Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38645235

ABSTRACT

Familial dilated cardiomyopathy (DCM) is frequently caused by autosomal dominant point mutations in genes involved in diverse cellular processes, including sarcomeric contraction. While patient studies have defined the genetic landscape of DCM, genetics are not currently used in patient care, and patients receive similar treatments regardless of the underlying mutation. It has been suggested that a precision medicine approach based on the molecular mechanism of the underlying mutation could improve outcomes; however, realizing this approach has been challenging due to difficulties linking genotype and phenotype and then leveraging this information to identify therapeutic approaches. Here, we used multiscale experimental and computational approaches to test whether knowledge of molecular mechanism could be harnessed to connect genotype, phenotype, and drug response for a DCM mutation in troponin T, deletion of K210. Previously, we showed that at the molecular scale, the mutation reduces thin filament activation. Here, we used computational modeling of this molecular defect to predict that the mutant will reduce cellular and tissue contractility, and we validated this prediction in human cardiomyocytes and engineered heart tissues. We then used our knowledge of molecular mechanism to computationally model the effects of a small molecule that can activate the thin filament. We demonstrate experimentally that the modeling correctly predicts that the small molecule can partially rescue systolic dysfunction at the expense of diastolic function. Taken together, our results demonstrate how molecular mechanism can be harnessed to connect genotype and phenotype and inspire strategies to optimize mechanism-based therapeutics for DCM.

2.
Cell Metab ; 34(10): 1499-1513.e8, 2022 10 04.
Article in English | MEDLINE | ID: mdl-36070756

ABSTRACT

Adipocytes transfer mitochondria to macrophages in white and brown adipose tissues to maintain metabolic homeostasis. In obesity, adipocyte-to-macrophage mitochondria transfer is impaired, and instead, adipocytes release mitochondria into the blood to induce a protective antioxidant response in the heart. We found that adipocyte-to-macrophage mitochondria transfer in white adipose tissue is inhibited in murine obesity elicited by a lard-based high-fat diet, but not a hydrogenated-coconut-oil-based high-fat diet, aging, or a corn-starch diet. The long-chain fatty acids enriched in lard suppress mitochondria capture by macrophages, diverting adipocyte-derived mitochondria into the blood for delivery to other organs, such as the heart. The depletion of macrophages rapidly increased the number of adipocyte-derived mitochondria in the blood. These findings suggest that dietary lipids regulate mitochondria uptake by macrophages locally in white adipose tissue to determine whether adipocyte-derived mitochondria are released into systemic circulation to support the metabolic adaptation of distant organs in response to nutrient stress.


Subject(s)
Adipose Tissue, White , Antioxidants , Adipocytes/metabolism , Adipose Tissue, White/metabolism , Animals , Antioxidants/metabolism , Diet, High-Fat , Fatty Acids/metabolism , Macrophages/metabolism , Mice , Mitochondria/metabolism , Obesity/metabolism , Starch/metabolism
3.
Circulation ; 146(8): 623-638, 2022 08 23.
Article in English | MEDLINE | ID: mdl-35880523

