Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 3475, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38658552

ABSTRACT

Somatic copy number alterations (SCNAs) are pervasive in advanced human cancers, but their prevalence and spatial distribution in early-stage, localized tumors and their surrounding normal tissues are poorly characterized. Here, we perform multi-region, single-cell DNA sequencing to characterize the SCNA landscape across tumor-rich and normal tissue in two male patients with localized prostate cancer. We identify two distinct karyotypes: 'pseudo-diploid' cells harboring few SCNAs and highly aneuploid cells. Pseudo-diploid cells form numerous small-sized subclones ranging from highly spatially localized to broadly spread subclones. In contrast, aneuploid cells do not form subclones and are detected throughout the prostate, including normal tissue regions. Highly localized pseudo-diploid subclones are confined within tumor-rich regions and carry deletions in multiple tumor-suppressor genes. Our study reveals that SCNAs are widespread in normal and tumor regions across the prostate in localized prostate cancer patients and suggests that a subset of pseudo-diploid cells drive tumorigenesis in the aging prostate.


Subject(s)
DNA Copy Number Variations , Prostatic Neoplasms , Single-Cell Analysis , Humans , Male , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Aneuploidy , Prostate/pathology , Prostate/metabolism , Clone Cells , Diploidy , Aged
2.
IEEE Int Conf Rehabil Robot ; 2023: 1-6, 2023 09.
Article in English | MEDLINE | ID: mdl-37941215

ABSTRACT

In this paper, we propose a task-generic learning-based model for the control of a powered ankle exoskeleton. In contrast to the traditional state machine-based control approaches that hard codes the transition heuristics for the different states and motion conditions during gait, we propose to learn the finer constraints of gait from multiple demonstrations of human gait. We validate our proposed approach on a dataset of ten subjects walking on various inclines and at multiple speeds. We deploy our model on an ankle exoskeleton, and conduct user studies on able-bodied subjects who perform gait scenarios across varying speeds and inclines. We conduct multiple online experiments to validate our learning-based approach for different motion conditions, e.g., normal walking, walking at different speeds and inclines, turns, cross-overs with variable speed and cadence, walking on a treadmill as well as on level ground. We find that our proposed learning-based model has the capability to extrapolate its learned decision rules to support untrained gait conditions, for, e.g., walking at higher speeds and inclines not seen during training. The subjects were able to adapt to the different gait scenarios comfortably without loss of stability.


Subject(s)
Bionics , Gait , Humans , Biomechanical Phenomena , Walking , Lower Extremity
3.
Nat Commun ; 14(1): 509, 2023 01 31.
Article in English | MEDLINE | ID: mdl-36720873

ABSTRACT

Spatially resolved transcriptomics has enabled precise genome-wide mRNA expression profiling within tissue sections. The performance of methods targeting the polyA tails of mRNA relies on the availability of specimens with high RNA quality. Moreover, the high cost of currently available spatial resolved transcriptomics assays requires a careful sample screening process to increase the chance of obtaining high-quality data. Indeed, the upfront analysis of RNA quality can show considerable variability due to sample handling, storage, and/or intrinsic factors. We present RNA-Rescue Spatial Transcriptomics (RRST), a workflow designed to improve mRNA recovery from fresh frozen specimens with moderate to low RNA quality. First, we provide a benchmark of RRST against the standard Visium spatial gene expression protocol on high RNA quality samples represented by mouse brain and prostate cancer samples. Then, we test the RRST protocol on tissue sections collected from five challenging tissue types, including human lung, colon, small intestine, pediatric brain tumor, and mouse bone/cartilage. In total, we analyze 52 tissue sections and demonstrate that RRST is a versatile, powerful, and reproducible protocol for fresh frozen specimens of different qualities and origins.


