151
|
Zadrag-Tecza R, Skoneczna A. Reproductive potential and instability of the rDNA region of the Saccharomyces cerevisiae yeast: Common or separate mechanisms of regulation? Exp Gerontol 2016; 84:29-39. [DOI: 10.1016/j.exger.2016.08.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 08/17/2016] [Indexed: 12/15/2022]
|
152
|
Wang Z, Xu Z, Zhu G. A Platinum(IV) Anticancer Prodrug Targeting Nucleotide Excision Repair To Overcome Cisplatin Resistance. Angew Chem Int Ed Engl 2016; 55:15564-15568. [PMID: 27736029 DOI: 10.1002/anie.201608936] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Indexed: 11/11/2022]
Abstract
DNA damage response plays a key role not only in maintaining genome integrity but also in mediating the antitumor efficacy of DNA-damaging antineoplastic drugs. Herein, we report the rational design and evaluation of a PtIV anticancer prodrug inhibiting nucleotide excision repair (NER), one of the most pivotal processes after the formation of cisplatin-induced DNA damage that deactivates the drug and leads to drug resistance in the clinic. This dual-action prodrug enters cells efficiently and causes DNA damage while simultaneously inhibiting NER to promote apoptotic response. The prodrug is strongly active against the proliferation of cisplatin-resistant human cancer cells with an up to 88-fold increase in growth inhibition compared with cisplatin, and the prodrug is much more active than a mixture of cisplatin and an NER inhibitor. Our study highlights the importance of targeting downstream pathways after the formation of Pt-induced DNA damage as a novel strategy to conquer cisplatin resistance.
Collapse
Affiliation(s)
- Zhigang Wang
- Department of Biology and Chemistry, City University of Hong Kong, 83 Tat Chee Ave, Kowloon Tong, Hong Kong SAR, P.R. China.,City University of Hong Kong, Shenzhen Research Institute, Shenzhen, P.R. China
| | - Zoufeng Xu
- Department of Biology and Chemistry, City University of Hong Kong, 83 Tat Chee Ave, Kowloon Tong, Hong Kong SAR, P.R. China.,City University of Hong Kong, Shenzhen Research Institute, Shenzhen, P.R. China
| | - Guangyu Zhu
- Department of Biology and Chemistry, City University of Hong Kong, 83 Tat Chee Ave, Kowloon Tong, Hong Kong SAR, P.R. China.,City University of Hong Kong, Shenzhen Research Institute, Shenzhen, P.R. China
| |
Collapse
|
153
|
Wang Z, Xu Z, Zhu G. A Platinum(IV) Anticancer Prodrug Targeting Nucleotide Excision Repair To Overcome Cisplatin Resistance. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201608936] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Zhigang Wang
- Department of Biology and Chemistry; City University of Hong Kong; 83 Tat Chee Ave Kowloon Tong Hong Kong SAR P.R. China
- City University of Hong Kong, Shenzhen Research Institute; Shenzhen P.R. China
| | - Zoufeng Xu
- Department of Biology and Chemistry; City University of Hong Kong; 83 Tat Chee Ave Kowloon Tong Hong Kong SAR P.R. China
- City University of Hong Kong, Shenzhen Research Institute; Shenzhen P.R. China
| | - Guangyu Zhu
- Department of Biology and Chemistry; City University of Hong Kong; 83 Tat Chee Ave Kowloon Tong Hong Kong SAR P.R. China
- City University of Hong Kong, Shenzhen Research Institute; Shenzhen P.R. China
| |
Collapse
|
154
|
Analysis of Repair Mechanisms following an Induced Double-Strand Break Uncovers Recessive Deleterious Alleles in the Candida albicans Diploid Genome. mBio 2016; 7:mBio.01109-16. [PMID: 27729506 PMCID: PMC5061868 DOI: 10.1128/mbio.01109-16] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The diploid genome of the yeast Candida albicans is highly plastic, exhibiting frequent loss-of-heterozygosity (LOH) events. To provide a deeper understanding of the mechanisms leading to LOH, we investigated the repair of a unique DNA double-strand break (DSB) in the laboratory C. albicans SC5314 strain using the I-SceI meganuclease. Upon I-SceI induction, we detected a strong increase in the frequency of LOH events at an I-SceI target locus positioned on chromosome 4 (Chr4), including events spreading from this locus to the proximal telomere. Characterization of the repair events by single nucleotide polymorphism (SNP) typing and whole-genome sequencing revealed a predominance of gene conversions, but we also observed mitotic crossover or break-induced replication events, as well as combinations of independent events. Importantly, progeny that had undergone homozygosis of part or all of Chr4 haplotype B (Chr4B) were inviable. Mining of genome sequencing data for 155 C. albicans isolates allowed the identification of a recessive lethal allele in the GPI16 gene on Chr4B unique to C. albicans strain SC5314 which is responsible for this inviability. Additional recessive lethal or deleterious alleles were identified in the genomes of strain SC5314 and two clinical isolates. Our results demonstrate that recessive lethal alleles in the genomes of C. albicans isolates prevent the occurrence of specific extended LOH events. While these and other recessive lethal and deleterious alleles are likely to accumulate in C. albicans due to clonal reproduction, their occurrence may in turn promote the maintenance of corresponding nondeleterious alleles and, consequently, heterozygosity in the C. albicans species. IMPORTANCE Recessive lethal alleles impose significant constraints on the biology of diploid organisms. Using a combination of an I-SceI meganuclease-mediated DNA DSB, a fluorescence-activated cell sorter (FACS)-optimized reporter of LOH, and a compendium of 155 genome sequences, we were able to unmask and identify recessive lethal and deleterious alleles in isolates of Candida albicans, a diploid yeast and the major fungal pathogen of humans. Accumulation of recessive deleterious mutations upon clonal reproduction of C. albicans could contribute to the maintenance of heterozygosity despite the high frequency of LOH events in this species.
Collapse
|
155
|
Venkannagari H, Verheugd P, Koivunen J, Haikarainen T, Obaji E, Ashok Y, Narwal M, Pihlajaniemi T, Lüscher B, Lehtiö L. Small-Molecule Chemical Probe Rescues Cells from Mono-ADP-Ribosyltransferase ARTD10/PARP10-Induced Apoptosis and Sensitizes Cancer Cells to DNA Damage. Cell Chem Biol 2016; 23:1251-1260. [DOI: 10.1016/j.chembiol.2016.08.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 07/22/2016] [Accepted: 08/31/2016] [Indexed: 12/21/2022]
|
156
|
Jang S, Zhou X, Ahn J. Substrate Specificity of SAMHD1 Triphosphohydrolase Activity Is Controlled by Deoxyribonucleoside Triphosphates and Phosphorylation at Thr592. Biochemistry 2016; 55:5635-5646. [PMID: 27588835 DOI: 10.1021/acs.biochem.6b00627] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The sterile alpha motif (SAM) and histidine-aspartate (HD) domain containing protein 1 (SAMHD1) constitute a triphosphohydrolase that converts deoxyribonucleoside triphosphates (dNTPs) into deoxyribonucleosides and triphosphates. SAMHD1 exists in multiple states. The monomer and apo- or GTP-bound dimer are catalytically inactive. Binding of dNTP at allosteric site 2 (AS2), adjacent to GTP-binding allosteric site 1 (AS1), induces formation of the tetramer, the catalytically active form. We have developed an enzyme kinetic assay, tailored to control specific dNTP binding at each site, allowing us to determine the kinetic binding parameters of individual dNTPs at both the AS2 and catalytic sites for all possible combinations of dNTP binding at both sites. Here, we show that the apparent Km values of dNTPs at AS2 vary in the order of dCTP < dGTP < dATP < dTTP. Interestingly, dCTP binding at AS2 significantly reduces the dCTP hydrolysis rate, which is restored to a rate comparable to that of other dNTPs upon dGTP, dATP, or dTTP binding at AS2. Strikingly, a phosphomimetic mutant, Thr592Asp SAMHD1 as well as phospho-Thr592, show a significantly altered substrate specificity, with the rate of dCTP hydrolysis being selectively reduced regardless of which dNTP binds at AS2. Furthermore, cyclin A2 binding at the C-terminus of SAMHD1 induces the disassembly of the SAMHD1 tetramer, suggesting an additional layer of SAMHD1 activity modulation by cyclin A2/CDK2 kinase. Together, our results reveal multiple allosteric mechanisms for controlling the rate of dNTP destruction by SAMHD1.
Collapse
Affiliation(s)
- Sunbok Jang
- Department of Structural Biology, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania 15260, United States
| | - Xiaohong Zhou
- Department of Structural Biology, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania 15260, United States
| | - Jinwoo Ahn
- Department of Structural Biology, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania 15260, United States
| |
Collapse
|
157
|
Suzuki T, Grúz P, Honma M, Adachi N, Nohmi T. The role of DNA polymerase ζ in translesion synthesis across bulky DNA adducts and cross-links in human cells. Mutat Res 2016; 791-792:35-41. [PMID: 27591392 DOI: 10.1016/j.mrfmmm.2016.08.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 08/17/2016] [Accepted: 08/24/2016] [Indexed: 06/06/2023]
Abstract
Translesion DNA synthesis (TLS) is a cellular defense mechanism against genotoxins. Defects or mutations in specialized DNA polymerases (Pols) involved in TLS are believed to result in hypersensitivity to various genotoxic stresses. Here, DNA polymerase ζ (Pol ζ)-deficient (KO: knockout) and Pol ζ catalytically dead (CD) human cells were established and their sensitivity towards cytotoxic activities of various genotoxins was examined. The CD cells were engineered by altering the DNA sequence encoding two amino acids essential for the catalytic activity of Pol ζ, i.e., D2781 and D2783, to alanines. Both Pol ζ KO and CD cells displayed a prolonged cell cycle and higher incidence of micronuclei formation than the wild-type (WT) cells in the absence of exogenous genotoxic treatments, and the order of abnormality was CD>KO>WT cells. Both KO and CD cells exhibited higher sensitivity towards the killing effects of benzo[a]pyrene diol epoxide, mitomycin C, potassium bromate, N-methyl-N'-nitro-N-nitrosoguanidine, and ultraviolet C irradiation than WT cells, and there were no differences between the sensitivities of KO and CD cells. Interestingly, neither KO nor CD cells were sensitive to the cytotoxic effects of hydrogen peroxide. Since KO and CD cells displayed similar sensitivities to the genotoxins, we employed only KO cells to further examine their sensitivity to other genotoxic agents. KO cells were more sensitive to the cytotoxicity of 4-nitroquinoline N-oxide, styrene oxide, cisplatin, methyl methanesulfonate, and ethyl methanesulfonate than WT cells. However, the KO cells displayed sensitivity camptothecin, etoposide, bleomycin, hydroxyurea, crotonealdehyde, and methylglyoxal in a manner similar to the WT cells. Our results suggest that Pol ζ plays an important role in the protection of human cells by carrying out TLS across bulky DNA adducts and cross-links, but has no or limited role in the protection against strand-breaks in DNA.
Collapse
Affiliation(s)
- Tetsuya Suzuki
- Division of Genetics and Mutagenesis, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501, Japan.
| | - Petr Grúz
- Division of Genetics and Mutagenesis, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501, Japan
| | - Masamitsu Honma
- Division of Genetics and Mutagenesis, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501, Japan
| | - Noritaka Adachi
- Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama 236-0027, Japan
| | - Takehiko Nohmi
- Division of Genetics and Mutagenesis, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501, Japan
| |
Collapse
|
158
|
Millet A, Martin AR, Ronco C, Rocchi S, Benhida R. Metastatic Melanoma: Insights Into the Evolution of the Treatments and Future Challenges. Med Res Rev 2016; 37:98-148. [PMID: 27569556 DOI: 10.1002/med.21404] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 06/28/2016] [Accepted: 07/06/2016] [Indexed: 02/06/2023]
Abstract
Melanoma is the deadliest form of skin cancer. While associated survival prognosis is good when diagnosed early, it dramatically drops when melanoma progresses into its metastatic form. Prior to 2011, the favored therapies include interleukin-2 and chemotherapies, regardless of their low efficiency and their toxicity. Following key biological findings, two new types of therapy have been approved. First, there are the targeted therapies, which rely on small molecule B-Raf and MEK inhibitors and allow the treatment of patients with B-Raf mutated melanoma. Second, there are the immunotherapies, with anti-CTLA-4 and anti-PD-1 antibodies that are used for patients harboring a B-Raf wild-type status. Both approaches have significantly improved patient survival, compared with alkylating agents, in the treatment of unresectable melanoma. Herein, we review the evolution of the treatment of melanoma starting from early discoveries to current therapies. A focus will be provided on drug discovery, synthesis, and mode of action of relevant drugs and the future directions of the domain to overcome the emergence of the resistance events.
