Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
Ann Diagn Pathol ; 55: 151813, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34509898

ABSTRACT

Malignant gastrointestinal neuroectodermal tumor (GNET) is a rare malignant primary gastrointestinal mesenchymal tumor which can be diagnosed via fine-needle aspiration (FNA) cytology. In the context of FNA, the diagnosis requires a cell block and the use of significant resources including immunohistochemical stains and molecular testing. The differential diagnosis of GNET includes clear cell sarcoma (CCS), gastrointestinal stromal tumor (GIST), gastric schwannoma, metastatic melanoma, malignant perivascular epithelioid cell tumor (PEComa) and granular cell tumor, among others. Here we describe a case which was initially diagnosed as malignant granular cell tumor by FNA which was later revised to GNET following the finding of an EWSR1-ATF1 fusion gene rearrangement.


Subject(s)
Gastrointestinal Tract/pathology , Neuroectodermal Tumors , Biomarkers, Tumor/analysis , Biomarkers, Tumor/metabolism , Biopsy, Fine-Needle , Calmodulin-Binding Proteins/analysis , Calmodulin-Binding Proteins/metabolism , Diagnosis, Differential , Female , Gastrointestinal Neoplasms/diagnosis , Gastrointestinal Neoplasms/metabolism , Gastrointestinal Neoplasms/pathology , Gastrointestinal Stromal Tumors/diagnosis , Gastrointestinal Stromal Tumors/metabolism , Gastrointestinal Stromal Tumors/pathology , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Melanoma/diagnosis , Melanoma/metabolism , Melanoma/pathology , Middle Aged , Neuroectodermal Tumors/diagnosis , Neuroectodermal Tumors/metabolism , Neuroectodermal Tumors/pathology , Sarcoma, Clear Cell/diagnosis , Sarcoma, Clear Cell/metabolism , Sarcoma, Clear Cell/pathology
2.
Mol Cancer Ther ; 20(9): 1680-1691, 2021 09.
Article in English | MEDLINE | ID: mdl-34158347

ABSTRACT

We analyzed the efficacy and mechanistic interactions of PARP inhibition (PARPi; olaparib) and CDK4/6 inhibition (CDK4/6i; palbociclib or abemaciclib) combination therapy in castration-resistant prostate cancer (CRPC) and neuroendocrine prostate cancer (NEPC) models. We demonstrated that combined olaparib and palbociblib or abemaciclib treatment resulted in synergistic suppression of the p-Rb1-E2F1 signaling axis at the transcriptional and posttranslational levels, leading to disruption of cell-cycle progression and inhibition of E2F1 gene targets, including genes involved in DDR signaling/damage repair, antiapoptotic BCL-2 family members (BCL-2 and MCL-1), CDK1, and neuroendocrine differentiation (NED) markers in vitro and in vivo In addition, olaparib + palbociclib or olaparib + abemaciclib combination treatment resulted in significantly greater growth inhibition and apoptosis than either single agent alone. We further showed that PARPi and CDK4/6i combination treatment-induced CDK1 inhibition suppressed p-S70-BCL-2 and increased caspase cleavage, while CDK1 overexpression effectively prevented the downregulation of p-S70-BCL-2 and largely rescued the combination treatment-induced cytotoxicity. Our study defines a novel combination treatment strategy for CRPC and NEPC and demonstrates that combination PARPi and CDK4/6i synergistically promotes suppression of the p-Rb1-E2F1 axis and E2F1 target genes, including CDK1 and NED proteins, leading to growth inhibition and increased apoptosis in vitro and in vivo Taken together, our results provide a molecular rationale for PARPi and CDK4/6i combination therapy and reveal mechanism-based clinical trial opportunities for men with NEPC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Differentiation , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Neuroectodermal Tumors/drug therapy , Poly(ADP-ribose) Polymerases/chemistry , Prostatic Neoplasms/drug therapy , Aminopyridines/administration & dosage , Animals , Apoptosis , Benzimidazoles/administration & dosage , Cell Cycle , Cell Proliferation , Humans , Male , Mice , Mice, Nude , Neuroectodermal Tumors/metabolism , Neuroectodermal Tumors/pathology , Phthalazines/administration & dosage , Piperazines/administration & dosage , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Pyridines/administration & dosage , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
Article in English | MEDLINE | ID: mdl-33028642

