Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
Add more filters










Publication year range
1.
Rozhl Chir ; 103(6): 202-207, 2024.
Article in English | MEDLINE | ID: mdl-38991783

ABSTRACT

Skin substitutes and covers are crucial across surgical disciplines, promoting interdisciplinary collaboration to meet varied clinical needs. While some medical professionals may encounter these products infrequently in their practice, understanding their properties and applications is paramount to provide optimal patient care. In this overview, we aim to provide healthcare professionals with essential information regarding skin substitutes and covers, equipping them with knowledge to navigate their use effectively across different clinical scenarios and to optimize patient outcomes. The speed of progress in tissue engineering and regenerative medicine is notable, driven by collaborative efforts among scientists, engineers, and clinicians. Technological advancements, increased funding, and a deeper understanding of cellular and molecular processes have accelerated research and development. However, challenges remain, such as achieving vascularization in engineered tissues, addressing immune responses, and ensuring long-term functionality of regenerated organs. Despite these hurdles, the field continues to evolve rapidly, offering hope for transformative medical solutions that may redefine the treatment landscape soon. In this article, we review the current selected commercially available epidermal, dermal, and total skin substitutes for wound healing.


Subject(s)
Skin, Artificial , Wound Healing , Humans , Tissue Engineering
2.
Acta Chir Plast ; 65(2): 79-83, 2023.
Article in English | MEDLINE | ID: mdl-37722905

ABSTRACT

Transection of the radial nerve is frequently associated with humeral shaft fractures that are part of a very complex upper extremity injury. In the presented case, a 19-year-old man with a 10-cm radial nerve defect with a need for nerve grafting to recover complete sensory and motor deficit of the radial nerve. In our case, at the same time we provided the tendon transfer of musculus (m.) pronator teres to m. extensor carpi radialis brevis, m. flexor carpi ulnaris to m. extensor digitorum communis, m. palmaris longus to m. extensor pollicis longus, and long sural nerve graft because of an extensive zone of the injury. The assumption was that if these two procedures are performed in one surgery, it will accelerate overall recovery, restore the functionality of the upper limb more quickly, and thus enable a faster recovery.


Subject(s)
Plastic Surgery Procedures , Radial Nerve , Male , Humans , Young Adult , Adult , Radial Nerve/surgery , Forearm , Upper Extremity , Tendons
3.
Acta Chir Plast ; 65(2): 84-85, 2023.
Article in English | MEDLINE | ID: mdl-37722907

Subject(s)
Burns , Child , Humans , Burns/mortality
4.
BMC Cancer ; 22(1): 1298, 2022 Dec 12.
Article in English | MEDLINE | ID: mdl-36503417

ABSTRACT

BACKGROUND: Metaplastic breast cancer (MpBC) is an aggressive subtype of breast carcinoma that is often resistant to conventional chemotherapy. Therefore, novel treatment strategies are urgently needed. Immune check point inhibitors have shown activity in programmed death-ligand 1 (PD-L1) - positive metastatic triple negative breast carcinoma (TNBC), which raises the possibility that immunotherapy may also be effective in MpBC as most of the MpBCs are triple negative. The aim of the present study was to assess genomic instability and immunogenicity in tumor specimens of patients with MpBC. METHODS: A total of 76 patients diagnosed with MpBC over a 15-year period were included in the study. We performed immunohistochemical analyses for tumor cell PD-L1, immune cell PD-L1 and p53 on tissue microarrays (TMAs), analyzed stromal and intratumoral tumor infiltrating lymphocytes (TILs) from hematoxylin and eosin-stained (H&E) slides and scored gamma-H2AX (γH2AX) and phosphorylated-RPA2 (pRPA2) from whole tissue sections. We correlated marker expression with clinicopathologic features and clinical outcome. RESULTS: All tumors expressed γH2AX and pRPA2 with median expressions of 43% and 44%. P53- (68%), tumor cell PD-L1- (59%) and immune cell PD-L1-positivity (62%) were common in MpBCs. Median stromal TIL and intratumoral TIL counts were 5% and 0. The spindle and squamous cell carcinomas expressed the highest levels of PD-L1 and TILs, and carcinoma with mesenchymal differentiation the lowest. CONCLUSIONS: MpBC appears to be an immunogenic cancer with high genomic instability and frequent PD-L1-positivity, implying that check point inhibitors might be effective in MpBC. Expression levels of PD-L1 and TILs varied across different histologic subtypes, suggesting that immunotherapy might be less effective in carcinoma with mesenchymal differentiation.


