Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
BMC Complement Altern Med ; 17(1): 342, 2017 Jun 29.
Article in English | MEDLINE | ID: mdl-28662725

ABSTRACT

BACKGROUND: Rhazya stricta Decne. is a medicinal plant that is widespread in Saudi Arabia and desert areas of the Arabian Peninsula. Its extract contains alkaloids, tannins, and flavonoids that are involved in different biological activities. The study aim was to evaluate the effects of Rhazya stricta plant extracts on the proliferation and differentiation of NTERA-2 (NT2) pluripotent embryonal carcinoma cells. METHODS: Soxhlet extraction was carried out using different solvents to extract stems, leaves and fruit parts of this plant. Cytotoxicity was evaluated by an MTS cell viability assay. The ability of the plant extract to induce cell differentiation was examined phenotypically using an inverted light microscope. The expression of pluripotency markers was investigated by reverse transcriptase polymerase chain reaction (RT-PCR) and immunocytochemistry. Phytochemical screening of chloroform stem extracts was carried out and a chromatographic fingerprint was generated using gas chromatography - mass spectrometry (GC-MS). RESULTS: Chloroform stem extract induced differentiation of NT2 cells at 5 µg/ml, and the differentiated cells exhibited neurite formation. Following induction of differentiation, there was significant down-regulation of the pluripotency marker genes Oct4 and Sox2. In addition, the surface antigen pluripotency marker, TRA-1-60, was strongly down-regulated. Phytochemical analysis of the extract showed the presence of alkaloids and saponins. The chromatogram revealed the presence of fifteen compounds with different retention times. CONCLUSION: Our results demonstrate for the first time that chloroform stem extract of R. stricta can induce neuronal differentiation of stem cells at an early stage and may contain potential therapeutic agent that can be used in neurodegenerative diseases.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apocynaceae/chemistry , Carcinoma, Embryonal/physiopathology , Cell Differentiation/drug effects , Plant Extracts/pharmacology , Antineoplastic Agents, Phytogenic/isolation & purification , Carcinoma, Embryonal/drug therapy , Carcinoma, Embryonal/genetics , Carcinoma, Embryonal/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Plant Extracts/isolation & purification , Plant Leaves/chemistry , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism
2.
J Neurosci Methods ; 204(1): 87-103, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22101194

ABSTRACT

P19 embryonal carcinoma (EC) cells are an invaluable tool for approximating the mechanisms that govern neuronal differentiation but with an enormous degree of simplification and have primarily been used to model the early stages of neurogenesis. However, they are often cultured under conditions that promote unrestricted non-neuronal growth that compromises neuronal viability. In this study we report an improved method to differentiate P19 EC cells that gives rise to high yields of functionally and morphologically mature neurons while significantly reducing the over-growth of non-neuronal cells in the cultures. In this protocol, P19 EC cells are induced in Minimum Essential Medium alpha supplemented with all-trans retinoic acid (RA) and 2.5% serum, and cultured as a monolayer. After RA-induction, cells are cultured on Matrigel coated-plates using defined media comprised of Neurobasal-A medium temporally supplemented with N2 and then B-27 for the remaining culture period. By treating the culture with Cytosine ß-d-arabinofuranoside and 2'-Deoxycytidine for five days, the cultures are reliably promoted toward the neuronal differentiation vs non-neuronal differentiation, this accounting for a progressive neuronal enrichment of the cultures reaching 56% after 20 days of culture. P19-derived neural progenitor cells progressively expressed neuronal markers such as NeuN, Calretinin, Calbindin and Synapsin I in close resemblance to that occurring in vivo in the central nervous system (CNS). Furthermore, RA-induced P19 EC cells progressively acquired functional neuronal traits and after approximately 3 weeks in culture revealed mature neurophysiological properties, characteristics of CNS neurons. This protocol allows for a more specific assessment of the neuronal differentiation processes in vitro.