ABSTRACT

We report a case of a DICER1-associated EWSR1-rearranged malignant primitive neuroectodermal tumor (PNET) arising in a patient with DICER1 tumor predisposition syndrome. A 16-yr-old female with a history of multinodular goiter presented with a widely metastatic abdominal small round blue cell tumor with neuroectodermal differentiation. EWSR1 gene rearrangement was identified in the tumor by fluorescence in situ hybridization (FISH). Genetic analysis revealed biallelic pathogenic DICER1 variation. The patient was treated with an aggressive course of chemotherapy, surgery, and radiation with complete pathologic response. We believe this case to represent a new expression of the DICER1 tumor predisposition syndrome, an entity caused by deleterious germline mutations in the DICER1 gene, encoding a ribonuclease active in the processing of miRNA. Patients with germline mutations in DICER1 develop a diverse group of benign and malignant tumors. Some of these tumors have been noted to have immature neuroepithelium as a component, including the ciliary body medulloepithelioma and the recently described DICER1-associated presacral malignant teratoid neoplasm. To our knowledge, abdominal sarcomas that resemble PNET histology with an EWSR1 rearrangement have not previously been described as a classical expression of the DICER1 syndrome phenotype.


Subject(s)
Neuroectodermal Tumors/genetics , RNA-Binding Protein EWS/genetics , Adolescent , DEAD-box RNA Helicases/metabolism , Female , Gene Rearrangement/genetics , Germ-Line Mutation , Humans , In Situ Hybridization, Fluorescence , Neoplasm Metastasis/genetics , Neuroectodermal Tumors/metabolism , RNA-Binding Protein EWS/metabolism , Ribonuclease III/metabolism , Sarcoma, Ewing/genetics , Sarcoma, Ewing/metabolism
4.
FEBS Lett ; 594(22): 3602-3618, 2020 11.
Article in English | MEDLINE | ID: mdl-32860713

ABSTRACT

Aberrant expression of glycosphingolipids (GSLs) is a unique feature of cancer and stromal cells in tumor microenvironments. Although the impact of GSLs on tumor progression remains largely unclear, anticancer immunotherapies directed against GSLs are attracting growing attention. Here, we focus on GD2, a disialoganglioside expressed in tumors of neuroectodermal origin, and Globo H ceramide (GHCer), the most prevalent cancer-associated GSL overexpressed in a variety of epithelial cancers. We first summarize recent advances on our understanding of GD2 and GHCer biology and then discuss the clinical development of the first immunotherapeutic agent targeting a glycolipid, the GD2-specific antibody dinutuximab, its approved indications, and new strategies to improve its efficacy for neuroblastoma. Next, we review ongoing clinical trials on Globo H-targeted immunotherapeutics. We end with highlighting how these studies provide sound scientific rationales for targeting GSLs in cancer and may facilitate a rational design of new GSL-targeted anticancer therapeutics.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Glycosphingolipids/metabolism , Neoplasms, Glandular and Epithelial/drug therapy , Neuroectodermal Tumors/drug therapy , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antigens, Tumor-Associated, Carbohydrate/metabolism , Antineoplastic Agents, Immunological/pharmacology , Clinical Trials as Topic , Gangliosides/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunotherapy , Neoplasms, Glandular and Epithelial/metabolism , Neuroectodermal Tumors/metabolism , Tumor Microenvironment/drug effects
5.
Proc Natl Acad Sci U S A ; 117(31): 18401-18411, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32690709

