Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Andrology ; 7(4): 516-526, 2019 07.
Article in English | MEDLINE | ID: mdl-31119900

ABSTRACT

BACKGROUND: Germ cells have a unique and critical role as the conduit for hereditary information and therefore employ multiple strategies to protect genomic integrity and avoid mutations. Unlike somatic cells, which often respond to DNA damage by arresting the cell cycle and conducting DNA repair, germ cells as well as long-lived pluripotent stem cells typically avoid the use of error-prone repair mechanisms and favor apoptosis, reducing the risk of genetic alterations. Testicular germ cell tumors, the most common cancers of young men, arise from pre-natal germ cells. OBJECTIVES: To summarize the current understanding of DNA damage response mechanisms in pre-meiotic germ cells and to discuss how they impact both the origins of testicular germ cell tumors and their remarkable responsiveness to genotoxic chemotherapy. MATERIALS AND METHODS: We conducted a review of literature gathered from PubMed regarding the DNA damage response properties of testicular germ cell tumors and the germ cells from which they arise, as well as the influence of these mechanisms on therapeutic responses by testicular germ cell tumors. RESULTS AND DISCUSSION: This review provides a comprehensive evaluation of how the developmental origins of male germ cells and their inherent germ cell-like DNA damage response directly impact the development and therapeutic sensitivity of testicular germ cell tumors. CONCLUSIONS: The DNA damage response of germ cells directly impacts the development and therapeutic sensitivity of testicular germ cell tumors. Recent advances in the study of primordial germ cells, post-natal mitotically dividing germ cells, and pluripotent stem cells will allow for new investigations into the initiation, progression, and treatment of testicular germ cell tumors.


Subject(s)
DNA Damage , Embryonic Germ Cells/physiology , Neoplasms, Germ Cell and Embryonal/etiology , Testicular Neoplasms/etiology , Animals , Drug Resistance, Neoplasm , Humans , Neoplasms, Germ Cell and Embryonal/drug therapy , Testicular Neoplasms/drug therapy
2.
Oncogene ; 35(5): 662-9, 2016 Feb 04.
Article in English | MEDLINE | ID: mdl-25915840

ABSTRACT

Cells are under constant attack from genotoxins and rely on a multifaceted DNA damage response (DDR) network to maintain genomic integrity. Central to the DDR are the ATM and ATR kinases, which respond primarily to double-strand DNA breaks (DSBs) and replication stress, respectively. Optimal ATR signaling requires the RAD9A-RAD1-HUS1 (9-1-1) complex, a toroidal clamp that is loaded at damage sites and scaffolds signaling and repair factors. Whereas complete ATR pathway inactivation causes embryonic lethality, partial Hus1 impairment has been accomplished in adult mice using hypomorphic (Hus1(neo)) and null (Hus1(Δ1)) Hus1 alleles, and here we use this system to define the tissue- and cell type-specific actions of the HUS1-mediated DDR in vivo. Hus1(neo/Δ1) mice showed hypersensitivity to agents that cause replication stress, including the crosslinking agent mitomycin C (MMC) and the replication inhibitor hydroxyurea, but not the DSB inducer ionizing radiation. Analysis of tissue morphology, genomic instability, cell proliferation and apoptosis revealed that MMC treatment caused severe damage in highly replicating tissues of mice with partial Hus1 inactivation. The role of the 9-1-1 complex in responding to MMC was partially ATR-independent, as a HUS1 mutant that was proficient for ATR-induced checkpoint kinase 1 phosphorylation nevertheless conferred MMC hypersensitivity. To assess the interplay between the ATM and ATR pathways in responding to replication stress in vivo, we used Hus1/Atm double mutant mice. Whereas Hus1(neo/neo) and Atm(-/-) single mutant mice survived low-dose MMC similar to wild-type controls, Hus1(neo/neo)Atm(-/-) double mutants showed striking MMC hypersensitivity, consistent with a model in which MMC exposure in the context of Hus1 dysfunction results in DSBs to which the ATM pathway normally responds. This improved understanding of the inter-dependency between two major DDR mechanisms during the response to a conventional chemotherapeutic illustrates how inhibition of checkpoint factors such as HUS1 may be effective for the treatment of ATM-deficient and other cancers.