Subject(s)
RNA , Transcriptome , Child , Male , Humans , Animals , Mice , Transcriptome/genetics , RNA, Messenger , Benchmarking , Biological Assay
4.
Nat Commun ; 13(1): 5475, 2022 09 17.
Article in English | MEDLINE | ID: mdl-36115838

ABSTRACT

The molecular mechanisms underlying lethal castration-resistant prostate cancer remain poorly understood, with intratumoral heterogeneity a likely contributing factor. To examine the temporal aspects of resistance, we analyze tumor heterogeneity in needle biopsies collected before and after treatment with androgen deprivation therapy. By doing so, we are able to couple clinical responsiveness and morphological information such as Gleason score to transcriptome-wide data. Our data-driven analysis of transcriptomes identifies several distinct intratumoral cell populations, characterized by their unique gene expression profiles. Certain cell populations present before treatment exhibit gene expression profiles that match those of resistant tumor cell clusters, present after treatment. We confirm that these clusters are resistant by the localization of active androgen receptors to the nuclei in cancer cells post-treatment. Our data also demonstrates that most stromal cells adjacent to resistant clusters do not express the androgen receptor, and we identify differentially expressed genes for these cells. Altogether, this study shows the potential to increase the power in predicting resistant tumors.


Subject(s)
Prostatic Neoplasms , Receptors, Androgen , Androgen Antagonists/pharmacology , Androgen Antagonists/therapeutic use , Androgens/metabolism , Clone Cells/metabolism , Humans , Male , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Spatio-Temporal Analysis
5.
Nature ; 608(7922): 360-367, 2022 08.
Article in English | MEDLINE | ID: mdl-35948708

ABSTRACT

Defining the transition from benign to malignant tissue is fundamental to improving early diagnosis of cancer1. Here we use a systematic approach to study spatial genome integrity in situ and describe previously unidentified clonal relationships. We used spatially resolved transcriptomics2 to infer spatial copy number variations in >120,000 regions across multiple organs, in benign and malignant tissues. We demonstrate that genome-wide copy number variation reveals distinct clonal patterns within tumours and in nearby benign tissue using an organ-wide approach focused on the prostate. Our results suggest a model for how genomic instability arises in histologically benign tissue that may represent early events in cancer evolution. We highlight the power of capturing the molecular and spatial continuums in a tissue context and challenge the rationale for treatment paradigms, including focal therapy.


Subject(s)
Clone Cells , DNA Copy Number Variations , Genomic Instability , Neoplasms , Spatial Analysis , Clone Cells/metabolism , Clone Cells/pathology , DNA Copy Number Variations/genetics , Early Detection of Cancer , Genome, Human , Genomic Instability/genetics , Genomics , Humans , Male , Models, Biological , Neoplasms/genetics , Neoplasms/pathology , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Transcriptome/genetics
7.
Cell Genom ; 1(3): 100065, 2021 Dec 08.
Article in English | MEDLINE | ID: mdl-36776149

ABSTRACT

Formalin-fixed paraffin embedding (FFPE) is the most widespread long-term tissue preservation approach. Here, we report a procedure to perform genome-wide spatial analysis of mRNA in FFPE-fixed tissue sections, using well-established, commercially available methods for imaging and spatial barcoding using slides spotted with barcoded oligo(dT) probes to capture the 3' end of mRNA molecules in tissue sections. We applied this method for expression profiling and cell type mapping in coronal sections from the mouse brain to demonstrate the method's capability to delineate anatomical regions from a molecular perspective. We also profiled the spatial composition of transcriptomic signatures in two ovarian carcinosarcoma samples, exemplifying the method's potential to elucidate molecular mechanisms in heterogeneous clinical samples. Finally, we demonstrate the applicability of the assay to characterize human lung and kidney organoids and a human lung biopsy specimen infected with SARS-CoV-2. We anticipate that genome-wide spatial gene expression profiling in FFPE biospecimens will be used for retrospective analysis of biobank samples, which will facilitate longitudinal studies of biological processes and biomarker discovery.