ABSTRACT

Disparities in cancer patient responses have prompted widespread searches to identify differences in sensitive vs. nonsensitive populations and form the basis of personalized medicine. This customized approach is dependent upon the development of pathway-specific therapeutics in conjunction with biomarkers that predict patient responses. Here, we show that Cdk5 drives growth in subgroups of patients with multiple types of neuroendocrine neoplasms. Phosphoproteomics and high throughput screening identified phosphorylation sites downstream of Cdk5. These phosphorylation events serve as biomarkers and effectively pinpoint Cdk5-driven tumors. Toward achieving targeted therapy, we demonstrate that mouse models of neuroendocrine cancer are responsive to selective Cdk5 inhibitors and biomimetic nanoparticles are effective vehicles for enhanced tumor targeting and reduction of drug toxicity. Finally, we show that biomarkers of Cdk5-dependent tumors effectively predict response to anti-Cdk5 therapy in patient-derived xenografts. Thus, a phosphoprotein-based diagnostic assay combined with Cdk5-targeted therapy is a rational treatment approach for neuroendocrine malignancies.


Subject(s)
Neoplasms/drug therapy , Neoplasms/metabolism , Neuroectodermal Tumors/drug therapy , Phosphoproteins/metabolism , Protein Kinase Inhibitors/administration & dosage , Animals , Biomarkers/analysis , Biomarkers/metabolism , Cyclin-Dependent Kinase 5/antagonists & inhibitors , Cyclin-Dependent Kinase 5/genetics , Cyclin-Dependent Kinase 5/metabolism , Heterografts , Humans , Mice , Neoplasms/genetics , Neuroectodermal Tumors/genetics , Neuroectodermal Tumors/metabolism , Phosphoproteins/analysis , Phosphoproteins/genetics , Phosphorylation
6.
Biomolecules ; 9(8)2019 07 27.
Article in English | MEDLINE | ID: mdl-31357634

ABSTRACT

Gangliosides, the glycosphingolipids carrying one or several sialic acid residues, are mostly localized at the plasma membrane in lipid raft domains and implicated in many cellular signaling pathways mostly by interacting with tyrosine kinase receptors. Gangliosides are divided into four series according to the number of sialic acid residues, which can be also modified by O-acetylation. Both ganglioside expression and sialic acid modifications can be modified in pathological conditions such as cancer, which can induce either pro-cancerous or anti-cancerous effects. In this review, we summarize the specific functions of gangliosides in neuro-ectodermal derived tumors, and their roles in reprogramming the lipidomic profile of cell membrane occurring with the induction of epithelial-mesenchymal transition.


Subject(s)
Gangliosides/metabolism , Neuroectodermal Tumors/metabolism , Acetylation , Cell Membrane/metabolism , Epithelial-Mesenchymal Transition , Gangliosides/chemistry , Humans , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction
7.
Cell Immunol ; 298(1-2): 96-103, 2015.
Article in English | MEDLINE | ID: mdl-26435344

ABSTRACT

Quantitative analysis of MUC1, a cell membrane associated mucin, expressed by intact cells of epithelial origin previously has been limited to flow cytometry, which requires using large quantities of cells and antibodies. Here, for the first time, we report the development of a novel Cellular-based Enzyme Linked Immunosorbent Assay (Cell ELISA) to quantify the expression of MUC1 by cell lines of epithelial and neuroectodermal origin using an antibody recognizing a specific tandem repeat found in the extracellular domain of MUC1. In contrast to flow cytometry, this method requires a much lower number of cells. We report here the results obtained from two variants of this Cell ELISA in live and fixed cells. We found that the Cell ELISA in live cells was not sensitive enough to detect a difference in MUC1 levels between the normal cells and tumor cells. However, we found that Cell ELISA in fixed cells followed by whole cell staining was a dependable method of MUC1 level detection in the normal and tumor cells showing significantly higher levels of MUC1 receptor in the tumor cells when compared to the normal controls. Therefore, we conclude that the Cell ELISA in fixed cells is an efficient method for quantifying the expression of MUC1 by epithelial and neuroectodermal cancer cell lines.