Subject(s)
Cell Cycle Proteins/metabolism , Mutagens/pharmacology , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Cycle Proteins/genetics , DNA Damage , Mice , Mutagenicity Tests , Signal Transduction
3.
Oncogene ; 34(16): 2011-21, 2015 Apr 16.
Article in English | MEDLINE | ID: mdl-24909171

ABSTRACT

Accurate DNA replication and repair is essential for proper development, growth and tumor-free survival in all multicellular organisms. A key requirement for the maintenance of genomic integrity is the availability of adequate and balanced pools of deoxyribonucleoside triphosphates (dNTPs), the building blocks of DNA. Notably, dNTP pool alterations lead to genomic instability and have been linked to multiple human diseases, including mitochondrial disorders, susceptibility to viral infection and cancer. In this review, we discuss how a key regulator of dNTP biosynthesis in mammals, the enzyme ribonucleotide reductase (RNR), impacts cancer susceptibility and serves as a target for anti-cancer therapies. Because RNR-regulated dNTP production can influence DNA replication fidelity while also supporting genome-protecting DNA repair, RNR has complex and stage-specific roles in carcinogenesis. Nevertheless, cancer cells are dependent on RNR for de novo dNTP biosynthesis. Therefore, elevated RNR expression is a characteristic of many cancers, and an array of mechanistically distinct RNR inhibitors serve as effective agents for cancer treatment. The dNTP metabolism machinery, including RNR, has been exploited for therapeutic benefit for decades and remains an important target for cancer drug development.


Subject(s)
Antineoplastic Agents/therapeutic use , Molecular Targeted Therapy , Neoplasms/drug therapy , Ribonucleotide Reductases/antagonists & inhibitors , Ribonucleotide Reductases/metabolism , Carcinogenesis/metabolism , DNA Repair/genetics , DNA Replication/genetics , Genomic Instability , Humans , Neoplasms/enzymology
4.
Oncogene ; 27(58): 7248-59, 2008 Dec 11.
Article in English | MEDLINE | ID: mdl-18794804

ABSTRACT

The Rad9-Rad1-Hus1 (9-1-1) cell cycle checkpoint complex plays a key role in the DNA damage response. Cells with a defective 9-1-1 complex have been shown to be sensitive to apoptosis induced by certain types of genotoxic stress. However, the mechanism linking the loss of a functional 9-1-1 complex to the cell death machinery has yet to be determined. Here, we report that etoposide treatment dramatically upregulates the BH3-only proteins, Bim and Puma, in Hus1-deficient cells. Inhibition of either Bim or Puma expression in Hus1-knockout cells confers significant resistance to etoposide-induced apoptosis, whereas knockdown of both proteins results in further resistance, suggesting that Bim and Puma cooperate in sensitizing Hus1-deficient cells to etoposide treatment. Moreover, we found that Rad9 collaborates with Bim and Puma to sensitize Hus1-deficient cells to etoposide-induced apoptosis. In response to DNA damage, Rad9 localizes to chromatin in Hus1-wild-type cells, whereas in Hus1-deficient cells, it is predominantly located in the cytoplasm where it binds to Bcl-2. Taken together, these results suggest that loss of Hus1 sensitizes cells to etoposide-induced apoptosis not only by inducing Bim and Puma expressions but also by releasing Rad9 into the cytosol to augment mitochondrial apoptosis.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Apoptosis/drug effects , Cell Cycle Proteins/genetics , Gene Expression Regulation , Membrane Proteins/genetics , Proto-Oncogene Proteins/genetics , Antineoplastic Agents, Phytogenic/pharmacology , Bcl-2-Like Protein 11 , DNA Damage , Etoposide/pharmacology , Humans
5.
Oncogene ; 27(10): 1412-20, 2008 Feb 28.
Article in English | MEDLINE | ID: mdl-17828302

ABSTRACT

While the process of homo-oligomer formation and disassembly into subunits represents a common strategy to regulate protein activity, reports of proteins in which the subunit and homo-oligomer perform independent functions are scarce. Tumorigenesis induced by the adenovirus E4-ORF1 oncoprotein depends on its binding to a select group of cellular PDZ proteins, including MUPP1, MAGI-1, ZO-2 and Dlg1. We report here that in cells E4-ORF1 exists as both a monomer and trimer and that monomers specifically bind and sequester MUPP1, MAGI-1 and ZO-2 within insoluble complexes whereas trimers specifically bind Dlg1 and promote its translocation to the plasma membrane. This work exposes a novel strategy wherein the oligomerization state of a protein not only determines the capacity to bind separate related targets but also couples the interactions to different functional consequences.