8.
Nat Commun ; 9(1): 2419, 2018 06 20.
Article in English | MEDLINE | ID: mdl-29925878

ABSTRACT

Intra-tumor heterogeneity is one of the biggest challenges in cancer treatment today. Here we investigate tissue-wide gene expression heterogeneity throughout a multifocal prostate cancer using the spatial transcriptomics (ST) technology. Utilizing a novel approach for deconvolution, we analyze the transcriptomes of nearly 6750 tissue regions and extract distinct expression profiles for the different tissue components, such as stroma, normal and PIN glands, immune cells and cancer. We distinguish healthy and diseased areas and thereby provide insight into gene expression changes during the progression of prostate cancer. Compared to pathologist annotations, we delineate the extent of cancer foci more accurately, interestingly without link to histological changes. We identify gene expression gradients in stroma adjacent to tumor regions that allow for re-stratification of the tumor microenvironment. The establishment of these profiles is the first step towards an unbiased view of prostate cancer and can serve as a dictionary for future studies.


Subject(s)
Adenocarcinoma/genetics , Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/genetics , Transcriptome/genetics , Adenocarcinoma/pathology , Adenocarcinoma/surgery , Computational Biology , Disease Progression , Gene Expression Profiling , Humans , Male , Prostate/cytology , Prostate/pathology , Prostate/surgery , Prostatectomy , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery , RNA, Messenger/genetics , Stromal Cells/pathology , Tumor Microenvironment/genetics
9.
Nat Commun ; 8(1): 374, 2017 08 29.
Article in English | MEDLINE | ID: mdl-28851861

ABSTRACT

Emerging data demonstrate homologous recombination (HR) defects in castration-resistant prostate cancers, rendering these tumours sensitive to PARP inhibition. Here we demonstrate a direct requirement for the androgen receptor (AR) to maintain HR gene expression and HR activity in prostate cancer. We show that PARP-mediated repair pathways are upregulated in prostate cancer following androgen-deprivation therapy (ADT). Furthermore, upregulation of PARP activity is essential for the survival of prostate cancer cells and we demonstrate a synthetic lethality between ADT and PARP inhibition in vivo. Our data suggest that ADT can functionally impair HR prior to the development of castration resistance and that, this potentially could be exploited therapeutically using PARP inhibitors in combination with androgen-deprivation therapy upfront in advanced or high-risk prostate cancer.Tumours with homologous recombination (HR) defects become sensitive to PARPi. Here, the authors show that androgen receptor (AR) regulates HR and AR inhibition activates the PARP pathway in vivo, thus inhibition of both AR and PARP is required for effective treatment of high risk prostate cancer.


Subject(s)
Collagen Type XI/metabolism , Prostatic Neoplasms, Castration-Resistant/genetics , Receptors, Androgen/metabolism , Synthetic Lethal Mutations , Collagen Type XI/genetics , Homologous Recombination , Humans , Male , Prostatic Neoplasms, Castration-Resistant/enzymology , Prostatic Neoplasms, Castration-Resistant/metabolism , Receptors, Androgen/genetics , Signal Transduction
12.
Cell Rep ; 14(2): 298-309, 2016 Jan 12.
Article in English | MEDLINE | ID: mdl-26748709

ABSTRACT

ATR and CHK1 maintain cancer cell survival under replication stress and inhibitors of both kinases are currently undergoing clinical trials. As ATR activity is increased after CHK1 inhibition, we hypothesized that this may indicate an increased reliance on ATR for survival. Indeed, we observe that replication stress induced by the CHK1 inhibitor AZD7762 results in replication catastrophe and apoptosis, when combined with the ATR inhibitor VE-821 specifically in cancer cells. Combined treatment with ATR and CHK1 inhibitors leads to replication fork arrest, ssDNA accumulation, replication collapse, and synergistic cell death in cancer cells in vitro and in vivo. Inhibition of CDK reversed replication stress and synthetic lethality, demonstrating that regulation of origin firing by ATR and CHK1 explains the synthetic lethality. In conclusion, this study exemplifies cancer-specific synthetic lethality between two proteins in the same pathway and raises the prospect of combining ATR and CHK1 inhibitors as promising cancer therapy.