Subject(s)
Enzyme-Linked Immunosorbent Assay/methods , Mucin-1/analysis , Neoplasms, Glandular and Epithelial/metabolism , Neuroectodermal Tumors/metabolism , Antibodies, Monoclonal/immunology , Cell Line, Tumor , Epithelial Cells/metabolism , Humans , Mucin-1/metabolism
8.
J Clin Oncol ; 33(15): 1688-96, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25800760

ABSTRACT

PURPOSE: The outcome for patients with metastatic or recurrent sarcoma remains poor. Adoptive therapy with tumor-directed T cells is an attractive therapeutic option but has never been evaluated in sarcoma. PATIENTS AND METHODS: We conducted a phase I/II clinical study in which patients with recurrent/refractory human epidermal growth factor receptor 2 (HER2) -positive sarcoma received escalating doses (1 × 10(4)/m(2) to 1 × 10(8)/m(2)) of T cells expressing an HER2-specific chimeric antigen receptor with a CD28.ζ signaling domain (HER2-CAR T cells). RESULTS: We enrolled 19 patients with HER2-positive tumors (16 osteosarcomas, one Ewing sarcoma, one primitive neuroectodermal tumor, and one desmoplastic small round cell tumor). HER2-CAR T-cell infusions were well tolerated with no dose-limiting toxicity. At dose level 3 (1 × 10(5)/m(2)) and above, we detected HER2-CAR T cells 3 hours after infusion by quantitative polymerase chain reaction in 14 of 16 patients. HER2-CAR T cells persisted for at least 6 weeks in seven of the nine evaluable patients who received greater than 1 × 10(6)/m(2) HER2-CAR T cells (P = .005). HER2-CAR T cells were detected at tumor sites of two of two patients examined. Of 17 evaluable patients, four had stable disease for 12 weeks to 14 months. Three of these patients had their tumor removed, with one showing ≥ 90% necrosis. The median overall survival of all 19 infused patients was 10.3 months (range, 5.1 to 29.1 months). CONCLUSION: This first evaluation of the safety and efficacy of HER2-CAR T cells in patients with cancer shows the cells can persist for 6 weeks without evident toxicities, setting the stage for studies that combine HER2-CAR T cells with other immunomodulatory approaches to enhance their expansion and persistence.


Subject(s)
Bone Neoplasms/therapy , Immunotherapy/methods , Receptor, ErbB-2/metabolism , Sarcoma/therapy , T-Lymphocytes/immunology , Adolescent , Adult , Bone Neoplasms/metabolism , Child , Female , Humans , Kaplan-Meier Estimate , Magnetic Resonance Imaging , Male , Maximum Tolerated Dose , Neoplasm Metastasis , Neuroectodermal Tumors/metabolism , Neuroectodermal Tumors/therapy , Osteosarcoma/metabolism , Osteosarcoma/therapy , Positron-Emission Tomography , Receptor, ErbB-2/genetics , Receptors, Antigen, T-Cell/chemistry , Recurrence , Sarcoma/metabolism , Sarcoma, Ewing/metabolism , Sarcoma, Ewing/therapy , Tomography, X-Ray Computed , Treatment Outcome , Young Adult
9.
Acta Histochem ; 117(4-5): 460-7, 2015.
Article in English | MEDLINE | ID: mdl-25765111

ABSTRACT

Cyclin D1 amplification and/or overexpression contribute to the loss of the regulatory circuits that govern G1-S transition phase of the cell cycle, playing pivotal roles in different human malignant tumors, including breast, colon, prostate cancer, lymphoma, melanoma and neuroblastoma. In vitro studies have shown that cyclin D1 is overexpressed in Ewing's sarcoma (EWS)/peripheral Primitive Neuroectodermal Tumor (pPNET), but not in rhabdomyosarcoma cell lines. Only a few immunohistochemical studies are available on cyclin D1 expression in EWS/pPNET, which confirmed its expression only in a limited number of cases. The aim of the present study was a comparative immunohistochemical analysis of the expression and distribution of cyclin D1 in a large series of pediatric/adolescent soft tissue EWS/pPNETs and rhabdomyosarcomas (both embryonal and alveolar subtypes) to assess its potential usefulness in their differential diagnosis. Notably cyclin D1 was strongly and diffusely expressed in all cases (20/20) of EWS/pPNET, while it was lacked in all cases (15/15) of rhabdomyosarcomas. Immunohistochemical overexpression of cyclin D1 in EWS/pPNET is a novel finding which could be exploitable as a diagnostic immunomarker for this tumor. Although highly sensitive, cyclin D1 is not specific for EWS/pPNET, and thus it should not be evaluated alone but in the context of a wide immunohistochemical panel. Accordingly, we first emphasize that when pathologists are dealing with a small round blue cell tumor of soft tissues in pediatric/adolescent patients, a strong and diffuse nuclear expression of cyclin D1 is of complementary diagnostic value to CD99 and FLI-1 in confirming diagnosis of EWS/pPNET and in ruling out rhabdomyosarcoma.


Subject(s)
Biomarkers, Tumor/biosynthesis , Cyclin D1/biosynthesis , Gene Expression Regulation, Neoplastic , Neuroectodermal Tumors/metabolism , Rhabdomyosarcoma/metabolism , Sarcoma, Ewing/metabolism , Adolescent , Cell Line, Tumor , Child , Child, Preschool , Female , Humans , Immunohistochemistry , Male , Neuroectodermal Tumors/pathology , Rhabdomyosarcoma/pathology , Sarcoma, Ewing/pathology
10.
Clin Lab ; 60(8): 1383-92, 2014.
Article in English | MEDLINE | ID: mdl-25185426

ABSTRACT

BACKGROUND: Small round blue-cell tumors (SRBCTs) of soft tissue, which mainly include rhabdomyosarcoma (RMS), synovial sarcoma (SS), and Ewing's sarcoma/peripheral primitive neuroectodermal tumors (EWS/ pPNETs), are malignancies with overlapping morphological and immunohistochemical characteristics. Immunohistochemistry is one of the most prevalent and convenient methods for pathological diagnosis; however, differentiation between SRBCT subtypes in the absence of valid diagnostic markers is still very challenging. The purpose of the present study was to investigate diagnostic immunohistochemistry for subtyping soft tissue SRBCTs. METHODS: Seventeen RMS, 25 SS, and 14 EWS/pPNETs were investigated. Reverse transcription RT-PCR and immunohistochemistry was performed to determine a diagnosis. Also, the expression of CD99, FLI1, PAX5, myogenin, and Keratin/EMA was assessed between subtypes. The sensitivity and specificity test was performed to evaluate their diagnostic significance. RESULTS: The sensitivity and specificity of the target markers were evaluated as follows. FLI1 and CD99 expression displayed strong associations in EWS/pPNETs, with OR (95% CI) and p values of 3.82 (1.23 - 11.94), p = 0.021 and 123.50 (12.63 - infinity), p < 0.001, respectively. Keratin/EMA expression did not support the diagnosis of EWS/pPNETs [OR (95% CI) = 0.06 (0.01 - 0.53), p = 0.011]. Myogenin expression displayed strong association with RMS, with high sensitivity and specificity of 94.1% and 100%, respectively. Membrane expression of CD99 did not support the diagnosis of RMS [OR (95% CI) = 0.09 (0.01 - 0.75), p = 0.026]. Keratin/EMA expression strongly indicated SS [OR (95% CI) = 345.00 (29.44 - infinity), p = 0.00011. A ROC curve value of 0.94 indicated that keratin/EMA expression might be a promising biomarker for SS, while separate expression of FLI1 and CD99 did not support the diagnosis of SS. Similarly, myogenin expression in RMS might be a promising biomarker for RMS with a ROC curve value of 0.97. CONCLUSIONS: Diagnosis of SRBCTs should be based on a comprehensive analysis involving morphology and immunoreactivity to a panel of markers.


Subject(s)
Immunohistochemistry/methods , Soft Tissue Neoplasms/diagnosis , Soft Tissue Neoplasms/metabolism , 12E7 Antigen , Adolescent , Adult , Aged , Antigens, CD/metabolism , Cell Adhesion Molecules/metabolism , Child , Child, Preschool , Female , Humans , Keratins/metabolism , Male , Middle Aged , Neuroectodermal Tumors/diagnosis , Neuroectodermal Tumors/metabolism , Odds Ratio , Proto-Oncogene Protein c-fli-1/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Rhabdomyosarcoma/diagnosis , Rhabdomyosarcoma/metabolism , Sarcoma, Ewing/diagnosis , Sarcoma, Ewing/metabolism , Sarcoma, Synovial/diagnosis , Sarcoma, Synovial/metabolism , Sensitivity and Specificity , Young Adult
13.
Tumour Biol ; 33(1): 229-39, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22108870

ABSTRACT

The aim of this study was to investigate the activity distribution in neouroendocrine tumors after diagnostic, or therapeutic, amounts of [(177)Lu-DOTA(0)-Tyr(3)]-octreotate and to investigate how the activity distribution influences the absorbed dose. Furthermore, the activity distribution of a second administration of radiolabeled octreotate was studied. Nude mice with subcutaneously grown human midgut carcinoid (GOT1) were injected intravenously with different amounts of (177)Lu-octreotate. At different time points thereafter (4 h to 13 days), a second injection of [(111)In-DOTA(0)-Tyr(3)]-octreotate was given to estimate the somatostatin receptor (sstr) expression. The activity distribution in the tumors was then determined. Monte Carlo simulations with PENELOPE were performed for dosimetry. Fifty-one out of 58 investigated tumors showed a lower activity concentration in the peripheral part than in the central part of the tumor. The amount of activity injected, or time after administration, did neither influence the relative activity nor the sstr distribution in the tumor. After an initial down-regulation (at 4-24 h), there was an up-regulation of sstr (1.5-2 times, at 7-14 days). Monte Carlo simulations demonstrated an inhomogeneous absorbed dose distribution in the tumor using (177)Lu, with twice as high absorbed dose centrally than peripherally. The high activity concentration centrally and the up-regulation of sstr demonstrated will facilitate fractionated therapy using radiolabeled somatostatin analogues if similar results will be obtained also in patients. The inhomogeneous activity distribution in the tumor has to be taken into account when the absorbed dose distribution in tumor is calculated.


Subject(s)
Carcinoid Tumor/diagnostic imaging , Carcinoid Tumor/diagnosis , Neuroectodermal Tumors/diagnostic imaging , Neuroectodermal Tumors/diagnosis , Octreotide/analogs & derivatives , Organometallic Compounds/pharmacokinetics , Receptors, Somatostatin/biosynthesis , Animals , Carcinoid Tumor/metabolism , Humans , Male , Mice , Mice, Nude , Neuroectodermal Tumors/metabolism , Octreotide/administration & dosage , Octreotide/pharmacokinetics , Organometallic Compounds/administration & dosage , Radionuclide Imaging , Tissue Distribution
16.
Curr Pharm Des ; 16(4): 440-50, 2010.
Article in English | MEDLINE | ID: mdl-20236073

ABSTRACT

Recent evidence suggests that nitric oxide (NO) has a remarkable anti-proliferative action towards dividing neural precursor cells as well as towards cells giving rise to neural-derived tumors. The present paper summarizes essential literature-derived information on this issue and provides novel experimental evidence for these NO-mediated actions regarding a well characterized population of neuronal precursors, the cerebellar granule cell precursors and a cell line of medulloblastoma, a pediatric tumor originating from these same precursor cells undergoing deregulated proliferation. Evidence is presented regarding the NO-mediated regulation of proliferation of neuronal precursor cells both during developmental and adult neurogenesis. Then, the role of NO in the control of proliferation of neural-derived tumor cells, such as PC12 and neuroblastoma cells, is discussed. Novel experimental data are provided documenting the anti-proliferative action of NO towards basal and mitogen-stimulated division of rat cerebellar granule cell precursors, as well as towards medulloblastoma DAOY cells. Finally, some molecular correlates of NO action on cell cycle regulation are discussed. Overall, the data presented and discussed here highlight similarities at the molecular level between physiologic processes regulating normal proliferation of neural precursors and pathologic deregulation of these processes leading to tumor formation.


Subject(s)
Cell Proliferation , Neuroectodermal Tumors/pathology , Neurons/cytology , Nitric Oxide/metabolism , Animals , Humans , Neuroectodermal Tumors/metabolism , Neurons/metabolism
17.
Cancer Biol Ther ; 9(4): 286-94, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20190567

ABSTRACT

There is now accumulating evidence showing that some tumors may arise from transformed stem cells. In this study we demonstrate that adult bone marrow- derived mesenchymal stem cells (MSCs) undergo neoplastic transformation induced by the human polyomavirus JCV, early protein, T-antigen, and are tumorigenic when transplanted into the flanks of Nude mice as compared to non-transformed MSCs. Histologically, the tumors are heterogeneous with mesenchymal and neural crest characteristics as evidenced by expression of the neural crest markers p75, SOX-10, and S-100, with populations of tumor cells exhibiting characteristics of primitive neuroectodermal cells. In addition, a subset of T-antigen positive tumor cells exhibit a high proliferation index as detected by Ki-67 labeling, and co-express CD133, a marker which is expressed on cancer stem cells. These results show that tumors with neuroectodermal characteristics may arise from transformation of MSCs, a globally accessible adult stem cell with multipotent differentiation capacity. In light of earlier reports on the association of JCV with a broad variety of human tumors, our data suggests that T-antigen transformation of adult stem cells with a multipotent capacity can serve as a possible common origin for some of these cancers, and offers a novel model for oncogenesis.


Subject(s)
Adult Stem Cells/pathology , Antigens, Viral, Tumor/metabolism , Cell Transformation, Neoplastic/pathology , JC Virus/metabolism , Mesenchymal Stem Cells/pathology , Neuroectodermal Tumors/pathology , Adult Stem Cells/metabolism , Animals , Antigens, Viral, Tumor/genetics , Cell Transformation, Neoplastic/metabolism , Humans , Mesenchymal Stem Cells/metabolism , Mice , Mice, Nude , Neoplasm Transplantation , Neuroectodermal Tumors/metabolism , Rats
18.
Oncogene ; 28(43): 3814-24, 2009 Oct 29.
Article in English | MEDLINE | ID: mdl-19668229

ABSTRACT

Reticulons are a family of highly conserved proteins, localized in the endoplasmic reticulum (ER) and involved in different cellular functions, such as intracellular membrane trafficking, apoptosis and nuclear envelope formation. The reticulon protein family consists of four members, but their specific functions are presently poorly understood. RTN-1C overexpression triggers apoptosis, regulating ER stress versus DNA damage-induced cell death in a mutually exclusive way. The different RTN isoforms share a C-terminal reticulon homology domain containing two hydrophobic segments and a 66-amino acid hydrophilic loop. In the C-terminal region of RTN-1C, a unique consensus sequence (GAKRH) has recently been identified, showing 100% identity with the DNA-binding domain of histone H4. In this study, we show that this sequence is essential for RTN-1C-mediated apoptosis. It is noteworthy that the lysine 204 present in this region is post-translationally modified by acetylation and that this event is associated with a significant decrease in histone deacetylase activity and contributes to RTN-1C binding to DNA. These data demonstrate a molecular mechanism by which RTN-1C controls apoptosis and indicate this protein to be a novel potential target for cancer therapy.


Subject(s)
Endoplasmic Reticulum/metabolism , Histone Deacetylase Inhibitors , Nerve Tissue Proteins/physiology , Neuroectodermal Tumors/metabolism , Acetylation , Apoptosis , Cell Line, Tumor , DNA/metabolism , Humans , Nerve Tissue Proteins/chemistry
19.
Turk Neurosurg ; 19(3): 260-4, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19621291

ABSTRACT

Glioblastoma multiforme (GB) is the most aggressive, and the most frequent primary tumor of the brain in adults. Presence of less-differentiated areas which exhibit a small cell morphology and neural immunophenotype is quite uncommon in GBs. Tumor tissue which had been determined in the frontotemporal region of a 61-year-old female patient and evaluated to be consistent with GB radiologically was subjected to total excision. Histopathological examination revealed two different components making up the tumor tissue. Using a morphological and immunophenotypic approach, the predominant component of the tumor was found to bear the properties of classic GB. The other component was composed of undifferentiated areas exhibiting small cell morphology and diffuse neuronal immunophenotype. The case was diagnosed as 'Glioblastoma with primitive neuroectodermal tumor-like component'. The patient who had been subjected to postoperative radiotherapy, showed no sign of recurrence during the follow-up examination performed on the 9th month. The histogenesis and prognostic significance of neuronal differentiation observed in glial tumors are not known yet. Inclusion of this component in pathological reports is important regarding formation of a database for future studies.


Subject(s)
Carcinoma, Small Cell/pathology , Glioblastoma/pathology , Neuroectodermal Tumors/pathology , Biomarkers, Tumor/metabolism , Biopsy , Carcinoma, Small Cell/metabolism , Carcinoma, Small Cell/surgery , Cell Differentiation , Female , Glioblastoma/metabolism , Glioblastoma/surgery , Humans , Immunophenotyping , Magnetic Resonance Imaging , Middle Aged , Neuroectodermal Tumors/metabolism , Neuroectodermal Tumors/surgery , Prognosis , Synaptophysin/metabolism
20.
Ann Clin Lab Sci ; 39(2): 160-6, 2009.
Article in English | MEDLINE | ID: mdl-19429803

ABSTRACT

In 3 patients with the Ewing family of tumors (EFT), morphoproteomic analyses of the tumors revealed constitutive activation of the mTOR, ERK, and NF-kappaB pathways, as evidenced by: (a) expression of phosphorylated (p)-mTOR, p-p70S6K, p-ERK 1/2, and p-NF-kappaB proteins using phosphospecific immunohistochemical probes directed against the activation sites; (b) nuclear translocation of p-p70S6K, p-ERK 1/2, and p-NF-kappaB p65; and (c) correlative expression of Ki-67 and Skp2 proteins consistent with cell cycling consequent to signal transduction by these pathways of convergence. This study examines the cytogenetic and molecular correlates and provides insight into therapeutic strategies relevant to this morphoproteomic profile. Based on a literature review, these observations appear to be the first morphoproteomic study of such pathways of convergence in tumors from EFT patients.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , NF-kappa B/metabolism , Neuroectodermal Tumors/metabolism , Protein Kinases/metabolism , Sarcoma, Ewing/metabolism , 12E7 Antigen , Antigens, CD/physiology , Cell Adhesion Molecules/physiology , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Nucleus/pathology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/genetics , Flavonoids/pharmacology , Gene Expression Profiling , Humans , NF-kappa B/genetics , Neuroectodermal Tumors/genetics , Phosphorylation , Protein Kinases/genetics , Proteome , RNA, Messenger/genetics , Sarcoma, Ewing/enzymology , Signal Transduction/physiology , TOR Serine-Threonine Kinases , Transforming Growth Factor beta/genetics , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL