Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
FASEB J ; 34(1): 631-647, 2020 01.
Article in English | MEDLINE | ID: mdl-31914608

ABSTRACT

In mammalian cells, the catabolic activity of the dNTP triphosphohydrolase SAMHD1 sets the balance and concentration of the four dNTPs. Deficiency of SAMHD1 leads to unequally increased pools and marked dNTP imbalance. Imbalanced dNTP pools increase mutation frequency in cancer cells, but it is not known if the SAMHD1-induced dNTP imbalance favors accumulation of somatic mutations in non-transformed cells. Here, we have investigated how fibroblasts from Aicardi-Goutières Syndrome (AGS) patients with mutated SAMHD1 react to the constitutive pool imbalance characterized by a huge dGTP pool. We focused on the effects on dNTP pools, cell cycle progression, dynamics and fidelity of DNA replication, and efficiency of UV-induced DNA repair. AGS fibroblasts entered senescence prematurely or upregulated genes involved in G1/S transition and DNA replication. The normally growing AGS cells exhibited unchanged DNA replication dynamics and, when quiescent, faster rate of excision repair of UV-induced DNA damages. To investigate whether the lack of SAMHD1 affects DNA replication fidelity, we compared de novo mutations in AGS and WT cells by exome next-generation sequencing. Somatic variant analysis indicated a mutator phenotype suggesting that SAMHD1 is a caretaker gene whose deficiency is per se mutagenic, promoting genome instability in non-transformed cells.


Subject(s)
Autoimmune Diseases of the Nervous System/genetics , Fibroblasts/metabolism , Mutation/genetics , Nervous System Malformations/genetics , SAM Domain and HD Domain-Containing Protein 1/deficiency , DNA Damage/genetics , DNA Replication/genetics , Humans , Monomeric GTP-Binding Proteins/metabolism , SAM Domain and HD Domain-Containing Protein 1/genetics
2.
Methods ; 156: 79-84, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30578845

ABSTRACT

A major concern of CRISPR and related genome engineering technologies is off-target mutagenesis from prolonged exposure to Cas9 and related editing enzymes. To help mitigate this concern we added a loxP site to the 3'-LTR of an HIV-based lentiviral vector capable of expressing Cas9/gRNA complexes in a wide variety of mammalian cell types. Transduction of susceptible target cells yields an integrated provirus that expresses the desired Cas9/gRNA complex. The reverse transcription process also results in duplication of the 3'-LTR such that the integrated provirus becomes flanked by loxP sites (floxed). Subsequent expression of Cre recombinase results in loxP-to-loxP site-specific recombination that deletes the Cas9/gRNA payload and effectively prevents additional Cas9-mediated mutations. This construct also expresses a gRNA with a single transcription termination sequence, which results in higher expression levels and more efficient genome engineering as evidenced by disruption of the SAMHD1 gene. This hit-and-run CRISPR approach was validated by recreating a natural APOBEC3B deletion and by disrupting the mismatch repair gene MSH2. This hit-and-run strategy may have broad utility in many areas and especially those where cell types are difficult to engineer by transient delivery of ribonucleoprotein complexes.


Subject(s)
CRISPR-Associated Protein 9/genetics , CRISPR-Cas Systems , Gene Editing/methods , Integrases/genetics , Lentivirus/genetics , RNA, Guide, Kinetoplastida/genetics , Recombination, Genetic , Base Pairing , Base Sequence , CRISPR-Associated Protein 9/metabolism , Cytidine Deaminase/deficiency , Cytidine Deaminase/genetics , Exons , Gene Deletion , Genetic Vectors/chemistry , Genetic Vectors/metabolism , HEK293 Cells , Humans , Integrases/metabolism , Introns , Lentivirus/metabolism , MCF-7 Cells , Minor Histocompatibility Antigens/genetics , MutS Homolog 2 Protein/deficiency , MutS Homolog 2 Protein/genetics , RNA, Guide, Kinetoplastida/metabolism , SAM Domain and HD Domain-Containing Protein 1/deficiency , SAM Domain and HD Domain-Containing Protein 1/genetics
3.
Cell Cycle ; 17(23): 2564-2576, 2018.
Article in English | MEDLINE | ID: mdl-30474474

