Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Nat Commun ; 14(1): 7714, 2023 Nov 24.
Article in English | MEDLINE | ID: mdl-38001070

ABSTRACT

Homologous recombination (HR)-deficiency induces a dependency on DNA polymerase theta (Polθ/Polq)-mediated end joining, and Polθ inhibitors (Polθi) are in development for cancer therapy. BRCA1 and BRCA2 deficient cells are thought to be synthetic lethal with Polθ, but whether distinct HR gene mutations give rise to equivalent Polθ-dependence, and the events that drive lethality, are unclear. In this study, we utilized mouse models with separate Brca1 functional defects to mechanistically define Brca1-Polθ synthetic lethality. Surprisingly, homozygous Brca1 mutant, Polq-/- cells were viable, but grew slowly and had chromosomal instability. Brca1 mutant cells proficient in DNA end resection were significantly more dependent on Polθ for viability; here, treatment with Polθi elevated RPA foci, which persisted through mitosis. In an isogenic system, BRCA1 null cells were defective, but PALB2 and BRCA2 mutant cells exhibited active resection, and consequently stronger sensitivity to Polθi. Thus, DNA end resection is a critical determinant of Polθi sensitivity in HR-deficient cells, and should be considered when selecting patients for clinical studies.


Subject(s)
BRCA1 Protein , Genes, BRCA2 , Mice , Animals , Humans , BRCA1 Protein/genetics , Mutation , Synthetic Lethal Mutations , DNA
2.
Nat Commun ; 12(1): 5016, 2021 08 18.
Article in English | MEDLINE | ID: mdl-34408138

ABSTRACT

DNA damage prompts a diverse range of alterations to the chromatin landscape. The RNF168 E3 ubiquitin ligase catalyzes the mono-ubiquitination of histone H2A at lysine (K)13/15 (mUb-H2A), forming a binding module for DNA repair proteins. BRCA1 promotes homologous recombination (HR), in part, through its interaction with PALB2, and the formation of a larger BRCA1-PALB2-BRCA2-RAD51 (BRCA1-P) complex. The mechanism by which BRCA1-P is recruited to chromatin surrounding DNA breaks is unclear. In this study, we reveal that an RNF168-governed signaling pathway is responsible for localizing the BRCA1-P complex to DNA damage. Using mice harboring a Brca1CC (coiled coil) mutation that blocks the Brca1-Palb2 interaction, we uncovered an epistatic relationship between Rnf168- and Brca1CC alleles, which disrupted development, and reduced the efficiency of Palb2-Rad51 localization. Mechanistically, we show that RNF168-generated mUb-H2A recruits BARD1 through a BRCT domain ubiquitin-dependent recruitment motif (BUDR). Subsequently, BARD1-BRCA1 accumulate PALB2-RAD51 at DNA breaks via the CC domain-mediated BRCA1-PALB2 interaction. Together, these findings establish a series of molecular interactions that connect the DNA damage signaling and HR repair machinery.


Subject(s)
BRCA1 Protein/metabolism , DNA Damage , Fanconi Anemia Complementation Group N Protein/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , BRCA1 Protein/genetics , Cell Nucleus/genetics , Cell Nucleus/metabolism , DNA/genetics , DNA/metabolism , Fanconi Anemia Complementation Group N Protein/genetics , Histones/genetics , Histones/metabolism , Humans , Mice , Protein Binding , Protein Transport , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Recombinational DNA Repair , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination
3.
Mol Cell ; 78(5): 951-959.e6, 2020 06 04.
Article in English | MEDLINE | ID: mdl-32359443

ABSTRACT

BRCA1 promotes the DNA end resection and RAD51 loading steps of homologous recombination (HR). Whether these functions can be uncoupled, and whether mutant proteins retaining partial activity can complement one another, is unclear and could affect the severity of BRCA1-associated Fanconi anemia (FA). Here we generated a Brca1CC mouse with a coiled-coil (CC) domain deletion. Brca1CC/CC mice are born at low frequencies, and post-natal mice have FA-like abnormalities, including bone marrow failure. Intercrossing with Brca1Δ11, which is homozygous lethal, generated Brca1CC/Δ11 mice at Mendelian frequencies that were indistinguishable from Brca1+/+ mice. Brca1CC and Brca1Δ11 proteins were individually responsible for counteracting 53BP1-RIF1-Shieldin activity and promoting RAD51 loading, respectively. Thus, Brca1CC and Brca1Δ11 alleles represent separation-of-function mutations that combine to provide a level of HR sufficient for normal development and hematopoiesis. Because BRCA1 activities can be genetically separated, compound heterozygosity for functional complementary mutations may protect individuals from FA.