Collapse
Affiliation(s)
- Antoine Millet
- Institut de Chimie de Nice UMR UNS-CNRS 7272, Nice, France
| | | | - Cyril Ronco
- Institut de Chimie de Nice UMR UNS-CNRS 7272, Nice, France
| | - Stéphane Rocchi
- INSERM, U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Equipe Biologie et Pathologie des cellules mélanocytaires: de la pigmentation cutanée au mélanome, Nice, France.,Université de Nice Sophia Antipolis, UFR de Médecine, Nice, France.,Service de Dermatologie, Hôpital Archet II, CHU Nice, France
| | - Rachid Benhida
- Institut de Chimie de Nice UMR UNS-CNRS 7272, Nice, France
| |
Collapse
|
159
|
Miles S, Breeden L. A common strategy for initiating the transition from proliferation to quiescence. Curr Genet 2016; 63:179-186. [PMID: 27544284 DOI: 10.1007/s00294-016-0640-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 08/04/2016] [Accepted: 08/06/2016] [Indexed: 10/21/2022]
Abstract
Development, tissue renewal and long term survival of multi-cellular organisms is dependent upon the persistence of stem cells that are quiescent, but retain the capacity to re-enter the cell cycle to self-renew, or to produce progeny that can differentiate and re-populate the tissue. Deregulated release of these cells from the quiescent state, or preventing them from entering quiescence, results in uncontrolled proliferation and cancer. Conversely, loss of quiescent cells, or their failure to re-enter cell division, disrupts organ development and prevents tissue regeneration and repair. Understanding the quiescent state and how cells control the transitions in and out of this state is of fundamental importance. Investigations into the mechanics of G1 arrest during the transition to quiescence continue to identify striking parallels between the strategies used by yeast and mammals to regulate this transition. When cells commit to a stable but reversible arrest, the G1/S genes responsible for promoting S phase must be inhibited. This process, from yeast to humans, involves the formation of quiescence-specific complexes on their promoters. In higher cells, these so-called DREAM complexes of E2F4/DP/RBL/MuvB recruit the highly conserved histone deacetylase HDAC1, which leads to local histone deacetylation and repression of S phase-promoting transcripts. Quiescent yeast cells also show pervasive histone deacetylation by the HDAC1 counterpart Rpd3. In addition, these cells contain quiescence-specific regulators of G1/S genes: Msa1 and Msa2, which can be considered components of the yeast equivalent of the DREAM complex. Despite a lack of physical similarities, the goals and the strategies used to achieve a reversible transition to quiescence are highly conserved. This motivates a detailed study of this process in the simple model organism: budding yeast.
Collapse
Affiliation(s)
- Shawna Miles
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA, 98109, USA
| | - Linda Breeden
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA, 98109, USA.
| |
Collapse
|
160
|
Gao Z, Putnam AA, Bowers HA, Guenther UP, Ye X, Kindsfather A, Hilliker AK, Jankowsky E. Coupling between the DEAD-box RNA helicases Ded1p and eIF4A. eLife 2016; 5. [PMID: 27494274 PMCID: PMC4990422 DOI: 10.7554/elife.16408] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Accepted: 08/04/2016] [Indexed: 11/13/2022] Open
Abstract
Eukaryotic translation initiation involves two conserved DEAD-box RNA helicases, eIF4A and Ded1p. Here we show that S. cerevisiae eIF4A and Ded1p directly interact with each other and simultaneously with the scaffolding protein eIF4G. We delineate a comprehensive thermodynamic framework for the interactions between Ded1p, eIF4A, eIF4G, RNA and ATP, which indicates that eIF4A, with and without eIF4G, acts as a modulator for activity and substrate preferences of Ded1p, which is the RNA remodeling unit in all complexes. Our results reveal and characterize an unexpected interdependence between the two RNA helicases and eIF4G, and suggest that Ded1p is an integral part of eIF4F, the complex comprising eIF4G, eIF4A, and eIF4E.
Collapse
Affiliation(s)
- Zhaofeng Gao
- Center for RNA Molecular Biology, Case Western Reserve University, Cleveland, United States.,Department of Biochemistry, Case Western Reserve University, Cleveland, United States.,School of Medicine, Case Western Reserve University, Cleveland, United States
| | - Andrea A Putnam
- Center for RNA Molecular Biology, Case Western Reserve University, Cleveland, United States.,Department of Biochemistry, Case Western Reserve University, Cleveland, United States.,School of Medicine, Case Western Reserve University, Cleveland, United States
| | - Heath A Bowers
- Center for RNA Molecular Biology, Case Western Reserve University, Cleveland, United States.,Department of Biochemistry, Case Western Reserve University, Cleveland, United States.,School of Medicine, Case Western Reserve University, Cleveland, United States
| | - Ulf-Peter Guenther
- Center for RNA Molecular Biology, Case Western Reserve University, Cleveland, United States.,Department of Biochemistry, Case Western Reserve University, Cleveland, United States.,School of Medicine, Case Western Reserve University, Cleveland, United States
| | - Xuan Ye
- Center for RNA Molecular Biology, Case Western Reserve University, Cleveland, United States.,Department of Biochemistry, Case Western Reserve University, Cleveland, United States.,School of Medicine, Case Western Reserve University, Cleveland, United States
| | | | - Angela K Hilliker
- Department of Biology, University of Richmond, Richmond, United States
| | - Eckhard Jankowsky
- Center for RNA Molecular Biology, Case Western Reserve University, Cleveland, United States.,Department of Biochemistry, Case Western Reserve University, Cleveland, United States.,School of Medicine, Case Western Reserve University, Cleveland, United States
| |
Collapse
|
161
|
Prakash C, Manjrekar J, Chattoo BB. Skp1, a component of E3 ubiquitin ligase, is necessary for growth, sporulation, development and pathogenicity in rice blast fungus (Magnaporthe oryzae). MOLECULAR PLANT PATHOLOGY 2016; 17:903-919. [PMID: 26575697 PMCID: PMC6638394 DOI: 10.1111/mpp.12336] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 10/15/2015] [Accepted: 10/15/2015] [Indexed: 05/31/2023]
Abstract
Ubiqitination is an important process in eukaryotic cells involving E3 ubiquitin ligase, which co-ordinates with cell cycle proteins and controls various cell functions. Skp1 (S-phase kinase-associated protein 1) is a core component of the SCF (Skp1-Cullin 1-F-box) E3 ubiquitin ligase complex necessary for protein degradation by the 26S proteasomal pathway. The rice blast fungus Magnaporthe oryzae has a single MoSKP1(MGG_04978) required for viability. Skp1 has multiple functions; however, its roles in growth, sporulation and appressorial development are not understood. MoSKP1 complements Skp1 function in the fission yeast temperature-sensitive mutant skp1 A7, restoring the normal length of yeast cells at restrictive temperature. The MoSkp1 protein in M. oryzae is present in spores and germ tubes, and is abundantly expressed in appressoria. Various RNA interference (RNAi) and antisense transformants of MoSKP1 in B157 show reduced sporulation, defective spore morphology, lesser septation and diffuse nuclei. Further, they show elongated germ tubes and are unable to form appressoria. Transformants arrested in G1/S stage during initial spore germination show a similar phenotype to wild-type spores treated with hydroxyurea (HU). Reduced MoSkp1 transcript and protein levels in knockdown transformants result in atypical germ tube development. MoSkp1 interacts with the putative F-box protein (MGG_06351) revealing the ability to form protein complexes. Our investigation of the role of MoSKP1 suggests that a decrease in MoSkp1 manifests in decreased total protein ubiquitination and, consequently, defective cell cycle and appressorial development. Thus, MoSKP1 plays important roles in growth, sporulation, appressorial development and pathogenicity of M. oryzae.
Collapse
Affiliation(s)
- Chandra Prakash
- Department of Microbiology and Biotechnology Centre, The Maharaja Sayajirao University of Baroda, Vadodara, 390 002, Gujarat, India
- Genome Research Centre, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, 390 002, Gujarat, India
| | - Johannes Manjrekar
- Department of Microbiology and Biotechnology Centre, The Maharaja Sayajirao University of Baroda, Vadodara, 390 002, Gujarat, India
| | - Bharat B Chattoo
- Department of Microbiology and Biotechnology Centre, The Maharaja Sayajirao University of Baroda, Vadodara, 390 002, Gujarat, India
- Genome Research Centre, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, 390 002, Gujarat, India
| |
Collapse
|
162
|
Peña JF, Alié A, Richter DJ, Wang L, Funayama N, Nichols SA. Conserved expression of vertebrate microvillar gene homologs in choanocytes of freshwater sponges. EvoDevo 2016; 7:13. [PMID: 27413529 PMCID: PMC4942974 DOI: 10.1186/s13227-016-0050-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Accepted: 06/28/2016] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND The microvillus is a versatile organelle that serves important functions in disparate animal cell types. However, from a molecular perspective, the microvillus has been well studied in only a few, predominantly vertebrate, contexts. Little is known about how differences in microvillar structure contribute to differences in function, and how these differences evolved. We sequenced the transcriptome of the freshwater sponge, Ephydatia muelleri, and examined the expression of vertebrate microvillar gene homologs in choanocytes-the only microvilli-bearing cell type present in sponges. Sponges offer a distant phylogenetic comparison with vertebrates, and choanocytes are central to discussions about early animal evolution due to their similarity with choanoflagellates, the single-celled sister lineage of modern animals. RESULTS We found that, from a genomic perspective, sponges have conserved homologs of most vertebrate microvillar genes, most of which are expressed in choanocytes, and many of which exhibit choanocyte-specific or choanocyte-enriched expression. Possible exceptions include the cadherins that form intermicrovillar links in the enterocyte brush border and hair cell stereocilia of vertebrates and cnidarians. No obvious orthologs of these proteins were detected in sponges, but at least four candidate cadherins were identified as choanocyte-enriched and might serve this function. In contrast to the evidence for conserved microvillar structure in sponges and vertebrates, we found that choanoflagellates and ctenophores lack homologs of many fundamental microvillar genes, suggesting that microvillar structure may diverge significantly in these lineages, warranting further study. CONCLUSIONS The available evidence suggests that microvilli evolved early in the prehistory of modern animals and have been repurposed to serve myriad functions in different cellular contexts. Detailed understanding of the sequence by which different microvilli-bearing cell/tissue types diversified will require further study of microvillar composition and development in disparate cell types and lineages. Of particular interest are the microvilli of choanoflagellates, ctenophores, and sponges, which collectively bracket the earliest events in animal evolution.
Collapse
Affiliation(s)
- Jesús F. Peña
- />Department of Biological Sciences, University of Denver, F.W. Olin Hall, Room 102, 2190 E. Iliff Ave., Denver, CO 80208 USA
| | - Alexandre Alié
- />Laboratoire de Biologie du Développement de Villefranche-sur-mer, CNRS, Sorbonne Universités, UPMC Univ Paris 06, Observatoire Océanographique, 06230 Villefranche-sur-mer, France
- />Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto, 606-8502 Japan
| | - Daniel J. Richter
- />Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720-3200 USA
- />UMR 7144, CNRS and Sorbonne Universités Université Pierre et Marie Curie (UPMC) Paris 06, Station Biologique de Roscoff, Place Georges Teissier, 29680 Roscoff, France
| | - Lingyu Wang
- />Department of Biology, University of Miami, 208 Cox Science Center, 1301 Memorial Drive, Coral Gables, FL 33124 USA
| | - Noriko Funayama
- />Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto, 606-8502 Japan
| | - Scott A. Nichols
- />Department of Biological Sciences, University of Denver, F.W. Olin Hall, Room 102, 2190 E. Iliff Ave., Denver, CO 80208 USA
| |
Collapse
|
163
|
Huang ME, Facca C, Fatmi Z, Baïlle D, Bénakli S, Vernis L. DNA replication inhibitor hydroxyurea alters Fe-S centers by producing reactive oxygen species in vivo. Sci Rep 2016; 6:29361. [PMID: 27405729 PMCID: PMC4942693 DOI: 10.1038/srep29361] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 06/17/2016] [Indexed: 01/05/2023] Open
Abstract
Redox homeostasis is tightly controlled in cells as it is critical for most cellular functions. Iron-Sulfur centers (Fe-S) are metallic cofactors with electronic properties that are associated with proteins and allow fine redox tuning. Following the observation that altered Fe-S biosynthesis is correlated with a high sensitivity to hydroxyurea (HU), a potent DNA replication blocking agent, we identified that oxidative stress response pathway under the control of the main regulator Yap1 attenuates HU deleterious effects, as it significantly increases resistance to HU, Fe-S biosynthesis and DNA replication kinetics in the presence of HU. Yap1 effect is mediated at least in part through up-regulation of two highly conserved genes controlling cytosolic Fe-S biosynthesis and oxidative stress, Dre2 and Tah18. We next observed that HU produces deleterious effects on cytosolic Fe-S clusters in proteins in vivo but not in vitro, suggesting that HU’s impact on Fe-S in vivo is mediated by cellular metabolism. Finally, we evidenced that HU exposure was accompanied by production of reactive oxygen species intracellularly. Altogether, this study provides mechanistic insight on the initial observation that mutants with altered Fe-S biosynthesis are highly sensitive to HU and uncovers a novel mechanism of action of this widely used DNA replication inhibitor.
