Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Genes (Basel) ; 14(10)2023 09 27.
Article in English | MEDLINE | ID: mdl-37895233

ABSTRACT

Prostate cancer (PCa) is the most common cause of cancer death among African men. The presence of tumor-specific variations in cell-free DNA (cfDNA), such as mutations, microsatellite instability, and DNA methylation, has been explored as a source of biomarkers for cancer diagnosis. In this study, we investigated the diagnostic role of cfDNA among South African PCa patients. We performed whole exome sequencing (WES) of urinary cfDNA. We identified a novel panel of 31 significantly deregulated somatic mutated genes between PCa and benign prostatic hyperplasia (BPH). Additionally, we performed whole-genome sequencing (WGS) on matching PCa and normal prostate tissue in an independent PCa cohort from South Africa. Our results suggest that the mutations are of germline origin as they were also found in the normal prostate tissue. In conclusion, our study contributes to the knowledge of cfDNA as a biomarker for diagnosing PCa in the South African population.


Subject(s)
Cell-Free Nucleic Acids , Prostatic Hyperplasia , Prostatic Neoplasms , Male , Humans , Cell-Free Nucleic Acids/genetics , South Africa , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/genetics , Prostatic Hyperplasia/diagnosis , Prostatic Hyperplasia/genetics , Mutation , Biomarkers
2.
Cancers (Basel) ; 15(15)2023 Aug 04.
Article in English | MEDLINE | ID: mdl-37568783

ABSTRACT

Prostate cancer (PCa) is the most common cause of cancer death among African men. The analysis of microRNAs (miRNAs) in plasma extracellular vesicles (EVs) can be utilized as a non-invasive tool for the diagnosis of PCa. In this study, we used small RNA sequencing to profile miRNAs cargo in plasma EVs from South African PCa patients. We evaluated the differential expression of miRNAs between low and high Gleason scores in the plasma EVs of South African patients and in the prostatic tissue from data available in the Cancer Genome Atlas (TCGA) Data Portal. We identified 7 miRNAs differently expressed in both EVs and prostatic tissues. We evaluated their expression using qPCR in a larger cohort of 10 patients with benign prostatic hyperplasia (BPH) and 24 patients with PCa. Here, we reported that the ratio between two of these miRNAs (i.e., miR-194-5p/miR-16-5p) showed a higher concentration in PCa compared to BPH and in metastatic PCa compared to localized PCa. We explored for the first time the profiling of miRNAs cargo in plasma EVs as a tool for the identification of putative markers in the South African population. Our finding indicated the ratio miR-194-5p/miR-16-5p as a non-invasive marker for the evaluation of PCa aggressiveness in this population.

3.
Methods Mol Biol ; 2675: 195-204, 2023.
Article in English | MEDLINE | ID: mdl-37258765

ABSTRACT

Nuclear magnetic resonance (NMR) spectroscopy enables the detection and the quantification of a large range of molecules, including low-molecular-weight metabolites and lipids. NMR spectroscopy is a powerful approach when applied to the high-throughput analysis of plasma or serum samples allowing, in addition, the detection of total proteins, lipoproteins, and signals arising from the glycosylation of circulating acute-phase proteins. Here, we describe the usage of NMR spectroscopy for profiling the plasma or serum of patients with prostate cancer.


Subject(s)
Metabolomics , Prostatic Neoplasms , Male , Humans , Metabolomics/methods , Magnetic Resonance Spectroscopy/methods , Prostatic Neoplasms/metabolism , Plasma/metabolism , Magnetic Resonance Imaging
4.
Front Mol Biosci ; 10: 1165720, 2023.
Article in English | MEDLINE | ID: mdl-36968275

ABSTRACT

[This corrects the article DOI: 10.3389/fmolb.2022.1070394.].

5.
Front Mol Biosci ; 9: 1070394, 2022.
Article in English | MEDLINE | ID: mdl-36733493

ABSTRACT

KODAMA is a valuable tool in metabolomics research to perform exploratory analysis. The advanced analytical technologies commonly used for metabolic phenotyping, mass spectrometry, and nuclear magnetic resonance spectroscopy push out a bunch of high-dimensional data. These complex datasets necessitate tailored statistical analysis able to highlight potentially interesting patterns from a noisy background. Hence, the visualization of metabolomics data for exploratory analysis revolves around dimensionality reduction. KODAMA excels at revealing local structures in high-dimensional data, such as metabolomics data. KODAMA has a high capacity to detect different underlying relationships in experimental datasets and correlate extracted features with accompanying metadata. Here, we describe the main application of KODAMA exploratory analysis in metabolomics research.

6.
Commun Biol ; 4(1): 119, 2021 01 26.
Article in English | MEDLINE | ID: mdl-33500545

ABSTRACT

Extracellular vesicles (EVs) are relevant means for transferring signals across cells and facilitate propagation of oncogenic stimuli promoting disease evolution and metastatic spread in cancer patients. Here, we investigated the release of miR-424 in circulating small EVs or exosomes from prostate cancer patients and assessed the functional implications in multiple experimental models. We found higher frequency of circulating miR-424 positive EVs in patients with metastatic prostate cancer compared to patients with primary tumors and BPH. Release of miR-424 in small EVs was enhanced in cell lines (LNCaPabl), transgenic mice (Pb-Cre4;Ptenflox/flox;Rosa26ERG/ERG) and patient-derived xenograft (PDX) models of aggressive disease. EVs containing miR-424 promoted stem-like traits and tumor-initiating properties in normal prostate epithelial cells while enhanced tumorigenesis in transformed prostate epithelial cells. Intravenous administration of miR-424 positive EVs to mice, mimicking blood circulation, promoted miR-424 transfer and tumor growth in xenograft models. Circulating miR-424 positive EVs from patients with aggressive primary and metastatic tumors induced stem-like features when supplemented to prostate epithelial cells. This study establishes that EVs-mediated transfer of miR-424 across heterogeneous cell populations is an important mechanism of tumor self-sustenance, disease recurrence and progression. These findings might indicate novel approaches for the management and therapy of prostate cancer.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cell-Derived Microparticles/metabolism , Extracellular Vesicles/metabolism , MicroRNAs/metabolism , Prostatic Neoplasms , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Cell-Derived Microparticles/genetics , Extracellular Vesicles/genetics , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , MicroRNAs/genetics , Models, Theoretical , Neoplasm Invasiveness , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology
7.
Cells ; 8(8)2019 08 09.
Article in English | MEDLINE | ID: mdl-31404988

ABSTRACT

Liquid biopsy technologies have the potential to transform cancer patient management as it offers non-invasive diagnosis and real-time monitoring of disease progression and treatment responses. The use of liquid biopsy for non-invasive cancer diagnosis can have pivotal importance for the African continent where access to medical infrastructures is limited, as it eliminates the need for surgical biopsies. To apply liquid biopsy technologies in the African setting, the influence of environmental and population genetic factors must be known. In this review, we discuss the use of circulating tumor cells, cell-free nucleic acids, extracellular vesicles, protein, and other biomolecules in liquid biopsy technology for cancer management with special focus on African studies. We discussed the prospect, barriers, and other aspects that pose challenges to the use of liquid biopsy in the African continent.


Subject(s)
Liquid Biopsy , Neoplasms/diagnosis , Africa , Biomarkers, Tumor/analysis , Humans , Neoplastic Cells, Circulating/pathology
8.
PLoS One ; 10(8): e0134783, 2015.
Article in English | MEDLINE | ID: mdl-26302043

ABSTRACT

Acute Lymphoblastic Leukemia (ALL) is the most frequent childhood malignancy. In the effort to find new anti-leukemic agents, we evaluated the small drug SB225002 (N-(2-hydroxy-4-nitrophenyl)-N'-(2-bromophenyl)urea). Although initially described as a selective antagonist of CXCR2, later studies have identified other cellular targets for SB225002, with potential medicinal use in cancer. We found that SB225002 has a significant pro-apoptotic effect against both B- and T-ALL cell lines. Cell cycle analysis demonstrated that treatment with SB225002 induces G2-M cell cycle arrest. Transcriptional profiling revealed that SB225002-mediated apoptosis triggered a transcriptional program typical of tubulin binding agents. Network analysis revealed the activation of genes linked to the JUN and p53 pathways and inhibition of genes linked to the TNF pathway. Early cellular effects activated by SB225002 included the up-regulation of GLIPR1, a p53-target gene shown to have pro-apoptotic activities in prostate and bladder cancer. Silencing of GLIPR1 in B- and T-ALL cell lines resulted in increased resistance to SB225002. Although SB225002 promoted ROS increase in ALL cells, antioxidant N-Acetyl Cysteine pre-treatment only modestly attenuated cell death, implying that the pro-apoptotic effects of SB225002 are not exclusively mediated by ROS. Moreover, GLIPR1 silencing resulted in increased ROS levels both in untreated and SB225002-treated cells. In conclusion, SB225002 induces cell cycle arrest and apoptosis in different B- and T-ALL cell lines. Inhibition of tubulin function with concurrent activation of the p53 pathway, in particular, its downstream target GLIPR1, seems to underlie the anti-leukemic effect of SB225002.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Cycle/drug effects , Cell Death/drug effects , Neoplasm Proteins/drug effects , Nerve Tissue Proteins/drug effects , Phenylurea Compounds/pharmacology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Animals , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing , Humans , Jurkat Cells , Membrane Proteins , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Oligonucleotide Array Sequence Analysis , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction
9.
PLoS One ; 10(7): e0134305, 2015.
Article in English | MEDLINE | ID: mdl-26222679