Subject(s)
Adenovirus E4 Proteins/chemistry , Adenovirus E4 Proteins/metabolism , Adenoviruses, Human/chemistry , Adenovirus E4 Proteins/genetics , Adenoviruses, Human/enzymology , Adenoviruses, Human/genetics , Amino Acid Sequence , Animals , Humans , Molecular Sequence Data , Protein Binding/physiology , Protein Structure, Quaternary , Protein Structure, Tertiary/physiology , Pyrophosphatases/chemistry , Pyrophosphatases/genetics
6.
Oncogene ; 25(13): 1980-3, 2006 Mar 23.
Article in English | MEDLINE | ID: mdl-16278671

ABSTRACT

Mammalian Hus1 plays an important role in maintaining genomic integrity. Cells lacking mouse Hus1 are hypersensitive to DNA damage inducers including UV and camptothecin (CPT). By using clonogenic assay, we show here that Hus1 deficient mouse cells are hypersensitive to ionizing radiation (IR) compared with their Hus1-positive counterparts. However, these cells show similar induction levels and similar rejoining rates of DNA double strand breaks (DSBs) following IR, indicating that the effect of Hus1 on cell radiosensitivity is independent of nonhomologous end-joining (NHEJ). By combining an I-SceI-induced-DNA DSBs system and a siRNA approach, we also show that knocking down Hus1 decreases the efficiency of homologous recombination repair (HRR), which is associated with the cellular sensitivity to IR-induced killing. Together, these results indicate that the role of Hus1 affecting the sensitivity of cells to IR-induced killing is independent of NHEJ but might be linked to HRR.


Subject(s)
Cell Cycle Proteins/physiology , DNA Repair , Radiation, Ionizing , Animals , Cell Cycle Proteins/genetics , Cell Line , DNA Damage , Mice , Radiation Tolerance
7.
EMBO J ; 20(20): 5578-86, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11598001

ABSTRACT

Adenovirus type 9 (Ad9) is distinct among human adenoviruses because it elicits solely mammary tumors in animals and its primary oncogenic determinant is the E4 region-encoded ORF1 (E4-ORF1) protein. We report here that the PDZ domain-containing protein ZO-2, which is a candidate tumor suppressor protein, is a cellular target for tumorigenic Ad9 E4-ORF1 but not for non-tumorigenic wild-type E4-ORF1 proteins encoded by adenovirus types 5 and 12. Complex formation was mediated by the C-terminal PDZ domain-binding motif of Ad9 E4- ORF1 and the first PDZ domain of ZO-2, and in cells this interaction resulted in aberrant sequestration of ZO-2 within the cytoplasm. Furthermore, transformation-defective Ad9 E4-ORF1 mutants exhibited impaired binding to and sequestration of ZO-2 in cells, and overexpression of wild-type ZO-2, but not mutant ZO-2 lacking the second and third PDZ domains, interfered with Ad9 E4-ORF1-induced focus formation. Our results suggest that the select capacity to complex with the candidate tumor suppressor protein ZO-2 is key to defining the unique transforming and tumorigenic properties of the Ad9 E4-ORF1 oncoprotein.


Subject(s)
Adenovirus E4 Proteins/metabolism , Adenoviruses, Human/pathogenicity , Cell Transformation, Viral/genetics , Mammary Neoplasms, Experimental/virology , Membrane Proteins/antagonists & inhibitors , Adenovirus E4 Proteins/chemistry , Adenovirus E4 Proteins/genetics , Adenoviruses, Human/genetics , Animals , COS Cells , Cell Compartmentation , Cell Line , Chlorocebus aethiops , Cytoplasm/metabolism , Fibroblasts , Genes, Tumor Suppressor , Humans , Macromolecular Substances , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Multigene Family , Open Reading Frames , Protein Binding , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Structure, Tertiary , Rats , Recombinant Fusion Proteins/physiology , Transfection , Zonula Occludens-2 Protein
8.
Genes Dev ; 14(15): 1886-98, 2000 Aug 01.
Article in English | MEDLINE | ID: mdl-10921903