ABSTRACT

Sterile alpha motif and HD domain-containing protein 1 (SAMHD1) is a mammalian dNTP hydrolase (dNTPase) and functions as a negative regulator in the efficacy of cytarabine treatment of acute myeloid leukemia (AML). We have reported that SAMHD1 knockout (KO) increased the activity of phosphoinositide 3-kinase (PI3K) in AML-derived THP-1 cells and attenuated their ability to form subcutaneous tumors in xenografted immunodeficient mice. However, the functional significance of SAMHD1 in controlling AML leukemogenesis remains unclear. Previous studies show that in vitro transformation of Madin-Darby canine kidney (MDCK) epithelial cells by the Jaagsiekte sheep retrovirus (JSRV) envelope protein requires activation of the PI3K/Akt oncogenic signaling pathway. Using this cell transformation model, we demonstrated that ectopic expression of wild-type human SAMHD1 or a dNTPase-defective SAMHD1 mutant (HD/AA) significantly inhibited MDCK cell transformation, but did not affect cell proliferation. To visualize and quantify THP-1 cell growth and metastasis in xenografted immunodeficient mice, we generated luciferase-expressing stable SAMHD1 KO THP-1 cells and control THP-1 cells, which were injected intravenously into immunodeficient mice. Bioluminescence imaging and quantification analysis of xenografted mice revealed that SAMHD1 KO cell-derived tumors had similar growth and metastatic potential compared with control cells at 35 days post-injection. However, mice xenografted with SAMHD1 KO cells showed greater survival compared with mice injected with control cells. Our data suggest that exogenous SAMHD1 expression suppresses in vitro cell transformation independently of its dNTPase activity, and that endogenous SAMHD1 affects AML tumorigenicity and disease progression in vivo.


Subject(s)
SAM Domain and HD Domain-Containing Protein 1/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Disease Progression , Dogs , G1 Phase Cell Cycle Checkpoints , Humans , Kaplan-Meier Estimate , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/pathology , Madin Darby Canine Kidney Cells , Mice , Mutagenesis , SAM Domain and HD Domain-Containing Protein 1/deficiency , SAM Domain and HD Domain-Containing Protein 1/genetics , Transplantation, Heterologous
4.
Mol Immunol ; 101: 450-460, 2018 09.
Article in English | MEDLINE | ID: mdl-30099227

ABSTRACT

Germline mutations in the human SAMHD1 gene cause the development of Aicardi-Goutières Syndrome (AGS), with a dominant feature being increased systemic type I interferon(IFN) production. Here we tested the state of type I IFN induction and response to, in SAMHD1 knockout (KO) human monocytic cells. SAMHD1 KO cells exhibited spontaneous transcription and translation of IFN-ß and subsequent interferon-stimulated genes (ISGs) as compared to parental wild-type cells. This elevation of IFN-ß and ISGs was abrogated via inhibition of the TBK1-IRF3 pathway in the SAMHD1 KO cells. In agreement, we found that SAMHD1 KO cells present high levels of phosphorylated TBK1 when compared to control cells. Moreover, addition of blocking antibody against type I IFN also reversed elevation of ISGs. These experiments suggested that SAMHD1 KO cells are persistently auto-stimulating the TBK1-IRF3 pathway, leading to an enhanced production of type I IFN and subsequent self-induction of ISGs.


Subject(s)
Interferon Type I/metabolism , Monocytes/metabolism , SAM Domain and HD Domain-Containing Protein 1/deficiency , Gene Expression Regulation/drug effects , Humans , Monocytes/drug effects , Protein Biosynthesis/drug effects , Pyrimidines/pharmacology , SAM Domain and HD Domain-Containing Protein 1/metabolism , Small Molecule Libraries/pharmacology , THP-1 Cells , Thiophenes/pharmacology , Transcription, Genetic/drug effects
5.
Nature ; 557(7703): 57-61, 2018 05.
Article in English | MEDLINE | ID: mdl-29670289

ABSTRACT

SAMHD1 was previously characterized as a dNTPase that protects cells from viral infections. Mutations in SAMHD1 are implicated in cancer development and in a severe congenital inflammatory disease known as Aicardi-Goutières syndrome. The mechanism by which SAMHD1 protects against cancer and chronic inflammation is unknown. Here we show that SAMHD1 promotes degradation of nascent DNA at stalled replication forks in human cell lines by stimulating the exonuclease activity of MRE11. This function activates the ATR-CHK1 checkpoint and allows the forks to restart replication. In SAMHD1-depleted cells, single-stranded DNA fragments are released from stalled forks and accumulate in the cytosol, where they activate the cGAS-STING pathway to induce expression of pro-inflammatory type I interferons. SAMHD1 is thus an important player in the replication stress response, which prevents chronic inflammation by limiting the release of single-stranded DNA from stalled replication forks.


Subject(s)
DNA Replication , Interferon Type I/metabolism , SAM Domain and HD Domain-Containing Protein 1/metabolism , Checkpoint Kinase 1/metabolism , Cytosol/metabolism , DNA, Single-Stranded/metabolism , HEK293 Cells , HeLa Cells , Humans , Inflammation/immunology , Inflammation/metabolism , Inflammation/prevention & control , Interferon Type I/immunology , MRE11 Homologue Protein/metabolism , Membrane Proteins/metabolism , Nucleotidyltransferases/metabolism , RecQ Helicases/metabolism , SAM Domain and HD Domain-Containing Protein 1/deficiency
6.
Sci Rep ; 8(1): 84, 2018 01 08.
Article in English | MEDLINE | ID: mdl-29311560

ABSTRACT

The autoimmune disorder Aicardi-Goutières syndrome (AGS) is characterized by a constitutive type I interferon response. SAMHD1 possesses both dNTPase and RNase activities and mutations in SAMHD1 cause AGS; however, how SAMHD1-deficiency causes the type I interferon response in patients with AGS remains unknown. Here, we show that endogenous RNA substrates accumulated in the absence of SAMHD1 act as a major immunogenic source for the type I interferon response. Reconstitution of SAMHD1-negative human cells with wild-type but not RNase-defective SAMHD1 abolishes spontaneous type I interferon induction. We further identify that the PI3K/AKT/IRF3 signaling pathway is essential for the type I interferon response in SAMHD1-deficient human monocytic cells. Treatment of PI3K or AKT inhibitors dramatically reduces the type I interferon signatures in SAMHD1-deficient cells. Moreover, SAMHD1/AKT1 double knockout relieves the type I interferon signatures to the levels observed for wild-type cells. Identification of AGS-related RNA sensing pathway provides critical insights into the molecular pathogenesis of the type I interferonopathies such as AGS and overlapping autoimmune disorders.


Subject(s)
Genetic Association Studies , Interferon Type I/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , SAM Domain and HD Domain-Containing Protein 1/deficiency , Signal Transduction , Animals , Cell Line , Humans , Interferon Regulatory Factor-3/metabolism , Mice , Monocytes/metabolism , Mutation , RNA/genetics , RNA/metabolism , Receptor, Interferon alpha-beta/metabolism , SAM Domain and HD Domain-Containing Protein 1/genetics , SAM Domain and HD Domain-Containing Protein 1/metabolism
7.
Cell Rep ; 20(8): 1921-1935, 2017 Aug 22.
Article in English | MEDLINE | ID: mdl-28834754

ABSTRACT

DNA double-strand break (DSB) repair by homologous recombination (HR) is initiated by CtIP/MRN-mediated DNA end resection to maintain genome integrity. SAMHD1 is a dNTP triphosphohydrolase, which restricts HIV-1 infection, and mutations are associated with Aicardi-Goutières syndrome and cancer. We show that SAMHD1 has a dNTPase-independent function in promoting DNA end resection to facilitate DSB repair by HR. SAMHD1 deficiency or Vpx-mediated degradation causes hypersensitivity to DSB-inducing agents, and SAMHD1 is recruited to DSBs. SAMHD1 complexes with CtIP via a conserved C-terminal domain and recruits CtIP to DSBs to facilitate end resection and HR. Significantly, a cancer-associated mutant with impaired CtIP interaction, but not dNTPase-inactive SAMHD1, fails to rescue the end resection impairment of SAMHD1 depletion. Our findings define a dNTPase-independent function for SAMHD1 in HR-mediated DSB repair by facilitating CtIP accrual to promote DNA end resection, providing insight into how SAMHD1 promotes genome integrity.


Subject(s)
DNA End-Joining Repair , Homologous Recombination , SAM Domain and HD Domain-Containing Protein 1/genetics , DNA Breaks, Double-Stranded , HCT116 Cells , HEK293 Cells , HeLa Cells , Humans , MCF-7 Cells , SAM Domain and HD Domain-Containing Protein 1/deficiency , SAM Domain and HD Domain-Containing Protein 1/metabolism , Transfection
8.
BMC Genomics ; 18(1): 53, 2017 01 07.
Article in English | MEDLINE | ID: mdl-28061811

ABSTRACT

BACKGROUND: Single-cell RNA-Seq can be a valuable and unbiased tool to dissect cellular heterogeneity, despite the transcriptome's limitations in describing higher functional phenotypes and protein events. Perhaps the most important shortfall with transcriptomic 'snapshots' of cell populations is that they risk being descriptive, only cataloging heterogeneity at one point in time, and without microenvironmental context. Studying the genetic ('nature') and environmental ('nurture') modifiers of heterogeneity, and how cell population dynamics unfold over time in response to these modifiers is key when studying highly plastic cells such as macrophages. RESULTS: We introduce the programmable Polaris™ microfluidic lab-on-chip for single-cell sequencing, which performs live-cell imaging while controlling for the culture microenvironment of each cell. Using gene-edited macrophages we demonstrate how previously unappreciated knockout effects of SAMHD1, such as an altered oxidative stress response, have a large paracrine signaling component. Furthermore, we demonstrate single-cell pathway enrichments for cell cycle arrest and APOBEC3G degradation, both associated with the oxidative stress response and altered proteostasis. Interestingly, SAMHD1 and APOBEC3G are both HIV-1 inhibitors ('restriction factors'), with no known co-regulation. CONCLUSION: As single-cell methods continue to mature, so will the ability to move beyond simple 'snapshots' of cell populations towards studying the determinants of population dynamics. By combining single-cell culture, live-cell imaging, and single-cell sequencing, we have demonstrated the ability to study cell phenotypes and microenvironmental influences. It's these microenvironmental components - ignored by standard single-cell workflows - that likely determine how macrophages, for example, react to inflammation and form treatment resistant HIV reservoirs.


Subject(s)
Gene-Environment Interaction , Macrophages/cytology , Sequence Analysis, RNA , Single-Cell Analysis , Gene Knockout Techniques , Humans , Macrophages/metabolism , Phenotype , SAM Domain and HD Domain-Containing Protein 1/deficiency , SAM Domain and HD Domain-Containing Protein 1/genetics
9.
Cell Rep ; 16(6): 1492-1501, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27477283

ABSTRACT

SAMHD1 is a restriction factor for HIV-1 infection. SAMHD1 mutations cause the autoinflammatory Aicardi-Goutières syndrome that is characterized by chronic type I interferon (IFN) secretion. We show that the spontaneous IFN response in SAMHD1-deficient cells and mice requires the cGAS/STING cytosolic DNA-sensing pathway. We provide genetic evidence that cell-autonomous control of lentivirus infection in myeloid cells by SAMHD1 limits virus-induced production of IFNs and the induction of co-stimulatory markers. This program of myeloid cell activation required reverse transcription, cGAS and STING, and signaling through the IFN receptor. Furthermore, SAMHD1 reduced the induction of virus-specific cytotoxic T cells in vivo. Therefore, virus restriction by SAMHD1 limits the magnitude of IFN and T cell responses. This demonstrates a competition between cell-autonomous virus control and subsequent innate and adaptive immune responses, a concept with important implications for the treatment of infection.


Subject(s)
Adaptive Immunity/immunology , HIV-1/immunology , Immunity, Innate/immunology , Membrane Proteins/metabolism , Nucleotidyltransferases/metabolism , SAM Domain and HD Domain-Containing Protein 1/metabolism , Animals , Dendritic Cells/immunology , HIV Infections/metabolism , HIV-1/genetics , Interferon-beta/metabolism , Mice , Mice, Inbred C57BL , SAM Domain and HD Domain-Containing Protein 1/deficiency , Virus Replication/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...