Subject(s)
Carcinoma, Squamous Cell , Triple Negative Breast Neoplasms , Humans , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Lymphocytes, Tumor-Infiltrating , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Prognosis , Biomarkers, Tumor/metabolism , Triple Negative Breast Neoplasms/therapy , Triple Negative Breast Neoplasms/drug therapy , Carcinoma, Squamous Cell/pathology
5.
Oncogene ; 36(17): 2405-2422, 2017 04 27.
Article in English | MEDLINE | ID: mdl-27841863

ABSTRACT

Chromatin DNA damage response (DDR) is orchestrated by the E3 ubiquitin ligase ring finger protein 168 (RNF168), resulting in ubiquitin-dependent recruitment of DDR factors and tumor suppressors breast cancer 1 (BRCA1) and p53 binding protein 1 (53BP1). This ubiquitin signaling regulates pathway choice for repair of DNA double-strand breaks (DSB), toxic lesions whose frequency increases during tumorigenesis. Recruitment of 53BP1 curbs DNA end resection, thereby limiting homologous recombination (HR) and directing DSB repair toward error-prone non-homologous end joining (NHEJ). Under cancer-associated ubiquitin starvation conditions reflecting endogenous or treatment-evoked proteotoxic stress, the ubiquitin-dependent accrual of 53BP1 and BRCA1 at the DNA damage sites is attenuated or lost. Challenging this current paradigm, here we identified diverse human cancer cell lines that display 53BP1 recruitment to DSB sites even under proteasome inhibitor-induced proteotoxic stress, that is, under substantial depletion of free ubiquitin. We show that central to this unexpected phenotype is overabundance of RNF168 that enables more efficient exploitation of the residual-free ubiquitin. Cells with elevated RNF168 are more resistant to combined treatment by ionizing radiation and proteasome inhibition, suggesting that such aberrant RNF168-mediated signaling might reflect adaptation to chronic proteotoxic and genotoxic stresses experienced by tumor cells. Moreover, the overabundant RNF168 and the ensuing unorthodox recruitment patterns of 53BP1, RIF1 and REV7 (monitored on laser micro-irradiation-induced DNA damage) shift the DSB repair balance from HR toward NHEJ, a scenario accompanied by enhanced chromosomal instability/micronuclei formation and sensitivity under replication stress-inducing treatments with camptothecin or poly(ADP-ribose) polymerase (PARP) inhibitor. Overall, our data suggest that the deregulated RNF168/53BP1 pathway could promote tumorigenesis by selecting for a more robust, better stress-adapted cancer cell phenotype, through altered DNA repair, fueling genomic instability and tumor heterogeneity. Apart from providing insights into cancer (patho)biology, the elevated RNF168, documented here also by immunohistochemistry on human clinical tumor specimens, may impact responses to standard-of-care and some emerging targeted cancer therapies.


Subject(s)
DNA Repair/genetics , Gene Expression Regulation, Neoplastic , Genomic Instability , Homeostasis/drug effects , Homeostasis/genetics , Mutagens/toxicity , Ubiquitin-Protein Ligases/genetics , Amino Acid Motifs , Carcinogenesis/drug effects , Carcinogenesis/genetics , Cell Line, Tumor , DNA Damage , DNA End-Joining Repair/drug effects , DNA End-Joining Repair/genetics , DNA Repair/drug effects , Genomic Instability/drug effects , Humans , Mutation , Phenotype , Protein Transport/drug effects , Protein Transport/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Tumor Suppressor p53-Binding Protein 1/chemistry , Tumor Suppressor p53-Binding Protein 1/genetics , Tumor Suppressor p53-Binding Protein 1/metabolism , Ubiquitin/metabolism
6.
Oncogene ; 34(46): 5699-708, 2015 Nov 12.
Article in English | MEDLINE | ID: mdl-25728682

ABSTRACT

Defining mechanisms that generate intratumour heterogeneity and branched evolution may inspire novel therapeutic approaches to limit tumour diversity and adaptation. SETD2 (Su(var), Enhancer of zeste, Trithorax-domain containing 2) trimethylates histone-3 lysine-36 (H3K36me3) at sites of active transcription and is mutated in diverse tumour types, including clear cell renal carcinomas (ccRCCs). Distinct SETD2 mutations have been identified in spatially separated regions in ccRCC, indicative of intratumour heterogeneity. In this study, we have addressed the consequences of SETD2 loss-of-function through an integrated bioinformatics and functional genomics approach. We find that bi-allelic SETD2 aberrations are not associated with microsatellite instability in ccRCC. SETD2 depletion in ccRCC cells revealed aberrant and reduced nucleosome compaction and chromatin association of the key replication proteins minichromosome maintenance complex component (MCM7) and DNA polymerase δ hindering replication fork progression, and failure to load lens epithelium-derived growth factor and the Rad51 homologous recombination repair factor at DNA breaks. Consistent with these data, we observe chromosomal breakpoint locations are biased away from H3K36me3 sites in SETD2 wild-type ccRCCs relative to tumours with bi-allelic SETD2 aberrations and that H3K36me3-negative ccRCCs display elevated DNA damage in vivo. These data suggest a role for SETD2 in maintaining genome integrity through nucleosome stabilization, suppression of replication stress and the coordination of DNA repair.