ABSTRACT

The eukaryotic cell cycle is overseen by regulatory mechanisms, termed checkpoints, that respond to DNA damage, mitotic spindle defects, and errors in the ordering of cell cycle events. The DNA replication and DNA damage cell cycle checkpoints of the fission yeast Schizosaccharomyces pombe require the hus1(+) (hydroxyurea sensitive) gene. To determine the role of the mouse homolog of hus1(+) in murine development and cell cycle checkpoint function, we produced a targeted disruption of mouse Hus1. Inactivation of Hus1 results in mid-gestational embryonic lethality due to widespread apoptosis and defective development of essential extra-embryonic tissues. DNA damage-inducible genes are up-regulated in Hus1-deficient embryos, and primary cells from Hus1-null embryos contain increased spontaneous chromosomal abnormalities, suggesting that loss of Hus1 leads to an accumulation of genome damage. Embryonic fibroblasts lacking Hus1 fail to proliferate in vitro, but inactivation of p21 allows for the continued growth of Hus1-deficient cells. Hus1(-/-)p21(-/-) cells display a unique profile of significantly heightened sensitivity to hydroxyurea, a DNA replication inhibitor, and ultraviolet light, but only slightly increased sensitivity to ionizing radiation. Taken together, these results indicate that mouse Hus1 functions in the maintenance of genomic stability and additionally identify an evolutionarily-conserved role for Hus1 in mediating cellular responses to genotoxins.


Subject(s)
Cell Cycle Proteins/genetics , Chromosome Aberrations , DNA Damage/genetics , Fetal Death/genetics , Nuclear Proteins , Animals , Apoptosis/genetics , Cell Cycle Proteins/metabolism , Cell Division/genetics , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Cyclins/metabolism , DNA Damage/radiation effects , Female , Fibroblasts/radiation effects , Gene Expression Regulation, Developmental , Gene Silencing , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2 , Schizosaccharomyces pombe Proteins , Stress, Physiological , Ultraviolet Rays
10.
Genomics ; 59(1): 32-9, 1999 Jul 01.
Article in English | MEDLINE | ID: mdl-10395797

ABSTRACT

Cell cycle checkpoints are regulatory mechanisms that arrest the cell cycle or initiate programmed cell death when critical events such as DNA replication fail to be completed or when DNA or spindle damage occurs. In fission yeast, cell cycle checkpoint responses to DNA replication blocks and DNA damage require the hus1+ gene. Mammalian homologs of hus1+ were recently identified, and here we report a detailed analysis of mouse Hus1. An approximately 4.2-kb full-length cDNA encoding the 32-kDa mouse Hus1 protein was isolated. The genomic structure and exon-intron boundary sequences of the gene were determined, and mouse Hus1 was found to consist of nine exons. Mouse Hus1 was mapped to the proximal end of chromosome 11 and is therefore a candidate gene for the mouse mutation germ cell deficient, which maps to the same genomic region. Finally, mouse Hus1 was found to be expressed in a variety of adult tissues and at several stages of embryonic development.


Subject(s)
Cell Cycle Proteins/genetics , Cell Cycle/genetics , Genes, Fungal/genetics , Schizosaccharomyces/genetics , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Chromosome Mapping , Crosses, Genetic , DNA, Complementary/chemistry , DNA, Complementary/genetics , DNA, Fungal/chemistry , DNA, Fungal/genetics , Embryo, Mammalian/metabolism , Embryonic and Fetal Development , Female , Gene Expression Regulation, Developmental , Gene Expression Regulation, Fungal , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Molecular Sequence Data , Muridae , RNA, Messenger/genetics , RNA, Messenger/metabolism , Schizosaccharomyces pombe Proteins , Sequence Analysis, DNA , Tissue Distribution
12.
J Virol ; 71(10): 7873-80, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9311876

ABSTRACT

Human adenovirus type 9 (Ad9) is unique among oncogenic adenoviruses in that it elicits exclusively mammary tumors in rats and requires the viral E4 region open reading frame 1 (9ORF1) gene for tumorigenicity. The 9ORF1 oncogenic determinant codes for a 14-kDa transforming protein, and three separate regions of this polypeptide, including one at the extreme C terminus, are necessary for transforming activity. In this study, we investigated whether the 9ORF1 transforming protein interacts with cellular factors. Following incubation with cell extracts, a glutathione S-transferase (GST)-9ORF1 fusion protein associated with several cellular phosphoproteins (p220, p180, p160, p155), whereas GST fusion proteins of transformation-defective 9ORF1 C-terminal mutants did not. Similar interactions requiring the 9ORF1 C terminus were revealed with protein-blotting assays, in which a GST-9ORF1 protein probe reacted specifically with cellular polypeptides having gel mobilities resembling those of the 9ORF1-associated cellular phosphoproteins, as well as with additional cellular polypeptides designated p140/p130. In addition, GST fusion proteins containing 9ORF1 C-terminal fragments associated with some of the 9ORF1-associated cellular polypeptides, as did GST fusion proteins of full-length wild-type Ad5 and Ad12 E4 ORF1 transforming proteins. Significantly, the results of coimmunoprecipitation analyses suggested that the same cellular polypeptides also associate with wild-type but not C-terminal-mutant 9ORF1 proteins in vivo. Together, these findings suggest that the 9ORF1 C terminus, which is essential for transformation, participates in specific and direct binding of the 9ORF1 oncoprotein to multiple cellular polypeptides. We propose that interactions with these cellular factors may be responsible, at least in part, for the transforming activity of the 9ORF1 viral oncoprotein.