ABSTRACT

Dipeptidyl peptidase IV (DPPIV/CD26) is a transmembrane glycoprotein that inactivates or degrades some bioactive peptides and chemokines. For this reason, it regulates cell proliferation, migration and adhesion, showing its role in cancer processes. This enzyme is found mainly anchored onto the cell membrane, although it also has a soluble form, an enzymatically active isoform. In the present study, we investigated DPPIV/CD26 activity and expression in cervical cancer cell lines (SiHa, HeLa and C33A) and non-tumorigenic HaCaT cells. The effect of the DPPIV/CD26 inhibitor (sitagliptin phosphate) on cell migration and adhesion was also evaluated. Cervical cancer cells and keratinocytes exhibited DPPIV/CD26 enzymatic activity both membrane-bound and in soluble form. DPPIV/CD26 expression was observed in HaCaT, SiHa and C33A, while in HeLa cells it was almost undetectable. We observed higher migratory capacity of HeLa, when compared to SiHa. But in the presence of sitagliptin SiHa showed an increase in migration, indicating that, at least in part, cell migration is regulated by DPPIV/CD26 activity. Furthermore, in the presence of sitagliptin phosphate, SiHa and HeLa cells exhibited a significant reduction in adhesion. However this mechanism seems to be mediated independent of DPPIV/CD26. This study demonstrates, for the first time, the activity and expression of DPPIV/CD26 in cervical cancer cells and the effect of sitagliptin phosphate on cell migration and adhesion.


Subject(s)
Carcinoma/metabolism , Carcinoma/pathology , Cell Movement/physiology , Dipeptidyl Peptidase 4/metabolism , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Movement/drug effects , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Female , HeLa Cells , Humans , Keratinocytes/drug effects , Keratinocytes/metabolism , Membrane Glycoproteins/metabolism , Sitagliptin Phosphate/pharmacology
10.
Mol Biol Cell ; 25(19): 2905-18, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25103241

ABSTRACT

In cervical cancer, HPV infection and disruption of mechanisms involving cell growth, differentiation, and apoptosis are strictly linked with tumor progression and invasion. Tumor microenvironment is ATP and adenosine rich, suggesting a role for purinergic signaling in cancer cell growth and death. Here we investigate the effect of extracellular ATP on human cervical cancer cells. We find that extracellular ATP itself has a small cytotoxic effect, whereas adenosine formed from ATP degradation by ectonucleotidases is the main factor responsible for apoptosis induction. The level of P2 × 7 receptor seemed to define the main cytotoxic mechanism triggered by ATP, since ATP itself eliminated a small subpopulation of cells that express high P2 × 7 levels, probably through its activation. Corroborating these data, blockage or knockdown of P2 × 7 only slightly reduced ATP cytotoxicity. On the other hand, cell viability was almost totally recovered with dipyridamole, an adenosine transporter inhibitor. Moreover, ATP-induced apoptosis and signaling-p53 increase, AMPK activation, and PARP cleavage-as well as autophagy induction were also inhibited by dipyridamole. In addition, inhibition of adenosine conversion into AMP also blocked cell death, indicating that metabolization of intracellular adenosine originating from extracellular ATP is responsible for the main effects of the latter in human cervical cancer cells.


Subject(s)
Adenosine Monophosphate/biosynthesis , Adenosine Triphosphate/pharmacology , Adenosine/metabolism , Apoptosis/drug effects , Uterine Cervical Neoplasms/drug therapy , AMP-Activated Protein Kinases/metabolism , Adenosine Triphosphate/metabolism , Autophagy/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dipyridamole/pharmacology , Female , HeLa Cells , Humans , Nucleoside Transport Proteins/antagonists & inhibitors , Poly(ADP-ribose) Polymerases/metabolism , RNA Interference , RNA, Messenger/biosynthesis , RNA, Small Interfering , Receptors, Purinergic P2X7/genetics , Receptors, Purinergic P2X7/metabolism , Tumor Microenvironment , Tumor Suppressor Protein p53/biosynthesis
11.
Mol Cancer Ther ; 13(7): 1929-1941, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24785412

ABSTRACT

Although early stages of clear cell renal cell carcinoma (ccRCC) are curable, survival outcome for metastatic ccRCC remains poor. We previously established a highly accurate signature of differentially expressed genes that distinguish ccRCC from normal kidney. The purpose of this study was to apply a new individualized bioinformatics analysis (IBA) strategy to these transcriptome data in conjunction with Gene Set Enrichment Analysis of the Connectivity Map (C-MAP) database to identify and reposition FDA-approved drugs for anticancer therapy. Here, we demonstrate that one of the drugs predicted to revert the RCC gene signature toward normal kidney, pentamidine, is effective against RCC cells in culture and in a RCC xenograft model. ccRCC-specific gene expression signatures of individual patients were used to query the C-MAP software. Eight drugs with negative correlation and P-value <0.05 were analyzed for efficacy against RCC in vitro and in vivo. Our data demonstrate consistency across most patients with ccRCC for the set of high-scoring drugs. Most of the selected high-scoring drugs potently induce apoptosis in RCC cells. Several drugs also demonstrate selectivity for Von Hippel-Lindau negative RCC cells. Most importantly, at least one of these drugs, pentamidine, slows tumor growth in the 786-O human ccRCC xenograft mouse model. Our findings suggest that pentamidine might be a new therapeutic agent to be combined with current standard-of-care regimens for patients with metastatic ccRCC and support our notion that IBA combined with C-MAP analysis enables repurposing of FDA-approved drugs for potential anti-RCC therapy.


Subject(s)
Carcinoma, Renal Cell/drug therapy , Pentamidine/pharmacology , Animals , Antineoplastic Agents/pharmacology , Antiprotozoal Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Renal Cell/pathology , Female , Humans , Male , Mice , Mice, Nude , Microarray Analysis
12.
Ann Hematol ; 91(2): 173-81, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21717102

ABSTRACT

Acute myeloid leukemia (AML) is a heterogeneous hematological malignancy. Treatment of patients suffering from high-risk AML as defined by clinical parameters, cytogenetics, and/or molecular analyses is often unsuccessful. OSI-461 is a pro-apoptotic compound that has been proposed as a novel therapeutic option for patients suffering from solid tumors like prostate or colorectal carcinoma. But little is known about its anti-proliferative potential in AML. Hence, we treated bone marrow derived CD34(+) selected blast cells from 20 AML patients and the five AML cell lines KG-1a, THP-1, HL-60, U-937, and MV4-11 with the physiologically achievable concentration of 1 µM OSI-461 or equal amounts of DMSO as a control. Following incubation with OSI-461, we found a consistent induction of apoptosis and an accumulation of cells in the G2/M phase of the cell cycle. In addition, we demonstrate that the OSI-461 mediated anti-proliferative effects observed in AML are associated with the induction of the pro-apoptotic cytokine mda-7/IL-24 and activation of the growth arrest and DNA-damage inducible genes (GADD) 45α and 45γ. Furthermore, OSI-461 treated leukemia cells did not regain their proliferative potential for up to 8 days after cessation of treatment following the initial 48 h treatment period with 1 µM OSI-461. This indicates sufficient targeting of the leukemia-initiating cells in our in vitro experiments through OSI-461. The AML samples tested in this study included samples from patients who were resistant to conventional chemotherapy and/or had FLT3-ITD mutations demonstrating the high potential of OSI-461 in human AML.


Subject(s)
Apoptosis/drug effects , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Sulindac/analogs & derivatives , Cell Cycle/drug effects , Cell Line, Tumor/drug effects , Clinical Trials as Topic , Gene Expression/drug effects , Humans , Phosphodiesterase Inhibitors/pharmacology , Phosphodiesterase Inhibitors/therapeutic use , Sulindac/pharmacology , Sulindac/therapeutic use
13.
Apoptosis ; 16(9): 889-901, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21739277

ABSTRACT

Acute myeloid leukemia is a heterogeneous disease with varying genetic and molecular pathologies. Non-steroidal anti-inflammatory drugs (NSAIDs) have been proven to possess significant anti-proliferative potential in various cancer cells in vitro and in vivo. Hence, treatment with these agents can be utilized to study disease specific anti-proliferative pathways. In this study, a total number of 42 bone marrow derived CD34(+) selected de novo AML patient samples and the AML cell lines THP-1 and HL-60 were treated with the NSAIDs Sulindac sulfide and Diclofenac. We analyzed viability, apoptosis, differentiation and addressed the molecular mechanisms involved. We found a consistent induction of apoptosis and to some extent an increased myeloid differentiation capacity in NSAID treated AML cells. Comprehensive protein and gene expression profiling of Diclofenac treated AML cells revealed transcriptional activation of GADD45α and its downstream MAPK/JNK pathway as well as increased protein levels of the caspase-3 precursor. This pointed towards a role of the c-Jun NH(2)-terminal kinase (JNK) in NSAID mediated apoptosis that we found indeed to be dependent on JNK activity as addition of a specific JNK-inhibitor abrogated apoptosis. Furthermore, the AP-1 transcription factor family members' c-Jun, JunB and Fra-2 were transcriptionally activated in NSAID treated AML cells and re-expression of these transcription factors led to activation of GADD45α with induction of apoptosis. Mechanistically, we demonstrate that NSAIDs induce apoptosis in AML through a novel pathway involving increased expression of AP-1 heterodimers, which by itself is sufficient to induce GADD45α expression with consecutive activation of JNK and induction of apoptosis.


Subject(s)
Apoptosis , Diclofenac/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Sulindac/analogs & derivatives , Transcription Factor AP-1/metabolism , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Caspase 3/metabolism , Cell Cycle Proteins/metabolism , Cell Differentiation , Cell Survival , Cloning, Molecular , Flow Cytometry , Fos-Related Antigen-2/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Genetic Vectors , HL-60 Cells , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-jun/metabolism , RNA Interference , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Sulindac/therapeutic use , Transcriptional Activation
14.
Cell Cycle ; 10(15): 2583-91, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21734453

ABSTRACT

The AP-1 transcription factor complex has been implicated in a variety of biological processes including cell differentiation, proliferation, apoptosis and oncogenic transformation. We previously established that activation of the AP-1 family member JunD contributes to deregulated expression of the anti-apoptotic IL-6 gene in prostate cancer cells. We now show that inhibition of JunD in prostate cancer cells results in GADD45α- and γ-dependent induction of cell death and inhibition of tumor growth that is mediated at least partially via c-Jun N-terminal kinase (JNK) and p38 kinase activation. Apoptosis induction by dominant negative JunD and JNK and p38 kinase activation are impeded upon knock down of GADD45α and γ expression by small interfering RNA, most vividly demonstrating the central role of GADD45α and γ in JunD-mediated escape of prostate cancer cells from programmed cell death.


Subject(s)
Apoptosis , Cell Cycle Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Line, Tumor , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Male , Mitogen-Activated Protein Kinase 8/metabolism , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-jun/antagonists & inhibitors , RNA Interference , RNA, Small Interfering/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
15.
Hum Gene Ther ; 13(15): 1833-44, 2002 Oct 10.
Article in English | MEDLINE | ID: mdl-12396616

ABSTRACT

Nucleotide excision repair (NER) is one of the most versatile DNA repair mechanisms, ensuring the proper functioning and trustworthy transmission of genetic information in all living cells. The phenotypic consequences caused by NER defects in humans are autosomal recessive diseases such as xeroderma pigmentosum (XP). This syndrome is the most sun-sensitive disorder leading to a high frequency of skin cancer. The majority of patients with XP carry mutations in the XPA or XPC genes that encode proteins involved in recognition of DNA damage induced by UV light at the beginning of the NER process. Cells cultured from XPA and XPC patients are hypersensitive to UV light, as a result of malfunctioning DNA repair. So far there is no effective long-term treatment for these patients. Skin cancer prevention can only be achieved by strict avoidance of sunlight exposure or by the use of sunscreen agents. We have constructed recombinant adenoviruses carrying the XPA and XPC genes that were used to infect XP-A and XP-C immortalized and primary fibroblast cell lines. UV survival curves and unscheduled DNA synthesis confirmed complete phenotypic reversion in XP DNA repair deficient cells with no trace of cytotoxicity. Moreover, transgene expression is stable for at least 60 days after infection. This efficient adenovirus gene delivery approach may be an important tool to better understand XP deficiency and the causes of DNA damage induced skin cancer.


Subject(s)
Adenoviruses, Human/genetics , DNA Repair/genetics , DNA-Binding Proteins/genetics , Genetic Vectors/genetics , Xeroderma Pigmentosum/pathology , Cell Line, Transformed , DNA Damage , DNA-Binding Proteins/physiology , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Fibroblasts/radiation effects , Genetic Complementation Test , Humans , Male , Phenotype , Polymerase Chain Reaction , Recombinant Fusion Proteins/physiology , Ultraviolet Rays/adverse effects , Xeroderma Pigmentosum/classification , Xeroderma Pigmentosum/genetics , Xeroderma Pigmentosum Group A Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...