Subject(s)
BRCA1 Protein/genetics , Homologous Recombination/genetics , Tumor Suppressor p53-Binding Protein 1/genetics , Animals , BRCA1 Protein/metabolism , DNA Breaks, Double-Stranded , DNA Repair , Exons , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Female , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mutation , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Tumor Suppressor p53-Binding Protein 1/metabolism
4.
Cancer Res ; 80(13): 2848-2860, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32213544

ABSTRACT

BRCA1 gene mutations impair homologous recombination (HR) DNA repair, resulting in cellular senescence and embryonic lethality in mice. Therefore, BRCA1-deficient cancers require adaptations that prevent excessive genomic alterations from triggering cell death. RNF168-mediated ubiquitination of γH2AX at K13/15 (ub-H2AX) serves as a recruitment module for the localization of 53BP1 to DNA break sites. Here, we found multiple BRCA1-mutant cancer cell lines and primary tumors with low levels of RNF168 protein expression. Overexpression of ectopic RNF168 or a ub-H2AX fusion protein induced cell death and delayed BRCA1-mutant tumor formation. Cell death resulted from the recruitment of 53BP1 to DNA break sites and inhibition of DNA end resection. Strikingly, reintroduction of BRCA1 or 53BP1 depletion restored HR and rescued the ability of cells to maintain RNF168 and ub-H2AX overexpression. Thus, downregulation of RNF168 protein expression is a mechanism for providing BRCA1-null cancer cell lines with a residual level of HR that is essential for viability. Overall, our work identifies loss of RNF168 ubiquitin signaling as a proteomic alteration that supports BRCA1-mutant carcinogenesis. We propose that restoring RNF168-ub-H2AX signaling, potentially through inhibition of deubiquitinases, could represent a new therapeutic approach. SIGNIFICANCE: This study explores the concept that homologous recombination DNA repair is not an all-or-nothing concept, but a spectrum, and that where a tumor stands on this spectrum may have therapeutic relevance.See related commentary by Wang and Wulf, p. 2720.


Subject(s)
Neoplasms , Ubiquitin , Animals , BRCA1 Protein/genetics , DNA Damage , Mice , Neoplasms/genetics , Proteomics , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
5.
Mol Cancer Ther ; 19(2): 602-613, 2020 02.
Article in English | MEDLINE | ID: mdl-31575654

ABSTRACT

Patients harboring germline breast cancer susceptibility genes 1 and 2 (BRCA1/2) mutations are predisposed to developing breast, pancreatic, and ovarian cancers. BRCA2 plays a critical role in homologous recombination (HR) DNA repair and deleterious mutations in BRCA2 confer sensitivity to PARP inhibition. Recently, the PARP inhibitors olaparib and rucaparib were FDA approved for the treatment of metastatic breast cancer and patients with recurrent ovarian cancer with mutations in BRCA1/2. Despite their initial antitumor activity, the development of resistance limits the clinical utility of PARP inhibitor therapy. Multiple resistance mechanisms have been described, including reversion mutations that restore the reading frame of the BRCA2 gene. In this study, we generated olaparib- and rucaparib-resistant BRCA2-mutant Capan1 cell lines. We did not detect secondary reversion mutations in the olaparib- or rucaparib-resistant clones. Several of the resistant clones had gene duplication and amplification of the mutant BRCA2 allele, with a corresponding increase in expression of a truncated BRCA2 protein. In addition, HR-mediated DNA repair was rescued, as evidenced by the restoration of RAD51 foci formation. Using mass spectrometry, we identified Disruptor Of Telomeric silencing 1-Like (DOT1L), as an interacting partner of truncated BRCA2. RNAi-mediated knockdown of BRCA2 or DOT1L was sufficient to resensitize cells to olaparib. The results demonstrate that independent of a BRCA2 reversion, mutation amplification of a mutant-carrying BRCA2 contributes to PARP inhibitor resistance.


Subject(s)
BRCA2 Protein/metabolism , Breast Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Rad51 Recombinase/metabolism , Cell Line, Tumor , Female , Humans , Mutation
6.
Nat Commun ; 10(1): 5661, 2019 12 11.
Article in English | MEDLINE | ID: mdl-31827092

ABSTRACT

BRCA1 mutant carcinomas are sensitive to PARP inhibitor (PARPi) therapy; however, resistance arises. BRCA1 BRCT domain mutant proteins do not fold correctly and are subject to proteasomal degradation, resulting in PARPi sensitivity. In this study, we show that cell lines and patient-derived tumors, with highly disruptive BRCT domain mutations, have readily detectable BRCA1 protein expression, and are able to proliferate in the presence of PARPi. Peptide analyses reveal that chemo-resistant cancers contain residues encoded by BRCA1 intron 15. Mechanistically, cancers with BRCT domain mutations harbor BRCA1 gene breakpoints within or adjacent to Alu elements in intron 15; producing partial gene duplications, inversions and translocations, and terminating transcription prior to the mutation-containing BRCT domain. BRCA1 BRCT domain-deficient protein isoforms avoid mutation-induced proteasomal degradation, support homology-dependent DNA repair, and promote PARPi resistance. Taken together, Alu-mediated BRCA1 gene rearrangements are responsible for generating hypomorphic proteins, and may represent a biomarker of PARPi resistance.


Subject(s)
Alu Elements , Antineoplastic Agents/administration & dosage , BRCA1 Protein/genetics , Breast Neoplasms/genetics , Gene Rearrangement , Introns , Poly(ADP-ribose) Polymerase Inhibitors/administration & dosage , Animals , BRCA1 Protein/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Chromosome Inversion , Drug Resistance, Neoplasm , Female , Humans , Mice , Mice, Nude , Translocation, Genetic
8.
Cell Rep ; 24(13): 3513-3527.e7, 2018 09 25.
Article in English | MEDLINE | ID: mdl-30257212

ABSTRACT

BRCA1 functions in homologous recombination (HR) both up- and downstream of DNA end resection. However, in cells with 53BP1 gene knockout (KO), BRCA1 is dispensable for the initiation of resection, but whether BRCA1 activity is entirely redundant after end resection is unclear. Here, we found that 53bp1 KO rescued the embryonic viability of a Brca1ΔC/ΔC mouse model that harbors a stop codon in the coiled-coil domain. However, Brca1ΔC/ΔC;53bp1-/- mice were susceptible to tumor formation, lacked Rad51 foci, and were sensitive to PARP inhibitor (PARPi) treatment, indicative of suboptimal HR. Furthermore, BRCA1 mutant cancer cell lines were dependent on truncated BRCA1 proteins that retained the ability to interact with PALB2 for 53BP1 KO induced RAD51 foci and PARPi resistance. Our data suggest that the overall efficiency of 53BP1 loss of function induced HR may be BRCA1 mutation dependent. In the setting of 53BP1 KO, hypomorphic BRCA1 proteins are active downstream of end resection, promoting RAD51 loading and PARPi resistance.


Subject(s)
BRCA1 Protein/genetics , Drug Resistance, Neoplasm , Homologous Recombination , Mammary Neoplasms, Experimental/genetics , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Tumor Suppressor p53-Binding Protein 1/genetics , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , BRCA1 Protein/metabolism , Fanconi Anemia Complementation Group N Protein/metabolism , Female , HEK293 Cells , Humans , Loss of Function Mutation , MCF-7 Cells , Mammary Neoplasms, Experimental/drug therapy , Mice , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Rad51 Recombinase/metabolism , Tumor Suppressor p53-Binding Protein 1/metabolism
9.
Cell Rep ; 23(11): 3127-3136, 2018 06 12.
Article in English | MEDLINE | ID: mdl-29898385

ABSTRACT

PARP inhibitors (PARPis) have been used to induce synthetic lethality in BRCA-deficient tumors in clinical trials with limited success. We hypothesized that RAD52-mediated DNA repair remains active in PARPi-treated BRCA-deficient tumor cells and that targeting RAD52 should enhance the synthetic lethal effect of PARPi. We show that RAD52 inhibitors (RAD52is) attenuated single-strand annealing (SSA) and residual homologous recombination (HR) in BRCA-deficient cells. Simultaneous targeting of PARP1 and RAD52 with inhibitors or dominant-negative mutants caused synergistic accumulation of DSBs and eradication of BRCA-deficient but not BRCA-proficient tumor cells. Remarkably, Parp1-/-;Rad52-/- mice are normal and display prolonged latency of BRCA1-deficient leukemia compared with Parp1-/- and Rad52-/- counterparts. Finally, PARPi+RAD52i exerted synergistic activity against BRCA1-deficient tumors in immunodeficient mice with minimal toxicity to normal cells and tissues. In conclusion, our data indicate that addition of RAD52i will improve therapeutic outcome of BRCA-deficient malignancies treated with PARPi.


Subject(s)
BRCA1 Protein/genetics , BRCA2 Protein/genetics , Poly (ADP-Ribose) Polymerase-1/genetics , Rad52 DNA Repair and Recombination Protein/genetics , Animals , BRCA1 Protein/deficiency , BRCA2 Protein/deficiency , DNA Repair/drug effects , Female , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Homologous Recombination/drug effects , Humans , Imatinib Mesylate/pharmacology , Kaplan-Meier Estimate , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/pathology , Male , Mice , Mice, Inbred NOD , Mice, Knockout , Phthalazines/pharmacology , Piperazines/pharmacology , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Poly (ADP-Ribose) Polymerase-1/deficiency , Rad52 DNA Repair and Recombination Protein/antagonists & inhibitors , Rad52 DNA Repair and Recombination Protein/deficiency , Synthetic Lethal Mutations , Tumor Suppressor p53-Binding Protein 1/deficiency , Tumor Suppressor p53-Binding Protein 1/genetics
10.
Cell Rep ; 17(9): 2367-2381, 2016 11 22.
Article in English | MEDLINE | ID: mdl-27880910

ABSTRACT

Although poly(ADP-ribose) polymerase (PARP) inhibitors are active in homologous recombination (HR)-deficient cancers, their utility is limited by acquired resistance after restoration of HR. Here, we report that dinaciclib, an inhibitor of cyclin-dependent kinases (CDKs) 1, 2, 5, and 9, additionally has potent activity against CDK12, a transcriptional regulator of HR. In BRCA-mutated triple-negative breast cancer (TNBC) cells and patient-derived xenografts (PDXs), dinaciclib ablates restored HR and reverses PARP inhibitor resistance. Additionally, we show that de novo resistance to PARP inhibition in BRCA1-mutated cell lines and a PDX derived from a PARP-inhibitor-naive BRCA1 carrier is mediated by residual HR and is reversed by CDK12 inhibition. Finally, dinaciclib augments the degree of response in a PARP-inhibitor-sensitive model, converting tumor growth inhibition to durable regression. These results highlight the significance of HR disruption as a therapeutic strategy and support the broad use of combined CDK12 and PARP inhibition in TNBC.


Subject(s)
BRCA1 Protein/metabolism , Cyclin-Dependent Kinases/antagonists & inhibitors , Drug Resistance, Neoplasm/drug effects , Mutation/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Triple Negative Breast Neoplasms/enzymology , Triple Negative Breast Neoplasms/pathology , Amino Acid Sequence , Animals , BRCA1 Protein/genetics , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Line, Tumor , Cyclic N-Oxides , Cyclin-Dependent Kinases/chemistry , Cyclin-Dependent Kinases/metabolism , DNA Damage/genetics , DNA Repair/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockout Techniques , Homologous Recombination/drug effects , Humans , Indolizines , Mice , Protein Kinase Inhibitors/pharmacology , Pyridinium Compounds/pharmacology , RNA, Small Interfering/metabolism , Transcription, Genetic/drug effects , Triple Negative Breast Neoplasms/genetics , Xenograft Model Antitumor Assays
11.
Oncotarget ; 7(47): 76590-76603, 2016 11 22.
Article in English | MEDLINE | ID: mdl-27740936

ABSTRACT

Cells that are deficient in homologous recombination, such as those that have mutations in any of the Fanconi Anemia (FA)/BRCA genes, are hypersensitive to inhibition of poly(ADP-ribose) polymerase (PARP). However, FA/BRCA-deficient tumors represent a small fraction of breast cancers, which might restrict the therapeutic utility of PARP inhibitor monotherapy. The gene encoding the serine-threonine protein kinase p21-activated kinase 1 (PAK1) is amplified and/or overexpressed in several human cancer types including 25-30% of breast tumors. This enzyme controls many cellular processes by phosphorylating both cytoplasmic and nuclear substrates. Here, we show that depletion or pharmacological inhibition of PAK1 down-regulated the expression of genes involved in the FA/BRCA pathway and compromised the ability of cells to repair DNA by Homologous Recombination (HR), promoting apoptosis and reducing colony formation. Combined inhibition of PAK1 and PARP in PAK1 overexpressing breast cancer cells had a synergistic effect, enhancing apoptosis, suppressing colony formation, and delaying tumor growth in a xenograft setting. Because reduced PAK1 activity impaired FA/BRCA function, inhibition of this kinase in PAK1 amplified and/or overexpressing breast cancer cells represents a plausible strategy for expanding the utility of PARP inhibitors to FA/BRCA-proficient cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Drug Resistance, Neoplasm , Fanconi Anemia Complementation Group Proteins/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , p21-Activated Kinases/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Chromosomes, Human, Pair 11/genetics , DNA Damage/drug effects , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Drug Synergism , Fanconi Anemia Complementation Group Proteins/deficiency , Female , Gene Amplification , Gene Expression Regulation, Neoplastic/drug effects , Homologous Recombination , Humans , Mice , Xenograft Model Antitumor Assays , p21-Activated Kinases/genetics
12.
J Clin Invest ; 126(8): 3145-57, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27454289

ABSTRACT

Patients with cancers that harbor breast cancer 1 (BRCA1) mutations initially respond well to platinum and poly(ADP-ribose) polymerase inhibitor (PARPi) therapy; however, resistance invariably arises in these patients and is a major clinical problem. The BRCA1185delAG allele is a common inherited mutation located close to the protein translation start site that is thought to produce a shortened, nonfunctional peptide. In this study, we investigated the mechanisms that lead to PARPi and platinum resistance in the SUM1315MO2 breast cancer cell line, which harbors a hemizygous BRCA1185delAG mutation. SUM1315MO2 cells were initially sensitive to PARPi and cisplatin but readily acquired resistance. PARPi- and cisplatin-resistant clones did not harbor secondary reversion mutations; rather, PARPi and platinum resistance required increased expression of a really interesting gene (RING) domain-deficient BRCA1 protein (Rdd-BRCA1). Initiation of translation occurred downstream of the frameshift mutation, probably at the BRCA1-Met-297 codon. In contrast to full-length BRCA1, Rdd-BRCA1 did not require BRCA1-associated RING domain 1 (BARD1) interaction for stability. Functionally, Rdd-BRCA1 formed irradiation-induced foci and supported RAD51 foci formation. Ectopic overexpression of Rdd-BRCA1 promoted partial PARPi and cisplatin resistance. Furthermore, Rdd-BRCA1 protein expression was detected in recurrent carcinomas from patients who carried germline BRCA1185delAG mutations. Taken together, these results indicate that RING-deficient BRCA1 proteins are hypomorphic and capable of contributing to PARPi and platinum resistance when expressed at high levels.


Subject(s)
BRCA1 Protein/metabolism , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm , Platinum/pharmacology , Animals , Antibodies, Monoclonal/chemistry , Breast Neoplasms/metabolism , CRISPR-Cas Systems , Cell Line, Tumor , Cell Nucleus/metabolism , Cisplatin/pharmacology , Exons , Female , Germ-Line Mutation , Humans , Mice , Mice, Inbred NOD , Mutation , Neoplasm Transplantation , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/metabolism , Protein Domains
13.
Cancer Res ; 76(9): 2778-90, 2016 05 01.
Article in English | MEDLINE | ID: mdl-27197267

ABSTRACT

Breast and ovarian cancer patients harboring BRCA1/2 germline mutations have clinically benefitted from therapy with PARP inhibitor (PARPi) or platinum compounds, but acquired resistance limits clinical impact. In this study, we investigated the impact of mutations on BRCA1 isoform expression and therapeutic response. Cancer cell lines and tumors harboring mutations in exon 11 of BRCA1 express a BRCA1-Δ11q splice variant lacking the majority of exon 11. The introduction of frameshift mutations to exon 11 resulted in nonsense-mediated mRNA decay of full-length, but not the BRCA1-Δ11q isoform. CRISPR/Cas9 gene editing as well as overexpression experiments revealed that the BRCA1-Δ11q protein was capable of promoting partial PARPi and cisplatin resistance relative to full-length BRCA1, both in vitro and in vivo Furthermore, spliceosome inhibitors reduced BRCA1-Δ11q levels and sensitized cells carrying exon 11 mutations to PARPi treatment. Taken together, our results provided evidence that cancer cells employ a strategy to remove deleterious germline BRCA1 mutations through alternative mRNA splicing, giving rise to isoforms that retain residual activity and contribute to therapeutic resistance. Cancer Res; 76(9); 2778-90. ©2016 AACR.


Subject(s)
BRCA1 Protein/genetics , Breast Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Ovarian Neoplasms/genetics , Alternative Splicing/genetics , Animals , BRCA1 Protein/metabolism , Blotting, Western , Cisplatin/pharmacology , Female , Fluorescent Antibody Technique , Germ-Line Mutation , Humans , Immunohistochemistry , Mice , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Polymerase Chain Reaction , Protein Isoforms , Xenograft Model Antitumor Assays
14.
PLoS One ; 10(5): e0125410, 2015.
Article in English | MEDLINE | ID: mdl-25933006

ABSTRACT

Two coding variants in the APOL1 gene (G1 and G2) explain most of the high rate of kidney disease in African Americans. APOL1-associated kidney disease risk inheritance follows an autosomal recessive pattern: The relative risk of kidney disease associated with inheritance of two high-risk variants is 7-30 fold, depending on the specific kidney phenotype. We wished to determine if the variability in phenotype might in part reflect structural differences in APOL1 gene. We analyzed sequence coverage from 1000 Genomes Project Phase 3 samples as well as exome sequencing data from African American kidney disease cases for copy number variation. 8 samples sequenced in the 1000 Genomes Project showed increased coverage over a ~100kb region that includes APOL2, APOL1 and part of MYH9, suggesting the presence of APOL1 copy number greater than 2. We reasoned that such duplications should be enriched in apparent G1 heterozygotes with kidney disease. Using a PCR-based assay, we observed the presence of this duplication in additional samples from apparent G0G1 or G0G2 individuals. The frequency of this APOL1 duplication was compared among cases (n = 123) and controls (n = 255) with apparent G0G1 heterozygosity. The presence of APOL1 duplication was observed in 4.06% of cases and 0.78% controls, preliminary evidence that this APOL1 duplication may alter susceptibility to kidney disease (p = 0.03). Taqman-based copy number assays confirmed the presence of 3 APOL1 copies in individuals positive for this specific duplication by PCR assay, but also identified a small number of individuals with additional APOL1 copies of presumably different structure. These observations motivate further studies to better assess the contribution of APOL1 copy number on kidney disease risk and on APOL1 function. Investigators and clinicians genotyping APOL1 should also consider whether the particular genotyping platform used is subject to technical errors when more than two copies of APOL1 are present.


Subject(s)
Apolipoproteins/genetics , DNA Copy Number Variations , Gene Duplication , Genetic Predisposition to Disease , Kidney Diseases/genetics , Lipoproteins, HDL/genetics , Black or African American , Apolipoprotein L1 , Apolipoproteins L , Case-Control Studies , Gene Expression , Gene Frequency , Genes, Recessive , Genetic Loci , Genome, Human , Heterozygote , Human Genome Project , Humans , Kidney/metabolism , Kidney/pathology , Kidney Diseases/diagnosis , Kidney Diseases/ethnology , Kidney Diseases/pathology , Molecular Motor Proteins/genetics , Myosin Heavy Chains/genetics , Pedigree , Risk
15.
Kidney Int ; 86(6): 1116-29, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24940801

ABSTRACT

Identifying new biomarkers and therapeutic targets for podocytopathies such as focal segmental glomerulosclerosis (FSGS) requires a detailed analysis of transcriptional changes in podocytes over the course of disease. Here we used translating ribosome affinity purification (TRAP) to isolate and profile podocyte-specific mRNA in two different models of FSGS. We expressed enhanced green fluorescent protein-tagged to ribosomal protein L10a in podocytes under the control of the collagen-1α1 promoter, enabling one-step podocyte-specific mRNA isolation over the course of disease. This TRAP protocol robustly enriched known podocyte-specific mRNAs. We crossed Col1α1-eGFP-L10a mice with the Actn4(-/-) and Actn4(+/K256E) models of FSGS and analyzed podocyte transcriptional profiles at 2, 6, and 44 weeks of age. Two upregulated podocyte genes in murine FSGS (CXCL1 and DMPK) were found to be upregulated at the protein level in biopsies from patients with FSGS, validating this approach. There was no dilution of podocyte-specific transcripts during disease. These are the first podocyte-specific RNA expression data sets during aging and in two models of FSGS. This approach identified new podocyte proteins that are upregulated in FSGS and defines novel biomarkers and therapeutic targets for human glomerular disease.


Subject(s)
Actinin/genetics , Aging/genetics , Glomerulosclerosis, Focal Segmental/genetics , Glomerulosclerosis, Focal Segmental/metabolism , Podocytes/metabolism , RNA, Messenger/analysis , Aging/metabolism , Animals , Biomarkers/metabolism , Chemokine CXCL1/genetics , Chemokine CXCL1/metabolism , Collagen Type I/genetics , Collagen Type I, alpha 1 Chain , Disease Models, Animal , Gene Expression Profiling/methods , Green Fluorescent Proteins/genetics , Humans , Mice , Mice, Knockout , Myotonin-Protein Kinase/genetics , Myotonin-Protein Kinase/metabolism , Neoplasm Proteins , Oligonucleotide Array Sequence Analysis , Protein Biosynthesis , Ribosomal Protein L10 , Ribosomal Proteins/genetics , Transcriptome
16.
Proc Natl Acad Sci U S A ; 110(42): 17041-6, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24085845

ABSTRACT

Breast Cancer Type 1 Susceptibility Protein (BRCA1)-deficient cells have compromised DNA repair and are sensitive to poly(ADP-ribose) polymerase (PARP) inhibitors. Despite initial responses, the development of resistance limits clinical efficacy. Mutations in the BRCA C-terminal (BRCT) domain of BRCA1 frequently create protein products unable to fold that are subject to protease-mediated degradation. Here, we show HSP90-mediated stabilization of a BRCT domain mutant BRCA1 protein under PARP inhibitor selection pressure. The stabilized mutant BRCA1 protein interacted with PALB2-BRCA2-RAD51, was essential for RAD51 focus formation, and conferred PARP inhibitor as well as cisplatin resistance. Treatment of resistant cells with the HSP90 inhibitor 17-dimethylaminoethylamino-17-demethoxygeldanamycin reduced mutant BRCA1 protein levels and restored their sensitivity to PARP inhibition. Resistant cells also acquired a TP53BP1 mutation that facilitated DNA end resection in the absence of a BRCA1 protein capable of binding CtIP. Finally, concomitant increased mutant BRCA1 and decreased 53BP1 protein expression occur in clinical samples of BRCA1-mutated recurrent ovarian carcinomas that have developed resistance to platinum. These results provide evidence for a two-event mechanism by which BRCA1-mutant tumors acquire anticancer therapy resistance.


Subject(s)
Antineoplastic Agents/pharmacology , BRCA1 Protein/metabolism , Cisplatin/pharmacology , Drug Resistance, Neoplasm/drug effects , Mutation , Ovarian Neoplasms/metabolism , Poly(ADP-ribose) Polymerase Inhibitors , BRCA1 Protein/genetics , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Benzoquinones/pharmacology , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Fanconi Anemia Complementation Group N Protein , Female , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Humans , Lactams, Macrocyclic/pharmacology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Platinum/pharmacology , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Protein Structure, Tertiary , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
17.
J Am Soc Nephrol ; 22(11): 2091-7, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21997398

ABSTRACT

African Americans have a markedly higher incidence of ESRD compared with other racial groups. Two variants in the APOL1 gene, to date observed only among individuals of recent African ancestry, associate with increased risk for renal disease among African Americans. Here, we investigated whether these risk alleles also associate with age at initiation of chronic hemodialysis. We performed a cross-sectional study of 407 nondiabetic African Americans with ESRD who participated in the Accelerated Mortality on Renal Replacement (ArMORR) study, a prospective cohort of incident chronic hemodialysis patients. African Americans carrying two copies of the G1 risk allele initiated chronic hemodialysis at a mean age of 49.0 ± 14.9 years, which was significantly younger than both subjects with one copy of the G1 allele (55.9 ± 16.7 years; P = 0.014) and subjects without either risk allele (61.8 ± 17.1 years; P = 6.2 × 10(-7)). The association between the presence of the G1 allele and age at initiation of hemodialysis remained statistically significant after adjusting for sociodemographic and other potential confounders. We did not detect an association between the G2 risk allele and age at initiation of hemodialysis, but the sample size was limited. In conclusion, genetic variations in APOL1 identify African Americans that initiate chronic hemodialysis at a younger age. Early interventions to prevent progression of kidney disease may benefit this high-risk population.


Subject(s)
Apolipoproteins/genetics , Black or African American/genetics , Kidney Failure, Chronic/genetics , Kidney Failure, Chronic/therapy , Lipoproteins, HDL/genetics , Renal Dialysis , Adolescent , Adult , Black or African American/statistics & numerical data , Age of Onset , Aged , Aged, 80 and over , Apolipoprotein L1 , Female , Genetic Variation , Humans , Incidence , Kidney Failure, Chronic/ethnology , Male , Middle Aged , Renal Dialysis/statistics & numerical data , Risk Factors , United States/epidemiology , Young Adult
18.
Kidney Int ; 78(7): 698-704, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20668430

ABSTRACT

Genetic variation at the MYH9 locus is linked to the high incidence of focal segmental glomerulosclerosis (FSGS) and non-diabetic end-stage renal disease among African Americans. To further define risk alleles with FSGS we performed a genome-wide association analysis using more than one million single-nucleotide polymorphisms in 56 African-American and 61 European-American patients with biopsy-confirmed FSGS. Results were compared to 1641 European Americans and 1800 African Americans as unselected controls. While no association was observed in the cohort of European Americans, the case-control comparison of African Americans found variants within a 60 kb region of chromosome 22 containing part of the APOL1 and MYH9 genes associated with increased risk of FSGS. This region spans different linkage disequilibrium blocks, and variants associating with disease within this region are in linkage disequilibrium with variants which have shown signals of natural selection. APOL1 is a strong candidate for a gene that has undergone recent natural selection and is known to be involved in the infection by Trypanosoma brucei, a parasite common in Africa that has recently adapted to infect human hosts. Further studies will be required to establish which variants are causally related to kidney disease, what mutations caused the selective sweep, and to ultimately determine if these are the same.


Subject(s)
Apolipoproteins/genetics , Black or African American/genetics , Glomerulosclerosis, Focal Segmental/genetics , Lipoproteins, HDL/genetics , Molecular Motor Proteins/genetics , Myosin Heavy Chains/genetics , Adult , Alleles , Apolipoprotein L1 , Female , Glomerulosclerosis, Focal Segmental/etiology , Humans , Linkage Disequilibrium , Male , Middle Aged , Polymorphism, Single Nucleotide , Risk
19.
Science ; 329(5993): 841-5, 2010 Aug 13.
Article in English | MEDLINE | ID: mdl-20647424

ABSTRACT

African Americans have higher rates of kidney disease than European Americans. Here, we show that, in African Americans, focal segmental glomerulosclerosis (FSGS) and hypertension-attributed end-stage kidney disease (H-ESKD) are associated with two independent sequence variants in the APOL1 gene on chromosome 22 {FSGS odds ratio = 10.5 [95% confidence interval (CI) 6.0 to 18.4]; H-ESKD odds ratio = 7.3 (95% CI 5.6 to 9.5)}. The two APOL1 variants are common in African chromosomes but absent from European chromosomes, and both reside within haplotypes that harbor signatures of positive selection. ApoL1 (apolipoprotein L-1) is a serum factor that lyses trypanosomes. In vitro assays revealed that only the kidney disease-associated ApoL1 variants lysed Trypanosoma brucei rhodesiense. We speculate that evolution of a critical survival factor in Africa may have contributed to the high rates of renal disease in African Americans.


Subject(s)
Apolipoproteins/genetics , Black or African American/genetics , Glomerulosclerosis, Focal Segmental/genetics , Kidney Failure, Chronic/genetics , Lipoproteins, HDL/genetics , Polymorphism, Single Nucleotide , Trypanosoma brucei rhodesiense/metabolism , Africa , Alleles , Apolipoprotein L1 , Apolipoproteins/blood , Apolipoproteins/metabolism , Case-Control Studies , Cohort Studies , Gene Frequency , Genetic Association Studies , Genetic Predisposition to Disease , Glomerulosclerosis, Focal Segmental/ethnology , Haplotypes , Humans , Hypertension/complications , Immunity, Innate , Kidney Failure, Chronic/ethnology , Kidney Failure, Chronic/etiology , Linkage Disequilibrium , Lipoproteins, HDL/blood , Lipoproteins, HDL/metabolism , Logistic Models , Molecular Motor Proteins/genetics , Myosin Heavy Chains/genetics , Recombinant Proteins/metabolism , Selection, Genetic , Sequence Deletion , Trypanosomiasis, African/genetics , Trypanosomiasis, African/parasitology
SELECTION OF CITATIONS
SEARCH DETAIL
...