Subject(s)
Adenoviridae/genetics , Cell Transformation, Neoplastic , Oncogene Proteins, Viral/metabolism , Adenoviridae/metabolism , Animals , Binding Sites , Cell Line , Glutathione Transferase , Humans , Membrane Proteins/isolation & purification , Membrane Proteins/metabolism , Oncogene Proteins, Viral/biosynthesis , Oncogene Proteins, Viral/isolation & purification , Open Reading Frames , Osteosarcoma , Phosphoproteins/isolation & purification , Phosphoproteins/metabolism , Protein Binding , Rats , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Tumor Cells, Cultured
13.
J Mol Cell Cardiol ; 29(9): 2525-32, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9299375

ABSTRACT

To determine the potential pathophysiologic role of alterations in sarcolemmal Ca2+ transport mechanisms, we investigated the effects of up to 120 min of global ischemia in the rabbit heart on the three major Ca2+ transport proteins in the sarcolemma: the Na+-Ca2+ exchanger, the ATP-dependent Ca2+ pump, and the L-type Ca2+ channel. We purified sarcolemmal vesicles from control rabbit hearts and rabbit hearts made ischaemic for 20, 30, 60, 90, and 120 min. Purification of K+-p-nitrophenylphosphatase activity was about 30-fold compared to the initial homogenate, and was the same for control and ischemic hearts. We measured the initial velocity of Na+-Ca2+ exchange and found no inhibition after 20 min of ischemia, a 22% reduction in Vmax after 30 min of ischemia, and approximately a 50% reduction in Vmax after 60, 90, and 120 min of ischemia. At no time was there any change in the Ca2+ affinity of the Na+-Ca2+ exchanger. Solubilization and reconstitution of the Na+-Ca2+ exchanger into asolectin vesicles restored the velocity to the same level as control reconstituted vesicles after 60 min of ischemia, but not after 90 or 120 min of ischemia. In contrast to Na+-Ca2+ exchange, the initial velocity of the sarcolemmal ATP-dependent Ca2+ pump was unaffected by up to 2 h of ischemia. The number of L-type Ca2+ channels, measured by nitrendipine binding, was reduced by 21% after 60 min of ischemia. Decreased Ca2+ efflux due to direct inhibition of the Na+-Ca2+ exchanger, as well as inhibition by low pH and increased intracellular Na2+ in ischemia, probably contribute to Ca2+ overload and irreversible myocyte injury. Conversely, decreased Ca2+ influx due to decreased availability of L-type Ca2+ channels, as well as decreased capacity for reversed Na+-Ca2+ exchange, could contribute to contractile dysfunction during ischemia and myocardial stunning following reperfusion.


Subject(s)
Calcium-Binding Proteins/metabolism , Calcium/metabolism , Myocardial Ischemia/metabolism , Sarcolemma/metabolism , 4-Nitrophenylphosphatase/metabolism , Adenosine Triphosphate/metabolism , Animals , Biological Transport , Calcium Channels/metabolism , Heart Ventricles/metabolism , Male , Nitrendipine/metabolism , Rabbits , Sodium/metabolism
14.
Proc Natl Acad Sci U S A ; 94(13): 6670-5, 1997 Jun 24.
Article in English | MEDLINE | ID: mdl-9192623