Collapse
Affiliation(s)
- Meng-Er Huang
- Institut Curie, PSL Research University, CNRS UMR3348, 91405 Orsay, France.,Institut Curie, CNRS UMR2027, 91405 Orsay, France.,Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, 91405 Orsay, France
| | - Céline Facca
- Institut Curie, CNRS UMR2027, 91405 Orsay, France
| | - Zakaria Fatmi
- Institut Curie, PSL Research University, CNRS UMR3348, 91405 Orsay, France.,Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, 91405 Orsay, France
| | - Dorothée Baïlle
- Institut Curie, PSL Research University, CNRS UMR3348, 91405 Orsay, France.,Institut Curie, CNRS UMR2027, 91405 Orsay, France.,Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, 91405 Orsay, France
| | | | - Laurence Vernis
- Institut Curie, PSL Research University, CNRS UMR3348, 91405 Orsay, France.,Institut Curie, CNRS UMR2027, 91405 Orsay, France.,Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, 91405 Orsay, France
| |
Collapse
|
164
|
Rybaczek D. Hydroxyurea-induced replication stress causes poly(ADP-ribose) polymerase-2 accumulation and changes its intranuclear location in root meristems of Vicia faba. JOURNAL OF PLANT PHYSIOLOGY 2016; 198:89-102. [PMID: 27155387 DOI: 10.1016/j.jplph.2016.03.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 03/29/2016] [Accepted: 03/30/2016] [Indexed: 06/05/2023]
Abstract
Replication stress induced by 24 and 48h exposure to 2.5mM hydroxyurea (HU) increased the activity of poly(ADP-ribose) polymerase-2 (PARP-2; EC 2.4.2.30) in root meristem cells of Vicia faba. An increase in the number of PARP-2 foci was accompanied by their delocalization from peripheral areas to the interior of the nucleus. Our results indicate that the increase in PARP-2 was connected with an increase in S139-phosphorylated H2AX histones. The findings suggest the possible role of PARP-2 in replication stress. We also confirm that the intranuclear location of PARP-2 depends on the duration of HU-induced replication stress, confirming the role of PARP-2 as an indicator of stress intensity. Finally, we conclude that the more intense the HU-mediated replication stress, the greater the probability of PARP-2 activation or H2AXS139 phosphorylation, but also the greater the chance of increasing the efficiency of repair processes and a return to normal cell cycle progression.
Collapse
Affiliation(s)
- Dorota Rybaczek
- Department of Cytophysiology, Institute of Experimental Biology, Faculty of Biology and Environmental Protection, University of Łódź, Pomorska 141/143, 90236 Łódź, Poland.
| |
Collapse
|
165
|
Sawant AA, Mukherjee PP, Jangid RK, Galande S, Srivatsan SG. A clickable UTP analog for the posttranscriptional chemical labeling and imaging of RNA. Org Biomol Chem 2016; 14:5832-42. [PMID: 27173127 DOI: 10.1039/c6ob00576d] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The development of robust tools and practical RNA labeling strategies that would facilitate the biophysical analysis of RNA in both cell-free and cellular systems will have profound implications in the discovery of new RNA diagnostic tools and therapeutic strategies. In this context, we describe the development of a new alkyne-modified UTP analog, 5-(1,7-octadinyl)uridine triphosphate (ODUTP), which serves as an efficient substrate for the introduction of a clickable alkyne label into RNA transcripts by bacteriophage T7 RNA polymerase and mammalian cellular RNA polymerases. The ODU-labeled RNA is effectively used by reverse transcriptase to produce cDNA, a property which could be utilized in expanding the chemical space of a RNA library in the aptamer selection scheme. Further, the alkyne label on RNA provides a convenient tool for the posttranscriptional chemical functionalization with a variety of biophysical tags (fluorescent, affinity, amino acid and sugar) by using alkyne-azide cycloaddition reaction. Importantly, the ability of endogenous RNA polymerases to specifically incorporate ODUTP into cellular RNA transcripts enabled the visualization of newly transcribing RNA in cells by microscopy using click reactions. In addition to a clickable alkyne group, ODU contains a Raman scattering label (internal disubstituted alkyne), which exhibits characteristic Raman shifts that fall in the Raman-silent region of cells. Our results indicate that an ODU label could potentially facilitate two-channel visualization of RNA in cells by using click chemistry and Raman spectroscopy. Taken together, ODU represents a multipurpose ribonucleoside tool, which is expected to provide new avenues to study RNA in cell-free and cellular systems.
Collapse
Affiliation(s)
- Anupam A Sawant
- Department of Chemistry, Indian Institute of Science Education and Research, Pune, Dr Homi Bhabha Road, Pashan, Pune 411008, India.
| | | | | | | | | |
Collapse
|
166
|
Bru S, Martínez-Laínez JM, Hernández-Ortega S, Quandt E, Torres-Torronteras J, Martí R, Canadell D, Ariño J, Sharma S, Jiménez J, Clotet J. Polyphosphate is involved in cell cycle progression and genomic stability in Saccharomyces cerevisiae. Mol Microbiol 2016; 101:367-80. [PMID: 27072996 DOI: 10.1111/mmi.13396] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 04/04/2016] [Accepted: 04/09/2016] [Indexed: 11/27/2022]
Abstract
Polyphosphate (polyP) is a linear chain of up to hundreds of inorganic phosphate residues that is necessary for many physiological functions in all living organisms. In some bacteria, polyP supplies material to molecules such as DNA, thus playing an important role in biosynthetic processes in prokaryotes. In the present study, we set out to gain further insight into the role of polyP in eukaryotic cells. We observed that polyP amounts are cyclically regulated in Saccharomyces cerevisiae, and those mutants that cannot synthesise (vtc4Δ) or hydrolyse polyP (ppn1Δ, ppx1Δ) present impaired cell cycle progression. Further analysis revealed that polyP mutants show delayed nucleotide production and increased genomic instability. Based on these findings, we concluded that polyP not only maintains intracellular phosphate concentrations in response to fluctuations in extracellular phosphate levels, but also muffles internal cyclic phosphate fluctuations, such as those produced by the sudden demand of phosphate to synthetize deoxynucleotides just before and during DNA duplication. We propose that the presence of polyP in eukaryotic cells is required for the timely and accurate duplication of DNA.
Collapse
Affiliation(s)
- Samuel Bru
- Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, Barcelona, Spain
| | | | - Sara Hernández-Ortega
- Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Eva Quandt
- Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Javier Torres-Torronteras
- Research Group on Neuromuscular and Mitochondrial Disorders, Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain.,Biomedical Network Research Centre on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Ramón Martí
- Research Group on Neuromuscular and Mitochondrial Disorders, Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain.,Biomedical Network Research Centre on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - David Canadell
- Department of Biochemistry and Molecular Biology and the Institute of Biotechnology and Biomedicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Joaquin Ariño
- Department of Biochemistry and Molecular Biology and the Institute of Biotechnology and Biomedicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Sushma Sharma
- Department of Medical Biochemistry and Biophysics, Umeå University, Sweden
| | - Javier Jiménez
- Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Josep Clotet
- Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, Barcelona, Spain
| |
Collapse
|
167
|
Mavor D, Barlow K, Thompson S, Barad BA, Bonny AR, Cario CL, Gaskins G, Liu Z, Deming L, Axen SD, Caceres E, Chen W, Cuesta A, Gate RE, Green EM, Hulce KR, Ji W, Kenner LR, Mensa B, Morinishi LS, Moss SM, Mravic M, Muir RK, Niekamp S, Nnadi CI, Palovcak E, Poss EM, Ross TD, Salcedo EC, See SK, Subramaniam M, Wong AW, Li J, Thorn KS, Conchúir SÓ, Roscoe BP, Chow ED, DeRisi JL, Kortemme T, Bolon DN, Fraser JS. Determination of ubiquitin fitness landscapes under different chemical stresses in a classroom setting. eLife 2016; 5. [PMID: 27111525 PMCID: PMC4862753 DOI: 10.7554/elife.15802] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Accepted: 04/06/2016] [Indexed: 12/31/2022] Open
Abstract
Ubiquitin is essential for eukaryotic life and varies in only 3 amino acid positions between yeast and humans. However, recent deep sequencing studies indicate that ubiquitin is highly tolerant to single mutations. We hypothesized that this tolerance would be reduced by chemically induced physiologic perturbations. To test this hypothesis, a class of first year UCSF graduate students employed deep mutational scanning to determine the fitness landscape of all possible single residue mutations in the presence of five different small molecule perturbations. These perturbations uncover 'shared sensitized positions' localized to areas around the hydrophobic patch and the C-terminus. In addition, we identified perturbation specific effects such as a sensitization of His68 in HU and a tolerance to mutation at Lys63 in DTT. Our data show how chemical stresses can reduce buffering effects in the ubiquitin proteasome system. Finally, this study demonstrates the potential of lab-based interdisciplinary graduate curriculum. DOI:http://dx.doi.org/10.7554/eLife.15802.001 The ability of an organism to grow and reproduce, that is, it’s “fitness”, is determined by how its genes interact with the environment. Yeast is a model organism in which researchers can control the exact mutations present in the yeast’s genes (its genotype) and the conditions in which the yeast cells live (their environment). This allows researchers to measure how a yeast cell’s genotype and environment affect its fitness. Ubiquitin is a protein that many organisms depend on to manage cell stress by acting as a tag that targets other proteins for degradation. Essential proteins such as ubiquitin often remain unchanged by mutation over long periods of time. As a result, these proteins evolve very slowly. Like all proteins, ubiquitin is built from a chain of amino acid molecules linked together, and the ubiquitin proteins of yeast and humans are made of almost identical sequences of amino acids. Although ubiquitin has barely changed its sequence over evolution, previous studies have shown that – under normal growth conditions in the laboratory – most amino acids in ubiquitin can be mutated without any loss of cell fitness. This led Mavor et al. to hypothesize that treating the yeast cells with chemicals that cause cell stress might lead to amino acids in ubiquitin becoming more sensitive to mutation. To test this idea, a class of graduate students at the University of California, San Francisco grew yeast cells with different ubiquitin mutations together, and with different chemicals that induce cell stress, and measured their growth rates. Sequencing the ubiquitin gene in the thousands of tested yeast cells revealed that three of the chemicals cause a shared set of amino acids in ubiquitin to become more sensitive to mutation. This result suggests that these amino acids are important for the stress response, possibly by altering the ability of yeast cells to target certain proteins for degradation. Conversely, another chemical causes yeast to become more tolerant to changes in the ubiquitin sequence. The experiments also link changes in particular amino acids in ubiquitin to specific stress responses. Mavor et al. show that many of ubquitin’s amino acids are sensitive to mutation under different stress conditions, while others can be mutated to form different amino acids without effecting fitness. By testing the effects of other chemicals, future experiments could further characterize how the yeast’s genotype and environment interact. DOI:http://dx.doi.org/10.7554/eLife.15802.002
Collapse
Affiliation(s)
- David Mavor
- Biophysics Graduate Group, University of California, San Francisco, San Francisco, United States
| | - Kyle Barlow
- Bioinformatics Graduate Group, University of California, San Francisco, San Francisco, United States
