced protein aggregation. The G3BP1 inhibition of protein aggregations targeted two aggregation-prone proteins, α-synuclein and CFTR-ΔF508, which are causative factors of PD and CF, respectively. G3BP1 depletion increased the amounts of ubiquitinated α-synuclein and CFTR-ΔF508 protein. A proteasome reporter indicated that G3BP1 depletion inhibits the proteasome activity. We herein present evidence that G3BP1, p62 and USP10 together control ubiquitinated protein toxicity by controlling both ubiquitination and aggregation. Taken together, these results suggest that G3BP1, p62 and USP10 could be therapeutic targets for ubiquitinated protein aggregation disorders, including PD and CF.


Subject(s)
DNA Helicases/deficiency , DNA Helicases/genetics , Gene Knockout Techniques , Poly-ADP-Ribose Binding Proteins/deficiency , Poly-ADP-Ribose Binding Proteins/genetics , RNA Helicases/deficiency , RNA Helicases/genetics , RNA Recognition Motif Proteins/deficiency , RNA Recognition Motif Proteins/genetics , RNA-Binding Proteins/metabolism , Ubiquitin Thiolesterase/metabolism , Ubiquitination , Cell Line , Humans , Proteasome Endopeptidase Complex/metabolism , alpha-Synuclein/metabolism
8.
Apoptosis ; 24(5-6): 404-413, 2019 06.
Article in English | MEDLINE | ID: mdl-30997620

ABSTRACT

Genotoxic anti-cancer therapies such as chemotherapy and radiotherapy can contribute to an increase in second malignancies in cancer survivors due to their oncogenic effects on non-cancerous cells. Inhibition of histone deacetylase (HDAC) proteins or the proteasome differ from chemotherapy in that they eliminate cancer cells by regulating gene expression or cellular protein equilibrium, respectively. As members of these drug classes have been approved for clinical use in recent times, we investigated whether these two drug classes exhibit similar mutagenic capabilities as chemotherapy. The HDAC inhibitors vorinostat/SAHA and romidepsin/FK288 were found to induce DNA damage, and mis-repair of this damage manifested into mutations in clonogenically viable surviving cells. DNA damage and mutations were also detected in cells treated with the proteasome inhibitor bortezomib. Exposure to both drug classes stimulated caspase activation consistent with apoptotic cell death. Inhibition of caspases protected cells from bortezomib-induced acute (but not clonogenic) death and mutagenesis, implying caspases were required for the mutagenic action of bortezomib. This was also observed for second generation proteasome inhibitors. Cells deficient in caspase-activated DNase (CAD) also failed to acquire DNA damage or mutations following treatment with bortezomib. Surprisingly, vorinostat and romidepsin maintained an equivalent level of killing and mutagenic ability regardless of caspase or CAD activity. Our findings indicate that both drug classes harbour mutagenic potential in vitro. If recapitulated in vivo, the mutagenicity of these agents may influence the treatment of cancer patients who are more susceptible to oncogenic mutations due to dysfunctional DNA repair pathways.


Subject(s)
Caspases/metabolism , Deoxyribonucleases/metabolism , Histone Deacetylase Inhibitors/pharmacology , Mutation/drug effects , Poly-ADP-Ribose Binding Proteins/metabolism , Proteasome Inhibitors/pharmacology , Apoptosis/drug effects , Bortezomib/pharmacology , Caspase Inhibitors/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , DNA Damage , Deoxyribonucleases/deficiency , Depsipeptides/pharmacology , Humans , Hypoxanthine Phosphoribosyltransferase/genetics , Hypoxanthine Phosphoribosyltransferase/metabolism , Mitochondria/drug effects , Mitochondria/microbiology , Mutagenesis , Poly-ADP-Ribose Binding Proteins/deficiency , Signal Transduction/drug effects , Vorinostat/pharmacology
9.
Cell Cycle ; 18(8): 880-888, 2019 04.
Article in English | MEDLINE | ID: mdl-30907229

ABSTRACT

The double-stranded telomeric binding protein TRF2 is expressed in many human cancers at elevated levels. Moreover, experimental overexpression of TRF2 in human cells causes replication stalling in telomeric tracts, which leads to drastic telomere shortening and fusion of deprotected chromosome ends. To understand which end joining pathway is involved in mediating these chromosome fusions, we overexpressed TRF2 in human HCT116 cell lines that were deficient for the DNA Ligase 4 (Lig4)-dependent classical non-homologous end joining (C-NHEJ) or the DNA Ligase 3 (Lig3)-dependent alternative non-homologous end joining (A-NHEJ) pathway. Surprisingly, abrogation of either Lig4 or nuclear Lig3 significantly reduced inter-chromosomal fusion of drastically shortened telomeres, suggesting that both the C-NHEJ and A-NHEJ pathways are involved in mediating this type of fusion. Fusion between deprotected sister chromatids, however, only required the Lig3-dependent A-NHEJ pathway. Interestingly, a previous study reported similar end joining pathway requirements for the fusion of critically shortened telomeres during a telomere attrition-based cellular crisis. We speculate that, as in cellular crisis, the same repair pathway(s) may drive clonal and genomic evolution in human cancers containing elevated TRF2 levels.


Subject(s)
Colorectal Neoplasms/metabolism , DNA End-Joining Repair/genetics , Telomere Shortening/genetics , Telomere/metabolism , Telomeric Repeat Binding Protein 2/metabolism , Chromatids/metabolism , DNA Ligase ATP/deficiency , Evolution, Molecular , Genetic Vectors , Genomic Instability/genetics , HCT116 Cells , Humans , Plasmids/genetics , Poly-ADP-Ribose Binding Proteins/deficiency , Sister Chromatid Exchange/genetics , Telomeric Repeat Binding Protein 2/genetics
10.
Sci Transl Med ; 10(456)2018 08 29.
Article in English | MEDLINE | ID: mdl-30158153

ABSTRACT

Cockayne syndrome (CS), a hereditary form of premature aging predominantly caused by mutations in the csb gene, affects multiple organs including skin where it manifests with hypersensitivity toward ultraviolet (UV) radiation and loss of subcutaneous fat. There is no curative treatment for CS, and its pathogenesis is only partially understood. Originally considered for its role in DNA repair, Cockayne syndrome group B (CSB) protein most likely serves additional functions. Using CSB-deficient human fibroblasts, Caenorhabditiselegans, and mice, we show that CSB promotes acetylation of α-tubulin and thereby regulates autophagy. At the organ level, chronic exposure of csbm/m mice to UVA radiation caused a severe skin phenotype with loss of subcutaneous fat, inflammation, and fibrosis. These changes in skin tissue were associated with an accumulation of autophagic/lysosomal proteins and reduced amounts of acetylated α-tubulin. At the cellular level, we found that CSB directly interacts with the histone deacetylase 6 (HDAC6) and the α-tubulin acetyltransferase MEC-17. Upon UVA irradiation, CSB is recruited to the centrosome where it colocalizes with dynein and HDAC6. Administration of the pan-HDAC inhibitor SAHA (suberoylanilide hydroxamic acid) enhanced α-tubulin acetylation, improved autophagic function in CSB-deficient models from all three species, and rescued the skin phenotype in csbm/m mice. HDAC inhibition may thus represent a therapeutic option for CS.


Subject(s)
Autophagy/drug effects , Cockayne Syndrome/pathology , Histone Deacetylase Inhibitors/pharmacology , Lysosomes/metabolism , Subcutaneous Fat/pathology , Acetylation , Animals , Autophagy/radiation effects , Autophagy-Related Proteins/metabolism , Caenorhabditis elegans/drug effects , Centrosome/drug effects , Centrosome/metabolism , Centrosome/radiation effects , DNA Helicases/deficiency , DNA Helicases/metabolism , DNA Repair Enzymes/deficiency , DNA Repair Enzymes/metabolism , Disease Models, Animal , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Fibroblasts/radiation effects , Lysosomes/drug effects , Lysosomes/radiation effects , Mice , Phenotype , Poly-ADP-Ribose Binding Proteins/deficiency , Poly-ADP-Ribose Binding Proteins/metabolism , Skin/pathology , Skin/radiation effects , Subcutaneous Fat/drug effects , Subcutaneous Fat/radiation effects , Tubulin/metabolism , Ubiquitinated Proteins/metabolism , Ultraviolet Rays , Vorinostat/pharmacology
11.
Genome Res ; 28(5): 666-675, 2018 05.
Article in English | MEDLINE | ID: mdl-29636374

ABSTRACT

Throughout their lifetime, cells are subject to extrinsic and intrinsic mutational processes leaving behind characteristic signatures in the genome. DNA mismatch repair (MMR) deficiency leads to hypermutation and is found in different cancer types. Although it is possible to associate mutational signatures extracted from human cancers with possible mutational processes, the exact causation is often unknown. Here, we use C. elegans genome sequencing of pms-2 and mlh-1 knockouts to reveal the mutational patterns linked to C. elegans MMR deficiency and their dependency on endogenous replication errors and errors caused by deletion of the polymerase ε subunit pole-4 Signature extraction from 215 human colorectal and 289 gastric adenocarcinomas revealed three MMR-associated signatures, one of which closely resembles the C. elegans MMR spectrum and strongly discriminates microsatellite stable and unstable tumors (AUC = 98%). A characteristic difference between human and C. elegans MMR deficiency is the lack of elevated levels of NCG > NTG mutations in C. elegans, likely caused by the absence of cytosine (CpG) methylation in worms. The other two human MMR signatures may reflect the interaction between MMR deficiency and other mutagenic processes, but their exact cause remains unknown. In summary, combining information from genetically defined models and cancer samples allows for better aligning mutational signatures to causal mutagenic processes.


Subject(s)
Adenocarcinoma/genetics , Caenorhabditis elegans/genetics , Colorectal Neoplasms/genetics , DNA Mismatch Repair , Mutation , Stomach Neoplasms/genetics , Adenocarcinoma/metabolism , Animals , Base Sequence , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , DNA Mutational Analysis/methods , DNA Polymerase II/deficiency , DNA Polymerase II/genetics , Humans , Mismatch Repair Endonuclease PMS2/deficiency , Mismatch Repair Endonuclease PMS2/genetics , MutL Protein Homolog 1/deficiency , MutL Protein Homolog 1/genetics , Poly-ADP-Ribose Binding Proteins/deficiency , Poly-ADP-Ribose Binding Proteins/genetics , Stomach Neoplasms/metabolism
12.
Exp Hematol ; 59: 40-50.e3, 2018 03.
Article in English | MEDLINE | ID: mdl-29288703

ABSTRACT

Self-renewing hematopoietic stem cells and multipotent progenitor cells are responsible for maintaining hematopoiesis throughout an individual's lifetime. For overall health and survival, it is critical that the genome stability of these cells is maintained and that the cell population is not exhausted. Previous reports have indicated that the DEK protein, a chromatin structural protein that functions in numerous nuclear processes, is required for DNA damage repair in vitro and long-term engraftment of hematopoietic stem cells in vivo. Therefore, we investigated the role of DEK in normal hematopoiesis and response to DNA damaging agents in vivo. Here, we report that hematopoiesis is largely unperturbed in DEK knockout mice compared with wild-type (WT) controls. However, DEK knockout mice have fewer radioprotective units, but increased capacity to survive repeated sublethal doses of radiation exposure compared with WT mice. Furthermore, this increased survival correlated with a sustained quiescent state in which DEK knockout restricted hematopoietic progenitor cells (HPC-1) were nearly three times more likely to be quiescent following irradiation compared with WT cells and were significantly more radioresistant during the early phases of myeloid reconstitution. Together, our studies indicate that DEK functions in the normal hematopoietic stress response to recurrent radiation exposure.


Subject(s)
DNA Damage , DNA-Binding Proteins/deficiency , Hematopoiesis/physiology , Hematopoietic Stem Cells/metabolism , Oncogene Proteins/deficiency , Poly-ADP-Ribose Binding Proteins/deficiency , Radiation Tolerance/physiology , Animals , Hematopoietic Stem Cells/cytology , Mice , Mice, Knockout
13.
Oncologist ; 22(5): 497-502, 2017 05.
Article in English | MEDLINE | ID: mdl-28465371

ABSTRACT

Deficiencies in DNA repair due to mutations in the exonuclease domain of DNA polymerase ɛ have recently been described in a subset of cancers characterized by an ultramutated and microsatellite stable (MSS) phenotype. This alteration in DNA repair is distinct from the better-known mismatch repair deficiencies which lead to microsatellite instability (MSI) and an increased tumor mutation burden. Instead, mutations in POLE lead to impaired proofreading intrinsic to Pol ɛ during DNA replication resulting in a dramatically increased mutation rate. Somatic mutations of Pol ɛ have been found most frequently in endometrial and colorectal cancers (CRC) and can lead to a unique familial syndrome in the case of germline mutations. While other key genomic abnormalities, such as MSI, have known prognostic and treatment implications, in this case it is less clear. As molecular genotyping of tumors becomes routine in the care of cancer patients, less common, but potentially actionable findings such as these POLE mutations could be overlooked unless appropriate algorithms are in place. We present two cases of metastatic CRC with a POLE mutation, both of which are ultramutated and MSS. The basic biochemical mechanisms leading to a unique phenotype in POLE deficiency as well as challenges faced with interpreting the genomic profiling of tumors in this important subset of patients and the potential clinical implications will be discussed here. The Oncologist 2017;22:497-502 KEY POINTS: Clinicians should recognize that tumors with high tumor mutation burden and that are microsatellite stable may harbor a POLE mutation, which is associated with an ultramutated phenotype.Work-up for POLE deficiency should indeed become part of the routine molecular testing paradigm for patients with colorectal cancer.This subset of patients may benefit from clinical trials where the higher number of mutation-associated neoantigens and defect in DNA repair may be exploited therapeutically.


Subject(s)
Colorectal Neoplasms/genetics , DNA Polymerase II/genetics , Microsatellite Instability , Poly-ADP-Ribose Binding Proteins/genetics , Prognosis , Colorectal Neoplasms/pathology , DNA Polymerase II/deficiency , DNA Repair/genetics , Genotype , Germ-Line Mutation/genetics , Humans , Male , Middle Aged , Neoplasm Metastasis , Phenotype , Poly-ADP-Ribose Binding Proteins/deficiency
14.
eNeuro ; 4(6)2017.
Article in English | MEDLINE | ID: mdl-29379870

ABSTRACT

Vital motor functions, such as respiration and locomotion, rely on the ability of spinal motor neurons (MNs) to acquire stereotypical positions in the ventral spinal cord and to project with high precision to their peripheral targets. These key properties of MNs emerge during development through transcriptional programs that dictate their subtype identity and connectivity; however, the molecular mechanisms that establish the transcriptional landscape necessary for MN specification are not fully understood. Here, we show that the enzyme topoisomerase IIß (Top2ß) controls MN migration and connectivity. Surprisingly, Top2ß is not required for MN generation or survival but has a selective role in columnar specification. In the absence of Top2ß, phrenic MN identity is eroded, while other motor columns are partially preserved but fail to cluster to their proper position. In Top2ß-/- mice, peripheral connectivity is impaired as MNs exhibit a profound deficit in terminal branching. These defects likely result from the insufficient activation of Hox/Pbx-dependent transcriptional programs as Hox and Pbx genes are downregulated in the absence of Top2ß. Top2ß mutants recapitulate many aspects of Pbx mutant mice, such as MN disorganization and defects in medial motor column (MMC) specification. Our findings indicate that Top2ß, a gene implicated in neurodevelopmental diseases such as autism spectrum disorders, plays a critical, cell-specific role in the assembly of motor circuits.


Subject(s)
DNA Topoisomerases, Type II/deficiency , Homeodomain Proteins/metabolism , Motor Neurons/enzymology , Motor Neurons/pathology , Poly-ADP-Ribose Binding Proteins/deficiency , Animals , Cell Movement/physiology , Cell Survival/physiology , DNA Topoisomerases, Type II/genetics , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Male , Mice, Transgenic , Neural Pathways/enzymology , Neural Pathways/pathology , Neural Stem Cells/enzymology , Neural Stem Cells/pathology , Neurogenesis/physiology , Peripheral Nerves/enzymology , Peripheral Nerves/growth & development , Peripheral Nerves/pathology , Poly-ADP-Ribose Binding Proteins/genetics , Spinal Cord/enzymology , Spinal Cord/growth & development , Spinal Cord/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...