Subject(s)
Protein Kinases/genetics , Apoptosis , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line, Tumor , Checkpoint Kinase 1 , DNA Damage , Humans , Protein Kinases/metabolism
13.
Sci Transl Med ; 7(312): 312re11, 2015 Nov 04.
Article in English | MEDLINE | ID: mdl-26537259

ABSTRACT

Chemical castration improves responses to radiotherapy in prostate cancer, but the mechanism is unknown. We hypothesized that this radiosensitization is caused by castration-mediated down-regulation of nonhomologous end joining (NHEJ) repair of DNA double-strand breaks (DSBs). To test this, we enrolled 48 patients with localized prostate cancer in two arms of the study: either radiotherapy first or radiotherapy after neoadjuvant castration treatment. We biopsied patients at diagnosis and before and after castration and radiotherapy treatments to monitor androgen receptor, NHEJ, and DSB repair in verified cancer tissue. We show that patients receiving neoadjuvant castration treatment before radiotherapy had reduced amounts of the NHEJ protein Ku70, impaired radiotherapy-induced NHEJ activity, and higher amounts of unrepaired DSBs, measured by γ-H2AX foci in cancer tissues. This study demonstrates that chemical castration impairs NHEJ activity in prostate cancer tissue, explaining the improved response of patients with prostate cancer to radiotherapy after chemical castration.


Subject(s)
DNA Breaks, Double-Stranded , Orchiectomy , Prostatic Neoplasms/radiotherapy , Radiation Tolerance , Radiation-Sensitizing Agents/pharmacology , Adult , Aged , Aged, 80 and over , DNA Repair Enzymes/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Histones/genetics , Humans , Male , Middle Aged , Prostatic Neoplasms/genetics , Prostatic Neoplasms/physiopathology , Radiation-Sensitizing Agents/therapeutic use , Receptors, Androgen/genetics , Young Adult
14.
Nature ; 508(7495): 215-21, 2014 Apr 10.
Article in English | MEDLINE | ID: mdl-24695224

ABSTRACT

Cancers have dysfunctional redox regulation resulting in reactive oxygen species production, damaging both DNA and free dNTPs. The MTH1 protein sanitizes oxidized dNTP pools to prevent incorporation of damaged bases during DNA replication. Although MTH1 is non-essential in normal cells, we show that cancer cells require MTH1 activity to avoid incorporation of oxidized dNTPs, resulting in DNA damage and cell death. We validate MTH1 as an anticancer target in vivo and describe small molecules TH287 and TH588 as first-in-class nudix hydrolase family inhibitors that potently and selectively engage and inhibit the MTH1 protein in cells. Protein co-crystal structures demonstrate that the inhibitors bind in the active site of MTH1. The inhibitors cause incorporation of oxidized dNTPs in cancer cells, leading to DNA damage, cytotoxicity and therapeutic responses in patient-derived mouse xenografts. This study exemplifies the non-oncogene addiction concept for anticancer treatment and validates MTH1 as being cancer phenotypic lethal.


Subject(s)
DNA Repair Enzymes/antagonists & inhibitors , Neoplasms/drug therapy , Neoplasms/metabolism , Nucleotides/metabolism , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Animals , Catalytic Domain , Cell Death/drug effects , Cell Survival/drug effects , Crystallization , DNA Damage , DNA Repair Enzymes/chemistry , DNA Repair Enzymes/metabolism , Deoxyguanine Nucleotides/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Female , Humans , Male , Mice , Models, Molecular , Molecular Conformation , Molecular Targeted Therapy , Neoplasms/pathology , Oxidation-Reduction/drug effects , Phosphoric Monoester Hydrolases/chemistry , Phosphoric Monoester Hydrolases/metabolism , Pyrimidines/chemistry , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Pyrophosphatases/antagonists & inhibitors , Reproducibility of Results , Xenograft Model Antitumor Assays , Nudix Hydrolases
15.
Clin Cancer Res ; 19(6): 1547-56, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23349316

ABSTRACT

PURPOSE: Neoadjuvant castration improves response to radiotherapy of prostate cancer. Here, we determine whether castration therapy impairs nonhomologous end-joining (NHEJ) repair of DNA double-strand breaks (DSB) by downregulating Ku70 protein expression. EXPERIMENTAL DESIGN: Twenty patients with locally advanced prostate cancer were enrolled, and 6 to 12 needle core biopsy specimens were taken from the prostate of each patient before treatment. Bilateral orchidectomy was conducted in eight patients and 12 patients were treated with a GnRH agonist. After castration, two to four similar biopsies were obtained, and the levels of Ku70 and γ-H2AX foci were determined by immunofluorescence in verified cancer tissues. RESULTS: We observed that the androgen receptor binds directly to Ku70 in prostate tissue. We also found a reduction of the Ku70 protein levels in the cell nuclei in 12 of 14 patients (P < 0.001) after castration. The reduction in Ku70 expression correlated significantly with decreased serum prostate-specific antigen (PSA) levels after castration, suggesting that androgen receptor activity regulates Ku70 protein levels in prostate cancer tissue. Furthermore, a significant correlation between the reductions of Ku70 after castration versus changes induced of castration of γ-H2AX foci could be seen implicating a functional linkage of decreased Ku70 levels and impaired DNA repair. CONCLUSIONS: Castration therapy results in decreased levels of the Ku70 protein in prostate cancer cells. Because the Ku70 protein is essential for the NHEJ repair of DSBs and its downregulation impairs DNA repair, this offers a possible explanation for the increased radiosensitivity of prostate cancer cells following castration.


Subject(s)
Antigens, Nuclear/genetics , Castration , DNA End-Joining Repair/genetics , DNA Repair/genetics , DNA-Binding Proteins/genetics , Prostatic Neoplasms/genetics , Aged , Biopsy, Needle , DNA Breaks, Double-Stranded , Gene Expression Regulation, Neoplastic , Humans , Ku Autoantigen , Male , Middle Aged , Prostate-Specific Antigen/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery , Receptors, Androgen
16.
Carcinogenesis ; 34(2): 325-30, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23125219

ABSTRACT

Although alcohol consumption is related to increased cancer risk, its molecular mechanism remains unclear. Here, we demonstrate that an intake of 10% alcohol for 4 weeks in rats is genotoxic due to induction of micronuclei. Acetaldehyde (AA), the first product of ethanol metabolism, is believed to be responsible for DNA damage induced by alcohol. Here, we observe that AA effectively blocks DNA replication elongation in mammalian cells, resulting in DNA double-strand breaks associated with replication. AA-induced DNA damage sites colocalize with the homologous recombination (HR) repair protein RAD51. HR measured in the hypoxhantineguaninefosforibosyltransferase (HPRT) gene is effectively induced by AA and recombination defective mammalian cells are hypersensitive to AA, clearly demonstrating that HR is essential in the repair of AA-induced DNA damage. Altogether, our data indicate that alcohol genotoxicity related to AA produces replication lesions on DNA triggering HR repair.


Subject(s)
Acetaldehyde/toxicity , Alcohols/toxicity , DNA Breaks, Double-Stranded/drug effects , DNA Replication/drug effects , Recombination, Genetic/drug effects , Recombinational DNA Repair/drug effects , Animals , CHO Cells , Cells, Cultured , Cricetinae , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Genomic Instability/drug effects , Lung/cytology , Lung/drug effects , Lung/metabolism , Male , Micronucleus Tests , Rad51 Recombinase/metabolism , Rats , Rats, Wistar
17.
Nucleic Acids Res ; 40(17): 8440-8, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22753029

ABSTRACT

Ultraviolet (UV)-induced DNA damage causes an efficient block of elongating replication forks. The checkpoint kinase, CHK1 has been shown to stabilize replication forks following hydroxyurea treatment. Therefore, we wanted to test if the increased UV sensitivity caused by the unspecific kinase inhibitor caffeine--inhibiting ATM and ATR amongst other kinases--is explained by inability to activate the CHK1 kinase to stabilize replicative structures. For this, we used cells deficient in polymerase η (Polη), a translesion synthesis polymerase capable of properly bypassing the UV-induced cis-syn TT pyrimidine dimer, which blocks replication. These cells accumulate gaps behind progressing replication forks after UV exposure. We demonstrate that both caffeine and CHK1 inhibition, equally retards continuous replication fork elongation after UV treatment. Interestingly, we found more pronounced UV-sensitization by caffeine than with the CHK1 inhibitor in clonogenic survival experiments. Furthermore, we demonstrate an increased collapse of replicative structures after caffeine treatment, but not after CHK1 inhibition, in UV-irradiated cells. This demonstrates that CHK1 activity is not required for stabilization of gaps induced during replication of UV-damaged DNA. These data suggest that elongation and stabilization of replicative structures at UV-induced DNA damage are distinct mechanisms, and that CHK1 is only involved in replication elongation.


Subject(s)
DNA Damage , DNA Replication , Protein Kinases/metabolism , Ultraviolet Rays , Caffeine/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Line, Transformed , Cell Survival , Checkpoint Kinase 1 , DNA Breaks, Double-Stranded , DNA Replication/drug effects , DNA Replication/radiation effects , DNA-Directed DNA Polymerase/deficiency , Humans , Protein Kinase Inhibitors/pharmacology , Signal Transduction/radiation effects
18.
Carcinogenesis ; 33(11): 2026-34, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22822095

ABSTRACT

DNA polymerase eta (pol η) is the only DNA polymerase causally linked to carcinogenesis in humans. Inherited deficiency of pol η in the variant form of xeroderma pigmentosum (XPV) predisposes to UV-light-induced skin cancer. Pol η-deficient cells demonstrate increased sensitivity to cisplatin and oxaliplatin chemotherapy. We have found that XP30R0 fibroblasts derived from a patient with XPV are more resistant to cell kill by ionising radiation (IR) than the same cells complemented with wild-type pol η. This phenomenon has been confirmed in Burkitt's lymphoma cells, which either expressed wild-type pol η or harboured a pol η deletion. Pol η deficiency was associated with accumulation of cells in S-phase, which persisted after IR. Cells deficient in pol η demonstrated increased homologous recombination (HR)-directed repair of double strand breaks created by IR. Depletion of the HR protein, X-ray repair cross-complementing protein 3 (XRCC3), abrogated the radioresistance observed in pol η-deficient cells as compared with pol η-complemented cells. These findings suggest that HR mediates S-phase-dependent radioresistance associated with pol η deficiency. We propose that pol η protein levels in tumours may potentially be used to identify patients who require treatment with chemo-radiotherapy rather than radiotherapy alone for adequate tumour control.


Subject(s)
DNA-Directed DNA Polymerase/genetics , Homologous Recombination/genetics , Radiation Tolerance/genetics , S Phase/physiology , Apoptosis , Blotting, Western , Cell Proliferation , Cells, Cultured , DNA Damage/radiation effects , DNA Repair/radiation effects , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Directed DNA Polymerase/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/radiation effects , Flow Cytometry , Gamma Rays , Humans , Neoplasms/metabolism , Neoplasms/pathology , Nucleic Acid Synthesis Inhibitors , RNA, Small Interfering/genetics , S Phase/radiation effects , Tumor Stem Cell Assay
19.
Int J Radiat Oncol Biol Phys ; 82(3): 1243-8, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22284032

ABSTRACT

BACKGROUND AND PURPOSE: Neoadjuvant androgen deprivation in combination with radiotherapy of prostate cancer is used to improve radioresponsiveness and local tumor control. Currently, the underlying mechanism is not well understood. Because hypoxia causes resistance to radiotherapy, we wanted to test whether castration affects the degree of hypoxia in prostate cancer. METHODS AND MATERIALS: In 14 patients with locally advanced prostate cancer, six to 12 prostatic needle core biopsy specimens were taken prior to castration therapy. Bilateral orchidectomy was performed in 7 patients, and 7 were treated with a GnRH-agonist (leuprorelin). After castrationm two to four prostatic core biopsy specimens were taken, and the level of hypoxia-inducible factor-1α (HIF-1α) in cancer was determined by immunofluorescence. RESULTS: Among biopsy specimens taken before castration, strong HIF-1α expression (mean intensity above 30) was shown in 5 patients, weak expression (mean intensity 10-30) in 3 patients, and background levels of HIF-1α (mean intensity 0-10) in 6 patients. Downregulation of HIF-1α expression after castration was observed in all 5 patients with strong HIF-1α precastration expression. HIF-1α expression was also reduced in 2 of 3 patients with weak HIF-1α precastration expression. CONCLUSIONS: Our data suggest that neoadjuvant castration decreases tumor cell hypoxia in prostate cancer, which may explain increased radiosensitivity after castration.


Subject(s)
Cell Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/analysis , Orchiectomy/methods , Prostatic Neoplasms/chemistry , Prostatic Neoplasms/surgery , Aged , Aged, 80 and over , Androgen Antagonists/therapeutic use , Biomarkers/analysis , Biomarkers/metabolism , Biopsy, Needle , Down-Regulation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Leuprolide/therapeutic use , Male , Middle Aged , Prostate/chemistry , Prostate/pathology , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Radiation Tolerance
20.
DNA Repair (Amst) ; 10(9): 961-9, 2011 Sep 05.
Article in English | MEDLINE | ID: mdl-21840775

ABSTRACT

CK2 phosphorylates the scaffold protein XRCC1, which is required for efficient DNA single-strand break (SSB) repair. Here, we express an XRCC1 protein (XRCC1(ckm)) that cannot be phosphorylated by CK2 in XRCC1 mutated EM9 cells and show that the role of this post-translational modification gives distinct phenotypes in SSB repair and base excision repair (BER). Interestingly, we find that fewer SSBs are formed during BER after treatment with the alkylating agent dimethyl sulfate (DMS) in EM9 cells expressing XRCC1(ckm) (CKM cells) or following inhibition with the CK2 inhibitor 2-dimethylamino-4,5,6,7-tetrabromo-1H-benzimidazole (DMAT). We also show that XRCC1(ckm) protein has a higher affinity for DNA than wild type XRCC1 protein and resides in an immobile fraction on DNA, in particular after damage. We propose a model whereby the increased affinity for DNA sequesters XRCC1(ckm) and the repair enzymes associated with it, at the repair site, which retards kinetics of BER. In conclusion, our results indicate that phosphorylation of XRCC1 by CK2 facilitates the BER incision step, likely by promoting dissociation from DNA.


Subject(s)
Casein Kinase II/metabolism , DNA Breaks, Single-Stranded/drug effects , DNA Repair , DNA-Binding Proteins/metabolism , DNA/metabolism , Alkylating Agents/pharmacology , Animals , CHO Cells , Cell Survival/genetics , Cricetinae , Cricetulus , DNA/drug effects , DNA-Binding Proteins/genetics , Gene Expression Regulation , Mutation Rate , Phosphorylation/drug effects , Rad51 Recombinase/metabolism , Sulfuric Acid Esters/pharmacology , Time Factors , X-ray Repair Cross Complementing Protein 1
SELECTION OF CITATIONS
SEARCH DETAIL
...