Subject(s)
Carcinoma, Renal Cell/genetics , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Kidney Neoplasms/genetics , Mutation , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , DNA Repair , DNA Replication , Genetic Heterogeneity , Histones/metabolism , Humans , Kidney Neoplasms/metabolism , Microsatellite Instability , Nucleosomes/pathology
7.
Cell Death Dis ; 5: e1351, 2014 Jul 24.
Article in English | MEDLINE | ID: mdl-25058425

ABSTRACT

Mammalian cells have mechanisms to counteract the effects of metabolic and exogenous stresses, many of that would be mutagenic if ignored. Damage arising during DNA replication is a major source of mutagenesis. The extent of damage dictates whether cells undergo transient cell cycle arrest and damage repair, senescence or apoptosis. Existing dogma defines these alternative fates as distinct choices. Here we show that immortalised breast epithelial cells are able to survive prolonged S phase arrest and subsequently re-enter cycle after many days of being in an arrested, senescence-like state. Prolonged cell cycle inhibition in fibroblasts induced DNA damage response and cell death. However, in immortalised breast epithelia, efficient S phase arrest minimised chromosome damage and protected sufficient chromatin-bound replication licensing complexes to allow cell cycle re-entry. We propose that our observation could have implications for the design of drug therapies for breast cancer.


Subject(s)
Breast Neoplasms/genetics , Cell Cycle , Cellular Senescence , DNA Replication , Epithelial Cells/cytology , Breast Neoplasms/physiopathology , Cell Line, Tumor , Cell Survival , DNA Damage , Female , Humans , Mammary Glands, Human/cytology , S Phase
8.
Ann Oncol ; 24(11): 2780-5, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23897704

ABSTRACT

BACKGROUND: Defective DNA repair is central to the progression and treatment of breast cancer. Immunohistochemically detected DNA repair markers may be good candidates for novel prognostic and predictive factors that could guide the selection of individualized treatment strategies. PATIENTS AND METHODS: We have analyzed nuclear immunohistochemical staining of BRCA1, FANCD2, RAD51, XPF, and PAR in relation to clinicopathological and survival data among 1240 paraffin-embedded breast tumors, and additional gene expression microarray data from 76 tumors. The antioxidant enzyme NQO1 was analyzed as a potential modifier of prognostic DNA repair markers. RESULTS: RAD51 [hazard ratio (HR) 0.81, 95% confidence interval (CI) 0.70-0.94, P = 0.0050] and FANCD2 expression (HR 1.50, 95% CI 1.28-1.76, P = 1.50 × 10(-7)) were associated with breast cancer survival. High FANCD2 expression correlated with markers of adverse prognosis but remained independently prognostic in multivariate analysis (HR 1.27, 95% CI 1.08-1.49, P = 0.0043). The FANCD2-associated survival effect was most pronounced in hormone receptor positive, HER2-negative tumors, and in tumors with above-median NQO1 expression. In the NQO1-high subset, patients belonging to the highest quartile of FANCD2 immunohistochemical scores had a threefold increased risk of metastasis or death (HR 3.10, 95% CI 1.96-4.92). Global gene expression analysis indicated that FANCD protein overabundance is associated with the upregulation of proliferation-related genes and a downregulated nucleotide excision repair pathway. CONCLUSION: FANCD2 immunohistochemistry is a sensitive, independent prognostic factor in breast cancer, particularly when standard markers indicate relatively favorable prognosis. Taken together, our results suggest that the prognostic effect is linked to proliferation, DNA damage, and oxidative stress; simultaneous detection of FANCD2 and NQO1 provides additional prognostic value.


Subject(s)
Breast Neoplasms/genetics , Fanconi Anemia Complementation Group D2 Protein/biosynthesis , NAD(P)H Dehydrogenase (Quinone)/biosynthesis , Prognosis , Biomarkers, Tumor , Breast Neoplasms/pathology , DNA Repair/genetics , Fanconi Anemia Complementation Group D2 Protein/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , NAD(P)H Dehydrogenase (Quinone)/genetics , Receptor, ErbB-2/genetics
9.
Cell Death Differ ; 20(11): 1485-97, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23852374

ABSTRACT

Oncogenic stimuli trigger the DNA damage response (DDR) and induction of the alternative reading frame (ARF) tumor suppressor, both of which can activate the p53 pathway and provide intrinsic barriers to tumor progression. However, the respective timeframes and signal thresholds for ARF induction and DDR activation during tumorigenesis remain elusive. Here, these issues were addressed by analyses of mouse models of urinary bladder, colon, pancreatic and skin premalignant and malignant lesions. Consistently, ARF expression occurred at a later stage of tumor progression than activation of the DDR or p16(INK4A), a tumor-suppressor gene overlapping with ARF. Analogous results were obtained in several human clinical settings, including early and progressive lesions of the urinary bladder, head and neck, skin and pancreas. Mechanistic analyses of epithelial and fibroblast cell models exposed to various oncogenes showed that the delayed upregulation of ARF reflected a requirement for a higher, transcriptionally based threshold of oncogenic stress, elicited by at least two oncogenic 'hits', compared with lower activation threshold for DDR. We propose that relative to DDR activation, ARF provides a complementary and delayed barrier to tumor development, responding to more robust stimuli of escalating oncogenic overload.