ABSTRACT

BACKGROUND: Cellular rejection after heart transplantation imparts significant morbidity and mortality. Current immunosuppressive strategies are imperfect, target recipient T cells, and have adverse effects. The innate immune response plays an essential role in the recruitment and activation of T cells. Targeting the donor innate immune response would represent the earliest interventional opportunity within the immune response cascade. There is limited knowledge about donor immune cell types and functions in the setting of cardiac transplantation, and no current therapeutics exist for targeting these cell populations. METHODS: Using genetic lineage tracing, cell ablation, and conditional gene deletion, we examined donor mononuclear phagocyte diversity and macrophage function during acute cellular rejection of transplanted hearts in mice. We performed single-cell RNA sequencing on donor and recipient macrophages and monocytes at multiple time points after transplantation. On the basis of our imaging and single-cell RNA sequencing data, we evaluated the functional relevance of donor CCR2+ (C-C chemokine receptor 2) and CCR2- macrophages using selective cell ablation strategies in donor grafts before transplant. Last, we performed functional validation that donor macrophages signal through MYD88 (myeloid differentiation primary response protein 88) to facilitate cellular rejection. RESULTS: Donor macrophages persisted in the rejecting transplanted heart and coexisted with recipient monocyte-derived macrophages. Single-cell RNA sequencing identified donor CCR2+ and CCR2- macrophage populations and revealed remarkable diversity among recipient monocytes, macrophages, and dendritic cells. Temporal analysis demonstrated that donor CCR2+ and CCR2- macrophages were transcriptionally distinct, underwent significant morphologic changes, and displayed unique activation signatures after transplantation. Although selective depletion of donor CCR2- macrophages reduced allograft survival, depletion of donor CCR2+ macrophages prolonged allograft survival. Pathway analysis revealed that donor CCR2+ macrophages are activated through MYD88/nuclear factor kappa light chain enhancer of activated B cells signaling. Deletion of MYD88 in donor macrophages resulted in reduced antigen-presenting cell recruitment, reduced ability of antigen-presenting cells to present antigen to T cells, decreased emergence of allograft-reactive T cells, and extended allograft survival. CONCLUSIONS: Distinct populations of donor and recipient macrophages coexist within the transplanted heart. Donor CCR2+ macrophages are key mediators of allograft rejection, and deletion of MYD88 signaling in donor macrophages is sufficient to suppress rejection and extend allograft survival. This highlights the therapeutic potential of donor heart-based interventions.


Subject(s)
Heart Transplantation , Animals , Graft Rejection/prevention & control , Heart Transplantation/adverse effects , Humans , Macrophages , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/genetics , Tissue Donors
4.
Development ; 149(8)2022 04 15.
Article in English | MEDLINE | ID: mdl-35178561

ABSTRACT

Tissue-resident macrophages are increasingly recognized as important determinants of organ homeostasis, tissue repair, remodeling and regeneration. Although the ontogeny and function of tissue-resident macrophages has been identified as distinct from postnatal hematopoiesis, the inability to specify, in vitro, similar populations that recapitulate these developmental waves has limited our ability to study their function and potential for regenerative applications. We took advantage of the concept that tissue-resident macrophages and monocyte-derived macrophages originate from distinct extra-embryonic and definitive hematopoietic lineages to devise a system to generate pure cultures of macrophages that resemble tissue-resident or monocyte-derived subsets. We demonstrate that human pluripotent stem cell-derived extra-embryonic-like and intra-embryonic-like hematopoietic progenitors differentiate into morphologically, transcriptionally and functionally distinct macrophage populations. Single-cell RNA sequencing of developing and mature cultures uncovered distinct developmental trajectories and gene expression programs of macrophages derived from extra-embryonic-like and intra-embryonic-like hematopoietic progenitors. These findings establish a resource for the generation of human tissue resident-like macrophages to study their specification and function under defined conditions and to explore their potential use in tissue engineering and regenerative medicine applications.


Subject(s)
Macrophages , Pluripotent Stem Cells , Cell Differentiation/genetics , Hematopoiesis , Homeostasis , Humans , Macrophages/metabolism
5.
JACC Basic Transl Sci ; 6(4): 331-345, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33681537

ABSTRACT

There is ongoing debate as to whether cardiac complications of coronavirus disease-2019 (COVID-19) result from myocardial viral infection or are secondary to systemic inflammation and/or thrombosis. We provide evidence that cardiomyocytes are infected in patients with COVID-19 myocarditis and are susceptible to severe acute respiratory syndrome coronavirus 2. We establish an engineered heart tissue model of COVID-19 myocardial pathology, define mechanisms of viral pathogenesis, and demonstrate that cardiomyocyte severe acute respiratory syndrome coronavirus 2 infection results in contractile deficits, cytokine production, sarcomere disassembly, and cell death. These findings implicate direct infection of cardiomyocytes in the pathogenesis of COVID-19 myocardial pathology and provides a model system to study this emerging disease.

6.
bioRxiv ; 2020 Nov 05.
Article in English | MEDLINE | ID: mdl-33173875

ABSTRACT

Epidemiological studies of the COVID-19 pandemic have revealed evidence of cardiac involvement and documented that myocardial injury and myocarditis are predictors of poor outcomes. Nonetheless, little is understood regarding SARS-CoV-2 tropism within the heart and whether cardiac complications result directly from myocardial infection. Here, we develop a human engineered heart tissue model and demonstrate that SARS-CoV-2 selectively infects cardiomyocytes. Viral infection is dependent on expression of angiotensin-I converting enzyme 2 (ACE2) and endosomal cysteine proteases, suggesting an endosomal mechanism of cell entry. After infection with SARS-CoV-2, engineered tissues display typical features of myocarditis, including cardiomyocyte cell death, impaired cardiac contractility, and innate immune cell activation. Consistent with these findings, autopsy tissue obtained from individuals with COVID-19 myocarditis demonstrated cardiomyocyte infection, cell death, and macrophage-predominate immune cell infiltrate. These findings establish human cardiomyocyte tropism for SARS-CoV-2 and provide an experimental platform for interrogating and mitigating cardiac complications of COVID-19.

7.
J Cell Biol ; 218(7): 2113-2123, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31123184

ABSTRACT

XRCC4-like factor (XLF) is a non-homologous end joining (NHEJ) DNA double strand break repair protein. However, XLF deficiency leads to phenotypes in mice and humans that are not necessarily consistent with an isolated defect in NHEJ. Here we show that XLF functions during DNA replication. XLF undergoes cell division cycle 7-dependent phosphorylation; associates with the replication factor C complex, a critical component of the replisome; and is found at replication forks. XLF deficiency leads to defects in replication fork progression and an increase in fork reversal. The additional loss of H2AX, which protects DNA ends from resection, leads to a requirement for ATR to prevent an MRE11-dependent loss of newly synthesized DNA and activation of DNA damage response. Moreover, H2ax-/-:Xlf-/- cells exhibit a marked dependence on the ATR kinase for survival. We propose that XLF and H2AX function in series to prevent replication stress induced by the MRE11-dependent resection of regressed arms at reversed replication forks.


Subject(s)
DNA-Binding Proteins/genetics , Histones/genetics , MRE11 Homologue Protein/genetics , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Cell Division/genetics , DNA Breaks, Double-Stranded , DNA Damage/genetics , DNA End-Joining Repair/genetics , DNA Repair/genetics , DNA Replication/genetics , Fibroblasts/metabolism , Mice , Phosphorylation/genetics
8.
Mol Cell ; 71(2): 332-342.e8, 2018 07 19.
Article in English | MEDLINE | ID: mdl-30017584

ABSTRACT

The modulator of retrovirus infection (MRI or CYREN) is a 30-kDa protein with a conserved N-terminal Ku-binding motif (KBM) and a C-terminal XLF-like motif (XLM). We show that MRI is intrinsically disordered and interacts with many DNA damage response (DDR) proteins, including the kinases ataxia telangiectasia mutated (ATM) and DNA-PKcs and the classical non-homologous end joining (cNHEJ) factors Ku70, Ku80, XRCC4, XLF, PAXX, and XRCC4. MRI forms large multimeric complexes that depend on its N and C termini and localizes to DNA double-strand breaks (DSBs), where it promotes the retention of DDR factors. Mice deficient in MRI and XLF exhibit embryonic lethality at a stage similar to those deficient in the core cNHEJ factors XRCC4 or DNA ligase IV. Moreover, MRI is required for cNHEJ-mediated DSB repair in XLF-deficient lymphocytes. We propose that MRI is an adaptor that, through multivalent interactions, increases the avidity of DDR factors to DSB-associated chromatin to promote cNHEJ.