ABSTRACT

The 9ORF1 gene encodes an adenovirus E4 region oncoprotein that requires a C-terminal region for transforming activity. Screening a lambdagt11 cDNA expression library with a 9ORF1 protein probe yielded a novel cellular PDZ domain-containing protein, 9BP-1, which binds to wild-type, but not a transformation-defective, C-terminal, mutant 9ORF1 protein. The fact that PDZ domains complex with specific sequences at the free C-terminal end of some proteins led to the recognition that the 9ORF1 C-terminal region contained such a consensus-binding motif. This discovery prompted investigations into whether the 9ORF1 protein associates with additional cellular proteins having PDZ domains. It was found that the 9ORF1 protein interacts directly, in vitro and in vivo, with the PDZ domain-containing protein hDlg/SAP97 (DLG), which is a mammalian homolog of the Drosophila discs large tumor suppressor protein and which also binds the adenomatous polyposis coli tumor suppressor protein. Of interest, in forming complexes, the 9ORF1 protein preferentially associated with the second PDZ domain of DLG, similar to adenomatous polyposis coli protein. Human T cell leukemia virus type 1 Tax and most oncogenic human papillomavirus E6 oncoproteins also possessed PDZ domain-binding motifs at their C termini and, significantly, human T cell leukemia virus type 1 Tax and human papillomavirus 18 E6 proteins bound DLG in vitro. Considering the requirement of the 9ORF1 C-terminal region in transformation, these findings suggest that interactions with the cellular factor DLG may contribute to the tumorigenic potentials of several different human virus oncoproteins.


Subject(s)
Oncogene Proteins, Viral/metabolism , Proteins/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Animals , Base Sequence , Binding Sites/genetics , Discs Large Homolog 1 Protein , Drosophila , Guanylate Kinases , Humans , Membrane Proteins , Mice , Molecular Sequence Data , Oncogene Proteins, Viral/genetics , Protein Binding , Proteins/genetics , Sequence Analysis
15.
J Virol ; 71(6): 4385-94, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9151828

ABSTRACT

Human adenovirus type 9 (Ad9) elicits exclusively estrogen-dependent mammary tumors in rats, and an essential oncogenic determinant for this virus is Ad9 E4 open reading frame 1 (9ORF1), which encodes a 125-residue cytoplasmic protein with cellular growth-transforming activity in vitro. In this study, we engineered 48 different mutant 9ORF1 genes in an attempt to identify regions of this viral protein essential for transformation of the established rat embryo fibroblast cell line CREF. In initial assays with CREF cells, 17 of the 48 mutant 9ORF1 genes proved to be severely defective for generating transformed foci but only 7 of these defective genes expressed detectable amounts of protein. To further examine the defects of the seven mutant proteins, we selected individual cell pools of stable CREF transformants for the wild-type and mutant 9ORF1 genes. Compared to cell pools expressing the wild-type 9ORF1 protein, most cell pools expressing mutant proteins displayed decreased growth in soft agar, and all generated significantly smaller tumors in syngeneic animals. The altered amino acid residues of the seven mutant 9ORF1 polypeptides clustered within three separate regions referred to as region I (residues 34 to 41), region II (residues 89 to 91), and C-terminal region III (residues 122 to 125). By using indirect immunofluorescence, we also assessed whether the mutant proteins localized properly to the cytoplasm of cells. The region I and region II mutants displayed approximately wild-type subcellular localizations, whereas most region III mutants aberrantly accumulated within the nucleus of cells. In summary, we have identified three 9ORF1 protein regions necessary for cellular transformation and have demonstrated that C-terminal region III sequences significantly influence the proper localization of the 9ORF1 polypeptide in cells.


Subject(s)
Adenoviruses, Human/genetics , Cell Transformation, Viral , Oncogene Proteins, Viral/genetics , Oncogenes , Adenoviruses, Human/pathogenicity , Amino Acid Sequence , Animals , Cells, Cultured , Cytoplasm/metabolism , Fibrosarcoma/microbiology , Genes, Viral , Molecular Sequence Data , Mutagenesis , Rats , Viral Structural Proteins/genetics
16.
J Pers Soc Psychol ; 72(4): 885-91, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9108701

ABSTRACT

Narrative accounts of bereaved partners of men with AIDS provided data regarding caregiving, bereavement, and the period immediately following bereavement. The findings of the Harvard Bereavement Study were used to develop the Bereavement Response Scale I (BR-I), containing 21 items within 6 categories. Total scores were correlated with scores from 4 mood measures taken at the time of bereavement and again 12 months following bereavement. A significant association was found with the Positive States of Mind Scale. Item analysis suggested that a shorter version containing 6 items, the Bereavement Response Scale II (BR-II), might be a more effective predictive instrument. Further development of the BR-I and BR-II is proposed as a means for using narrative data to predict bereavement outcomes.


Subject(s)
Acquired Immunodeficiency Syndrome/psychology , Adaptation, Psychological , Bereavement , Homosexuality, Male/psychology , Personality Inventory/statistics & numerical data , Adult , Follow-Up Studies , Grief , Humans , Male , Psychometrics , Reproducibility of Results
17.
J Virol ; 71(3): 1857-70, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9032316

ABSTRACT

An essential oncogenic determinant of subgroup D human adenovirus type 9 (Ad9), which uniquely elicits estrogen-dependent mammary tumors in rats, is encoded by early region 4 open reading frame 1 (E4 ORF1). Whereas Ad9 E4 ORF1 efficiently induces transformed foci on the established rat embryo fibroblast cell line CREF, the related subgroup A Ad12 and subgroup C Ad5 E4 ORF1s do not (R. T. Javier, J. Virol. 68:3917-3924, 1994). In this study, we found that the lack of transforming activity associated with non-subgroup D adenovirus E4 ORF1s in CREF cells correlated with significantly reduced protein levels compared to Ad9 E4 ORF1 in these cells. In the human cell line TE85, however, the non-subgroup D adenovirus E4 ORF1s produced protein levels higher than those seen in CREF cells as well as transforming activities similar to that of Ad9 E4 ORF1, suggesting that all adenovirus E4 ORF1 polypeptides possess comparable cellular growth-transforming activities. In addition, searches for known proteins related to these novel viral transforming proteins revealed that the E4 ORF1 proteins had weak sequence similarity, over the entire length of the E4 ORF1 polypeptides, with a variety of organismal and viral dUTP pyrophosphatase (dUTPase) enzymes. Even though adenovirus E4 ORF1 proteins lacked conserved protein motifs of dUTPase enzymes or detectable enzymatic activity, E4 ORF1 and dUTPase proteins were predicted to possess strikingly similar secondary structure arrangements. It was also established that an avian adenovirus protein, encoded within a genomic location analogous to that of the human adenovirus E4 ORF1s, was a genuine dUTPase enzyme. Although no functional similarity was found for the E4 ORF1 and dUTPase proteins, we propose that human adenovirus E4 ORF1 genes have evolved from an ancestral adenovirus dUTPase and, from this structural framework, developed novel transforming properties.


Subject(s)
Adenovirus E4 Proteins/genetics , Adenoviruses, Human/metabolism , Genes, Viral , Open Reading Frames , Pyrophosphatases/genetics , Transforming Growth Factors/genetics , Adenovirus E4 Proteins/chemistry , Adenovirus E4 Proteins/classification , Adenovirus E4 Proteins/metabolism , Adenoviruses, Human/genetics , Amino Acid Sequence , Animals , Cell Line , Evolution, Molecular , Humans , Molecular Sequence Data , Phylogeny , Pyrophosphatases/chemistry , Pyrophosphatases/classification , Sequence Homology, Amino Acid , Transforming Growth Factors/chemistry , Transforming Growth Factors/classification , Transforming Growth Factors/metabolism
18.
J Virol ; 70(2): 862-72, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8551625

ABSTRACT

The induction of estrogen-dependent rat mammary tumors by human adenovirus type 9 (Ad9) requires the Ad9 E4 open reading frame 1 (9ORF1) protein, which alone can transform that rat embryo fibroblast cell line CREF in vitro. In the present study, independent pools of both 9ORF1-expressing and control CREF cells were generated by selection with G418 and compared with respect to transformed properties. Indirect immunofluorescence analyses revealed that more than 99% of the cells that made up the 9ORF1-transfected pools expressed 9ORF1 protein and, together with confocal laser scanning microscopy, indicated that this E4 protein was located predominantly within the cytoplasm of cells. With regard to transformation, cells of the 9ORF1-expressing pools differed from those of control pools by forming foci, displaying morphological alterations, growing more efficiently in soft agar, and reaching higher saturation densities. Following injection into immunocompetent syngeneic rats, the 9ORF1-expressing pool cells exhibited greatly enhanced oncogenicity compared with control pool cells. These results show that 9ORF1 protein (i) localizes predominantly within the cytoplasm, (ii) confers multiple general transformed characteristics to CREF cells in vitro, and (iii) increases the tumorigenic properties of these cells in vivo.


Subject(s)
Adenovirus E4 Proteins/physiology , Adenoviruses, Human/physiology , Cell Transformation, Neoplastic , Cell Transformation, Viral , Open Reading Frames , Adenovirus E4 Proteins/genetics , Adenoviruses, Human/genetics , Animals , Base Sequence , Cell Adhesion , Cell Count , Cell Line , DNA, Viral , Humans , Molecular Sequence Data , Neoplasms, Experimental/etiology , Rats
19.
Oncogene ; 8(9): 2457-67, 1993 Sep.
Article in English | MEDLINE | ID: mdl-8103211

ABSTRACT

Fibroblast cultures were derived from mouse embryos containing either one (p53+/-) or two (p53-/-) inactivated p53 alleles and compared to normal embryo fibroblasts for a number of growth parameters. Early passage p53-deficient embryo fibroblasts (p53-/-) divided faster than normal embryo fibroblasts, achieved higher confluent densities, and had a higher fraction of division-competent cells under conditions of low cell density. Flow cytometry studies of early passage embryo fibroblasts showed that the percent of p53-deficient cells in G0/G1 was lower than in normal cells, consistent with the argument that p53 mediates a G1 block. When p53-deficient and normal cells were passaged for long periods of time, the homozygote (p53-/-) fibroblasts grew at a high rate for over 50 passages and never entered a non-growing senescent phase characteristic of the heterozygote (p53+/-) and normal (p53+/+) cells. The p53-deficient fibroblasts were genetically unstable during passaging, with the p53-/- cells showing a high degree of aneuploidy and the p53+/- cells displaying a moderate level of chromosomal abnormalities by passage 25. Surprisingly, the heterozygote cells lost their single wild type allele very early during culturing and in spite of this loss most heterozygote lines entered into senescence. We conclude that the loss of p53 by itself is insufficient to confer immortality on a cell, but does confer a growth advantage. Taken together, the findings confirm that the absence of p53 promotes genomic instability, which in turn may result in genetic alterations which directly produce immortality.


Subject(s)
Cell Cycle , Genes, p53 , Tumor Suppressor Protein p53/deficiency , Animals , Base Sequence , Creatine Kinase/genetics , Fibroblasts/cytology , Gene Expression , Karyotyping , Mice/embryology , Molecular Sequence Data , Nuclear Proteins/genetics , Oligodeoxyribonucleotides/chemistry , Proliferating Cell Nuclear Antigen , RNA, Messenger/genetics
20.
Am J Physiol ; 260(3 Pt 1): C433-8, 1991 Mar.
Article in English | MEDLINE | ID: mdl-2003570

ABSTRACT

Lysophosphoglyceride accumulation in ischemic myocardium has been hypothesized to be a mechanism for altered sarcolemmal properties that underlie electrophysiological changes and Ca2+ accumulation in ischemia. We find that in vitro application of lysophosphatidylcholine to normal canine sarcolemmal vesicles at a concentration of 0.3 mumol/mg sarcolemmal protein inhibits Na(+)-Ca2+ exchange. Both maximum velocity (Vmax) for Ca2+ transport and Ca2+ affinity are reduced by lysophosphatidylcholine, whereas in ischemia only Vmax is reduced [M. M. Bersohn, K. D. Philipson, and J. Y. Fukushima. Am. J. Physiol. 242 (Cell Physiol. 11): C288-C295, 1982]. This amount of lysophosphatidylcholine does not affect sarcolemmal passive permeability to either Ca2+ or Na+. Treatment of sarcolemma with phospholipase A2 sufficient to inhibit Na(+)-Ca2+ exchange velocity by 50% causes large increases in sarcolemmal lysophosphatidylcholine and lysophosphatidylethanolamine. On the other hand, 1 h of ischemia in rabbit hearts does not affect sarcolemmal phospholipid composition. Thus, although in vitro treatment with lysophosphatidylcholine or phospholipase A2 has profound effects on sarcolemmal properties, sarcolemmal accumulation of lysophosphatidylcholine cannot account for the effects of ischemia as measured in highly purified sarcolemmal vesicles from ischemic hearts.


Subject(s)
Calcium/metabolism , Carrier Proteins/metabolism , Coronary Disease/metabolism , Lysophosphatidylcholines/pharmacology , Myocardium/metabolism , Sarcolemma/metabolism , Sodium/metabolism , Animals , Dogs , Kinetics , Membrane Lipids/analysis , Phospholipases A , Phospholipases A2 , Phospholipids/analysis , Sarcolemma/chemistry , Sarcolemma/drug effects , Sodium-Calcium Exchanger
SELECTION OF CITATIONS
SEARCH DETAIL
...