Subject(s)
Carcinogenesis/genetics , DNA Damage , Neoplasms/genetics , Tumor Suppressor Protein p14ARF/genetics , Amino Acid Sequence , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , Gene Expression , Heterografts , Humans , Immunohistochemistry , Mice , Molecular Sequence Data , Neoplasms/metabolism , Neoplasms/pathology , Oncogenes , Transfection , Tumor Suppressor Protein p14ARF/metabolism
10.
Oncogene ; 32(31): 3577-86, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-22926521

ABSTRACT

One major challenge in cancer research is to understand the complex interplay between the DNA damage response (DDR), genomic integrity, and tumor development. To address these issues, we analyzed 43 bladder tumor genomes from 22 patients using single nucleotide polymorphism (SNP) arrays, and tissue expression of multiple DDR proteins, including Timeless and its interaction partner Tipin. The SNP profiles confirmed and extended known copy number alterations (CNAs) at high resolution, showed clustering of CNAs at nine common fragile sites, and revealed that most metachronous tumors were clonally related. The occurrence of many novel uniparental disomy regions (UPDs) was of potential functional importance in some tumors because UPDs spanned mutated FGFR3 and PIK3CA alleles, and also homozygous deletion of the CDKN2A tumor suppressor locus. The DDR signaling as evaluated by phospho-epitope-specific antibodies against Ser139-phosphorylated H2A histone family member X (γH2AX), ataxia telangiectasia mutated (ATM), and ATM- and Rad3-related (ATR) was commonly activated in tumors with both moderate and high extent of accumulated genomic aberrations, the latter tumors showing a more frequent loss of ATM expression. Strikingly, the tumor genomes exhibiting the most complex alterations were associated with a high Ki67-proliferation index, abundant Timeless but not Tipin expression, aberrant p53 expression, and homozygous CDKN2A deletions. Of clinical relevance, evaluation of a tissue microarray (TMA; n=319) showed that abundant Timeless expression was associated with risk of progression to muscle-invasive disease (P<0.0005; hazard ratio, 2.4; 95% confidence interval, 1.6-3.8) and higher T stage (P<0.05). Univariate analysis confirmed this association (P=0.006) in an independent cohort (n=241) but statistical significance was not reached in a multivariate model. Overall, our results are consistent with DDR activation preceding the accumulation of genomic aberrations. Tumors with extensive genomic rearrangements were associated with inactivation of CDKN2A, excessive proliferation, and robust Timeless expression, the latter also correlating with the risk of disease progression. Moreover, we provide evidence to suggest that UPDs likely contribute to bladder tumorigenesis.


Subject(s)
DNA Damage/genetics , Genomics/methods , Urinary Bladder Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Carrier Proteins/genetics , Cell Cycle Proteins/genetics , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA-Binding Proteins , Disease Progression , Female , Genetic Predisposition to Disease/genetics , Homozygote , Humans , Intracellular Signaling Peptides and Proteins/genetics , Male , Middle Aged , Nuclear Proteins/genetics , Oligonucleotide Array Sequence Analysis , Polymorphism, Single Nucleotide , Sequence Deletion , Signal Transduction/genetics , Transcriptome , Tumor Suppressor Protein p53/metabolism , Uniparental Disomy , Urinary Bladder Neoplasms/pathology
11.
Int J Androl ; 34(4 Pt 2): e103-13, 2011 Aug.
Article in English | MEDLINE | ID: mdl-20695923

ABSTRACT

Heterochromatinization has been implicated in fundamental biological and pathological processes including differentiation, senescence, ageing and tumourigenesis; however, little is known about its regulation and roles in human cells and tissues in vivo. Here, we show distinct cell-type- and cancer-stage-associated patterns of key heterochromatin marks: histone H3 trimethylated at lysine 9 (H3K9me3) and heterochromatic adaptor proteins HP1α and HP1γ, compared with the γH2AX marker of endogenously activated DNA damage response (DDR) and proliferation markers in normal human foetal (n=4) and adult (n=29) testes, pre-invasive carcinoma in situ (CIS; n=26) lesions and a series of overt germ cell tumours, including seminomas (n=26), embryonal carcinomas (n=18) and teratomas (n=11). Among striking findings were high levels of HP1γ in foetal gonocytes, CIS and seminomas; enhanced multimarker heterochromatinization without DDR activation in CIS; and enhanced HP1α in teratoma structures with epithelial and neuronal differentiation. Differential expression of the three heterochromatin markers suggests their partly non-overlapping roles, and separation of heterochromatinization from DDR activation highlights distinct responses of germ cells vs. somatic tissues in early tumourigenesis. Conceptually interesting findings were that subsets of human cells in vivo proliferate despite enhanced heterochromatinization, and that cells can strongly express even multiple heterochromatin features in the absence of functional retinoblastoma protein and without DDR activation. Overall, these results provide novel insights into cell-related and tumour-related diversity of heterochromatin in human tissues in vivo, relevant for andrology and intrinsic anti-tumour defence roles attributed to activated DDR and cellular senescence.