| | - Samuel Thompson
- Biophysics Graduate Group, University of California, San Francisco, San Francisco, United States
| | - Benjamin A Barad
- Biophysics Graduate Group, University of California, San Francisco, San Francisco, United States
| | - Alain R Bonny
- Biophysics Graduate Group, University of California, San Francisco, San Francisco, United States
| | - Clinton L Cario
- Bioinformatics Graduate Group, University of California, San Francisco, San Francisco, United States
| | - Garrett Gaskins
- Bioinformatics Graduate Group, University of California, San Francisco, San Francisco, United States
| | - Zairan Liu
- Biophysics Graduate Group, University of California, San Francisco, San Francisco, United States
| | - Laura Deming
- Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, United States
| | - Seth D Axen
- Bioinformatics Graduate Group, University of California, San Francisco, San Francisco, United States
| | - Elena Caceres
- Bioinformatics Graduate Group, University of California, San Francisco, San Francisco, United States
| | - Weilin Chen
- Bioinformatics Graduate Group, University of California, San Francisco, San Francisco, United States
| | - Adolfo Cuesta
- Chemistry and Chemical Biology Graduate Program, University of California, San Francisco, San Francisco, United States
| | - Rachel E Gate
- Bioinformatics Graduate Group, University of California, San Francisco, San Francisco, United States
| | - Evan M Green
- Biophysics Graduate Group, University of California, San Francisco, San Francisco, United States
| | - Kaitlin R Hulce
- Chemistry and Chemical Biology Graduate Program, University of California, San Francisco, San Francisco, United States
| | - Weiyue Ji
- Biophysics Graduate Group, University of California, San Francisco, San Francisco, United States
| | - Lillian R Kenner
- Biophysics Graduate Group, University of California, San Francisco, San Francisco, United States
| | - Bruk Mensa
- Chemistry and Chemical Biology Graduate Program, University of California, San Francisco, San Francisco, United States
| | - Leanna S Morinishi
- Bioinformatics Graduate Group, University of California, San Francisco, San Francisco, United States
| | - Steven M Moss
- Chemistry and Chemical Biology Graduate Program, University of California, San Francisco, San Francisco, United States
| | - Marco Mravic
- Biophysics Graduate Group, University of California, San Francisco, San Francisco, United States
| | - Ryan K Muir
- Chemistry and Chemical Biology Graduate Program, University of California, San Francisco, San Francisco, United States
| | - Stefan Niekamp
- Biophysics Graduate Group, University of California, San Francisco, San Francisco, United States
| | - Chimno I Nnadi
- Chemistry and Chemical Biology Graduate Program, University of California, San Francisco, San Francisco, United States
| | - Eugene Palovcak
- Biophysics Graduate Group, University of California, San Francisco, San Francisco, United States
| | - Erin M Poss
- Chemistry and Chemical Biology Graduate Program, University of California, San Francisco, San Francisco, United States
| | - Tyler D Ross
- Biophysics Graduate Group, University of California, San Francisco, San Francisco, United States
| | - Eugenia C Salcedo
- Chemistry and Chemical Biology Graduate Program, University of California, San Francisco, San Francisco, United States
| | - Stephanie K See
- Chemistry and Chemical Biology Graduate Program, University of California, San Francisco, San Francisco, United States
| | - Meena Subramaniam
- Bioinformatics Graduate Group, University of California, San Francisco, San Francisco, United States
| | - Allison W Wong
- Chemistry and Chemical Biology Graduate Program, University of California, San Francisco, San Francisco, United States
| | - Jennifer Li
- UCSF Science and Health Education Partnership, University of California, San Francisco, San Francisco, United States
| | - Kurt S Thorn
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States
| | - Shane Ó Conchúir
- Department of Bioengineering and Therapeutic Sciences, California Institute for Quantitative Biology, University of California, San Francisco, San Francisco, United States
| | - Benjamin P Roscoe
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, United States
| | - Eric D Chow
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States.,Center for Advanced Technology, University of California, San Francisco, San Francisco, United States
| | - Joseph L DeRisi
- Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, United States.,Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States
| | - Tanja Kortemme
- Department of Bioengineering and Therapeutic Sciences, California Institute for Quantitative Biology, University of California, San Francisco, San Francisco, United States
| | - Daniel N Bolon
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, United States
| | - James S Fraser
- Department of Bioengineering and Therapeutic Sciences, California Institute for Quantitative Biology, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
168
|
The Protein Arginine Methylase 5 (PRMT5/SKB1) Gene Is Required for the Maintenance of Root Stem Cells in Response to DNA Damage. J Genet Genomics 2016; 43:187-97. [DOI: 10.1016/j.jgg.2016.02.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 02/06/2016] [Accepted: 02/15/2016] [Indexed: 11/23/2022]
|
169
|
A novel transcription factor gene FHS1 is involved in the DNA damage response in Fusarium graminearum. Sci Rep 2016; 6:21572. [PMID: 26888604 PMCID: PMC4757864 DOI: 10.1038/srep21572] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 01/27/2016] [Indexed: 12/13/2022] Open
Abstract
Cell cycle regulation and the maintenance of genome integrity are crucial for the development and virulence of the pathogenic plant fungus Fusarium graminearum. To identify transcription factors (TFs) related to these processes, four DNA-damaging agents were applied to screen a F. graminearum TF mutant library. Sixteen TFs were identified to be likely involved in DNA damage responses. Fhs1 is a fungal specific Zn(II)2Cys6 TF that localises exclusively to nuclei. fhs1 deletion mutants were hypersensitive to hydroxyurea and defective in mitotic cell division. Moreover, deletion of FHS1 resulted in defects in perithecia production and virulence and led to the accumulation of DNA damage. Our genetic evidence demonstrated that the FHS1-associated signalling pathway for DNA damage response is independent of the ATM or ATR pathways. This study identified sixteen genes involved in the DNA damage response and is the first to characterise the novel transcription factor gene FHS1, which is involved in the DNA damage response. The results provide new insights into mechanisms underlying DNA damage responses in fungi, including F. graminearum.
Collapse
|
170
|
Ivanova IG, Maringele L. Polymerases ε and ∂ repair dysfunctional telomeres facilitated by salt. Nucleic Acids Res 2016; 44:3728-38. [PMID: 26883631 PMCID: PMC4856982 DOI: 10.1093/nar/gkw071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 02/01/2016] [Indexed: 12/19/2022] Open
Abstract
Damaged DNA can be repaired by removal and re-synthesis of up to 30 nucleotides during base or nucleotide excision repair. An important question is what happens when many more nucleotides are removed, resulting in long single-stranded DNA (ssDNA) lesions. Such lesions appear on chromosomes during telomere damage, double strand break repair or after the UV damage of stationary phase cells. Here, we show that long single-stranded lesions, formed at dysfunctional telomeres in budding yeast, are re-synthesized when cells are removed from the telomere-damaging environment. This process requires Pol32, an accessory factor of Polymerase δ. However, re-synthesis takes place even when the telomere-damaging conditions persist, in which case the accessory factors of both polymerases δ and ε are required, and surprisingly, salt. Salt added to the medium facilitates the DNA synthesis, independently of the osmotic stress responses. These results provide unexpected insights into the DNA metabolism and challenge the current view on cellular responses to telomere dysfunction.
Collapse
Affiliation(s)
- Iglika G Ivanova
- Institute for Cell and Molecular Biosciences (ICaMB), Newcastle University, Newcastle upon Tyne, NE2 44H, UK
| | - Laura Maringele
- Institute for Cell and Molecular Biosciences (ICaMB), Newcastle University, Newcastle upon Tyne, NE2 44H, UK
| |
Collapse
|
171
|
Panek J, Frąc M, Bilińska-Wielgus N. Comparison of Chemical Sensitivity of Fresh and Long-Stored Heat Resistant Neosartorya fischeri Environmental Isolates Using BIOLOG Phenotype MicroArray System. PLoS One 2016; 11:e0147605. [PMID: 26815302 PMCID: PMC4729462 DOI: 10.1371/journal.pone.0147605] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 01/06/2016] [Indexed: 12/23/2022] Open
Abstract
Spoilage of heat processed food and beverage by heat resistant fungi (HRF) is a major problem for food industry in many countries. Neosartorya fischeri is the leading source of spoilage in thermally processed products. Its resistance to heat processing and toxigenicity makes studies about Neosartorya fischeri metabolism and chemical sensitivity essential. In this study chemical sensitivity of two environmental Neosartorya fischeri isolates were compared. One was isolated from canned apples in 1923 (DSM3700), the other from thermal processed strawberry product in 2012 (KC179765), used as long-stored and fresh isolate, respectively. The study was conducted using Biolog Phenotype MicroArray platforms of chemical sensitivity panel and traditional hole-plate method. The study allowed for obtaining data about Neosartorya fischeri growth inhibitors. The fresh isolate appeared to be much more resistant to chemical agents than the long-stored isolate. Based on phenotype microarray assay nitrogen compounds, toxic cations and membrane function compounds were the most effective in growth inhibition of N. fischeri isolates. According to the study zaragozic acid A, thallium(I) acetate and sodium selenate were potent and promising N. fischeri oriented fungicides which was confirmed by both chemical sensitivity microplates panel and traditional hole-plate methods.
Collapse
Affiliation(s)
- Jacek Panek
- Institute of Agrophysics, Polish Academy of Sciences, Department of Plant and Soil System, Laboratory of Molecular and Environmental Microbiology, Doświadczalna 4, 20–290 Lublin, Poland
| | - Magdalena Frąc
- Institute of Agrophysics, Polish Academy of Sciences, Department of Plant and Soil System, Laboratory of Molecular and Environmental Microbiology, Doświadczalna 4, 20–290 Lublin, Poland
- * E-mail:
| | - Nina Bilińska-Wielgus
- Institute of Agrophysics, Polish Academy of Sciences, Department of Plant and Soil System, Laboratory of Molecular and Environmental Microbiology, Doświadczalna 4, 20–290 Lublin, Poland
| |
Collapse
|
172
|
Chen PW, Fonseca LL, Hannun YA, Voit EO. Analysis of the Involvement of Different Ceramide Variants in the Response to Hydroxyurea Stress in Baker's Yeast. PLoS One 2016; 11:e0146839. [PMID: 26784947 PMCID: PMC4718512 DOI: 10.1371/journal.pone.0146839] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 12/21/2015] [Indexed: 12/11/2022] Open
Abstract
Sphingolipids have been identified as important signaling compounds in stress responses. However, it is not always clear how different sphingolipid profiles are achieved in a particular stress situation. Here we propose a detailed mass action model, containing 42 dependent variables and 137 reactions, that offers explanations of the roles of variant ceramides species, which differ in the lengths of their fatty acyl chains and their saturation state, in the response to hydroxyurea stress. The simulations demonstrate that the cells manage to achieve hydroxyurea tolerance through a well-coordinated, differential usage of the variant ceramide species. Moreover, the results suggest that key enzymes have different affinities toward saturated and unsaturated fatty acyl chains, which implies that the saturation state affords the cells with an additional mode of regulation that had not been recognized so far. These conclusions from our computational analysis are yet to be validated experimentally.