Subject(s)
DNA Breaks, Double-Stranded , DNA End-Joining Repair , Animals , Cell Cycle Proteins/metabolism , Chromatin/genetics , Chromatin/metabolism , DNA Ligase ATP/genetics , DNA Repair , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Ku Autoantigen/genetics , Mice
9.
SLAS Discov ; 23(7): 624-633, 2018 08.
Article in English | MEDLINE | ID: mdl-29232168

ABSTRACT

DNA double-strand breaks (DSBs) are repaired primarily by homologous recombination (HR) or nonhomologous end joining (NHEJ). Compounds that modulate HR have shown promise as cancer therapeutics. The V(D)J recombination reaction, which assembles antigen receptor genes in lymphocytes, is initiated by the introduction of DNA DSBs at two recombining gene segments by the RAG endonuclease, followed by the NHEJ-mediated repair of these DSBs. Here, using HyperCyt automated flow cytometry, we develop a robust high-throughput screening (HTS) assay for NHEJ that utilizes engineered pre-B-cell lines where the V(D)J recombination reaction can be induced and monitored at a single-cell level. This approach, novel in processing four 384-well plates at a time in parallel, was used to screen the National Cancer Institute NeXT library to identify compounds that inhibit V(D)J recombination and NHEJ. Assessment of cell light scattering characteristics at the primary HTS stage (83,536 compounds) enabled elimination of 60% of apparent hits as false positives. Although all the active compounds that we identified had an inhibitory effect on RAG cleavage, we have established this as an approach that could identify compounds that inhibit RAG cleavage or NHEJ using new chemical libraries.


Subject(s)
DNA End-Joining Repair/drug effects , Drug Discovery , High-Throughput Screening Assays , DNA Breaks, Double-Stranded/drug effects , Dose-Response Relationship, Drug , Drug Discovery/methods , Flow Cytometry , Homologous Recombination , Humans , Molecular Structure , Precursor Cells, B-Lymphoid/immunology , Precursor Cells, B-Lymphoid/metabolism , V(D)J Recombination
10.
Cell Cycle ; 16(3): 286-295, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27830975

ABSTRACT

Non-homologous end joining (NHEJ) is a major DNA double-strand break (DSB) repair pathway that functions in all phases of the cell cycle. NHEJ repairs genotoxic and physiological DSBs, such as those generated by ionizing radiation and during V(D)J recombination at antigen receptor loci, respectively. DNA end joining by NHEJ relies on the core factors Ku70, Ku80, XRCC4, and DNA Ligase IV. Additional proteins also play important roles in NHEJ. The XRCC4-like factor (XLF) participates in NHEJ through its interaction with XRCC4, and XLF deficiency in humans leads to immunodeficiency and increased sensitivity to ionizing radiation. However, XLF is dispensable for NHEJ-mediated DSB repair during V(D)J recombination in murine lymphocytes, where it may have redundant functions with other DSB repair factors. Paralog of XRCC4 and XLF (PAXX) is a recently identified NHEJ factor that has structural similarity to XRCC4 and XLF. Here we show that PAXX is also dispensable for NHEJ during V(D)J recombination and during the repair of genotoxic DSBs in lymphocytes. However, a combined deficiency of PAXX and XLF blocks NHEJ with a severity comparable to that observed in DNA Ligase IV-deficient cells. Similar to XLF, PAXX interacts with Ku through its C-terminal region, and mutations that disrupt Ku binding prevent PAXX from promoting NHEJ in XLF-deficient lymphocytes. Our findings suggest that the PAXX and XLF proteins may have redundant functions during NHEJ.


Subject(s)
B-Lymphocytes/metabolism , DNA Breaks, Double-Stranded , DNA End-Joining Repair , DNA-Binding Proteins/deficiency , Animals , DNA Repair , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Mice , Mutant Proteins/metabolism , Protein Domains , V(D)J Recombination
11.
Curr Biol ; 25(1): R45-6, 2015 Jan 05.
Article in English | MEDLINE | ID: mdl-25562302

ABSTRACT

The RAG endonuclease generates DNA double strand breaks during antigen receptor gene assembly, an essential process for B- and T-lymphocyte development. However, a recent study reveals that RAG endonuclease activity affects natural killer cell function, demonstrating that such double strand breaks, and the responses they elicit, may have broad cellular effects.


Subject(s)
DNA-Binding Proteins/metabolism , Homeodomain Proteins/metabolism , Killer Cells, Natural/immunology , Animals
12.
Mol Cell ; 56(6): 808-18, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25435138

ABSTRACT

The structure of broken DNA ends is a critical determinant of the pathway used for DNA double-strand break (DSB) repair. Here, we develop an approach involving the hairpin capture of DNA end structures (HCoDES), which elucidates chromosomal DNA end structures at single-nucleotide resolution. HCoDES defines structures of physiologic DSBs generated by the RAG endonuclease, as well as those generated by nucleases widely used for genome editing. Analysis of G1 phase cells deficient in H2AX or 53BP1 reveals DNA ends that are frequently resected to form long single-stranded overhangs that can be repaired by mutagenic pathways. In addition to 3' overhangs, many of these DNA ends unexpectedly form long 5' single-stranded overhangs. The divergence in DNA end structures resolved by HCoDES suggests that H2AX and 53BP1 may have distinct activities in end protection. Thus, the high-resolution end structures obtained by HCoDES identify features of DNA end processing during DSB repair.


Subject(s)
Chromosomes, Human/genetics , Sequence Analysis, DNA/methods , Base Sequence , Cells, Cultured , DNA Breaks, Double-Stranded , Humans
13.
Mol Cell Biol ; 34(15): 2811-21, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24842905

ABSTRACT

The resection of broken DNA ends is required for DNA double-strand break (DSB) repair by homologous recombination (HR) but can inhibit normal repair by nonhomologous end joining (NHEJ), the main DSB repair pathway in G1-phase cells. Antigen receptor gene assembly proceeds through DNA DSB intermediates generated in G1-phase lymphocytes by the RAG endonuclease. These DSBs activate ATM, which phosphorylates H2AX, forming γ-H2AX in flanking chromatin. γ-H2AX prevents CtIP from initiating resection of RAG DSBs. Whether there are additional proteins required to promote resection of these DNA ends is not known. KRAB-associated protein 1 (KAP-1) (TRIM28) is a transcriptional repressor that modulates chromatin structure and has been implicated in the repair of DNA DSBs in heterochromatin. Here, we show that in murine G1-phase lymphocytes, KAP-1 promotes resection of DSBs that are not protected by H2AX and its downstream effector 53BP1. In these murine cells, KAP-1 activity in DNA end resection is attenuated by a single-amino-acid change that reflects a KAP-1 polymorphism between primates and other mammalian species. These findings establish KAP-1 as a component of the machinery that can resect DNA ends in G1-phase cells and suggest that there may be species-specific features to this activity.


Subject(s)
DNA End-Joining Repair/genetics , DNA-Binding Proteins/metabolism , G1 Phase/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lymphocytes/metabolism , Animals , Cells, Cultured , DNA/genetics , DNA Breaks, Double-Stranded , DNA-Binding Proteins/genetics , Heterochromatin/metabolism , Histones/genetics , Histones/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Inbred C57BL , Phosphorylation/genetics
14.
Proc Natl Acad Sci U S A ; 111(1): 391-6, 2014 Jan 07.
Article in English | MEDLINE | ID: mdl-24371308

ABSTRACT

Deregulated activation of ß-catenin in cancer has been correlated with genomic instability. During thymocyte development, ß-catenin activates transcription in partnership with T-cell-specific transcription factor 1 (Tcf-1). We previously reported that targeted activation of ß-catenin in thymocytes (CAT mice) induces lymphomas that depend on recombination activating gene (RAG) and myelocytomatosis oncogene (Myc) activities. Here we show that these lymphomas have recurring Tcra/Myc translocations that resulted from illegitimate RAG recombination events and resembled oncogenic translocations previously described in human T-ALL. We therefore used the CAT animal model to obtain mechanistic insights into the transformation process. ChIP-seq analysis uncovered a link between Tcf-1 and RAG2 showing that the two proteins shared binding sites marked by trimethylated histone-3 lysine-4 (H3K4me3) throughout the genome, including near the translocation sites. Pretransformed CAT thymocytes had increased DNA damage at the translocating loci and showed altered repair of RAG-induced DNA double strand breaks. These cells were able to survive despite DNA damage because activated ß-catenin promoted an antiapoptosis gene expression profile. Thus, activated ß-catenin promotes genomic instability that leads to T-cell lymphomas as a consequence of altered double strand break repair and increased survival of thymocytes with damaged DNA.


Subject(s)
Genomic Instability , Lymphocyte Activation , Lymphoma/genetics , T-Lymphocytes/cytology , beta Catenin/metabolism , Animals , Apoptosis , Base Sequence , Cell Survival , DNA Breaks, Double-Stranded , DNA Methylation , DNA Repair , Disease Models, Animal , Genes, RAG-1/genetics , Hepatocyte Nuclear Factor 1-alpha , Histones/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Sequence Data , Recombination, Genetic , T Cell Transcription Factor 1/metabolism , Thymocytes/cytology , Translocation, Genetic , beta Catenin/genetics
15.
Science ; 337(6090): 104-9, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22628553

ABSTRACT

Tumors exhibit numerous recurrent hemizygous focal deletions that contain no known tumor suppressors and are poorly understood. To investigate whether these regions contribute to tumorigenesis, we searched genetically for genes with cancer-relevant properties within these hemizygous deletions. We identified STOP and GO genes, which negatively and positively regulate proliferation, respectively. STOP genes include many known tumor suppressors, whereas GO genes are enriched for essential genes. Analysis of their chromosomal distribution revealed that recurring deletions preferentially overrepresent STOP genes and underrepresent GO genes. We propose a hypothesis called the cancer gene island model, whereby gene islands encompassing high densities of STOP genes and low densities of GO genes are hemizygously deleted to maximize proliferative fitness through cumulative haploinsufficiencies. Because hundreds to thousands of genes are hemizygously deleted per tumor, this mechanism may help to drive tumorigenesis across many cancer types.


Subject(s)
Cell Proliferation , Cell Transformation, Neoplastic , Genes, Neoplasm , Haploinsufficiency , Neoplasms/genetics , Neoplasms/pathology , Sequence Deletion , Cell Line , Cell Line, Tumor , Chromosome Mapping , Genes, Essential , Genes, Recessive , Genes, Tumor Suppressor , Hemizygote , Humans , Models, Genetic , Oncogenes
16.
Mol Cell ; 40(4): 645-57, 2010 Nov 24.
Article in English | MEDLINE | ID: mdl-21055985

ABSTRACT

Replication stress involving collision of replisomes with camptothecin (CPT)-stabilized DNA-Topoisomerase I adducts activates an ATR-dependent pathway to promote repair by homologous recombination. To identify human genes that protect cells from such replication stress, we performed a genome-wide CPT sensitivity screen. Among numerous candidate genes are two previously unstudied proteins: the ankyrin repeat protein NFKBIL2 and C6ORF167 (MMS22L), distantly related to yeast replication stress regulator Mms22p. MMS22L and NFKBIL2 interact with each other and with FACT (facilitator of chromatin transcription) and MCM (minichromosome maintenance) complexes. Cells depleted of NFKBIL2 or MMS22L are sensitive to DNA-damaging agents, load phosphorylated RPA onto chromatin in a CTIP-dependent manner, activate the ATR/ATRIP-CHK1 and double-strand break repair signaling pathways, and are defective in HR. This study identifies MMS22L-NFKBIL2 as components of the replication stress control pathway and provides a resource for discovery of additional components of this pathway.


Subject(s)
Camptothecin/pharmacology , DNA-Binding Proteins/metabolism , Genetic Testing , Genome, Human/genetics , Genomic Instability/drug effects , NF-kappa B/metabolism , Nuclear Proteins/metabolism , DNA Damage , DNA Repair/drug effects , DNA Replication/drug effects , DNA-Directed DNA Polymerase/metabolism , Drug Resistance, Neoplasm/drug effects , HeLa Cells , Histones/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Multienzyme Complexes/metabolism , NF-kappa B/deficiency , Phosphorylation/drug effects , Protein Binding/drug effects , RNA, Small Interfering/metabolism , Recombination, Genetic/drug effects , Recombination, Genetic/genetics , Replication Protein A/metabolism , Reproducibility of Results , Stress, Physiological/drug effects , Tumor Suppressor p53-Binding Protein 1
17.
Nat Immunol ; 11(3): 207-15, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20081848

ABSTRACT

Hematopoietic stem cell (HSC) differentiation is regulated by cell-intrinsic and cell-extrinsic cues. In addition to transcriptional regulation, post-translational regulation may also control HSC differentiation. To test this hypothesis, we visualized the ubiquitin-regulated protein stability of a single transcription factor, c-Myc. The stability of c-Myc protein was indicative of HSC quiescence, and c-Myc protein abundance was controlled by the ubiquitin ligase Fbw7. Fine changes in the stability of c-Myc protein regulated the HSC gene-expression signature. Using whole-genome genomic approaches, we identified specific regulators of HSC function directly controlled by c-Myc binding; however, adult HSCs and embryonic stem cells sensed and interpreted c-Myc-regulated gene expression in distinct ways. Our studies show that a ubiquitin ligase-substrate pair can orchestrate the molecular program of HSC differentiation.


Subject(s)
Cell Differentiation/physiology , Hematopoietic Stem Cells/cytology , Ubiquitin-Protein Ligases/immunology , Animals , Cell Cycle/genetics , Cell Cycle/immunology , Cell Cycle Proteins/immunology , Cell Differentiation/genetics , Chromatin Immunoprecipitation , Flow Cytometry , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Mice , Mice, Knockout , Proto-Oncogene Proteins c-myc/immunology
18.
J Exp Med ; 206(12): 2625-39, 2009 Nov 23.
Article in English | MEDLINE | ID: mdl-19887394

ABSTRACT

The H2AX core histone variant is phosphorylated in chromatin around DNA double strand breaks (DSBs) and functions through unknown mechanisms to suppress antigen receptor locus translocations during V(D)J recombination. Formation of chromosomal coding joins and suppression of translocations involves the ataxia telangiectasia mutated and DNA-dependent protein kinase catalytic subunit serine/threonine kinases, each of which phosphorylates H2AX along cleaved antigen receptor loci. Using Abelson transformed pre-B cell lines, we find that H2AX is not required for coding join formation within chromosomal V(D)J recombination substrates. Yet we show that H2AX is phosphorylated along cleaved Igkappa DNA strands and prevents their separation in G1 phase cells and their progression into chromosome breaks and translocations after cellular proliferation. We also show that H2AX prevents chromosome breaks emanating from unrepaired RAG endonuclease-generated TCR-alpha/delta locus coding ends in primary thymocytes. Our data indicate that histone H2AX suppresses translocations during V(D)J recombination by creating chromatin modifications that stabilize disrupted antigen receptor locus DNA strands to prevent their irreversible dissociation. We propose that such H2AX-dependent mechanisms could function at additional chromosomal locations to facilitate the joining of DNA ends generated by other types of DSBs.


Subject(s)
Chromatin/metabolism , Chromosome Breakage , DNA Breaks, Double-Stranded , Histones/metabolism , Recombination, Genetic/physiology , VDJ Exons/physiology , Animals , Ataxia Telangiectasia Mutated Proteins , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/immunology , Cell Cycle Proteins/metabolism , Cell Line, Transformed , Chromatin/genetics , Chromatin/immunology , G1 Phase/physiology , Histones/genetics , Histones/immunology , Mice , Mice, Knockout , Phosphorylation/physiology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , Quantitative Trait Loci/physiology , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/immunology , Receptors, Antigen, B-Cell/metabolism , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism
19.
Genes Dev ; 23(20): 2415-25, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19793862

ABSTRACT

The integrity of genomic DNA is continuously challenged by the presence of DNA base lesions or DNA strand breaks. Here we report the identification of a new DNA damage response protein, SMARCAL1 (SWI/SNF-related, matrix associated, actin-dependent regulator of chromatin, subfamily a-like 1), which is a member of the SNF2 family and is mutated in Schimke immunoosseous dysplasia (SIOD). We demonstrate that SMARCAL1 directly interacts with Replication protein A (RPA) and is recruited to sites of DNA damage in an RPA-dependent manner. SMARCAL1-depleted cells display sensitivity to DNA-damaging agents that induce replication fork collapse, and exhibit slower fork recovery and delayed entry into mitosis following S-phase arrest. Furthermore, SIOD patient fibroblasts reconstituted with SMARCAL1 exhibit faster cell cycle progression after S-phase arrest. Thus, the symptoms of SIOD may be caused, at least in part, by defects in the cellular response to DNA replication stress.


Subject(s)
DNA Helicases/metabolism , Osteochondrodysplasias/physiopathology , Replication Protein A/metabolism , Amino Acid Sequence , Cell Cycle , Cell Line , DNA Damage , DNA Helicases/chemistry , DNA Replication , Humans , Molecular Sequence Data , Osteochondrodysplasias/genetics , Sequence Alignment
20.
Proc Natl Acad Sci U S A ; 106(43): 18339-44, 2009 Oct 27.
Article in English | MEDLINE | ID: mdl-19820166

ABSTRACT

Canonical chromosomal translocations juxtaposing antigen receptor genes and oncogenes are a hallmark of many lymphoid malignancies. These translocations frequently form through the joining of DNA ends from double-strand breaks (DSBs) generated by the recombinase activating gene (RAG)-1 and -2 proteins at lymphocyte antigen receptor loci and breakpoint targets near oncogenes. Our understanding of chromosomal breakpoint target selection comes primarily from the analyses of these lesions, which are selected based on their transforming properties. RAG DSBs are rarely resolved aberrantly in wild-type developing lymphocytes. However, in ataxia telangiectasia mutated (ATM)-deficient lymphocytes, RAG breaks are frequently joined aberrantly, forming chromosomal lesions such as translocations that predispose (ATM)-deficient mice and humans to the development of lymphoid malignancies. Here, an approach that minimizes selection biases is used to isolate a large cohort of breakpoint targets of aberrantly resolved RAG DSBs in Atm-deficient lymphocytes. Analyses of this cohort revealed that frequently, the breakpoint targets for aberrantly resolved RAG breaks are other DSBs. Moreover, these nonselected lesions exhibit a bias for using breakpoints in cis, forming small chromosomal deletions, rather than breakpoints in trans, forming chromosomal translocations.


Subject(s)
Chromosome Breakage , DNA Breaks, Double-Stranded , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Homeodomain Proteins/genetics , Lymphocytes/metabolism , Protein Serine-Threonine Kinases/deficiency , Translocation, Genetic , Tumor Suppressor Proteins/deficiency , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , Chromosomes, Mammalian/metabolism , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , Homeodomain Proteins/immunology , Homeodomain Proteins/metabolism , Lymphocytes/immunology , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptors, Antigen/immunology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...