Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 2210, 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38472229

ABSTRACT

The ATR-CHK1 DNA damage response pathway becomes activated by the exposure of RPA-coated single-stranded DNA (ssDNA) that forms as an intermediate during DNA damage and repair, and as a part of the replication stress response. Here, we identify ZNF827 as a component of the ATR-CHK1 kinase pathway. We demonstrate that ZNF827 is a ssDNA binding protein that associates with RPA through concurrent binding to ssDNA intermediates. These interactions are dependent on two clusters of C2H2 zinc finger motifs within ZNF827. We find that ZNF827 accumulates at stalled forks and DNA damage sites, where it activates ATR and promotes the engagement of homologous recombination-mediated DNA repair. Additionally, we demonstrate that ZNF827 depletion inhibits replication initiation and sensitizes cancer cells to the topoisomerase inhibitor topotecan, revealing ZNF827 as a therapeutic target within the DNA damage response pathway.


Subject(s)
Protein Kinases , Signal Transduction , Protein Kinases/metabolism , Phosphorylation , Replication Protein A/metabolism , Ataxia Telangiectasia Mutated Proteins/metabolism , DNA-Binding Proteins/metabolism , DNA Replication , DNA Damage , DNA, Single-Stranded , DNA Repair
2.
Nat Commun ; 15(1): 1385, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38360978

ABSTRACT

The Eyes Absent proteins (EYA1-4) are a biochemically unique group of tyrosine phosphatases known to be tumour-promoting across a range of cancer types. To date, the targets of EYA phosphatase activity remain largely uncharacterised. Here, we identify Polo-like kinase 1 (PLK1) as an interactor and phosphatase substrate of EYA4 and EYA1, with pY445 on PLK1 being the primary target site. Dephosphorylation of pY445 in the G2 phase of the cell cycle is required for centrosome maturation, PLK1 localization to centrosomes, and polo-box domain (PBD) dependent interactions between PLK1 and PLK1-activation complexes. Molecular dynamics simulations support the rationale that pY445 confers a structural impairment to PBD-substrate interactions that is relieved by EYA-mediated dephosphorylation. Depletion of EYA4 or EYA1, or chemical inhibition of EYA phosphatase activity, dramatically reduces PLK1 activation, causing mitotic defects and cell death. Overall, we have characterized a phosphotyrosine signalling network governing PLK1 and mitosis.


Subject(s)
Cell Cycle Proteins , Protein Serine-Threonine Kinases , Humans , Protein Serine-Threonine Kinases/metabolism , Cell Cycle Proteins/metabolism , Tyrosine/metabolism , Mitosis , Centrosome/metabolism , Phosphoric Monoester Hydrolases/metabolism , HeLa Cells , Nuclear Proteins/metabolism , Protein Tyrosine Phosphatases/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Trans-Activators/metabolism
3.
iScience ; 27(1): 108655, 2024 Jan 19.
Article in English | MEDLINE | ID: mdl-38213617

ABSTRACT

Alternative lengthening of telomeres (ALT) is a homology-directed repair mechanism that becomes activated in a subset of cancers to maintain telomere length. One of the defining features of ALT cells is the prevalence of extrachromosomal telomeric repeat (ECTR) DNA. Here, we identify that ALT cells engage in two modes of telomere synthesis. Non-productive telomere synthesis occurs during the G2 phase of the cell cycle and is characterized by newly synthesized internal telomeric regions that are not retained in the subsequent G1, coinciding with an induction of ECTR DNA. Productive telomere synthesis occurs specifically during the transition from G2 to mitosis and is defined as the extension of the telomere termini. While many proteins associated with break-induced telomere synthesis function in both non-productive and productive telomere synthesis, POLH specifically promotes productive telomere lengthening and suppresses non-productive telomere synthesis. These findings delineate the mechanism and cell cycle regulation of ALT-mediated telomere synthesis and extension.

4.
Nat Cancer ; 5(1): 47-65, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37904045

ABSTRACT

Telomerase enables replicative immortality in most cancers including acute myeloid leukemia (AML). Imetelstat is a first-in-class telomerase inhibitor with clinical efficacy in myelofibrosis and myelodysplastic syndromes. Here, we develop an AML patient-derived xenograft resource and perform integrated genomics, transcriptomics and lipidomics analyses combined with functional genetics to identify key mediators of imetelstat efficacy. In a randomized phase II-like preclinical trial in patient-derived xenografts, imetelstat effectively diminishes AML burden and preferentially targets subgroups containing mutant NRAS and oxidative stress-associated gene expression signatures. Unbiased, genome-wide CRISPR/Cas9 editing identifies ferroptosis regulators as key mediators of imetelstat efficacy. Imetelstat promotes the formation of polyunsaturated fatty acid-containing phospholipids, causing excessive levels of lipid peroxidation and oxidative stress. Pharmacological inhibition of ferroptosis diminishes imetelstat efficacy. We leverage these mechanistic insights to develop an optimized therapeutic strategy using oxidative stress-inducing chemotherapy to sensitize patient samples to imetelstat causing substantial disease control in AML.


Subject(s)
Ferroptosis , Leukemia, Myeloid, Acute , Oligonucleotides , Telomerase , Humans , Telomerase/genetics , Telomerase/metabolism , Leukemia, Myeloid, Acute/drug therapy , Fatty Acids
5.
Cell Chem Biol ; 30(12): 1652-1665.e6, 2023 12 21.
Article in English | MEDLINE | ID: mdl-38065101

ABSTRACT

The TRF2 shelterin component is an essential regulator of telomere homeostasis and genomic stability. Mutations in the TRF2TRFH domain physically impair t-loop formation and prevent the recruitment of several factors that promote efficient telomere replication, causing telomeric DNA damage. Here, we design, synthesize, and biologically test covalent cyclic peptides that irreversibly target the TRF2TRFH domain. We identify APOD53 as our most promising compound, as it consistently induces a telomeric DNA damage response in cancer cell lines. APOD53 forms a covalent adduct with a reactive cysteine residue present in the TRF2TRFH domain and induces phenotypes consistent with TRF2TRFH domain mutants. These include induction of a telomeric DNA damage response, increased telomeric replication stress, and impaired recruitment of RTEL1 and SLX4 to telomeres. We demonstrate that APOD53 impairs cancer cell growth and find that co-treatment with APOD53 can exacerbate telomere replication stress caused by the G4 stabilizer RHPS4 and low dose aphidicolin (APH).


Subject(s)
Peptides, Cyclic , Telomeric Repeat Binding Protein 2 , DNA Damage , Peptides, Cyclic/pharmacology , Telomere , Telomeric Repeat Binding Protein 2/antagonists & inhibitors , Telomeric Repeat Binding Protein 2/chemistry , Telomeric Repeat Binding Protein 2/genetics , Protein Domains , Cell Line, Tumor
6.
EMBO Mol Med ; 14(10): e15859, 2022 10 10.
Article in English | MEDLINE | ID: mdl-35920001

ABSTRACT

In some types of cancer, telomere length is maintained by the alternative lengthening of telomeres (ALT) mechanism. In many ALT cancers, the α-thalassemia/mental retardation syndrome X-linked (ATRX) gene is mutated leading to the conclusion that the ATRX complex represses ALT. Here, we report that most high-grade pediatric osteosarcomas maintain their telomeres by ALT, and that the majority of these ALT tumors are ATRX wild-type (wt) and instead carry an amplified 17p11.2 chromosomal region containing TOP3A. We found that TOP3A was overexpressed in the ALT-positive ATRX-wt tumors consistent with its amplification. We demonstrated the functional significance of these results by showing that TOP3A overexpression in ALT cancer cells countered ATRX-mediated ALT inhibition and that TOP3A knockdown disrupted the ALT phenotype in ATRX-wt cells. Moreover, we report that TOP3A is required for proper BLM localization and promotes ALT DNA synthesis in ALT cell lines. Collectively, our results identify TOP3A as a major ALT player and potential therapeutic target.


Subject(s)
DNA Topoisomerases, Type I , Osteosarcoma , X-linked Nuclear Protein , DNA , DNA Helicases/genetics , DNA Topoisomerases, Type I/genetics , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Osteosarcoma/genetics , Telomere/genetics , Telomere/metabolism , Telomere Homeostasis , X-linked Nuclear Protein/genetics
7.
Front Cell Dev Biol ; 8: 493, 2020.
Article in English | MEDLINE | ID: mdl-32612998

ABSTRACT

Telomeres are repetitive regions of DNA bound by specialized proteins at the termini of linear chromosomes that prevent the natural chromosome ends from being recognized as DNA double strand breaks. Telomeric DNA is gradually eroded with each round of cell division, resulting in the accumulation of critically short or dysfunctional telomeres that eventually trigger cellular senescence. Consequently, telomere length is indicative of the proliferative capacity of a cell. Multiple methods exist to measure telomere length and telomere content, but a simple and reliable technique to accurately measure individual telomere lengths is currently lacking. We have developed the Telomere length Combing Assay (TCA) to measure telomere length on stretched DNA fibers. We used TCA to measure telomere erosion in primary human fibroblasts, and to detect telomere lengthening in response to activation of telomere maintenance pathways. TCA was also used to accurately measure telomere length in healthy individuals, and to identify critically short telomeres in patients with telomere biology disorders. TCA is performed on isolated DNA, negating the need for cycling cells. TCA is amenable to semi-automated image analysis, and can be fully automated using the Genomic Vision molecular combing platform. This not only precludes sampling bias, but also provides the potential for high-throughput applications and clinical development. TCA is a simple and versatile technique to measure the distribution of individual telomere lengths in a cell population, offering improved accuracy, and more detailed biological insight for telomere length measurement applications.

8.
Curr Opin Genet Dev ; 60: 25-30, 2020 02.
Article in English | MEDLINE | ID: mdl-32119936

ABSTRACT

Telomere maintenance is essential for the continued proliferation of mitotically active cells. Alternative Lengthening of Telomeres (ALT) is a recombination-dependent pathway of telomere maintenance analogous to break-induced replication (BIR) [1] that becomes activated in approximately 10-15% of human cancers. ALT is prevalent in tumours of mesenchymal or neuroepithelial origin, and typically confers a poor prognosis. The aggressiveness and lack of effective strategies to treat these cancers make the ALT pathway a compelling potential therapeutic target to prevent tumour formation and/or the appearance of secondary malignancies after conventional chemotherapy [2]. While the precise initiator of ALT during tumourigenesis remains elusive, substantial progress has been made in interrogating the underlying homology-directed repair mechanisms that converge at telomeres to enable telomere length maintenance. Here, we describe recent advances in our understanding of the ALT mechanism and highlight potential therapeutic targets that may offer future promise in the treatment of ALT cancers.


Subject(s)
Cell Transformation, Neoplastic/pathology , DNA Replication , Neoplasms/genetics , Neoplasms/pathology , Recombination, Genetic , Telomere Homeostasis , Telomere , Cell Transformation, Neoplastic/genetics , Humans
9.
Nat Commun ; 10(1): 5345, 2019 Nov 20.
Article in English | MEDLINE | ID: mdl-31745078

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

10.
Sci Adv ; 5(10): eaav4409, 2019 10.
Article in English | MEDLINE | ID: mdl-31616780

ABSTRACT

Telomerase is a ribonucleoprotein complex that catalyzes addition of telomeric DNA repeats to maintain telomeres in replicating cells. Here, we demonstrate that the telomerase protein hTERT performs an additional role at telomeres that is independent of telomerase catalytic activity yet essential for telomere integrity and cell proliferation. Short-term depletion of endogenous hTERT reduced the levels of heat shock protein 70 (Hsp70-1) and the telomere protective protein Apollo at telomeres, and induced telomere deprotection and cell cycle arrest, in the absence of telomere shortening. Short-term expression of hTERT promoted colocalization of Hsp70-1 with telomeres and Apollo and reduced numbers of deprotected telomeres, in a manner independent of telomerase catalytic activity. These data reveal a previously unidentified noncanonical function of hTERT that promotes formation of a telomere protective complex containing Hsp70-1 and Apollo and is essential for sustained proliferation of telomerase-positive cancer cells, likely contributing to the known cancer-promoting effects of both hTERT and Hsp70-1.


Subject(s)
HSP70 Heat-Shock Proteins/metabolism , Neoplasms/metabolism , Telomerase/metabolism , Telomere/metabolism , Cell Line, Tumor , DNA Damage , Gene Expression Regulation , HSP70 Heat-Shock Proteins/genetics , Humans , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Neoplasms/genetics , Telomerase/genetics
11.
Nat Commun ; 10(1): 2252, 2019 05 28.
Article in English | MEDLINE | ID: mdl-31138797

ABSTRACT

The collapse of stalled replication forks is a major driver of genomic instability. Several committed mechanisms exist to resolve replication stress. These pathways are particularly pertinent at telomeres. Cancer cells that use Alternative Lengthening of Telomeres (ALT) display heightened levels of telomere-specific replication stress, and co-opt stalled replication forks as substrates for break-induced telomere synthesis. FANCM is a DNA translocase that can form independent functional interactions with the BLM-TOP3A-RMI (BTR) complex and the Fanconi anemia (FA) core complex. Here, we demonstrate that FANCM depletion provokes ALT activity, evident by increased break-induced telomere synthesis, and the induction of ALT biomarkers. FANCM-mediated attenuation of ALT requires its inherent DNA translocase activity and interaction with the BTR complex, but does not require the FA core complex, indicative of FANCM functioning to restrain excessive ALT activity by ameliorating replication stress at telomeres. Synthetic inhibition of FANCM-BTR complex formation is selectively toxic to ALT cancer cells.


Subject(s)
Carrier Proteins/metabolism , DNA Helicases/metabolism , DNA Topoisomerases, Type I/metabolism , DNA-Binding Proteins/metabolism , Neoplasms/metabolism , Nuclear Proteins/metabolism , RecQ Helicases/metabolism , Telomere Homeostasis , Telomere/metabolism , Cell Line, Tumor , DNA Replication , HCT116 Cells , HEK293 Cells , HeLa Cells , Humans
12.
J Cell Physiol ; 233(4): 3262-3273, 2018 04.
Article in English | MEDLINE | ID: mdl-28884822

ABSTRACT

RNA-binding proteins (RBP) are important facilitators of post-transcriptional gene regulation. We have previously established that nuclear overexpression of the RBP Musashi-2 (MSI2) during male germ cell maturation is detrimental to sperm cell development and fertility. Herein we determine the genes and pathways impacted by the upregulation of Msi2. Microarray analysis and qPCR confirmed differential gene expression in factors fundamental to the cell cycle, cellular proliferation, and cell death. Similarly, comparative protein expression analysis via iTRAQ, immunoblot, and immunolocalization, identified differential expression and localization of important regulators of transcription, translation, RNA processing, and spermatogenesis. Specifically, the testis-expressed transcription factor, Tbx1, and the piRNA regulator of gamete development, Piwil1, were both found to be targeted for translational repression by MSI2. This study provides key evidence to support a fundamental role for MSI2 in post-transcriptional regulation during male gamete development.


Subject(s)
Argonaute Proteins/metabolism , RNA-Binding Proteins/metabolism , Spermatogenesis , T-Box Domain Proteins/metabolism , Animals , Argonaute Proteins/genetics , Gene Expression Regulation , Male , Mice, Transgenic , Models, Biological , Proteome/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Spermatids/metabolism , Spermatogenesis/genetics , T-Box Domain Proteins/genetics
13.
Trends Genet ; 33(12): 921-932, 2017 12.
Article in English | MEDLINE | ID: mdl-28969871

ABSTRACT

Telomeres shorten during each cellular division, with cumulative attrition resulting in telomeric damage and replicative senescence. Bypass of replicative senescence precipitates catastrophic telomere shortening or crisis, and is characterized by widespread genomic instability. Activation of a telomere maintenance mechanism (TMM) is necessary to stabilise the genome and establish cellular immortality through the reconstitution of telomere capping function. The alternative lengthening of telomeres (ALT) pathway is a TMM frequently activated in tumors of mesenchymal or neuroepithelial origin. ALT is a homology-directed recombination-dependent replication pathway that utilizes telomeric templates for synthesis; however, its precise protein requirements have remained elusive. Recently, several developments have shed light on the DNA repair pathways that become engaged at ALT telomeres, implicating ALT telomeres as DNA repair hot spots. Here, we review recent discoveries regarding the ALT mechanism, and discuss how DNA repair pathways converge to maintain the length and functional integrity of telomeres in ALT cancers.


Subject(s)
DNA Repair/genetics , Telomere Homeostasis/genetics , Telomere/genetics , Animals , DNA Replication/genetics , Homologous Recombination/genetics , Humans
14.
EMBO J ; 36(19): 2907-2919, 2017 10 02.
Article in English | MEDLINE | ID: mdl-28877996

ABSTRACT

Alternative lengthening of telomeres (ALT) is a telomere lengthening pathway that predominates in aggressive tumors of mesenchymal origin; however, the underlying mechanism of telomere synthesis is not fully understood. Here, we show that the BLM-TOP3A-RMI (BTR) dissolvase complex is required for ALT-mediated telomere synthesis. We propose that recombination intermediates formed during strand invasion are processed by the BTR complex, initiating rapid and extensive POLD3-dependent telomere synthesis followed by dissolution, with no overall exchange of telomeric DNA. This process is counteracted by the SLX4-SLX1-ERCC4 complex, which promotes resolution of the recombination intermediate, resulting in telomere exchange in the absence of telomere extension. Our data are consistent with ALT being a conservative DNA replication process, analogous to break-induced replication, which is dependent on BTR and counteracted by SLX4 complex-mediated resolution events.


Subject(s)
DNA Replication/genetics , RecQ Helicases/physiology , Recombinases/physiology , Recombination, Genetic/genetics , Telomere Homeostasis/genetics , Cells, Cultured , DNA Topoisomerases, Type I/metabolism , DNA Topoisomerases, Type I/physiology , DNA-Directed DNA Polymerase/metabolism , DNA-Directed DNA Polymerase/physiology , Humans , Multienzyme Complexes/metabolism , Multienzyme Complexes/physiology , RecQ Helicases/metabolism , Recombinases/metabolism , Telomere/metabolism
15.
Cell Rep ; 19(12): 2544-2556, 2017 06 20.
Article in English | MEDLINE | ID: mdl-28636942

ABSTRACT

Acquisition of replicative immortality is currently regarded as essential for malignant transformation. This is achieved by activating a telomere lengthening mechanism (TLM), either telomerase or alternative lengthening of telomeres, to counter normal telomere attrition. However, a substantial proportion of some cancer types, including glioblastomas, liposarcomas, retinoblastomas, and osteosarcomas, are reportedly TLM-negative. As serial samples of human tumors cannot usually be obtained to monitor telomere length changes, it has previously been impossible to determine whether tumors are truly TLM-deficient, there is a previously unrecognized TLM, or the assay results are false-negative. Here, we show that a subset of high-risk neuroblastomas (with ∼50% 5-year mortality) lacked significant TLM activity. Cancer cells derived from these highly aggressive tumors initially had long telomeres and proliferated for >200 population doublings with ever-shorter telomeres. This indicates that prevention of telomere shortening is not always required for oncogenesis, which has implications for inhibiting TLMs for cancer therapy.


Subject(s)
Cell Proliferation , Telomere Shortening , Cell Line, Tumor , Enzyme Activation , Gene Amplification , Humans , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/genetics , Neuroblastoma/pathology , Telomerase/metabolism
16.
Biol Reprod ; 94(2): 39, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26764348

ABSTRACT

The theory of fetal origins of adult disease was first proposed in 1989, and in the decades since, a wide range of other diseases from obesity to asthma have been found to originate in early development. Because mammalian oocyte development begins in fetal life it has been suggested that environmental and lifestyle factors of the mother could directly impact the fertility of subsequent generations. Cigarette smoke is a known ovotoxicant in active smokers, yet disturbingly 13% of Australian and 12% of US women continue to smoke throughout pregnancy. The focus of our investigation was to characterize the adverse effects of smoking on ovary and oocyte quality in female offspring exposed in utero. Pregnant mice were nasally exposed to cigarette smoke for 12 wk throughout pregnancy/lactation, and ovary and oocyte quality of the F1 (maternal smoke exposed) generation was examined. Neonatal ovaries displayed abnormal somatic cell proliferation and increased apoptosis, leading to a reduction in follicle numbers. Further investigation found that altered somatic cell proliferation and reduced follicle number continued into adulthood; however, apoptosis did not. This reduction in follicles resulted in decreased oocyte numbers, with these oocytes found to have elevated levels of oxidative stress, altered metaphase II spindle, and reduced sperm-egg interaction. These ovarian and oocyte changes ultimately lead to subfertility, with maternal smoke-exposed animals having smaller litters and also taking longer to conceive. In conclusion, our results demonstrate that in utero and lactational exposure to cigarette smoke can have long-lasting effects on the fertility of the next generation of females.


Subject(s)
Fertility/drug effects , Maternal Exposure , Oocytes/drug effects , Ovarian Follicle/drug effects , Ovary/drug effects , Smoke/adverse effects , Animals , Female , Mice , Oocytes/metabolism , Ovarian Follicle/metabolism , Ovary/metabolism , Oxidative Stress/drug effects , Pregnancy , Sperm-Ovum Interactions
17.
Biomolecules ; 5(3): 1228-44, 2015 Jun 26.
Article in English | MEDLINE | ID: mdl-26131972

ABSTRACT

Characterizing the mechanisms underlying follicle development in the ovary is crucial to understanding female fertility and is an area of increasing research interest. The RNA binding protein Musashi is essential for post-transcriptional regulation of oocyte maturation in Xenopus and is expressed during ovarian development in Drosophila. In mammals Musashi is important for spermatogenesis and male fertility, but its role in the ovary has yet to be characterized. In this study we determined the expression of mammalian Musashi proteins MSI1 and MSI2 during mouse folliculogenesis, and through the use of a MSI2-specific knockout mouse model we identified that MSI2 is essential for normal follicle development. Time-course characterization of MSI1 and MSI2 revealed distinct differences in steady-state mRNA levels and protein expression/localization at important developmental time-points during folliculogenesis. Using a gene-trap mouse model that inactivates Msi2, we observed a significant decrease in ovarian mass, and change in follicle-stage composition due to developmental blocking of antral stage follicles and pre-antral follicle loss through atresia. We also confirmed that hormonally stimulated Msi2-deficient mice produce significantly fewer MII oocytes (60.9% less than controls, p < 0.05). Furthermore, the majority of these oocytes are of poor viability (62.2% non-viable/apoptotic, p < 0.05), which causes a reduction in female fertility evidenced by decreased litter size in Msi2-deficient animals (33.1% reduction to controls, p < 0.05). Our findings indicate that MSI1 and MSI2 display distinct expression profiles during mammalian folliculogenesis and that MSI2 is required for pre-antral follicle development.


Subject(s)
Gene Knockout Techniques , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Ovarian Follicle/growth & development , RNA-Binding Proteins/genetics , Animals , Female , Gene Expression Regulation, Developmental , Mice , Ovarian Follicle/metabolism , RNA-Binding Proteins/metabolism
18.
PLoS Genet ; 11(6): e1005286, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26053551

ABSTRACT

The TERT-CLPTM1L region of chromosome 5p15.33 is a multi-cancer susceptibility locus that encodes the reverse transcriptase subunit, hTERT, of the telomerase enzyme. Numerous cancer-associated single-nucleotide polymorphisms (SNPs), including rs10069690, have been identified within the hTERT gene. The minor allele (A) at rs10069690 creates an additional splice donor site in intron 4 of hTERT, and is associated with an elevated risk of multiple cancers including breast and ovarian carcinomas. We previously demonstrated that the presence of this allele resulted in co-production of full length (FL)-hTERT and an alternatively spliced, INS1b, transcript. INS1b does not encode the reverse transcriptase domain required for telomerase enzyme activity, but we show here that INS1b protein retains its ability to bind to the telomerase RNA subunit, hTR. We also show that INS1b expression results in decreased telomerase activity, telomere shortening, and an increased telomere-specific DNA damage response (DDR). We employed antisense oligonucleotides to manipulate endogenous transcript expression in favor of INS1b, which resulted in a decrease in telomerase activity. These data provide the first detailed mechanistic insights into a cancer risk-associated SNP in the hTERT locus, which causes cell type-specific expression of INS1b transcript from the presence of an additional alternative splice site created in intron 4 by the risk allele. We predict that INS1b expression levels cause subtle inadequacies in telomerase-mediated telomere maintenance, resulting in an increased risk of genetic instability and therefore of tumorigenesis.


Subject(s)
Alleles , Breast Neoplasms/genetics , Carcinoma/genetics , Ovarian Neoplasms/genetics , Telomerase/genetics , Alternative Splicing , Female , Genes, Dominant , HEK293 Cells , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , MCF-7 Cells , Polymorphism, Single Nucleotide , Telomerase/metabolism , Telomere Shortening
19.
FASEB J ; 29(7): 2759-68, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25782991

ABSTRACT

Controlled gene regulation during gamete development is vital for maintaining reproductive potential. During the process of gamete development, male germ cells experience extended periods of inactive transcription despite requirements for continued growth and differentiation. Spermatogenesis therefore provides an ideal model to study the effects of posttranscriptional control on gene regulation. During spermatogenesis posttranscriptional regulation is orchestrated by abundantly expressed RNA-binding proteins. One such group of RNA-binding proteins is the Musashi family, previously identified as a critical regulator of testis germ cell development and meiosis in Drosophila and also shown to be vital to sperm development and reproductive potential in the mouse. We focus in depth on the role and function of the vertebrate Musashi ortholog Musashi-1 (MSI1). Through detailed expression studies and utilizing our novel transgenic Msi1 testis-specific overexpression model, we have identified 2 unique RNA-binding targets of MSI1 in spermatogonia, Msi2 and Erh, and have demonstrated a role for MSI1 in translational regulation. We have also provided evidence to suggest that nuclear import protein, IPO5, facilitates the nuclear translocation of MSI1 to the transcriptionally silenced XY chromatin domain in meiotic pachytene spermatocytes, resulting in the release of MSI1 RNA-binding targets. This firmly establishes MSI1 as a master regulator of posttranscriptional control during early spermatogenesis and highlights the significance of the subcellular localization of RNA binding proteins in relation to their function.


Subject(s)
Cell Cycle Proteins/metabolism , Nerve Tissue Proteins/metabolism , RNA-Binding Proteins/metabolism , Spermatogenesis/physiology , Transcription Factors/metabolism , beta Karyopherins/metabolism , Active Transport, Cell Nucleus , Amino Acid Sequence , Animals , Cell Cycle Proteins/genetics , Gene Expression Regulation, Developmental , Male , Mice , Mice, Transgenic , Models, Biological , Molecular Sequence Data , Nerve Tissue Proteins/genetics , RNA-Binding Proteins/genetics , Spermatocytes/metabolism , Spermatogonia/metabolism , Testis/cytology , Testis/growth & development , Testis/metabolism , Transcription Factors/genetics , beta Karyopherins/genetics
20.
Biol Reprod ; 92(1): 27, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25472923

ABSTRACT

Chlamydia trachomatis infections are increasingly prevalent worldwide. Male chlamydial infections are associated with urethritis, epididymitis, and orchitis; however, the role of Chlamydia in prostatitis and male factor infertility remains controversial. Using a model of Chlamydia muridarum infection in male C57BL/6 mice, we investigated the effects of chlamydial infection on spermatogenesis and determined the potential of immune T cells to prevent infection-induced outcomes. Antigen-specific CD4 T cells significantly reduced the infectious burden in the penile urethra, epididymis, and vas deferens. Infection disrupted seminiferous tubules, causing loss of germ cells at 4 and 8 wk after infection, with the most severely affected tubules containing only Sertoli cells. Increased mitotic proliferation, DNA repair, and apoptosis in spermatogonial cells and damaged germ cells were evident in atrophic tubules. Activated caspase 3 (casp3) staining revealed increased (6-fold) numbers of Sertoli cells with abnormal morphology that were casp3 positive in tubules of infected mice, indicating increased levels of apoptosis. Sperm count and motility were both decreased in infected mice, and there was a significant decrease in morphologically normal spermatozoa. Assessment of the spermatogonial stem cell population revealed a decrease in promyelocytic leukemia zinc finger (PLZF)-positive cells in the seminiferous tubules. Interestingly, adoptive transfer of antigen-specific CD4 cells, particularly T-helper 2-like cells, prior to infection prevented these effects in spermatogenesis and Sertoli cells. These data suggest that chlamydial infection adversely affects spermatogenesis and male fertility, and that vaccination can potentially prevent the spread of infection and these adverse outcomes.


Subject(s)
Apoptosis , Bacterial Outer Membrane Proteins/immunology , CD4-Positive T-Lymphocytes/physiology , Chlamydia Infections/immunology , Chlamydia muridarum/immunology , Cytoprotection/immunology , Sertoli Cells/physiology , Spermatozoa/physiology , Animals , Apoptosis/immunology , Chlamydia Infections/pathology , Chlamydia muridarum/pathogenicity , Infertility, Male/microbiology , Male , Mice , Mice, Inbred C57BL , Spermatogenesis/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...