Collapse
Affiliation(s)
- Po-Wei Chen
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States of America
- The George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Luis L. Fonseca
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Yusuf A. Hannun
- The Cancer Center at Stony Brook Medicine, Stony Brook University, Health Science Center, Stony Brook, New York, United States of America
| | - Eberhard O. Voit
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
173
|
Kuang YT, Bhat R, Davies R. Mechanisms of Repair of Low Water Activity and pH-Injured Z
ygosaccharomyces rouxii
YSa40 in Glycerol and Sucrose/CPB Liquid Holding System. J FOOD PROCESS PRES 2015. [DOI: 10.1111/jfpp.12328] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Affiliation(s)
- Yeoh Tow Kuang
- School of Hospitality, Tourism and Culinary Arts; Taylor's University; Lakeside Campus Subang Jaya 47500 Selangor Malaysia
| | - Rajeev Bhat
- Food Technology Division; School of Industrial Technology; Universiti Sains Malaysia; Penang 11800 Malaysia
| | - Roland Davies
- Department of Food and Nutritional Sciences; University of Reading; Reading United Kingdom
| |
Collapse
|
174
|
Affected chromosome homeostasis and genomic instability of clonal yeast cultures. Curr Genet 2015; 62:405-18. [PMID: 26581629 PMCID: PMC4826422 DOI: 10.1007/s00294-015-0537-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 11/02/2015] [Accepted: 11/03/2015] [Indexed: 01/08/2023]
Abstract
Yeast cells originating from one single colony are considered genotypically and phenotypically identical. However, taking into account the cellular heterogeneity, it seems also important to monitor cell-to-cell variations within a clone population. In the present study, a comprehensive yeast karyotype screening was conducted using single chromosome comet assay. Chromosome-dependent and mutation-dependent changes in DNA (DNA with breaks or with abnormal replication intermediates) were studied using both single-gene deletion haploid mutants (bub1, bub2, mad1, tel1, rad1 and tor1) and diploid cells lacking one active gene of interest, namely BUB1/bub1, BUB2/bub2, MAD1/mad1, TEL1/tel1, RAD1/rad1 and TOR1/tor1 involved in the control of cell cycle progression, DNA repair and the regulation of longevity. Increased chromosome fragility and replication stress-mediated chromosome abnormalities were correlated with elevated incidence of genomic instability, namely aneuploid events—disomies, monosomies and to a lesser extent trisomies as judged by in situ comparative genomic hybridization (CGH). The tor1 longevity mutant with relatively balanced chromosome homeostasis was found the most genomically stable among analyzed mutants. During clonal yeast culture, spontaneously formed abnormal chromosome structures may stimulate changes in the ploidy state and, in turn, promote genomic heterogeneity. These alterations may be more accented in selected mutated genetic backgrounds, namely in yeast cells deficient in proper cell cycle regulation and DNA repair.
Collapse
|
175
|
PTEN Controls the DNA Replication Process through MCM2 in Response to Replicative Stress. Cell Rep 2015; 13:1295-1303. [DOI: 10.1016/j.celrep.2015.10.016] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 09/08/2015] [Accepted: 10/05/2015] [Indexed: 01/07/2023] Open
|
176
|
Sawant AA, Tanpure AA, Mukherjee PP, Athavale S, Kelkar A, Galande S, Srivatsan SG. A versatile toolbox for posttranscriptional chemical labeling and imaging of RNA. Nucleic Acids Res 2015; 44:e16. [PMID: 26384420 PMCID: PMC4737177 DOI: 10.1093/nar/gkv903] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 08/31/2015] [Indexed: 12/21/2022] Open
Abstract
Cellular RNA labeling strategies based on bioorthogonal chemical reactions are much less developed in comparison to glycan, protein and DNA due to its inherent instability and lack of effective methods to introduce bioorthogonal reactive functionalities (e.g. azide) into RNA. Here we report the development of a simple and modular posttranscriptional chemical labeling and imaging technique for RNA by using a novel toolbox comprised of azide-modified UTP analogs. These analogs facilitate the enzymatic incorporation of azide groups into RNA, which can be posttranscriptionally labeled with a variety of probes by click and Staudinger reactions. Importantly, we show for the first time the specific incorporation of azide groups into cellular RNA by endogenous RNA polymerases, which enabled the imaging of newly transcribing RNA in fixed and in live cells by click reactions. This labeling method is practical and provides a new platform to study RNA in vitro and in cells.
Collapse
Affiliation(s)
- Anupam A Sawant
- Department of Chemistry, Indian Institute of Science Education and Research, Pune, Dr. Homi Bhabha Road, Pashan, Pune 411008, India
| | - Arun A Tanpure
- Department of Chemistry, Indian Institute of Science Education and Research, Pune, Dr. Homi Bhabha Road, Pashan, Pune 411008, India
| | - Progya P Mukherjee
- Department of Chemistry, Indian Institute of Science Education and Research, Pune, Dr. Homi Bhabha Road, Pashan, Pune 411008, India
| | - Soumitra Athavale
- Center of Excellence in Epigenetics, Indian Institute of Science Education and Research, Pune, Dr. Homi Bhabha Road, Pashan, Pune 411008, India
| | - Ashwin Kelkar
- Center of Excellence in Epigenetics, Indian Institute of Science Education and Research, Pune, Dr. Homi Bhabha Road, Pashan, Pune 411008, India
| | - Sanjeev Galande
- Center of Excellence in Epigenetics, Indian Institute of Science Education and Research, Pune, Dr. Homi Bhabha Road, Pashan, Pune 411008, India National Centre for Cell Science, Ganeshkhind, Pune 411007, India
| | - Seergazhi G Srivatsan
- Department of Chemistry, Indian Institute of Science Education and Research, Pune, Dr. Homi Bhabha Road, Pashan, Pune 411008, India
| |
Collapse
|
177
|
Abstract
INTRODUCTION Sickle cell anemia (SCA) is a severe, inherited hemoglobin disorder affecting 100,000 persons in the US and millions worldwide. Hydroxyurea, a once daily oral medication, has emerged as the primary disease-modifying therapy for SCA. The accumulated body of evidence over 30 years demonstrates that hydroxyurea is a safe and effective therapy for SCA, but hydroxyurea remains underutilized for a variety of reasons. AREAS COVERED In this review, we summarize the available evidence regarding the pharmacology, clinical, and laboratory benefits, and safety of hydroxyurea therapy for the treatment of SCA. The purpose of this review is to provide the reader a comprehensive understanding of hydroxyurea and to reinforce the fact that hydroxyurea is a safe and effective medication for the treatment of SCA. EXPERT OPINION In our opinion, hydroxyurea therapy should be considered standard-of-care for SCA, representing an essential component of patient management. Early initiation and broader use of hydroxyurea will alter the natural history of SCA, so affected children can live longer and healthier lives. In addition, hydroxyurea use should be extended to low-resource settings such as sub-Saharan Africa, where the burden of SCA and the need for hydroxyurea is arguably the greatest.
Collapse
Affiliation(s)
- Patrick T McGann
- a Division of Hematology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center , Cincinnati , OH, USA
| | - Russell E Ware
- a Division of Hematology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center , Cincinnati , OH, USA
| |
Collapse
|
178
|
Complementation of Yeast Genes with Human Genes as an Experimental Platform for Functional Testing of Human Genetic Variants. Genetics 2015; 201:1263-74. [PMID: 26354769 PMCID: PMC4649650 DOI: 10.1534/genetics.115.181099] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Accepted: 09/01/2015] [Indexed: 12/11/2022] Open
Abstract
While the pace of discovery of human genetic variants in tumors, patients, and diverse populations has rapidly accelerated, deciphering their functional consequence has become rate-limiting. Using cross-species complementation, model organisms like the budding yeast, Saccharomyces cerevisiae, can be utilized to fill this gap and serve as a platform for testing human genetic variants. To this end, we performed two parallel screens, a one-to-one complementation screen for essential yeast genes implicated in chromosome instability and a pool-to-pool screen that queried all possible essential yeast genes for rescue of lethality by all possible human homologs. Our work identified 65 human cDNAs that can replace the null allele of essential yeast genes, including the nonorthologous pair yRFT1/hSEC61A1. We chose four human cDNAs (hLIG1, hSSRP1, hPPP1CA, and hPPP1CC) for which their yeast gene counterparts function in chromosome stability and assayed in yeast 35 tumor-specific missense mutations for growth defects and sensitivity to DNA-damaging agents. This resulted in a set of human–yeast gene complementation pairs that allow human genetic variants to be readily characterized in yeast, and a prioritized list of somatic mutations that could contribute to chromosome instability in human tumors. These data establish the utility of this cross-species experimental approach.
Collapse
|
179
|
Wemhoff S, Klassen R, Meinhardt F. DNA damage induced by the anticodon nuclease from a Pichia acaciae killer strain is linked to ribonucleotide reductase depletion. Cell Microbiol 2015; 18:211-22. [PMID: 26247322 DOI: 10.1111/cmi.12496] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 07/24/2015] [Accepted: 07/26/2015] [Indexed: 11/30/2022]
Abstract
Virus like element (VLE) encoded killer toxins of Pichia acaciae and Kluyveromyces lactis kill target cells through anticodon nuclease (ACNase) activity directed against tRNA(Gln) and tRNA(Glu) respectively. Not only does tRNA cleavage disable translation, it also affects DNA integrity as well. Consistent with DNA damage, which is involved in toxicity, target cells' mutation frequencies are elevated upon ACNase exposure, suggesting a link between translational integrity and genome surveillance. Here, we analysed whether ACNase action impedes the periodically and highly expressed S-phase specific ribonucleotide reductase (RNR) and proved that RNR expression is severely affected by PaT. Because RNR catalyses the rate-limiting step in dNTP synthesis, mutants affected in dNTP synthesis were scrutinized with respect to ACNase action. Mutations elevating cellular dNTPs antagonized the action of both the above ACNases, whereas mutations lowering dNTPs aggravated toxicity. Consistently, prevention of tRNA cleavage in elp3 or trm9 mutants, which both affect the wobble uridine modification of the target tRNA, suppressed the toxin hypersensitivity of a dNTP synthesis mutant. Moreover, dNTP synthesis defects exacerbated the PaT ACNase sensitivity of cells defective in homologous recombination, proving that dNTP depletion is responsible for subsequent DNA damage.
Collapse
Affiliation(s)
- Sabrina Wemhoff
- Institut für Molekulare Mikrobiologie und Biotechnologie, Westfälische Wilhelms-Universität Münster, 48149, Münster, Germany
| | - Roland Klassen
- Institut für Molekulare Mikrobiologie und Biotechnologie, Westfälische Wilhelms-Universität Münster, 48149, Münster, Germany
| | - Friedhelm Meinhardt
- Institut für Molekulare Mikrobiologie und Biotechnologie, Westfälische Wilhelms-Universität Münster, 48149, Münster, Germany
| |
Collapse
|
180
|
Fojtikova V, Stranava M, Vos MH, Liebl U, Hranicek J, Kitanishi K, Shimizu T, Martinkova M. Kinetic Analysis of a Globin-Coupled Histidine Kinase, AfGcHK: Effects of the Heme Iron Complex, Response Regulator, and Metal Cations on Autophosphorylation Activity. Biochemistry 2015. [PMID: 26212354 DOI: 10.1021/acs.biochem.5b00517] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The globin-coupled histidine kinase, AfGcHK, is a part of the two-component signal transduction system from the soil bacterium Anaeromyxobacter sp. Fw109-5. Activation of its sensor domain significantly increases its autophosphorylation activity, which targets the His183 residue of its functional domain. The phosphate group of phosphorylated AfGcHK is then transferred to the cognate response regulator. We investigated the effects of selected variables on the autophosphorylation reaction's kinetics. The kcat values of the heme Fe(III)-OH(-), Fe(III)-cyanide, Fe(III)-imidazole, and Fe(II)-O2 bound active AfGcHK forms were 1.1-1.2 min(-1), and their Km(ATP) values were 18.9-35.4 μM. However, the active form bearing a CO-bound Fe(II) heme had a kcat of 1.0 min(-1) but a very high Km(ATP) value of 357 μM, suggesting that its active site structure differs strongly from the other active forms. The Fe(II) heme-bound inactive form had kcat and Km(ATP) values of 0.4 min(-1) and 78 μM, respectively, suggesting that its low activity reflects a low affinity for ATP relative to that of the Fe(III) form. The heme-free form exhibited low activity, with kcat and Km(ATP) values of 0.3 min(-1) and 33.6 μM, respectively, suggesting that the heme iron complex is essential for high catalytic activity. Overall, our results indicate that the coordination and oxidation state of the sensor domain heme iron profoundly affect the enzyme's catalytic activity because they modulate its ATP binding affinity and thus change its kcat/Km(ATP) value. The effects of the response regulator and different divalent metal cations on the autophosphorylation reaction are also discussed.
Collapse
Affiliation(s)
| | | | - Marten H Vos
- §Laboratoire d'Optique et Biosciences, INSERM U1182-CNRS UMR7645, Ecole Polytechnique, 91128 Palaiseau Cedex, France
| | - Ursula Liebl
- §Laboratoire d'Optique et Biosciences, INSERM U1182-CNRS UMR7645, Ecole Polytechnique, 91128 Palaiseau Cedex, France
| | | | | | | | | |
Collapse
|
181
|
Verma G, Mishra S, Sangwan N, Sharma S. Reactive oxygen species mediate axis-cotyledon signaling to induce reserve mobilization during germination and seedling establishment in Vigna radiata. JOURNAL OF PLANT PHYSIOLOGY 2015; 184:79-88. [PMID: 26241759 DOI: 10.1016/j.jplph.2015.07.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Revised: 07/03/2015] [Accepted: 07/03/2015] [Indexed: 05/18/2023]
Abstract
Seeds represent an excellent opportunity to investigate the role of reactive oxygen species (ROS) in control of metabolism during germination and seedling establishment. Cotyledons, the storage organs in Vigna, do not display growth/cell division while the embryonic axis shows rapid growth and intense metabolic activity. The present study investigates the possibility of ROS generated during respiration in the axis serving as messengers guiding storage reserve mobilization from cotyledons at the pre-greening stage. Seeds were germinated in the presence of hydroxyurea to halt cell division in the S-phase and separately in Edaravone, a potent free radical scavenger. Both treatments caused a decrease in germination percentage, seedling growth and protein mobilization. In the growing axis, both treatments resulted in a decrease in hydrogen peroxide (H2O2), total ROS, MDA and protein carbonyls. The picture in cotyledons was quite different, owing to the physiological dissimilarities between the tissues. The status of redox as evident by GSH/GSSG ratios tended toward oxidizing in axis in comparison to the highly reducing environment found in cotyledons. This is construed as a tendency to maintain redox buffering on the oxidizing side in the axis, to facilitate the passage of ROS message. These results strongly indicate that suppression of cell division or scavenging of ROS adversely affects protein reserve mobilization. It is proposed that apart from H2O2 being a transportable signal, the final message perceived in cotyledons also comprises lipid peroxidation, protein carbonylation and alteration of redox status of the glutathione pool.
Collapse
Affiliation(s)
- Giti Verma
- Department of Biochemistry, University of Lucknow, Lucknow 226007, India.
| | - Sujata Mishra
- Plant Diversity, Systematics and Herbarium Division, CSIR-National Botanical Research Institute, Lucknow 226001, India.
| | - Neelam Sangwan
- Metabolic and Structural Biology Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226015, India.
| | - Samir Sharma
- Department of Biochemistry, University of Lucknow, Lucknow 226007, India.
| |
Collapse
|
182
|
Affiliation(s)
- Wenjian Gan
- a Department of Pathology; Beth Israel Deaconess Medical Center; Harvard Medical School ; Boston , MA USA
| | | | | |
Collapse
|
183
|
Skoneczna A, Kaniak A, Skoneczny M. Genetic instability in budding and fission yeast-sources and mechanisms. FEMS Microbiol Rev 2015; 39:917-67. [PMID: 26109598 PMCID: PMC4608483 DOI: 10.1093/femsre/fuv028] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/26/2015] [Indexed: 12/17/2022] Open
Abstract
Cells are constantly confronted with endogenous and exogenous factors that affect their genomes. Eons of evolution have allowed the cellular mechanisms responsible for preserving the genome to adjust for achieving contradictory objectives: to maintain the genome unchanged and to acquire mutations that allow adaptation to environmental changes. One evolutionary mechanism that has been refined for survival is genetic variation. In this review, we describe the mechanisms responsible for two biological processes: genome maintenance and mutation tolerance involved in generations of genetic variations in mitotic cells of both Saccharomyces cerevisiae and Schizosaccharomyces pombe. These processes encompass mechanisms that ensure the fidelity of replication, DNA lesion sensing and DNA damage response pathways, as well as mechanisms that ensure precision in chromosome segregation during cell division. We discuss various factors that may influence genome stability, such as cellular ploidy, the phase of the cell cycle, transcriptional activity of a particular region of DNA, the proficiency of DNA quality control systems, the metabolic stage of the cell and its respiratory potential, and finally potential exposure to endogenous or environmental stress. The stability of budding and fission yeast genomes is influenced by two contradictory factors: (1) the need to be fully functional, which is ensured through the replication fidelity pathways of nuclear and mitochondrial genomes through sensing and repairing DNA damage, through precise chromosome segregation during cell division; and (2) the need to acquire changes for adaptation to environmental challenges.
Collapse
Affiliation(s)
- Adrianna Skoneczna
- Laboratory of Mutagenesis and DNA Repair, Institute of Biochemistry and Biophysics, Polish Academy of Science, 02-106 Warsaw, Poland
| | - Aneta Kaniak
- Laboratory of Mutagenesis and DNA Repair, Institute of Biochemistry and Biophysics, Polish Academy of Science, 02-106 Warsaw, Poland
| | - Marek Skoneczny
- Department of Genetics, Institute of Biochemistry and Biophysics, Polish Academy of Science, 02-106 Warsaw, Poland
| |
Collapse
|
184
|
Martín Sánchez C, Pérez Martín JM, Jin JS, Dávalos A, Zhang W, de la Peña G, Martínez-Botas J, Rodríguez-Acebes S, Suárez Y, Hazen MJ, Gómez-Coronado D, Busto R, Cheng YC, Lasunción MA. Disruption of the mevalonate pathway induces dNTP depletion and DNA damage. Biochim Biophys Acta Mol Cell Biol Lipids 2015; 1851:1240-53. [PMID: 26055626 DOI: 10.1016/j.bbalip.2015.06.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 05/23/2015] [Accepted: 06/01/2015] [Indexed: 12/21/2022]
Abstract
The mevalonate pathway is tightly linked to cell division. Mevalonate derived non-sterol isoprenoids and cholesterol are essential for cell cycle progression and mitosis completion respectively. In the present work, we studied the effects of fluoromevalonate, a competitive inhibitor of mevalonate diphosphate decarboxylase, on cell proliferation and cell cycle progression in both HL-60 and MOLT-4 cells. This enzyme catalyzes the synthesis of isopentenyl diphosphate, the first isoprenoid in the cholesterol biosynthesis pathway, consuming ATP at the same time. Inhibition of mevalonate diphosphate decarboxylase was followed by a rapid accumulation of mevalonate diphosphate and the reduction of ATP concentrations, while the cell content of cholesterol was barely affected. Strikingly, mevalonate diphosphate decarboxylase inhibition also resulted in the depletion of dNTP pools, which has never been reported before. These effects were accompanied by inhibition of cell proliferation and cell cycle arrest at S phase, together with the appearance of γ-H2AX foci and Chk1 activation. Inhibition of Chk1 in cells treated with fluoromevalonate resulted in premature entry into mitosis and massive cell death, indicating that the inhibition of mevalonate diphosphate decarboxylase triggered a DNA damage response. Notably, the supply of exogenously deoxyribonucleosides abolished γ-H2AX formation and prevented the effects of mevalonate diphosphate decarboxylase inhibition on DNA replication and cell growth. The results indicate that dNTP pool depletion caused by mevalonate diphosphate decarboxylase inhibition hampered DNA replication with subsequent DNA damage, which may have important consequences for replication stress and genomic instability.
Collapse
Affiliation(s)
- Covadonga Martín Sánchez
- Servicio de Bioquímica-Investigación, Hospital Universitario Ramón y Cajal, IRyCIS, 28034 Madrid, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Spain.
| | - José Manuel Pérez Martín
- Departamento de Biología, Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain.
| | - Jong-Sik Jin
- Department of Pharmacology, Section of Medical Oncology, Yale School of Medicine, New Haven, CT 06520, USA; Department of Oriental Medicine Resources, College of Environmental & Bioresource Sciences, Chonbuk National University, Jeonju, Jeonbuk, Republic of Korea.
| | - Alberto Dávalos
- Laboratory of Functional Foods, IMDEA-Food, 28036 Madrid, Spain.
| | - Wei Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau, China.
| | - Gema de la Peña
- Servicio de Bioquímica-Investigación, Hospital Universitario Ramón y Cajal, IRyCIS, 28034 Madrid, Spain.
| | - Javier Martínez-Botas
- Servicio de Bioquímica-Investigación, Hospital Universitario Ramón y Cajal, IRyCIS, 28034 Madrid, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Spain.
| | - Sara Rodríguez-Acebes
- DNA Replication Group, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain.
| | - Yajaira Suárez
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Section of Comparative Medicine, Department of Pathology and the Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA.
| | - María José Hazen
- Departamento de Biología, Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain.
| | - Diego Gómez-Coronado
- Servicio de Bioquímica-Investigación, Hospital Universitario Ramón y Cajal, IRyCIS, 28034 Madrid, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Spain.
| | - Rebeca Busto
- Servicio de Bioquímica-Investigación, Hospital Universitario Ramón y Cajal, IRyCIS, 28034 Madrid, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Spain.
| | - Yung-Chi Cheng
- Department of Pharmacology, Section of Medical Oncology, Yale School of Medicine, New Haven, CT 06520, USA.
| | - Miguel A Lasunción
- Servicio de Bioquímica-Investigación, Hospital Universitario Ramón y Cajal, IRyCIS, 28034 Madrid, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Spain.
| |
Collapse
|
185
|
Molina B, Marchetti F, Gómez L, Ramos S, Torres L, Ortiz R, Altamirano-Lozano M, Carnevale A, Frias S. Hydroxyurea induces chromosomal damage in G2 and enhances the clastogenic effect of mitomycin C in Fanconi anemia cells. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2015; 56:457-467. [PMID: 25663157 DOI: 10.1002/em.21938] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 01/05/2015] [Indexed: 06/04/2023]
Abstract
Fanconi's anemia (FA) is a recessive disease; 16 genes are currently recognized in FA. FA proteins participate in the FA/BRCA pathway that plays a crucial role in the repair of DNA damage induced by crosslinking compounds. Hydroxyurea (HU) is an agent that induces replicative stress by inhibiting ribonucleotide reductase (RNR), which synthesizes deoxyribonucleotide triphosphates (dNTPs) necessary for DNA replication and repair. HU is known to activate the FA pathway; however, its clastogenic effects are not well characterized. We have investigated the effects of HU treatment alone or in sequential combination with mitomycin-C (MMC) on FA patient-derived lymphoblastoid cell lines from groups FA-A, B, C, D1/BRCA2, and E and on lymphocytes from two unclassified FA patients. All FA cells showed a significant increase (P < 0.05) in chromosomal aberrations following treatment with HU during the last 3 h before mitosis. Furthermore, when FA cells previously exposed to MMC were treated with HU, we observed an increase of MMC-induced DNA damage that was characterized by high occurrence of DNA breaks and a reduction in rejoined chromosomal aberrations. These findings show that exposure to HU during G2 induces chromosomal aberrations by a mechanism that is independent of its well-known role in replication fork stalling during S-phase and that HU interfered mainly with the rejoining process of DNA damage. We suggest that impaired oxidative stress response, lack of an adequate amount of dNTPs for DNA repair due to RNR inhibition, and interference with cell cycle control checkpoints underlie the clastogenic activity of HU in FA cells. Environ. Mol. Mutagen. 56:457-467, 2015. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Bertha Molina
- Laboratorio de Citogenética, Instituto Nacional de Pediatría, México
| | | | - Laura Gómez
- Laboratorio de Citogenética, Instituto Nacional de Pediatría, México
| | - Sandra Ramos
- Laboratorio de Citogenética, Instituto Nacional de Pediatría, México
| | - Leda Torres
- Laboratorio de Citogenética, Instituto Nacional de Pediatría, México
| | - Rocio Ortiz
- Laboratorio de Citometría de Flujo, Universidad Autónoma Metropolitana, Iztapalapa, Mexico
| | | | - Alessandra Carnevale
- Subdirección de Genómica Poblacional, Instituto Nacional de Medicina Genómica, México
| | - Sara Frias
- Laboratorio de Citogenética, Instituto Nacional de Pediatría, México
- Departamento de Medicina, Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, UNAM, México
| |
Collapse
|
186
|
Schneider LS, von Schwarzenberg K, Lehr T, Ulrich M, Kubisch-Dohmen R, Liebl J, Trauner D, Menche D, Vollmar AM. Vacuolar-ATPase Inhibition Blocks Iron Metabolism to Mediate Therapeutic Effects in Breast Cancer. Cancer Res 2015; 75:2863-74. [DOI: 10.1158/0008-5472.can-14-2097] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 04/28/2015] [Indexed: 11/16/2022]
|
187
|
Chen X, Bosques L, Sung P, Kupfer GM. A novel role for non-ubiquitinated FANCD2 in response to hydroxyurea-induced DNA damage. Oncogene 2015; 35:22-34. [PMID: 25893307 DOI: 10.1038/onc.2015.68] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 01/30/2015] [Accepted: 02/02/2015] [Indexed: 12/20/2022]
Abstract
Fanconi anemia (FA) is a genetic disease of bone marrow failure, cancer susceptibility, and sensitivity to DNA crosslinking agents. FANCD2, the central protein of the FA pathway, is monoubiquitinated upon DNA damage, such as crosslinkers and replication blockers such as hydroxyurea (HU). Even though FA cells demonstrate unequivocal sensitivity to crosslinkers, such as mitomycin C (MMC), we find that they are largely resistant to HU, except for cells absent for expression of FANCD2. FANCD2, RAD51 and RAD18 form a complex, which is enhanced upon HU exposure. Surprisingly, although FANCD2 is required for this enhanced interaction, its monoubiquitination is not. Similarly, non-ubiquitinated FANCD2 can still support proliferation cell nuclear antigen (PCNA) monoubiquitination. RAD51, but not BRCA2, is also required for PCNA monoubiquitination in response to HU, suggesting that this function is independent of homologous recombination (HR). We further show that translesion (TLS) polymerase PolH chromatin localization is decreased in FANCD2 deficient cells, FANCD2 siRNA knockdown cells and RAD51 siRNA knockdown cells, and PolH knockdown results in HU sensitivity only. Our data suggest that FANCD2 and RAD51 have an important role in PCNA monoubiquitination and TLS in a FANCD2 monoubiquitination and HR-independent manner in response to HU. This effect is not observed with MMC treatment, suggesting a non-canonical function for the FA pathway in response to different types of DNA damage.
Collapse
Affiliation(s)
- X Chen
- Department of Pediatrics, Section of Hematology/Oncology, Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA.,Department of Pathology, Section of Hematology/Oncology, Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | - L Bosques
- Department of Pediatrics, Section of Hematology/Oncology, Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA.,Department of Pathology, Section of Hematology/Oncology, Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | - P Sung
- Department of Molecular, Cellular, and Developmental Biology, Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | - G M Kupfer
- Department of Pediatrics, Section of Hematology/Oncology, Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA.,Department of Pathology, Section of Hematology/Oncology, Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
188
|
Narukawa-Nara M, Sasaki K, Ishii A, Baba K, Amano K, Kuroki M, Saitoh KI, Kamakura T. Identification and characterization of a novel gene encoding the NBS1 protein in Pyricularia oryzae. Biosci Biotechnol Biochem 2015; 79:1183-90. [PMID: 25774746 DOI: 10.1080/09168451.2015.1015951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The ascomycete Pyricularia oryzae (teleomorph: Magnaporthe oryzae) causes one of the most serious diseases known as rice blast. The Nijmegen breakage syndrome protein (NBS1) is essential for DNA repair; thus, we studied the P. oryzae NBS1 homolog (PoNBS1). A PoNBS1 null mutant exhibited high sensitivity to DNA damage-inducing agents. The mutant also exhibited the retarded hyphal growth, and induced abnormal conidial germination and shape, but showed normal appressorium formation. The phenotypes of the null mutant were complemented by introducing the cDNA of PoNBS1 driven by a TrpC promoter of Aspergillus nidulans. In addition, the null mutant similarly complemented with the PoNBS1 cDNA lacking the FHA domain that had a normal phenotype except for hyphal growth. These results suggest that PoNBS1 is involved in DNA repair and normal development in P. oryzae. Moreover, the FHA domain of PoNBS1 participates in normal hyphal growth.
Collapse
Affiliation(s)
- Megumi Narukawa-Nara
- a Department of Applied Biological Science, Faculty of Science and Technology , Tokyo University of Science , Noda , Japan
| | | | | | | | | | | | | | | |
Collapse
|
189
|
Kaur S, Srivastava G, Sharma AN, Jolly RS. Novel immunosuppressive agent caerulomycin A exerts its effect by depleting cellular iron content. Br J Pharmacol 2015; 172:2286-99. [PMID: 25537422 DOI: 10.1111/bph.13051] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 12/03/2014] [Accepted: 12/14/2014] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND AND PURPOSE Recently, we have described the use of caerulomycin A (CaeA) as a potent novel immunosuppressive agent. Immunosuppressive drugs are crucial for long-term graft survival following organ transplantation and treatment of autoimmune diseases, inflammatory disorders, hypersensitivity to allergens, etc. The objective of this study was to identify cellular targets of CaeA and decipher its mechanism of action. EXPERIMENTAL APPROACH Jurkat cells were treated with CaeA and cellular iron content, iron uptake/release, DNA content and deoxyribonucleoside triphosphate pool determined. Activation of MAPKs; expression level of transferrin receptor 1, ferritin and cell cycle control molecules; reactive oxygen species (ROS) and cell viability were measured using Western blotting, qRT-PCR or flow cytometry. KEY RESULTS CaeA caused intracellular iron depletion by reducing its uptake and increasing its release by cells. CaeA caused cell cycle arrest by (i) inhibiting ribonucleotide reductase (RNR) enzyme, which catalyses the rate-limiting step in the synthesis of DNA; (ii) stimulating MAPKs signalling transduction pathways that play an important role in cell growth, proliferation and differentiation; and (iii) by targeting cell cycle control molecules such as cyclin D1, cyclin-dependent kinase 4 and p21(CIP1/WAF1) . The effect of CaeA on cell proliferation was reversible. CONCLUSIONS AND IMPLICATIONS CaeA exerts its immunosuppressive effect by targeting iron. The effect is reversible, which makes CaeA an attractive candidate for development as a potent immunosuppressive drug, but also indicates that iron chelation can be used as a rationale approach to selectively suppress the immune system, because compared with normal cells, rapidly proliferating cells require a higher utilization of iron.
Collapse
Affiliation(s)
- Suneet Kaur
- Department of Chemistry, CSIR-Institute of Microbial Technology, Chandigarh, India
| | | | | | | |
Collapse
|
190
|
Guan Z, Wang X, Dong Y, Xu L, Zhu Z, Wang J, Zhang T, Niu B. dNTP deficiency induced by HU via inhibiting ribonucleotide reductase affects neural tube development. Toxicology 2014; 328:142-51. [PMID: 25527867 DOI: 10.1016/j.tox.2014.12.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 11/12/2014] [Accepted: 12/01/2014] [Indexed: 12/22/2022]
Abstract
Exposure to environmental toxic chemicals in utero during the neural tube development period can cause developmental disorders. To evaluate the disruption of neural tube development programming, the murine neural tube defects (NTDs) model was induced by interrupting folate metabolism using methotrexate in our previous study. The present study aimed to examine the effects of dNTP deficiency induced by hydroxyurea (HU), a specific ribonucleotide reductase (RNR) inhibitor, during murine neural tube development. Pregnant C57BL/6J mice were intraperitoneally injected with various doses of HU on gestation day (GD) 7.5, and the embryos were checked on GD 11.5. RNR activity and deoxynucleoside triphosphate (dNTP) levels were measured in the optimal dose. Additionally, DNA damage was examined by comet analysis and terminal deoxynucleotidyl transferase mediated dUTP nick end-labeling (TUNEL) assay. Cellular behaviors in NTDs embryos were evaluated with phosphorylation of histone H3 (PH-3) and caspase-3 using immunohistochemistry and western blot analysis. The results showed that NTDs were observed mostly with HU treatment at an optimal dose of 225 mg/kg b/w. RNR activity was inhibited and dNTP levels were decreased in HU-treated embryos with NTDs. Additionally, increased DNA damage, decreased proliferation, and increased caspase-3 were significant in NTDs embryos compared to the controls. Results indicated that HU induced murine NTDs model by disturbing dNTP metabolism and further led to the abnormal cell balance between proliferation and apoptosis.
Collapse
Affiliation(s)
- Zhen Guan
- Department of Biotechnology, Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Xiuwei Wang
- Department of Biotechnology, Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Yanting Dong
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China
| | - Lin Xu
- Department of Biotechnology, Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Zhiqiang Zhu
- Department of Biotechnology, Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Jianhua Wang
- Department of Biotechnology, Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Ting Zhang
- Department of Biotechnology, Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China.
| | - Bo Niu
- Department of Biotechnology, Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China.
| |
Collapse
|
191
|
Lim EK, Kim T, Paik S, Haam S, Huh YM, Lee K. Nanomaterials for Theranostics: Recent Advances and Future Challenges. Chem Rev 2014; 115:327-94. [DOI: 10.1021/cr300213b] [Citation(s) in RCA: 916] [Impact Index Per Article: 91.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Eun-Kyung Lim
- Department
of Radiology, Yonsei University, Seoul 120-752, Korea
- BioNanotechnology
Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Korea
| | - Taekhoon Kim
- Department
of Chemistry, Korea University, Seoul 136-701, Korea
- Electronic
Materials Laboratory, Samsung Advanced Institute of Technology, Mt. 14-1,
Nongseo-Ri, Giheung-Eup, Yongin-Si, Gyeonggi-Do 449-712, Korea
| | - Soonmyung Paik
- Severance
Biomedical Research Institute, Yonsei University College of Medicine, Seoul 120-749, Korea
- Division
of Pathology, NSABP Foundation, Pittsburgh, Pennsylvania 15212, United States
| | - Seungjoo Haam
- Department
of Chemical and Biomolecular Engineering, Yonsei University, Seoul 120-749, Korea
| | - Yong-Min Huh
- Department
of Radiology, Yonsei University, Seoul 120-752, Korea
| | - Kwangyeol Lee
- Department
of Chemistry, Korea University, Seoul 136-701, Korea
| |
Collapse
|
192
|
Tang M, Li Y, Zhang X, Deng T, Zhou Z, Ma W, Songyang Z. Structural maintenance of chromosomes flexible hinge domain containing 1 (SMCHD1) promotes non-homologous end joining and inhibits homologous recombination repair upon DNA damage. J Biol Chem 2014; 289:34024-32. [PMID: 25294876 DOI: 10.1074/jbc.m114.601179] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Structural maintenance of chromosomes flexible hinge domain containing 1 (SMCHD1) has been shown to be involved in gene silencing and DNA damage. However, the exact mechanisms of how SMCHD1 participates in DNA damage remains largely unknown. Here we present evidence that SMCHD1 recruitment to DNA damage foci is regulated by 53BP1. Knocking out SMCHD1 led to aberrant γH2AX foci accumulation and compromised cell survival upon DNA damage, demonstrating the critical role of SMCHD1 in DNA damage repair. Following DNA damage induction, SMCHD1 depletion resulted in reduced 53BP1 foci and increased BRCA1 foci, as well as less efficient non-homologous end joining (NHEJ) and elevated levels of homologous recombination (HR). Taken together, these results suggest an important function of SMCHD1 in promoting NHEJ and repressing HR repair in response to DNA damage.
Collapse
Affiliation(s)
- Mengfan Tang
- From the Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory for Biocontrol, SYSU-Baylor College of Medicine Joint Research Center for Biomedical Sciences, School of Life Sciences, Sun Yat-sen University, Guangzhou, China, 510275 and
| | - Yujing Li
- From the Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory for Biocontrol, SYSU-Baylor College of Medicine Joint Research Center for Biomedical Sciences, School of Life Sciences, Sun Yat-sen University, Guangzhou, China, 510275 and
| | - Xiya Zhang
- From the Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory for Biocontrol, SYSU-Baylor College of Medicine Joint Research Center for Biomedical Sciences, School of Life Sciences, Sun Yat-sen University, Guangzhou, China, 510275 and
| | - Tingting Deng
- From the Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory for Biocontrol, SYSU-Baylor College of Medicine Joint Research Center for Biomedical Sciences, School of Life Sciences, Sun Yat-sen University, Guangzhou, China, 510275 and
| | - Zhifen Zhou
- From the Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory for Biocontrol, SYSU-Baylor College of Medicine Joint Research Center for Biomedical Sciences, School of Life Sciences, Sun Yat-sen University, Guangzhou, China, 510275 and
| | - Wenbin Ma
- From the Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory for Biocontrol, SYSU-Baylor College of Medicine Joint Research Center for Biomedical Sciences, School of Life Sciences, Sun Yat-sen University, Guangzhou, China, 510275 and
| | - Zhou Songyang
- From the Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory for Biocontrol, SYSU-Baylor College of Medicine Joint Research Center for Biomedical Sciences, School of Life Sciences, Sun Yat-sen University, Guangzhou, China, 510275 and Verna and Marrs Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas 77030
| |
Collapse
|
193
|
DeBaun MR. Hydroxyurea therapy contributes to infertility in adult men with sickle cell disease: a review. Expert Rev Hematol 2014; 7:767-73. [PMID: 25242414 DOI: 10.1586/17474086.2014.959922] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Hydroxyurea therapy, a chemotherapeutic agent, is the only US FDA approved therapy for the prevention of vaso-occlusive pain in sickle cell disease (SCD). The National Institutes of Health has sponsored two Phase III randomized, placebo-controlled trials, initially in adults, and subsequently in children with sickle cell anemia (SCA). Despite the overwhelming evidence that hydroxyurea therapy is beneficial to children and adults with SCA, individuals with SCA and their families express reservations about its use, in part because of the concerns about fertility, particularly in men. As adolescent boys with SCD are now expected to reach their reproductive years, a new concern is emerging about the role of hydroxyurea therapy as a barrier to their progeny. This review will systemically evaluate compromised fertility in men with SCD, and the evidence that hydroxyurea therapy is associated with further decreasing fertility in men with SCD.
Collapse
Affiliation(s)
- Michael R DeBaun
- Department of Pediatrics, Division of Hematology/Oncology, Vanderbilt-Meharry-Matthew Walker Center for Excellence in Sickle Cell Disease, Vanderbilt University Medical Center 2200 Children's Way, Room 11206 DOT, Nashville, TN 37232-9000, USA
| |
Collapse
|
194
|
Roy K, Chanfreau G. Stress-induced nuclear RNA degradation pathways regulate yeast bromodomain factor 2 to promote cell survival. PLoS Genet 2014; 10:e1004661. [PMID: 25232960 PMCID: PMC4169253 DOI: 10.1371/journal.pgen.1004661] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 08/11/2014] [Indexed: 11/23/2022] Open
Abstract
Bromodomain proteins are key regulators of gene expression. How the levels of these factors are regulated in specific environmental conditions is unknown. Previous work has established that expression of yeast Bromodomain factor 2 (BDF2) is limited by spliceosome-mediated decay (SMD). Here we show that BDF2 is subject to an additional layer of post-transcriptional control through RNase III-mediated decay (RMD). We found that the yeast RNase III Rnt1p cleaves a stem-loop structure within the BDF2 mRNA to down-regulate its expression. However, these two nuclear RNA degradation pathways play distinct roles in the regulation of BDF2 expression, as we show that the RMD and SMD pathways of the BDF2 mRNA are differentially activated or repressed in specific environmental conditions. RMD is hyper-activated by salt stress and repressed by hydroxyurea-induced DNA damage while SMD is inactivated by salt stress and predominates during DNA damage. Mutations of cis-acting signals that control SMD and RMD rescue numerous growth defects of cells lacking Bdf1p, and show that SMD plays an important role in the DNA damage response. These results demonstrate that specific environmental conditions modulate nuclear RNA degradation pathways to control BDF2 expression and Bdf2p-mediated gene regulation. Moreover, these results show that precise dosage of Bromodomain factors is essential for cell survival in specific environmental conditions, emphasizing their importance for controlling chromatin structure and gene expression in response to environmental stress. Cells adapt to changes in the environment through modulating gene expression at both the RNA and protein levels. RNA degradation plays a central role in this adaption response, by controlling the stability of specific mRNAs to optimize protein production in different conditions. In this study, we show that the gene encoding Bromodomain Factor 2 (BDF2) is tightly regulated according to environmental conditions by two distinct RNA degradation mechanisms. We show that these RNA degradation pathways are critical for cell growth in specific conditions. Our study suggests that environmental modulation of nuclear RNA degradation pathways is a previously unappreciated aspect of gene expression control.
Collapse
Affiliation(s)
- Kevin Roy
- Department of Chemistry and Biochemistry and the Molecular Biology Institute, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Guillaume Chanfreau
- Department of Chemistry and Biochemistry and the Molecular Biology Institute, University of California, Los Angeles, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
195
|
Hirota K, Tsuda M, Murai J, Takagi T, Keka IS, Narita T, Fujita M, Sasanuma H, Kobayashi J, Takeda S. SUMO-targeted ubiquitin ligase RNF4 plays a critical role in preventing chromosome loss. Genes Cells 2014; 19:743-54. [PMID: 25205350 DOI: 10.1111/gtc.12173] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 07/26/2014] [Indexed: 01/08/2023]
Abstract
RING finger protein 4 (RNF4) represents a subclass of ubiquitin ligases that target proteins modified by the small ubiquitin-like modifier (SUMO) for ubiquitin-mediated degradation. We disrupted the RNF4 gene in chicken DT40 cells and found that the resulting RNF4(-/-) cells gradually lost proliferation capability. Strikingly, this compromised proliferation was associated with an unprecedented cellular effect: the gradual decrease in the number of intact chromosomes. In the 6 weeks after gene targeting, there was a 25% reduction in the DNA content of the RNF4(-/-) cells. Regarding trisomic chromosome 2, 60% of the RNF4(-/-) cells lost one homologue, suggesting that DNA loss was mediated by whole chromosome loss. To determine the cause of this chromosome loss, we examined cell-cycle checkpoint pathways. RNF4(-/-) cells showed a partial defect in the spindle assembly checkpoint, premature dissociation of sister chromatids, and a marked increase in the number of lagging chromosomes at anaphase. Thus, combined defects in SAC and sister chromatid cohesion may result in increased lagging chromosomes, leading to chromosome loss without accompanying chromosome gain in RNF4(-/-) cells. We therefore propose that RNF4 plays a novel role in preventing the loss of intact chromosomes and ensures the maintenance of chromosome integrity.
Collapse
Affiliation(s)
- Kouji Hirota
- Department of Radiation Genetics, Graduate School of Medicine, Kyoto University, Yoshidakonoe, Sakyo-ku, Kyoto, 606-8501, Japan; Department of Chemistry, Graduate School of Science and Engineering, Tokyo Metropolitan University, Minamiosawa 1-1, Hachioji-shi, Tokyo, 192-0397, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
196
|
Bryan DS, Ransom M, Adane B, York K, Hesselberth JR. High resolution mapping of modified DNA nucleobases using excision repair enzymes. Genome Res 2014; 24:1534-42. [PMID: 25015380 PMCID: PMC4158761 DOI: 10.1101/gr.174052.114] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 07/08/2014] [Indexed: 12/20/2022]
Abstract
The incorporation and creation of modified nucleobases in DNA have profound effects on genome function. We describe methods for mapping positions and local content of modified DNA nucleobases in genomic DNA. We combined in vitro nucleobase excision with massively parallel DNA sequencing (Excision-seq) to determine the locations of modified nucleobases in genomic DNA. We applied the Excision-seq method to map uracil in E. coli and budding yeast and discovered significant variation in uracil content, wherein uracil is excluded from the earliest and latest replicating regions of the genome, possibly driven by changes in nucleotide pool composition. We also used Excision-seq to identify sites of pyrimidine dimer formation induced by UV light exposure, where the method could distinguish between sites of cyclobutane and 6-4 photoproduct formation. These UV mapping data enabled analysis of local sequence bias around pyrimidine dimers and suggested a preference for an adenosine downstream from 6-4 photoproducts. The Excision-seq method is broadly applicable for high precision, genome-wide mapping of modified nucleobases with cognate repair enzymes.
Collapse
Affiliation(s)
- D Suzi Bryan
- Department of Biochemistry and Molecular Genetics, Program in Molecular Biology, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| | - Monica Ransom
- Department of Biochemistry and Molecular Genetics, Program in Molecular Biology, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| | - Biniam Adane
- Department of Biochemistry and Molecular Genetics, Program in Molecular Biology, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| | - Kerri York
- Department of Biochemistry and Molecular Genetics, Program in Molecular Biology, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| | - Jay R Hesselberth
- Department of Biochemistry and Molecular Genetics, Program in Molecular Biology, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| |
Collapse
|
197
|
Kessler Z, Yanowitz J. Methodological considerations for mutagen exposure in C. elegans. Methods 2014; 68:441-9. [PMID: 24768858 PMCID: PMC5449201 DOI: 10.1016/j.ymeth.2014.04.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 04/10/2014] [Accepted: 04/11/2014] [Indexed: 11/15/2022] Open
Abstract
Maintenance of the genome requires the continual repair of DNA lesions. Exposure of nematodes to DNA damage-inducing agents is a powerful method to rapidly ascribe a role for specific genes in DNA repair and to define epistatic relationships to other repair genes which allows for the construction of repair pathways. Despite the extensive use of these agents, however, differences in dosing, timing, and handling makes it difficult to compare results across laboratories. We provide herein a consideration of the parameters that influence the results of these exposures and detailed protocols for the exposure to mutagenic inducing agents.
Collapse
Affiliation(s)
- Zebulin Kessler
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh School of Medicine, 204 Craft Avenue, Pittsburgh, PA 15213, United States
| | - Judith Yanowitz
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh School of Medicine, 204 Craft Avenue, Pittsburgh, PA 15213, United States.
| |
Collapse
|
198
|
Heo SH, Cha Y, Park KS. Hydroxyurea induces a hypersensitive apoptotic response in mouse embryonic stem cells through p38-dependent acetylation of p53. Stem Cells Dev 2014; 23:2435-42. [PMID: 24836177 DOI: 10.1089/scd.2013.0608] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
While hydroxyurea (HU) is well known to deplete dNTP pools and lead to replication fork arrest in the cell, the mechanisms by which it exerts a cell response are poorly understood. Here, our results suggest that mouse embryonic stem cells (mESCs), unlike terminally differentiated cells such as mouse embryonic fibroblasts (MEFs), rapidly respond to low concentrations of HU by p53 acetylation, leading to activation of the caspase-dependent apoptotic pathway. We show that HU treatment induces the production of nitric oxide (NO), which plays a central role in the rapid induction of apoptosis in mESCs. By contrast, reactive oxygen species, which are expressed at significantly higher levels in mESCs compared with MEFs, are not related to the HU response. Furthermore, on exposure to HU, the p38 signaling pathway becomes activated in a dose-dependent manner, and chemical inhibition of the p38 pathway attenuates HU-dependent apoptosis in mESCs. Our data reveal that acetylation of p53 as a result of HU-dependent NO production plays a key role in the induction of the apoptotic response in mESCs. Finally, p38 signaling appears to be the main pathway underlying the activation of apoptosis in mESCs in response to HU exposure.
Collapse
Affiliation(s)
- Sun-Hee Heo
- 1 Department of Biomedical Science, College of Life Science, CHA University , Seoul, Korea
| | | | | |
Collapse
|
199
|
|
200
|
Guarino E, Salguero I, Kearsey SE. Cellular regulation of ribonucleotide reductase in eukaryotes. Semin Cell Dev Biol 2014; 30:97-103. [PMID: 24704278 DOI: 10.1016/j.semcdb.2014.03.030] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Accepted: 03/26/2014] [Indexed: 12/22/2022]
Abstract
Synthesis of deoxynucleoside triphosphates (dNTPs) is essential for both DNA replication and repair and a key step in this process is catalyzed by ribonucleotide reductases (RNRs), which reduce ribonucleotides (rNDPs) to their deoxy forms. Tight regulation of RNR is crucial for maintaining the correct levels of all four dNTPs, which is important for minimizing the mutation rate and avoiding genome instability. Although allosteric control of RNR was the first discovered mechanism involved in regulation of the enzyme, other controls have emerged in recent years. These include regulation of expression of RNR genes, proteolysis of RNR subunits, control of the cellular localization of the small RNR subunit, and regulation of RNR activity by small protein inhibitors. This review will focus on these additional mechanisms of control responsible for providing a balanced supply of dNTPs.
Collapse
Affiliation(s)
- Estrella Guarino
- Tinbergen Building, Department of Zoology, University of Oxford, South Parks Road, Oxford OX1 3PS, United Kingdom.
| | - Israel Salguero
- Tinbergen Building, Department of Zoology, University of Oxford, South Parks Road, Oxford OX1 3PS, United Kingdom.
| | - Stephen E Kearsey
- Tinbergen Building, Department of Zoology, University of Oxford, South Parks Road, Oxford OX1 3PS, United Kingdom.
| |
Collapse
|