Subject(s)
Chromosomal Proteins, Non-Histone/biosynthesis , Histones/metabolism , Neoplasms, Germ Cell and Embryonal/pathology , Testicular Neoplasms/pathology , Testis/embryology , Antibodies, Monoclonal , Cell Line , Cellular Senescence/genetics , Chromobox Protein Homolog 5 , DNA Damage , DNA Repair , Fluorescent Antibody Technique , Heterochromatin/metabolism , Histones/biosynthesis , Histones/genetics , Histones/immunology , Humans , Immunoblotting , Male , Methylation , Neoplasm Staging , Neoplasms, Germ Cell and Embryonal/genetics , Testicular Neoplasms/genetics , Testicular Neoplasms/metabolism , Testis/metabolism
12.
Oncogene ; 29(36): 5095-102, 2010 Sep 09.
Article in English | MEDLINE | ID: mdl-20581868

ABSTRACT

Malignant gliomas, the deadliest of brain neoplasms, show rampant genetic instability and resistance to genotoxic therapies, implicating potentially aberrant DNA damage response (DDR) in glioma pathogenesis and treatment failure. Here, we report on gross, aberrant constitutive activation of DNA damage signalling in low- and high-grade human gliomas, and analyze the sources of such endogenous genotoxic stress. Based on analyses of human glioblastoma multiforme (GBM) cell lines, normal astrocytes and clinical specimens from grade II astrocytomas (n=41) and grade IV GBM (n=60), we conclude that the DDR machinery is constitutively activated in gliomas, as documented by phosphorylated histone H2AX (gammaH2AX), activation of the ATM-Chk2-p53 pathway, 53BP1 foci and other markers. Oxidative DNA damage (8-oxoguanine) was high in some GBM cell lines and many GBM tumors, while it was low in normal brain and grade II astrocytomas, despite the degree of DDR activation was higher in grade II tumors. Markers indicative of ongoing DNA replication stress (Chk1 activation, Rad17 phosphorylation, replication protein A foci and single-stranded DNA) were present in GBM cells under high- or low-oxygen culture conditions and in clinical specimens of both low- and high-grade tumors. The observed global checkpoint signaling, in contrast to only focal areas of overabundant p53 (indicative of p53 mutation) in grade II astrocytomas, are consistent with DDR activation being an early event in gliomagenesis, initially limiting cell proliferation (low Ki-67 index) and selecting for mutations of p53 and likely other genes that allow escape (higher Ki-67 index) from the checkpoint and facilitate tumor progression. Overall, these results support the potential role of the DDR machinery as a barrier to gliomagenesis and indicate that replication stress, rather than oxidative stress, fuels the DNA damage signalling in early stages of astrocytoma development.


Subject(s)
Brain Neoplasms/genetics , DNA Damage/physiology , DNA Replication/physiology , Glioma/genetics , Oxidative Stress/physiology , Stress, Physiological/physiology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , DNA Replication/genetics , Glioma/metabolism , Glioma/pathology , Histones/metabolism , Humans , Ki-67 Antigen/metabolism , Signal Transduction/genetics , Tumor Suppressor Protein p53/metabolism
13.
Oncogene ; 27(17): 2501-6, 2008 Apr 10.
Article in English | MEDLINE | ID: mdl-17982490

ABSTRACT

The ataxia-telangiectasia-mutated (ATM) kinase is a key transducer of DNA damage signals within the genome maintenance machinery and a tumour suppressor whose germline mutations predispose to familial breast cancer. ATM signalling is constitutively activated in early stages of diverse types of human malignancies and cell culture models in response to oncogene-induced DNA damage providing a barrier against tumour progression. As BRCA1 and BRCA2 are also components of the genome maintenance network and their mutations predispose to breast cancer, we have examined the ATM expression in human breast carcinomas of BRCA1/2 mutation carriers, sporadic cases and familial non-BRCA1/2 patients. Our results show that ATM protein expression is aberrantly reduced more frequently among BRCA1 (33%; P=0.0003) and BRCA2 (30%; P=0.0009) tumours than in non-BRCA1/2 tumours (10.7%). Furthermore, the non-BRCA1/2 tumours with reduced ATM expression were more often estrogen receptor (ER) negative (P=0.0002), progesterone receptor (PR) negative (P=0.004) and were of higher grade (P=0.0004). In our series of 1013 non-BRCA1/2 cases, ATM was more commonly deficient (20%; P=0.0006) and p53 was overabundant (47%; P<0.0000000001) among the difficult-to-treat ER/PR/ERBB2-triple-negative subset of tumours compared with cases that expressed at least one of these receptors (10 and 16% of aberrant ATM and p53, respectively). We propose a model of 'conditional haploinsufficiency' for BRCA1/2 under conditions of enhanced DNA damage in precancerous lesions resulting in more robust activation and hence increased selection for inactivation or loss of ATM in tumours of BRCA1/2 mutation carriers, with implications for genomic instability and curability of diverse subsets of human breast cancer.


Subject(s)
BRCA1 Protein/deficiency , BRCA2 Protein/deficiency , Breast Neoplasms/metabolism , DNA Damage , DNA-Binding Proteins/deficiency , Protein Serine-Threonine Kinases/deficiency , Receptor, ErbB-2/deficiency , Receptors, Estrogen/deficiency , Receptors, Progesterone/deficiency , Tumor Suppressor Proteins/deficiency , Ataxia Telangiectasia Mutated Proteins , BRCA1 Protein/metabolism , BRCA2 Protein/metabolism , Breast Neoplasms/genetics , Cell Cycle Proteins/metabolism , DNA Damage/genetics , DNA-Binding Proteins/metabolism , Humans , Protein Serine-Threonine Kinases/metabolism , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism
14.
Oncogene ; 26(56): 7773-9, 2007 Dec 10.
Article in English | MEDLINE | ID: mdl-18066090

ABSTRACT

DNA damage response (DDR), the guardian of genomic integrity, emerges as an oncogene-inducible biological barrier against progression of cancer beyond its early stages. Recent evidence from both cell culture and animal models as well as analyses of clinical specimens show that activation of numerous oncogenes and loss of some tumour suppressors result in DNA replication stress and DNA damage that alarm the cellular DDR machinery, a multifaceted response orchestrated by the ATR-Chk1 and ATM-Chk2 kinase signalling pathways. Such activation of the DDR network leads to cellular senescence or death of oncogene-transformed cells, resulting in delay or prevention of tumorigenesis. At the same time, the ongoing chronic DDR activation creates selective pressure that eventually favours outgrowth of malignant clones with genetic or epigenetic defects in the genome maintenance machinery, such as aberrations in the ATM-Chk2-p53 cascade and other DDR components. Furthermore, the executive DDR machinery is shared by at least two anticancer barriers, as both the oncogene-induced DNA replication stress and telomere shortening impact the cell fate decisions through convergence on DNA damage signalling. In this study, we highlight recent advances in this rapidly evolving area of cancer research, with particular emphasis on mechanistic insights, emerging issues of special conceptual significance and discussion of major remaining challenges and implications of the concept of DDR as a tumorigenesis barrier for experimental and clinical oncology.


Subject(s)
DNA Damage/genetics , DNA Repair , Genes, Tumor Suppressor , Neoplasms/prevention & control , Oncogenes/genetics , Animals , Cellular Senescence , Humans , Neoplasms/genetics , Signal Transduction
15.
Oncogene ; 26(53): 7414-22, 2007 Nov 22.
Article in English | MEDLINE | ID: mdl-17546051

ABSTRACT

MDC1 and 53BP1 are critical components of the DNA damage response (DDR) machinery that protects genome integrity and guards against cancer, yet the tissue expression patterns and involvement of these two DDR adaptors/mediators in human tumours remain largely unknown. Here we optimized immunohistochemical analyses of human 53BP1 and MDC1 proteins in situ and identified their virtually ubiquitous expression, both in proliferating and quiescent, differentiated tissues. Focus formation by 53BP1 and/or MDC1 in human spermatogenesis and subsets of breast and lung carcinomas indicated physiological and 'pathological' activation of the DDR, respectively. Furthermore, aberrant reduction or lack of either protein in significant proportions of carcinomas supported the candidacy of 53BP1 and MDC1 for tumour suppressors. Contrary to carcinomas, almost no activation or loss of MDC1 or 53BP1 were found among testicular germ-cell tumours (TGCTs), a tumour type with unique biology and exceptionally low incidence of p53 mutations. Such concomitant presence (in carcinomas) or absence (in TGCTs) of DDR activation and DDR aberrations supports the roles of MDC1 and 53BP1 within the ATM/ATR-regulated checkpoint network which, when activated, provides an early anti-cancer barrier the pressure of which selects for DDR defects such as p53 mutations or loss of 53BP1/MDC1 during cancer progression.


Subject(s)
Breast Neoplasms/metabolism , DNA Damage , Intracellular Signaling Peptides and Proteins/metabolism , Lung Neoplasms/metabolism , Neoplasms, Germ Cell and Embryonal/metabolism , Nuclear Proteins/biosynthesis , Testicular Neoplasms/metabolism , Trans-Activators/biosynthesis , Adaptor Proteins, Signal Transducing , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Cycle Proteins , Cell Line, Tumor , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Humans , Immunohistochemistry , Intracellular Signaling Peptides and Proteins/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Neoplasms, Germ Cell and Embryonal/genetics , Neoplasms, Germ Cell and Embryonal/pathology , Nuclear Proteins/genetics , Testicular Neoplasms/genetics , Testicular Neoplasms/pathology , Trans-Activators/genetics , Tumor Suppressor p53-Binding Protein 1
16.
Int J Androl ; 30(4): 282-91; discussion 291, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17573848

ABSTRACT

DNA damage response (DDR) is emerging as a physiological anti-cancer barrier in early stages of cancer development, as shown for several types of solid cancers derived from somatic cells. Here we discuss our recently published and unpublished results on the exceptional paucity of such constitutive activation of the DDR machinery in human testicular germ cell tumours (TGCTs), including their common pre-invasive stage of carcinoma in situ (CIS). Our conclusions are supported by immunohistochemical analyses of multiple markers of activated DNA damage signalling, such as the phosphorylated ATM and Chk2 checkpoint kinases and phosphorylated histone H2AX. We propose that the unique lack of DDR activation in TGCTs reflects the biology of their cell of origin, the gonocyte. Furthermore, we propose that the lack of DDR activation avoids the pressure to select for mutations in DDR genes such as p53 or ATM, and the resulting intact DDR machinery may have implications for the exceptional curability of TGCTs by DNA damaging therapies.


Subject(s)
DNA Damage , Neoplasms, Germ Cell and Embryonal/genetics , Testicular Neoplasms/genetics , Carcinoma in Situ/genetics , Chromosome Aberrations , Humans , Male , Mutation , Neoplasm Invasiveness , Neoplasms, Germ Cell and Embryonal/pathology , Neoplasms, Germ Cell and Embryonal/therapy , Reference Values , Testicular Neoplasms/pathology , Testicular Neoplasms/therapy , Testis/physiology
17.
J Pathol ; 209(4): 512-21, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16739112

ABSTRACT

Centrosome abnormalities are observed in human cancers and have been associated with aneuploidy, a driving force in tumour progression. However, the exact pathways that tend to cause centrosome abnormalities have not been fully elucidated in human tumours. Using a series of 68 non-small-cell lung carcinomas and an array of in vitro experiments, the relationship between centrosome abnormalities, aneuploidy, and the status of key G1 to S-phase transition cell-cycle molecules, involved in the regulation of centrosome duplication, was investigated. Centrosome amplification and structural abnormalities were common (53%), were strongly related to aneuploidy, and, surprisingly, were even seen in adjacent hyperplastic regions, suggesting the possibility that these are early lesions in lung carcinogenesis. Cyclin E and E2F1 overexpression, but not p53 mutation, was observed to correlate with centrosome abnormalities in vivo (p = 0.029 and p = 0.015, respectively). This was further strengthened by the observation that cyclin E was specifically present in the nucleus and/or cytoplasm of the cells that contained centrosome aberrations. The cytoplasmic cyclin E signal may be attributed, in part, to the presence of truncated low-molecular-weight isoforms of cyclin E. In order to isolate the effect of cyclin E on the appearance of centrosome abnormalities, a U2OS tetracycline-repressible cyclin E cell line that has a normal centrosome profile by default was used. With this system, it was confirmed in vitro that persistent cyclin E overexpression is sufficient to cause the appearance of centrosome abnormalities.


Subject(s)
Aneuploidy , Carcinoma, Non-Small-Cell Lung/pathology , Centrosome/ultrastructure , Cyclin E/genetics , Lung Neoplasms/pathology , Aged , Carcinoma, Non-Small-Cell Lung/genetics , Chi-Square Distribution , E2F1 Transcription Factor/genetics , Electrophoresis, Polyacrylamide Gel , Female , Gene Expression , Gene Expression Regulation, Neoplastic , Genes, p53 , Humans , In Situ Hybridization , Lung Neoplasms/genetics , Male , Middle Aged , Statistics, Nonparametric
18.
Histopathology ; 42(3): 217-26, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12605640

ABSTRACT

AIMS: Spermatocytic seminoma is a rare germ cell derived tumour of the testis that occurs mainly in older men. We analysed the expression of recently discovered markers for germ cell differentiation and the mitosis-meiosis transition in order to define the antigen profile for diagnostic purposes and to clarify the biology and histogenesis of spermatocytic seminoma. METHODS AND RESULTS: Twenty-five spermatocytic seminomas were examined for immunohistochemical expression of germ cell-specific onco-fetal antigens and proteins involved in regulation of germ cell division, DNA repair and differentiation. The panel included Chk2, p19INK4d, p53, MAGE-A4, KIT, TRA-1-60, neurone-specific enolase and placental-like alkaline phosphatase. Four of these proteins/antigens have never before been investigated in spermatocytic seminoma. Proteins highly expressed in gonocytes and spermatogonia, such as Chk2, MAGE-A4 and neurone-specific enolase, were consistently present in spermatocytic seminoma. Antigens expressed in embryonic germ cells but not in the normal adult testis, e.g. TRA-1-60, were undetectable, with the exception of p53 protein, which was demonstrated in 80% of cases. A proto-oncogene p19INK4d, which is involved in the transition from mitotic to meiotic division in germ cells, was not detected in spermatocytic seminoma. CONCLUSIONS: The investigation provided new information concerning the expression of Chk2, MAGE-A4, neurone-specific enolase and p19INK4d in spermatocytic seminoma. The pattern of expression is highly consistent with the origin of spermatocytic seminoma from a premeiotic germ cell, which has lost embryonic traits and has committed to spermatogenic lineage but has not yet passed the meiotic checkpoint, most probably from the spermatogonium of the adult testis.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Cycle Proteins , Neoplasm Proteins/metabolism , Protein Serine-Threonine Kinases , Seminoma/metabolism , Testicular Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Antigens, Neoplasm/metabolism , Checkpoint Kinase 2 , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p19 , Humans , Immunoenzyme Techniques , Male , Middle Aged , Phosphopyruvate Hydratase/metabolism , Protein Kinases/metabolism , Proto-Oncogene Mas , Seminoma/etiology , Seminoma/pathology , Testicular Neoplasms/etiology , Testicular Neoplasms/pathology , Tumor Suppressor Protein p53/metabolism
19.
Oncogene ; 20(41): 5897-902, 2001 Sep 13.
Article in English | MEDLINE | ID: mdl-11593395

ABSTRACT

Chk2 is a transducer of DNA damage signals and a tumour suppressor whose germ-line mutations predispose to diverse tumour types. Unlike its downstream targets such as the p53 tumour suppressor, the expression patterns of Chk2 in tissues and tumours remain unknown. As DNA breaks occur commonly during gametogenesis, and p53 is wild-type and overexpressed in testicular cancer, we examined abundance and localisation of the Chk2 protein during normal development of human testes, and at various stages of germ-cell tumour (GCT) pathogenesis. Our results show that Chk2 is abundant in foetal germ cells and adult spermatogonia, yet only weakly expressed or lacking during the meiotic and later stages of spermatogenesis. High levels of Chk2 are detected in the majority of GCTs including all pre-invasive carcinoma-in-situ lesions, contrary to variable expression and even lack of Chk2 in subsets of invasive GCTs and some teratoma structures, respectively. Together with our analyses of cell culture models, these results indicate that downmodulation or lack of Chk2 is not simply attributable to quiescence or differentiation, they suggest a role for Chk2 in mitotic rather than meiotic divisions, support the concept of foetal origin of GCTs, and have implications for protein-based screening for tumour-associated aberrations of Chk2.


Subject(s)
Carcinoma in Situ/metabolism , Germinoma/metabolism , Neoplasm Proteins/metabolism , Protein Kinases/metabolism , Protein Serine-Threonine Kinases , Spermatogenesis/physiology , Testicular Neoplasms/metabolism , Testis/metabolism , Checkpoint Kinase 2 , Genes, Tumor Suppressor , Humans , Male , Spermatozoa/metabolism , Testis/embryology , Testis/growth & development
20.
Virchows Arch ; 439(2): 132-40, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11561753

ABSTRACT

The p21/WAF1/Cipl antibody, DCS-60, was characterized by means of immunoblotting and immunofluorescence on a variety of human breast cancer cell lines. Heterogeneous staining of nuclei was observed with strong staining of cells in early G1. p21/WAF1/Cipl expression in invasive ductal, not otherwise specified breast carcinomas was determined using immunohistochemistry with this antibody and computerized image analysis. Two hundred and twenty-two tumors, including 130 from patients with no axillary node involvement, were examined. p21-positive tumor cell nuclei were found in 30% of the breast carcinomas. The percentage of tumor cell nuclei that were positive ranged from less than 1% to greater than 10%. In the whole cohort of patients, p21 expression was significantly associated with a low histological grade. In the node-negative group, there was a significant negative correlation between p21 positivity and a high (>10%) MIB-1 score. The mean MIB-1 score was significantly lower in p21-positive tumors in the whole cohort of patients (P=0.03) and in the nodenegative group (P=0.02). No association was found between p21 expression and overall survival at 5 years. With respect to p21/p53 phenotype, the significant difference in survival was noted only for the group of patients treated with adjuvant chemotherapy. The p21- p53+ phenotype had the worst survival (58% surviving 5 years), while the p21+ p53- phenotype had good survival (83% surviving 5 years; P<0.05). The results seem to suggest a correlation between p21/p53 phenotype and response to adjuvant chemotherapy.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/metabolism , Cyclins/metabolism , Tumor Suppressor Protein p53/metabolism , Antibodies, Monoclonal , Antigens, Nuclear , Biomarkers/analysis , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Carcinoma, Ductal, Breast/mortality , Carcinoma, Ductal, Breast/secondary , Carcinoma, Ductal, Breast/therapy , Cell Division , Chemotherapy, Adjuvant , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/immunology , Disease-Free Survival , Female , Humans , Immunoenzyme Techniques , Ki-67 Antigen , Lymph Nodes/pathology , Lymphatic Metastasis , Nuclear Proteins/metabolism , Survival Rate , Tumor Cells, Cultured/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL