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










Publication year range
1.
Mol Cancer Res ; 19(6): 1015-1025, 2021 06.
Article in English | MEDLINE | ID: mdl-33619228

ABSTRACT

FANCJ (BRIP1/BACH1) is a hereditary breast and ovarian cancer (HBOC) gene encoding a DNA helicase. Similar to HBOC genes, BRCA1 and BRCA2, FANCJ is critical for processing DNA inter-strand crosslinks (ICL) induced by chemotherapeutics, such as cisplatin. Consequently, cells deficient in FANCJ or its catalytic activity are sensitive to ICL-inducing agents. Unfortunately, the majority of FANCJ clinical mutations remain uncharacterized, limiting therapeutic opportunities to effectively use cisplatin to treat tumors with mutated FANCJ. Here, we sought to perform a comprehensive screen to identify FANCJ loss-of-function (LOF) mutations. We developed a FANCJ lentivirus mutation library representing approximately 450 patient-derived FANCJ nonsense and missense mutations to introduce FANCJ mutants into FANCJ knockout (K/O) HeLa cells. We performed a high-throughput screen to identify FANCJ LOF mutants that, as compared with wild-type FANCJ, fail to robustly restore resistance to ICL-inducing agents, cisplatin or mitomycin C (MMC). On the basis of the failure to confer resistance to either cisplatin or MMC, we identified 26 missense and 25 nonsense LOF mutations. Nonsense mutations elucidated a relationship between location of truncation and ICL sensitivity, as the majority of nonsense mutations before amino acid 860 confer ICL sensitivity. Further validation of a subset of LOF mutations confirmed the ability of the screen to identify FANCJ mutations unable to confer ICL resistance. Finally, mapping the location of LOF mutations to a new homology model provides additional functional information. IMPLICATIONS: We identify 51 FANCJ LOF mutations, providing important classification of FANCJ mutations that will afford additional therapeutic strategies for affected patients.


Subject(s)
BRCA1 Protein/genetics , DNA Helicases/genetics , DNA Mutational Analysis/methods , Fanconi Anemia Complementation Group Proteins/genetics , Mutation/genetics , Neoplasms/genetics , RNA Helicases/genetics , Cell Line, Tumor , Cisplatin/pharmacology , Codon, Nonsense , Cross-Linking Reagents/pharmacology , Gene Knockout Techniques , HeLa Cells , Humans , Loss of Function Mutation , Mitomycin/pharmacology , Mutation/drug effects , Mutation, Missense , Neoplasms/pathology
2.
Expert Opin Ther Targets ; 25(1): 27-36, 2021 01.
Article in English | MEDLINE | ID: mdl-33416413

ABSTRACT

Introduction: Translesion synthesis (TLS) is a DNA damage tolerance (DDT) mechanism that employs error-prone polymerases to bypass replication blocking DNA lesions, contributing to a gain in mutagenesis and chemo-resistance. However, recent findings illustrate an emerging role for TLS in replication gap suppression (RGS), distinct from its role in post-replication gap filling. Here, TLS protects cells from replication stress (RS)-induced toxic single-stranded DNA (ssDNA) gaps that accumulate in the wake of active replication. Intriguingly, TLS-mediated RGS is specifically observed in several cancer cell lines and contributes to their survival. Thus, targeting TLS has the potential to uniquely eradicate tumors without harming non-cancer tissues. Areas Covered: This review provides an innovative perspective on the role of TLS beyond its canonical function of lesion bypass or post-replicative gap filling. We provide a comprehensive analysis that underscores the emerging role of TLS as a cancer adaptation necessary to overcome the replication stress response (RSR), an anti-cancer barrier. Expert Opinion: TLS RGS is critical for tumorigenesis and is a new hallmark of cancer. Although the exact mechanism and extent of TLS dependency in cancer is still emerging, TLS inhibitors have shown promise as an anti-cancer therapy in selectively targeting this unique cancer vulnerability.


Subject(s)
DNA Damage/genetics , Molecular Targeted Therapy , Neoplasms/therapy , Animals , DNA Replication/genetics , DNA, Single-Stranded/genetics , Humans , Neoplasms/genetics , Neoplasms/pathology
3.
Cancer Res ; 81(5): 1388-1397, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33184108

ABSTRACT

Defects in DNA repair and the protection of stalled DNA replication forks are thought to underlie the chemosensitivity of tumors deficient in the hereditary breast cancer genes BRCA1 and BRCA2 (BRCA). Challenging this assumption are recent findings that indicate chemotherapies, such as cisplatin used to treat BRCA-deficient tumors, do not initially cause DNA double-strand breaks (DSB). Here, we show that ssDNA replication gaps underlie the hypersensitivity of BRCA-deficient cancer and that defects in homologous recombination (HR) or fork protection (FP) do not. In BRCA-deficient cells, ssDNA gaps developed because replication was not effectively restrained in response to stress. Gap suppression by either restoration of fork restraint or gap filling conferred therapy resistance in tissue culture and BRCA patient tumors. In contrast, restored FP and HR could be uncoupled from therapy resistance when gaps were present. Moreover, DSBs were not detected after therapy when apoptosis was inhibited, supporting a framework in which DSBs are not directly induced by genotoxic agents, but rather are induced from cell death nucleases and are not fundamental to the mechanism of action of genotoxic agents. Together, these data indicate that ssDNA replication gaps underlie the BRCA cancer phenotype, "BRCAness," and we propose they are fundamental to the mechanism of action of genotoxic chemotherapies. SIGNIFICANCE: This study suggests that ssDNA replication gaps are fundamental to the toxicity of genotoxic agents and underlie the BRCA-cancer phenotype "BRCAness," yielding promising biomarkers, targets, and opportunities to resensitize refractory disease.See related commentary by Canman, p. 1214.


Subject(s)
BRCA2 Protein , DNA Replication , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , BRCA2 Protein/genetics , DNA Breaks, Double-Stranded , DNA Repair/genetics , Genes, BRCA2 , Homologous Recombination , Humans
4.
Sci Adv ; 6(24): eaaz7808, 2020 06.
Article in English | MEDLINE | ID: mdl-32577513

ABSTRACT

The replication stress response, which serves as an anticancer barrier, is activated not only by DNA damage and replication obstacles but also oncogenes, thus obscuring how cancer evolves. Here, we identify that oncogene expression, similar to other replication stress-inducing agents, induces single-stranded DNA (ssDNA) gaps that reduce cell fitness. DNA fiber analysis and electron microscopy reveal that activation of translesion synthesis (TLS) polymerases restricts replication fork slowing, reversal, and fork degradation without inducing replication gaps despite the continuation of replication during stress. Consistent with gap suppression (GS) being fundamental to cancer, we demonstrate that a small-molecule inhibitor targeting the TLS factor REV1 not only disrupts DNA replication and cancer cell fitness but also synergizes with gap-inducing therapies such as inhibitors of ATR or Wee1. Our work illuminates that GS during replication is critical for cancer cell fitness and therefore a targetable vulnerability.

5.
Oncotarget ; 8(40): 68707-68720, 2017 Sep 15.
Article in English | MEDLINE | ID: mdl-28978150

ABSTRACT

Alkylating agents are commonly used to treat cancer. Although base excision repair (BER) is a major pathway for repairing DNA alkylation damage, under certain conditions, the initiation of BER produces toxic repair intermediates that damage healthy tissues. The initiation of BER by the alkyladenine DNA glycosylase (AAG, a.k.a. MPG) can mediate alkylation-induced cytotoxicity in specific cells in the retina and cerebellum of male mice. Cytotoxicity in both wild-type and Aag-transgenic (AagTg) mice is abrogated in the absence of Poly(ADP-ribose) polymerase-1 (PARP1). Here, we tested whether PARP inhibitors can also prevent alkylation-induced retinal and cerebellar degeneration in male and female WT and AagTg mice. Importantly, we found that WT mice display sex-dependent alkylation-induced retinal damage (but not cerebellar damage), with WT males being more sensitive than females. Accordingly, estradiol treatment protects males against alkylation-induced retinal degeneration. In AagTg male and female mice, the alkylation-induced tissue damage in both the retina and cerebellum is exacerbated and the sex difference in the retina is abolished. PARP inhibitors, much like Parp1 gene deletion, protect against alkylation-induced AAG-dependent neuronal degeneration in WT and AagTg mice, regardless of the gender, but their efficacy in preventing alkylation-induced neuronal degeneration depends on PARP inhibitor characteristics and doses. The recent surge in the use of PARP inhibitors in combination with cancer chemotherapeutic alkylating agents might represent a powerful tool for obtaining increased therapeutic efficacy while avoiding the collateral effects of alkylating agents in healthy tissues.

6.
Article in English | MEDLINE | ID: mdl-29472318

ABSTRACT

The BRCA-Fanconi anemia (FA) pathway preserves the genome and suppresses cancer and is a main determinant of chemotherapeutic efficacy. The hereditary breast cancer genes BRCA1 and BRCA2 function in DNA double-strand break repair mediating distinct steps of homologous recombination (HR). More recently, independent of DNA repair, functions in the replication stress response have come to light, providing insight as to how the BRCA-FA pathway also balances genome preservation with proliferation. The BRCA-FA proteins associate with the replisome and contribute to the efficiency and recovery of replication following perturbations that slow or arrest DNA replication. Although the full repertoire of functions in the replication stress response remains to be elucidated, the function of BRCA1 and BRCA2 in protecting stalled replication forks contributes along with HR to the sensitivity of BRCA-associated tumors to chemotherapy. Moreover, chemoresistance evolves from restoration of either HR and/or fork protection. Although mechanisms underlying the restoration of HR have been characterized, it remains less clear how restoration of fork protection is achieved. Here, we outline mechanisms of "rewired" fork protection and chemotherapy resistance in BRCA cancer. We propose that mechanisms are linked to permissive replication that limits fork remodeling and therefore opportunities for fork degradation. Combating this chemoresistance mechanism will require drugs that inactivate replication bypass mechanisms.

8.
Nature ; 535(7612): 382-7, 2016 07 21.
Article in English | MEDLINE | ID: mdl-27443740

ABSTRACT

Cells deficient in the Brca1 and Brca2 genes have reduced capacity to repair DNA double-strand breaks by homologous recombination and consequently are hypersensitive to DNA-damaging agents, including cisplatin and poly(ADP-ribose) polymerase (PARP) inhibitors. Here we show that loss of the MLL3/4 complex protein, PTIP, protects Brca1/2-deficient cells from DNA damage and rescues the lethality of Brca2-deficient embryonic stem cells. However, PTIP deficiency does not restore homologous recombination activity at double-strand breaks. Instead, its absence inhibits the recruitment of the MRE11 nuclease to stalled replication forks, which in turn protects nascent DNA strands from extensive degradation. More generally, acquisition of PARP inhibitors and cisplatin resistance is associated with replication fork protection in Brca2-deficient tumour cells that do not develop Brca2 reversion mutations. Disruption of multiple proteins, including PARP1 and CHD4, leads to the same end point of replication fork protection, highlighting the complexities by which tumour cells evade chemotherapeutic interventions and acquire drug resistance.


Subject(s)
DNA Replication/physiology , Drug Resistance, Neoplasm/drug effects , Gene Deletion , Genes, BRCA1 , Genes, BRCA2 , Neoplasms/pathology , Nuclear Proteins/deficiency , Animals , Carrier Proteins/genetics , Cell Line, Tumor , Cisplatin/pharmacology , DNA/biosynthesis , DNA/metabolism , DNA Breaks, Double-Stranded , DNA Damage/drug effects , DNA Damage/genetics , DNA Helicases/genetics , DNA Repair/drug effects , DNA Repair/genetics , DNA Repair Enzymes/antagonists & inhibitors , DNA Repair Enzymes/metabolism , DNA Replication/drug effects , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Drug Resistance, Neoplasm/genetics , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Female , Homologous Recombination , MRE11 Homologue Protein , Mice , Neoplasms/genetics , Nuclear Proteins/genetics , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/genetics
9.
Oncotarget ; 7(29): 44950-44965, 2016 Jul 19.
Article in English | MEDLINE | ID: mdl-27391435

ABSTRACT

Nephrotoxicity is a common toxic side-effect of chemotherapeutic alkylating agents. Although the base excision repair (BER) pathway is essential in repairing DNA alkylation damage, under certain conditions the initiation of BER produces toxic repair intermediates that damage healthy tissues. We have shown that the alkyladenine DNA glycosylase, Aag (a.k.a. Mpg), an enzyme that initiates BER, mediates alkylation-induced whole-animal lethality and cytotoxicity in the pancreas, spleen, retina, and cerebellum, but not in the kidney. Cytotoxicity in both wild-type and Aag-transgenic mice (AagTg) was abrogated in the absence of Poly(ADP-ribose) polymerase-1 (Parp1). Here we report that Parp1-deficient mice expressing increased Aag (AagTg/Parp1-/-) develop sex-dependent kidney failure upon exposure to the alkylating agent, methyl methanesulfonate (MMS), and suffer increased whole-animal lethality compared to AagTg and wild-type mice. Macroscopic, histological, electron microscopic and immunohistochemical analyses revealed morphological kidney damage including dilated tubules, proteinaceous casts, vacuolation, collapse of the glomerular tuft, and deterioration of podocyte structure. Moreover, mice exhibited clinical signs of kidney disease indicating functional damage, including elevated blood nitrogen urea and creatinine, hypoproteinemia and proteinuria. Pharmacological Parp inhibition in AagTg mice also resulted in sensitivity to MMS-induced nephrotoxicity. These findings provide in vivo evidence that Parp1 modulates Aag-dependent MMS-induced nephrotoxicity in a sex-dependent manner and highlight the critical roles that Aag-initiated BER and Parp1 may play in determining the side-effects of chemotherapeutic alkylating agents.


Subject(s)
Antineoplastic Agents, Alkylating/adverse effects , DNA Glycosylases/metabolism , Kidney/drug effects , Poly (ADP-Ribose) Polymerase-1/metabolism , Alkylation , Animals , DNA Damage/drug effects , DNA Repair/drug effects , Female , Male , Mice , Mice, Transgenic , Sex Characteristics
10.
Oncotarget ; 7(19): 28624-36, 2016 May 10.
Article in English | MEDLINE | ID: mdl-27086921

ABSTRACT

Much of the global cancer burden is associated with longstanding inflammation accompanied by release of DNA-damaging reactive oxygen and nitrogen species. Here, we report that the Mbd4 DNA glycosylase is protective in the azoxymethane/dextran sodium sulfate (AOM/DSS) mouse model of inflammation-driven colon cancer. Mbd4 excises T and U from T:G and U:G mismatches caused by deamination of 5-methylcytosine and cytosine. Since the rate of deamination is higher in inflamed tissues, we investigated the role of Mbd4 in inflammation-driven tumorigenesis. In the AOM/DSS assay, Mbd4-/- mice displayed more severe clinical symptoms, decreased survival, and a greater tumor burden than wild-type (WT) controls. The increased tumor burden in Mbd4-/- mice did not arise from impairment of AOM-induced apoptosis in the intestinal crypt. Histopathological analysis indicated that the colonic epithelium of Mbd4-/- mice is more vulnerable than WT to DSS-induced tissue damage. We investigated the role of the Mbd4-/- immune system in AOM/DSS-mediated carcinogenesis by repeating the assay on WT and Mbd4-/- mice transplanted with WT bone marrow. Mbd4-/- mice with WT bone marrow behaved similarly to Mbd4-/- mice. Together, our results indicate that the colonic epithelium of Mbd4-/- mice is more vulnerable to DSS-induced injury, which exacerbates inflammation-driven tissue injury and cancer.


Subject(s)
Colon/metabolism , Colonic Neoplasms/metabolism , DNA Glycosylases/metabolism , Endodeoxyribonucleases/metabolism , Inflammation/metabolism , Animals , Apoptosis/genetics , Azoxymethane , Colon/pathology , Colonic Neoplasms/genetics , DNA Glycosylases/genetics , Dextran Sulfate , Endodeoxyribonucleases/genetics , Humans , Inflammation/chemically induced , Inflammation/genetics , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Kaplan-Meier Estimate , Mice, Knockout , Tumor Burden/genetics
11.
Proc Natl Acad Sci U S A ; 111(45): E4878-86, 2014 Nov 11.
Article in English | MEDLINE | ID: mdl-25349415

ABSTRACT

Inflammation is accompanied by the release of highly reactive oxygen and nitrogen species (RONS) that damage DNA, among other cellular molecules. Base excision repair (BER) is initiated by DNA glycosylases and is crucial in repairing RONS-induced DNA damage; the alkyladenine DNA glycosylase (Aag/Mpg) excises several DNA base lesions induced by the inflammation-associated RONS release that accompanies ischemia reperfusion (I/R). Using mouse I/R models we demonstrate that Aag(-/-) mice are significantly protected against, rather than sensitized to, I/R injury, and that such protection is observed across three different organs. Following I/R in liver, kidney, and brain, Aag(-/-) mice display decreased hepatocyte death, cerebral infarction, and renal injury relative to wild-type. We infer that in wild-type mice, Aag excises damaged DNA bases to generate potentially toxic abasic sites that in turn generate highly toxic DNA strand breaks that trigger poly(ADP-ribose) polymerase (Parp) hyperactivation, cellular bioenergetics failure, and necrosis; indeed, steady-state levels of abasic sites and nuclear PAR polymers were significantly more elevated in wild-type vs. Aag(-/-) liver after I/R. This increase in PAR polymers was accompanied by depletion of intracellular NAD and ATP levels plus the translocation and extracellular release of the high-mobility group box 1 (Hmgb1) nuclear protein, activating the sterile inflammatory response. We thus demonstrate the detrimental effects of Aag-initiated BER during I/R and sterile inflammation, and present a novel target for controlling I/R-induced injury.


Subject(s)
Brain/enzymology , DNA Glycosylases/metabolism , DNA Repair , Kidney/enzymology , Liver/enzymology , Reperfusion Injury/enzymology , Acute Kidney Injury/enzymology , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Animals , Brain/pathology , Brain Infarction/enzymology , Brain Infarction/genetics , Brain Infarction/pathology , Cell Death , DNA Damage , DNA Glycosylases/genetics , Enzyme Induction/genetics , HMGB1 Protein/genetics , HMGB1 Protein/metabolism , Hepatocytes/enzymology , Hepatocytes/pathology , Inflammation/enzymology , Inflammation/genetics , Inflammation/pathology , Kidney/pathology , Liver/pathology , Mice , Mice, Knockout , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Reperfusion Injury/genetics , Reperfusion Injury/pathology
12.
DNA Repair (Amst) ; 21: 78-86, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24994062

ABSTRACT

The accumulation of DNA damage is thought to contribute to the physiological decay associated with the aging process. Here, we report the results of a large-scale study examining longevity in various mouse models defective in the repair of DNA alkylation damage, or defective in the DNA damage response. We find that the repair of spontaneous DNA damage by alkyladenine DNA glycosylase (Aag/Mpg)-initiated base excision repair and O(6)-methylguanine DNA methyltransferase (Mgmt)-mediated direct reversal contributes to maximum life span in the laboratory mouse. We also uncovered important genetic interactions between Aag, which excises a wide variety of damaged DNA bases, and the DNA damage sensor and signaling protein, Atm. We show that Atm plays a role in mediating survival in the face of both spontaneous and induced DNA damage, and that Aag deficiency not only promotes overall survival, but also alters the tumor spectrum in Atm(-/-) mice. Further, the reversal of spontaneous alkylation damage by Mgmt interacts with the DNA mismatch repair pathway to modulate survival and tumor spectrum. Since these aging studies were performed without treatment with DNA damaging agents, our results indicate that the DNA damage that is generated endogenously accumulates with age, and that DNA alkylation repair proteins play a role in influencing longevity.


Subject(s)
DNA Glycosylases/genetics , DNA Repair , Longevity/genetics , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , DNA Glycosylases/metabolism , DNA Modification Methylases/genetics , DNA Modification Methylases/metabolism , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , Mice , Mice, Inbred C57BL , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
13.
PLoS Genet ; 9(4): e1003413, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23593019

ABSTRACT

Alkylating agents comprise a major class of front-line cancer chemotherapeutic compounds, and while these agents effectively kill tumor cells, they also damage healthy tissues. Although base excision repair (BER) is essential in repairing DNA alkylation damage, under certain conditions, initiation of BER can be detrimental. Here we illustrate that the alkyladenine DNA glycosylase (AAG) mediates alkylation-induced tissue damage and whole-animal lethality following exposure to alkylating agents. Aag-dependent tissue damage, as observed in cerebellar granule cells, splenocytes, thymocytes, bone marrow cells, pancreatic ß-cells, and retinal photoreceptor cells, was detected in wild-type mice, exacerbated in Aag transgenic mice, and completely suppressed in Aag⁻/⁻ mice. Additional genetic experiments dissected the effects of modulating both BER and Parp1 on alkylation sensitivity in mice and determined that Aag acts upstream of Parp1 in alkylation-induced tissue damage; in fact, cytotoxicity in WT and Aag transgenic mice was abrogated in the absence of Parp1. These results provide in vivo evidence that Aag-initiated BER may play a critical role in determining the side-effects of alkylating agent chemotherapies and that Parp1 plays a crucial role in Aag-mediated tissue damage.


Subject(s)
Antineoplastic Agents, Alkylating , DNA Glycosylases , Neoplasms/drug therapy , Poly(ADP-ribose) Polymerases , Alkylation/drug effects , Alkylation/genetics , Animals , Antineoplastic Agents, Alkylating/adverse effects , Antineoplastic Agents, Alkylating/therapeutic use , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , DNA Glycosylases/genetics , DNA Glycosylases/metabolism , DNA Repair/drug effects , DNA Repair/genetics , Humans , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Mice , Mice, Transgenic/genetics , Mice, Transgenic/injuries , Neoplasms/genetics , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Thymocytes/cytology , Thymocytes/drug effects
14.
J Clin Invest ; 122(7): 2680-9, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22684101

ABSTRACT

More than 15% of cancer deaths worldwide are associated with underlying infections or inflammatory conditions, therefore understanding how inflammation contributes to cancer etiology is important for both cancer prevention and treatment. Inflamed tissues are known to harbor elevated etheno-base (ε-base) DNA lesions induced by the lipid peroxidation that is stimulated by reactive oxygen and nitrogen species (RONS) released from activated neutrophils and macrophages. Inflammation contributes to carcinogenesis in part via RONS-induced cytotoxic and mutagenic DNA lesions, including ε-base lesions. The mouse alkyl adenine DNA glycosylase (AAG, also known as MPG) recognizes such base lesions, thus protecting against inflammation-associated colon cancer. Two other DNA repair enzymes are known to repair ε-base lesions, namely ALKBH2 and ALKBH3; thus, we sought to determine whether these DNA dioxygenase enzymes could protect against chronic inflammation-mediated colon carcinogenesis. Using established chemically induced colitis and colon cancer models in mice, we show here that ALKBH2 and ALKBH3 provide cancer protection similar to that of the DNA glycosylase AAG. Moreover, Alkbh2 and Alkbh3 each display apparent epistasis with Aag. Surprisingly, deficiency in all 3 DNA repair enzymes confers a massively synergistic phenotype, such that animals lacking all 3 DNA repair enzymes cannot survive even a single bout of chemically induced colitis.


Subject(s)
Colitis/genetics , DNA Glycosylases/genetics , DNA Repair Enzymes/genetics , DNA Repair , Dioxygenases/genetics , Pancreatitis/genetics , AlkB Homolog 2, Alpha-Ketoglutarate-Dependent Dioxygenase , AlkB Homolog 3, Alpha-Ketoglutarate-Dependent Dioxygenase , Animals , Azoxymethane/pharmacology , Carcinogens/pharmacology , Colitis/chemically induced , Colitis/metabolism , Colon/immunology , Colon/pathology , Colorectal Neoplasms/chemically induced , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , DNA Glycosylases/metabolism , DNA Repair Enzymes/metabolism , Dextran Sulfate/pharmacology , Dioxygenases/metabolism , Epistasis, Genetic , Female , Genetic Predisposition to Disease , Kaplan-Meier Estimate , Lethal Dose 50 , Lipopolysaccharides/pharmacology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Pancreas/immunology , Pancreas/pathology , Pancreatitis/chemically induced , Pancreatitis/metabolism
15.
Nat Rev Cancer ; 12(2): 104-20, 2012 Jan 12.
Article in English | MEDLINE | ID: mdl-22237395

ABSTRACT

Alkylating agents constitute a major class of frontline chemotherapeutic drugs that inflict cytotoxic DNA damage as their main mode of action, in addition to collateral mutagenic damage. Numerous cellular pathways, including direct DNA damage reversal, base excision repair (BER) and mismatch repair (MMR), respond to alkylation damage to defend against alkylation-induced cell death or mutation. However, maintaining a proper balance of activity both within and between these pathways is crucial for a favourable response of an organism to alkylating agents. Furthermore, the response of an individual to alkylating agents can vary considerably from tissue to tissue and from person to person, pointing to genetic and epigenetic mechanisms that modulate alkylating agent toxicity.


Subject(s)
Alkylating Agents/toxicity , DNA Damage , DNA Repair , Base Pair Mismatch , Humans
16.
J Biol Chem ; 285(29): 22619-29, 2010 Jul 16.
Article in English | MEDLINE | ID: mdl-20418374

ABSTRACT

Estrogen-related receptor gamma (ERRgamma) regulates the perinatal switch to oxidative metabolism in the myocardium. We wanted to understand the significance of induction of ERRgamma expression in skeletal muscle by exercise. Muscle-specific VP16ERRgamma transgenic mice demonstrated an increase in exercise capacity, mitochondrial enzyme activity, and enlarged mitochondria despite lower muscle weights. Furthermore, peak oxidative capacity was higher in the transgenics as compared with control littermates. In contrast, mice lacking one copy of ERRgamma exhibited decreased exercise capacity and muscle mitochondrial function. Interestingly, we observed that increased ERRgamma in muscle generates a gene expression profile that closely overlays that of red oxidative fiber-type muscle. We further demonstrated that a small molecule agonist of ERRbeta/gamma can increase mitochondrial function in mouse myotubes. Our data indicate that ERRgamma plays an important role in causing a shift toward slow twitch muscle type and, concomitantly, a greater capacity for endurance exercise. Thus, the activation of this nuclear receptor provides a potential node for therapeutic intervention for diseases such as obesity, which is associated with reduced oxidative metabolism and a lower type I fiber content in skeletal muscle.


Subject(s)
Mitochondria/metabolism , Muscle, Skeletal/metabolism , Receptors, Estrogen/metabolism , Animals , Cells, Cultured , Down-Regulation/drug effects , Gene Expression Profiling , Heterozygote , Hydrazines/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/enzymology , Mitochondria/genetics , Mitochondria/ultrastructure , Models, Biological , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/ultrastructure , Muscle Fibers, Slow-Twitch/drug effects , Muscle Fibers, Slow-Twitch/metabolism , Muscle Fibers, Slow-Twitch/ultrastructure , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Muscle, Skeletal/ultrastructure , Oxidation-Reduction/drug effects , Physical Conditioning, Animal , Receptors, Estrogen/agonists , Up-Regulation/drug effects
17.
Proc Natl Acad Sci U S A ; 106(2): 576-81, 2009 Jan 13.
Article in English | MEDLINE | ID: mdl-19124772

ABSTRACT

Alkylation-induced O(6)-methylguanine (O(6)MeG) DNA lesions can be mutagenic or cytotoxic if unrepaired by the O(6)MeG-DNA methyltransferase (Mgmt) protein. O(6)MeG pairs with T during DNA replication, and if the O(6)MeG:T mismatch persists, a G:C to A:T transition mutation is fixed at the next replication cycle. O(6)MeG:T mismatch detection by MutSalpha and MutLalpha leads to apoptotic cell death, but the mechanism by which this occurs has been elusive. To explore how mismatch repair mediates O(6)MeG-dependent apoptosis, we used an Mgmt-null mouse model combined with either the Msh6-null mutant (defective in mismatch recognition) or the Exo1-null mutant (impaired in the excision step of mismatch repair). Mouse embryonic fibroblasts and bone marrow cells derived from Mgmt-null mice were much more alkylation-sensitive than wild type, as expected. However, ablation of either Msh6 or Exo1 function rendered these Mgmt-null cells just as resistant to alkylation-induced cytotoxicity as wild-type cells. Rapidly proliferating tissues in Mgmt-null mice (bone marrow, thymus, and spleen) are extremely sensitive to apoptosis induced by O(6)MeG-producing agents. Here, we show that ablation of either Msh6 or Exo1 function in the Mgmt-null mouse renders these rapidly proliferating tissues alkylation-resistant. However, whereas the Msh6 defect confers total alkylation resistance, the Exo1 defect leads to a variable tissue-specific alkylation resistance phenotype. Our results indicate that Exo1 plays an important role in the induction of apoptosis by unrepaired O(6)MeGs.


Subject(s)
Apoptosis/genetics , Base Pair Mismatch , DNA-Binding Proteins/physiology , Exodeoxyribonucleases/physiology , Guanine/analogs & derivatives , Alkylation , Animals , Bone Marrow Cells/cytology , Cell Proliferation , Fibroblasts/cytology , Guanine/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , O(6)-Methylguanine-DNA Methyltransferase , Spleen/cytology , Thymus Gland/cytology
18.
Proc Natl Acad Sci U S A ; 106(3): 888-93, 2009 Jan 20.
Article in English | MEDLINE | ID: mdl-19139400

ABSTRACT

Vision loss affects >3 million Americans and many more people worldwide. Although predisposing genes have been identified their link to known environmental factors is unclear. In wild-type animals DNA alkylating agents induce photoreceptor apoptosis and severe retinal degeneration. Alkylation-induced retinal degeneration is totally suppressed in the absence of the DNA repair protein alkyladenine DNA glycosylase (Aag) in both differentiating and postmitotic retinas. Moreover, transgenic expression of Aag activity restores the alkylation sensitivity of photoreceptors in Aag null animals. Aag heterozygotes display an intermediate level of retinal degeneration, demonstrating haploinsufficiency and underscoring that Aag expression confers a dominant retinal degeneration phenotype.


Subject(s)
Alkylating Agents/toxicity , DNA Glycosylases/physiology , DNA Repair , Retinal Degeneration/chemically induced , Animals , Apoptosis , DNA Modification Methylases/physiology , DNA Repair Enzymes/physiology , Methyl Methanesulfonate/toxicity , Methylnitrosourea/toxicity , Mice , Photoreceptor Cells, Vertebrate/drug effects , Tumor Suppressor Proteins/physiology
19.
J Appl Physiol (1985) ; 104(5): 1304-12, 2008 May.
Article in English | MEDLINE | ID: mdl-18239076

ABSTRACT

The induction of peroxisome proliferator-activated receptor-gamma coactivator-1alpha (PGC-1alpha), a key regulator of mitochondriogenesis, is well-established under multiple physical exercise regimens, including, endurance, resistance, and sprint training. We wanted to determine if increased expression of PGC-1alpha in muscle is sufficient to improve performance during exercise in vivo. We demonstrate that muscle-specific expression of PGC-1alpha improves the performance during voluntary as well as forced exercise challenges. Additionally, PGC-1alpha transgenic mice exhibit an enhanced performance during a peak oxygen uptake exercise test, demonstrating an increased peak oxidative capacity, or whole body oxygen uptake. This increased ability to perform in multiple exercise paradigms is supported by enhanced mitochondrial function as suggested by increased mitochondrial gene expression, mitochondrial DNA, and mitochondrial enzyme activity. Thus this study demonstrates that upregulation of PGC-1alpha in muscle in vivo is sufficient to greatly improve exercise performance under various exercise paradigms as well as increase peak oxygen uptake.


Subject(s)
Anaerobic Threshold/physiology , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , Oxygen Consumption/physiology , PPAR gamma/physiology , Physical Conditioning, Animal/physiology , Trans-Activators/biosynthesis , Trans-Activators/physiology , Animals , Citrate (si)-Synthase/biosynthesis , DNA, Mitochondrial/biosynthesis , DNA, Mitochondrial/genetics , Glucose Intolerance/physiopathology , Glycogen/metabolism , Insulin Resistance/physiology , Male , Mice , Muscle, Skeletal/enzymology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Pulmonary Gas Exchange/physiology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Running/physiology , Transcription Factors
20.
Mol Cell Biol ; 26(21): 8022-31, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16954387

ABSTRACT

Recent work with mouse models and human leukemic samples has shown that gain-of-function mutation(s) in Notch1 is a common genetic event in T-cell acute lymphoblastic leukemia (T-ALL). The Notch1 receptor signals through a gamma-secretase-dependent process that releases intracellular Notch1 from the membrane to the nucleus, where it forms part of a transcriptional activator complex. To identify Notch1 target genes in leukemia, we developed mouse T-cell leukemic lines that express intracellular Notch1 in a doxycycline-dependent manner. Using gene expression profiling and chromatin immunoprecipitation, we identified c-myc as a novel, direct, and critical Notch1 target gene in T-cell leukemia. c-myc mRNA levels are increased in primary mouse T-cell tumors that harbor Notch1 mutations, and Notch1 inhibition decreases c-myc mRNA levels and inhibits leukemic cell growth. Retroviral expression of c-myc, like intracellular Notch1, rescues the growth arrest and apoptosis associated with gamma-secretase inhibitor treatment or Notch1 inhibition. Consistent with these findings, retroviral insertional mutagenesis screening of our T-cell leukemia mouse model revealed common insertions in either notch1 or c-myc genes. These studies define the Notch1 molecular signature in mouse T-ALL and importantly provide mechanistic insight as to how Notch1 contributes to human T-ALL.


Subject(s)
Gene Expression Regulation, Neoplastic , Leukemia, T-Cell/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Receptor, Notch1/metabolism , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Animals , Apoptosis/physiology , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Cycle/physiology , Cell Line, Tumor , Enzyme Inhibitors/metabolism , Gene Expression Profiling , Humans , Mice , Mutagenesis, Insertional , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-myc/genetics , Receptor, Notch1/genetics , Retroviridae/genetics , Retroviridae/metabolism , T-Cell Acute Lymphocytic Leukemia Protein 1
SELECTION OF CITATIONS
SEARCH DETAIL
...