Subject(s)
Carcinoma, Embryonal/pathology , Carcinoma, Embryonal/physiopathology , Cell Culture Techniques/methods , Neurogenesis/physiology , Neurons/cytology , Neurons/physiology , Tretinoin/pharmacology , Animals , Cell Differentiation , Cell Line , Cell Line, Tumor , Cell Proliferation , Mice
3.
BMC Cancer ; 9: 383, 2009 Oct 29.
Article in English | MEDLINE | ID: mdl-19874621

ABSTRACT

BACKGROUND: Testicular germ cell tumors (TGCTs) are classified as seminonas or non-seminomas of which a major subset is embryonal carcinoma (EC) that can differentiate into diverse tissues. The pluripotent nature of human ECs resembles that of embryonic stem (ES) cells. Many Wnt signalling species are regulated during differentiation of TGCT-derived EC cells. This study comprehensively investigated expression profiles of Wnt signalling components regulated during induced differentiation of EC cells and explored the role of key components in maintaining pluripotency. METHODS: Human embryonal carcinoma cells were stably infected with a lentiviral construct carrying a canonical Wnt responsive reporter to assess Wnt signalling activity following induced differentiation. Cells were differentiated with all-trans retinoic acid (RA) or by targeted repression of pluripotency factor, POU5F1. A Wnt pathway real-time-PCR array was used to evaluate changes in gene expression as cells differentiated. Highlighted Wnt pathway genes were then specifically repressed using siRNA or stable shRNA and transfected EC cells were assessed for proliferation, differentiation status and levels of core pluripotency genes. RESULTS: Canonical Wnt signalling activity was low basally in undifferentiated EC cells, but substantially increased with induced differentiation. Wnt pathway gene expression levels were compared during induced differentiation and many components were altered including ligands (WNT2B), receptors (FZD5, FZD6, FZD10), secreted inhibitors (SFRP4, SFRP1), and other effectors of Wnt signalling (FRAT2, DAAM1, PITX2, Porcupine). Independent repression of FZD5, FZD7 and WNT5A using transient as well as stable methods of RNA interference (RNAi) inhibited cell growth of pluripotent NT2/D1 human EC cells, but did not appreciably induce differentiation or repress key pluripotency genes. Silencing of FZD7 gave the greatest growth suppression in all human EC cell lines tested including NT2/D1, NT2/D1-R1, Tera-1 and 833K cells. CONCLUSION: During induced differentiation of human EC cells, the Wnt signalling pathway is reprogrammed and canonical Wnt signalling induced. Specific species regulating non-canonical Wnt signalling conferred growth inhibition when targeted for repression in these EC cells. Notably, FZD7 repression significantly inhibited growth of human EC cells and is a promising therapeutic target for TGCTs.


Subject(s)
Carcinoma, Embryonal/metabolism , Cell Differentiation , Signal Transduction , Wnt Proteins/metabolism , Carcinoma, Embryonal/drug therapy , Carcinoma, Embryonal/genetics , Carcinoma, Embryonal/physiopathology , Cell Line , Gene Expression Regulation, Neoplastic , Humans , Wnt Proteins/antagonists & inhibitors , Wnt Proteins/genetics
4.
Neurochem Res ; 33(10): 2118-25, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18465225

ABSTRACT

Nervous system development at early stage is in hypoxic environment. Very little is known about the role of hypoxia in neuronal development. P19 embryonal carcinoma (EC) cells are a widely used model for studying early neuronal development. In this study we investigated the roles of hypoxia in differentiation of dopaminergic neurons derived from P19 EC cells. Results demonstrate that hypoxia increases the percentage of differentiated neurons, especially neurons of dopaminergic phenotype. To investigate the potential mechanism involved in hypoxia promoted differentiation of dopaminergic neurons, we measured the expression of hypoxia-inducible factor 1alpha (HIF-1alpha), based on its characteristic response to hypoxia. The result shows that HIF-1alpha mRNA level in P19 EC cells increases after hypoxia treatment. It is known that HIF-1alpha regulates the expression of tyrosine hydroxylase (TH) gene through binding to its promoter. Therefore, we propose that the underlying mechanism for hypoxia promoted differentiation of dopaminergic neurons was mediated by HIF-1alpha up-regulation under hypoxia.


Subject(s)
Carcinoma, Embryonal/physiopathology , Cell Hypoxia/physiology , Dopamine/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Neurons/cytology , Animals , Carcinoma, Embryonal/pathology , Cell Differentiation , Mice , Pluripotent Stem Cells/physiology , RNA, Messenger/metabolism , Teratocarcinoma/pathology , Teratocarcinoma/physiopathology , Tumor Cells, Cultured , Tyrosine 3-Monooxygenase/biosynthesis , Up-Regulation
5.
Brain ; 130(Pt 4): 1076-88, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17301082

ABSTRACT

Although it is well known that cisplatin causes a sensory neuropathy, the primary site of involvement is not established. The clinical symptoms localized in a stocking-glove distribution may be explained by a length dependent neuronopathy or by a distal axonopathy. To study whether the whole neuron or the distal axon was primarily affected, we have carried out serial clinical and electrophysiological studies in 16 males with testicular cancer before or early and late during and after treatment with cisplatin, etoposide and bleomycin at limited (<400 mg/m2 cisplatin), conventional (approximately 400 mg/m2 cisplatin) or high (>400 mg/m2 cisplatin) doses. At cumulative doses of cisplatin higher than 300 mg/m2 the patients lost distal tendon and H-reflexes and displayed reduced vibration sense in the feet and the fingers. The amplitudes of sensory nerve action potentials (SNAP) from the fingers innervated by the median nerve and the dorsolateral side of the foot innervated by the sural nerve were 50-60% reduced, whereas no definite changes occurred at lower doses. The SNAP conduction velocities were reduced by 10-15% at cumulative doses of 400-700 mg/m2 consistent with loss of large myelinated fibres. SNAPs from primarily Pacinian corpuscles in digit 3 and the dorsolateral side of the foot evoked by a tactile probe showed similar changes to those observed in SNAPs evoked by electrical stimulation. At these doses, somatosensory evoked potentials (SEPs) from the tibial nerve had increased latencies of peripheral, spinal and central responses suggesting loss of central processes of large dorsal root ganglion cells. Motor conduction studies, autonomic function and warm and cold temperature sensation remained unchanged at all doses of cisplatin treatment. The results of these studies are consistent with degeneration of large sensory neurons whereas there was no evidence of distal axonal degeneration even at the lowest toxic doses of cisplatin.


Subject(s)
Antineoplastic Agents/adverse effects , Carcinoma, Embryonal/drug therapy , Cisplatin/adverse effects , Neurons, Afferent/drug effects , Peripheral Nervous System Diseases/chemically induced , Testicular Neoplasms/drug therapy , Action Potentials/physiology , Adult , Bleomycin/adverse effects , Carcinoma, Embryonal/physiopathology , Etoposide/adverse effects , Evoked Potentials, Somatosensory/physiology , Humans , Longitudinal Studies , Male , Middle Aged , Neural Conduction/physiology , Neurons, Afferent/physiology , Peripheral Nervous System Diseases/physiopathology , Prospective Studies , Reflex/physiology , Seminoma/drug therapy , Seminoma/physiopathology , Sensation Disorders/chemically induced , Sensation Disorders/physiopathology , Sensory Thresholds/physiology , Testicular Neoplasms/complications , Touch/physiology
6.
Cancer Res ; 66(2): 820-7, 2006 Jan 15.
Article in English | MEDLINE | ID: mdl-16424014

ABSTRACT

Adult male germ cell tumors (GCTs) comprise distinct groups: seminomas and nonseminomas, which include pluripotent embryonal carcinomas as well as other histologic subtypes exhibiting various stages of differentiation. Almost all GCTs show 12p gain, but the target genes have not been clearly defined. To identify 12p target genes, we examined Affymetrix (Santa Clara, CA) U133A+B microarray ( approximately 83% coverage of 12p genes) expression profiles of 17 seminomas, 84 nonseminoma GCTs, and 5 normal testis samples. Seventy-three genes on 12p were significantly overexpressed, including GLUT3 and REA (overexpressed in all GCTs) and CCND2 and FLJ22028 (overexpressed in all GCTs, except choriocarcinomas). We characterized a 200-kb gene cluster at 12p13.31 that exhibited coordinated overexpression in embryonal carcinomas and seminomas, which included the known stem cell genes NANOG, STELLA, and GDF3 and two previously uncharacterized genes. A search for other coordinately regulated genomic clusters of stem cell genes did not reveal any genomic regions similar to that at 12p13.31. Comparison of embryonal carcinoma with seminomas revealed relative overexpression of several stem cell-associated genes in embryonal carcinoma, including several core "stemness" genes (EBAF, TDGF1, and SOX2) and several downstream targets of WNT, NODAL, and FGF signaling (FGF4, NODAL, and ZFP42). Our results indicate that 12p gain is a functionally relevant change leading to activation of proliferation and reestablishment/maintenance of stem cell function through activation of key stem cell genes. Furthermore, the differential expression of core stem cell genes may explain the differences in pluripotency between embryonal carcinomas and seminomas.


Subject(s)
Chromosomes, Human, Pair 12 , Gene Expression Profiling , Neoplasms, Germ Cell and Embryonal/genetics , Pluripotent Stem Cells/physiology , Testicular Neoplasms/genetics , Carcinoma, Embryonal/genetics , Carcinoma, Embryonal/physiopathology , Cell Proliferation , Down-Regulation , Humans , Male , Multigene Family , Neoplasms, Germ Cell and Embryonal/physiopathology , Oligonucleotide Array Sequence Analysis , Prohibitins , Seminoma/genetics , Seminoma/physiopathology , Testicular Neoplasms/physiopathology
7.
Br J Cancer ; 93(12): 1382-7, 2005 Dec 12.
Article in English | MEDLINE | ID: mdl-16333245

ABSTRACT

Testicular neoplasms are comprised of a variety of histologically different forms, and their pathogenesis has not been elucidated. Dysadherin is a recently described cell membrane glycoprotein, which has an anticell-cell adhesion function and downregulates E-cadherin. In this study, we examined immunohistochemically the expression of E-cadherin and dysadherin in 120 testicular neoplasms (37 seminomas-26 classic, five spermatocytic and six anaplastic-, 45 embryonal carcinomas, 10 mixed germ cell tumours, two yolk sac tumours, 10 mature and eight immature teratomas and eight non-Hodgkin B-cell lymphomas), clinical stage I. The intensity, the expression pattern and the percentage of neoplastic cell staining was recorded and correlated with the histologic type and vascular/lymphatic invasion. Dysadherin was not expressed in non-neoplastic germ cells, neither in CIS/ITGCNU, but it was highly expressed in all types of germ cell tumours, that demonstrated either embryonic phenotype or somatic differentiation, in most terminally differentiated neoplasms, and in all lymphomas. Dysadherin expression did not correlate with vascular invasion. Increased dysadherin expression was correlated with aberrant E-cadherin expression in most tumours. In 17% of embryonal carcinomas colocalisation of dysadherin and membranous E-cadherin staining was noted. This is the first report on dysadherin expression and its association with E-cadherin in testicular tumours. Since dysadherin is not normally expressed in non-neoplastic testis, it is conceivable that it plays a role in the neoplastic transformation of germ cells. In testicular tumours, as in other neoplasms, dysadherin downregulates E-cadherin expression, at least in part.


Subject(s)
Cadherins/biosynthesis , Carcinoma, Embryonal/genetics , Carcinoma, Embryonal/physiopathology , Lymphoma, Non-Hodgkin/genetics , Lymphoma, Non-Hodgkin/physiopathology , Membrane Glycoproteins/biosynthesis , Neoplasm Proteins/biosynthesis , Testicular Neoplasms/genetics , Testicular Neoplasms/physiopathology , Adolescent , Adult , Aged , Cadherins/physiology , Cell Adhesion , Gene Expression Profiling , Humans , Immunohistochemistry , Ion Channels , Male , Membrane Glycoproteins/physiology , Microfilament Proteins , Middle Aged , Neoplasm Invasiveness , Neoplasm Proteins/physiology
8.
Biochim Biophys Acta ; 1731(1): 48-56, 2005 Oct 15.
Article in English | MEDLINE | ID: mdl-16168501

ABSTRACT

Testicular germ cell tumors (TGCTs) are the most common carcinomas of young men aged 15-35. The molecular events involved in TGCT genesis are poorly understood. TGCTs have near universal amplification of the short arm of chromosome 12, however positional cloning efforts have not identified causative genes on 12p involved in formation or progression of TGCTs. Human embryonal carcinoma (EC) are the stem cells of TGCTs and are pluripotent. EC cells terminally differentiate toward a neuronal lineage with all-trans retinoic acid (RA) treatment resulting in a concomitant G1 cell cycle arrest and loss of tumorigenicity. Our efforts to define the molecular mechanisms of RA-mediated tumor cell differentiation at a critical "commitment to differentiate" window has identified a cassette of genes on 12p that are repressed with RA precisely as EC cells lose tumorigenic potential. These are Nanog, CD9, EDR1 (PHC1), SCNN1A, GDF3, Glut3 and Stella. The master pluripotency regulator Oct4 is located on chromosome 6 and is also repressed by RA. Notably, knockdown of Oct4 with siRNA results in repression of basal Nanog, EDR1, GDF3 and Stella gene expression. Nanog has recently been identified to play a role in maintenance of the pluripotency of mouse embryonic stem cells and CD9, EDR1, GDF3, and Stella have each been implicated as stem cell markers. Since RA suppresses the tumorigenicity of EC cells, these genes may have a critical role in the etiology of TGCTs, suggesting a link between enforced pluripotency and transformation.


Subject(s)
Carcinoma, Embryonal/physiopathology , Chromosomes, Human, Pair 12/genetics , Pluripotent Stem Cells/physiology , Tretinoin/pharmacology , Adolescent , Adult , Cell Differentiation , Cell Line, Tumor , Chromosomes, Human, Pair 12/drug effects , Humans , Male , Octamer Transcription Factor-3/genetics , Oligonucleotide Array Sequence Analysis , Pluripotent Stem Cells/drug effects , RNA, Small Interfering/physiology , Testicular Neoplasms/physiopathology
9.
Urol Res ; 32(5): 332-7, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15118852

ABSTRACT

In situ end-labeling (ISEL) of internucleosomal 3' DNA strand breaks and the morphological proof of nuclear chromatin condensation are two widely used methods to investigate and quantify apoptosis. However, it is still unclear whether both processes are linked with each other and if quantifying apoptosis by both methods leads to comparable results. Therefore, internucleosomal DNA fragmentation and chromatin condensation were measured simultaneously on double-fluorescence-labeled sections of 62 testicular tumors (47 nonseminomatous tumors and 15 seminomas) using immunofluorescence microscopy. Different apoptotic indices (AI), based on DNA fragmentation and/or morphological criteria were determined. The AI were quantified. Morphologically obtained AI ranged between 1.99% for non-seminomatous tumors and 0.88% for seminomas. The detection of DNA fragmentation values ranged between 8.15% for non-seminomatous tumors and 2.70% for seminomas. Only about 30% of all apoptotic cells could be detected with the morphological method compared to 80% using ISEL in both tumor entities. Therefore, the equivalence of investigations using different apoptosis detection methods in human testicular cancer seems questionable.


Subject(s)
Apoptosis , Carcinoma, Embryonal/physiopathology , Neoplasms, Germ Cell and Embryonal/physiopathology , Seminoma/physiopathology , Testicular Neoplasms/physiopathology , Carcinoma, Embryonal/pathology , DNA Fragmentation , Humans , Male , Neoplasms, Germ Cell and Embryonal/pathology , Seminoma/pathology , Testicular Neoplasms/pathology
10.
Nihon Hinyokika Gakkai Zasshi ; 95(3): 621-5, 2004 Mar.
Article in Japanese | MEDLINE | ID: mdl-15103926

ABSTRACT

A 28-year-old man, who had undergone right orchiectomy and prophylactic irradiation for stage I seminoma 6 years ago, developed left testicular tumor. Since the secondary tumor was localized in the lower pole of the testis, partial orchiectomy was performed with an attempt to preserve the testicular function. The pathological finding of the surgical specimen was a mixed type testicular tumor consisting of seminoma, embryonal carcinoma and teratoma elements. Postoperative chemotherapy with 3 courses of BEP regimen resulted in azoospermia, but the impaired spermatogenesis recovered to a normal range within 18 months with no evidence of tumor recurrence and his wife delivered a healthy baby 2 years later. For the synchronous or metachronous bilateral testicular tumors, the combination of organ sparing surgery and chemotherapy could be a treatment of choice.


Subject(s)
Carcinoma, Embryonal/surgery , Fertility , Neoplasms, Second Primary , Orchiectomy/methods , Seminoma/surgery , Teratoma/surgery , Testicular Neoplasms/surgery , Adult , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bleomycin/administration & dosage , Bleomycin/adverse effects , Carcinoma, Embryonal/physiopathology , Cisplatin/administration & dosage , Cisplatin/adverse effects , Combined Modality Therapy , Etoposide/administration & dosage , Etoposide/adverse effects , Humans , Male , Oligospermia/chemically induced , Seminoma/physiopathology , Testicular Neoplasms/physiopathology , Testis/physiology , Testis/physiopathology , Treatment Outcome
11.
Urol Res ; 32(1): 28-35, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14579108

ABSTRACT

Testis tumors of embryonal origin (ten metastasized, six non-metastasized) and 17 mixed testis cell carcinomas (eight metastasized, nine non-metastasized) were examined. A triple immunofluorescence microscopic labeling procedure allowed the simultaneous detection of two features of apoptosis, namely morphological changes in the nucleus (DNA condensation visualized by DAPI staining) and the process of DNA fragmentation (TdT-assay) in tumor cells as well as T-cells (recognized by their CD45RO epitope). Both methods for apoptosis detection showed similar apoptotic indices (AI) only in 2.6% of all tumors. Most tumors (81.6%) showed more cells with DNA fragments than condensed chromatin, but in a number of cases (10.5%) the opposite pattern was found. These data add to the few published in vivo examinations of apoptosis using different methods and help to explain differences in the judgment of apoptosis significance for tumor prognosis. With regard to tumorigenesis, non-metastasized testis tumors were characterized by higher AIs of tumor cells and T-cells compared with metastasized tumors, which could be interpreted as a characteristic of tumors in an earlier stage of their development into an apoptosis-resistant phenotype. For the first time, in metastasized tumors a 5 to 25-fold increase of the T-cell's AIs over the corresponding AIs of tumor cells was shown. This suggests a successful counterattack of tumor cells, thus supporting the process of metastasis. However, only ten out of 33 tumors revealed these AI changes, which again highlights that tumor biology cannot be predicted by a single parametric approach. It remains to be seen whether these characteristics might be suitable for a reliable prediction of metastasis.


Subject(s)
Apoptosis , Carcinoma, Embryonal/physiopathology , Carcinoma/physiopathology , Testicular Neoplasms/physiopathology , Adult , Carcinoma/secondary , Carcinoma, Embryonal/secondary , Cell Nucleus/ultrastructure , Chromatin/ultrastructure , DNA Fragmentation , Fluorescent Antibody Technique , Humans , Male , T-Lymphocytes
12.
Brain Res Dev Brain Res ; 140(1): 45-56, 2003 Jan 10.
Article in English | MEDLINE | ID: mdl-12524175

ABSTRACT

Trip15/CSN2 is a transcriptional corepressor/a component of COP9 signalosome (CSN) and participates in various signaling pathways. However, participation of Trip15/CSN2 in neural differentiation is still obscure. Here, we show that Trip15/CSN2 plays a critical role in neuronal differentiation. The expression of Trip15/CSN2 mRNA was induced at an early stage of neuronal differentiation in the retinoic acid (RA)-treated P19 cells, but not in the triiodothyronine (T3)-primed cardiac muscular cell differentiation. The expression of Trip15/CSN2 mRNA in the rat brain was detected at E14 and the protein was localized in the nuclei of neonatal rat CNS neurons. Enforced expression of sense rat Trip15/CSN2 mRNA caused the downregulation of Oct-3/4 mRNA expression and was sufficient to convert P19 cells into neurons, but not glial cells, only after the aggregation without RA. In the presence of RA, exogenous expression of the sense mRNA caused the intense and rapid induction of neurogenic Brn-2 and Mash-1 mRNA expressions accompanying the strong downregulation of Oct-3/4 mRNA expression, and stimulated both neuronal and glial cell differentiations of P19 cells. In contrast, enforced expression of the antisense mRNA suppressed the commitment of RA-treated aggregation form of P19 cells to neuronal lineage. These data strongly suggest that Trip15/CSN2 could implicate in the commitment of multipotent embryonal carcinoma (EC) cells to neuronal fate through the downregulation of Oct-3/4 which suppresses neurogenic genes. Moreover, in addition to Trip15/CSN2, RA-regulated other factor(s) may be required for glial cell differentiation.


Subject(s)
Carcinoma, Embryonal/physiopathology , Cell Differentiation/physiology , Gene Expression Regulation, Neoplastic , Neurons/cytology , Nuclear Proteins , Receptors, Thyroid Hormone/genetics , Transcription Factors/genetics , Animals , Base Sequence , Blotting, Northern , COP9 Signalosome Complex , Cloning, Molecular , DNA Primers , Female , Immunohistochemistry , Male , Mice , RNA, Messenger/genetics , Rats , Rats, Inbred F344 , Transcription, Genetic , Tumor Cells, Cultured
13.
Histol Histopathol ; 16(3): 909-22, 2001 07.
Article in English | MEDLINE | ID: mdl-11510982

ABSTRACT

Mouse F9 embryocarcinoma (EC) cells constitute a well established cell-autonomous model system for investigating retinoid signaling in vitro as, depending on culture conditions, retinoic acid (RA) can induce their differentiation into either primitive, parietal or visceral extraembryonic endoderm-like cells. These RA-induced differentiations are accompanied by decreases in proliferation rates, modifications of expression of subsets of RA-target genes, and induction of apoptosis. To elucidate the roles played by the multiple retinoid receptors (RARs and RXRs) in response to RA treatments, F9 EC cells lacking one or several RARs or RXRs were engineered through homologous recombination. Mutated RARs and/or RXRs were then reexpressed in given RAR or RXR null backgrounds. WT and mutant cells were also treated with different combinations of ligands selective for RXRs and/or for each of the three RAR isotypes. These studies lead to the conclusion that most RA-induced events (e.g. primitive and visceral differentiation, growth arrest, apoptosis and activation of expression of a number of genes) are transduced by RARgamma/RXRalpha heterodimers, whereas some other events (e.g. parietal differentiation) are mediated by RARalpha/RXRalpha. heterodimers. They also demonstrate that both AF-1 and AF-2 activation functions of RARs and RXRs, as well as their phosphorylation, are differentially required in these RA-induced events. In RARgamma/RXRalpha heterodimers, the phosphorylation of RARgamma is necessary for triggering primitive differentiation, while that of RXRalpha is required for growth arrest. On the other hand, phosphorylation of RARalpha is necessary for parietal differentiation. Thus, retinoid receptors are sophisticated signal integrators that transduce not only the effects of their cognate ligands, but also those of ligands that bind to membrane receptors.


Subject(s)
Carcinoma, Embryonal/pathology , Carcinoma, Embryonal/physiopathology , Receptors, Retinoic Acid/physiology , Retinoids/physiology , Transcription Factors/physiology , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Carcinoma, Embryonal/drug therapy , Carcinoma, Embryonal/genetics , Cell Differentiation/drug effects , Cell Division/drug effects , DNA, Neoplasm/genetics , Genetic Engineering , Mice , Models, Biological , Molecular Sequence Data , Phosphorylation , Receptors, Retinoic Acid/deficiency , Receptors, Retinoic Acid/genetics , Retinoid X Receptors , Signal Transduction , Transcription Factors/deficiency , Transcription Factors/genetics , Tretinoin/pharmacology , Tumor Cells, Cultured
14.
Urol Res ; 27(5): 368-75, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10550526

ABSTRACT

Disorders in the regulation of apoptotic cell death may contribute to cancer. Furthermore, lymphocytes are supposed to play a role in counteracting tumorigenesis by inducing apoptosis in different human tumors. In this study, for the first time, tumor cell and lymphocyte apoptosis were investigated systematically in human embryonal cell carcinoma. DNA fragmentation and DNA condensation were measured simultaneously on double-fluorescence-labeled testis tumor sections using immunofluorescence microscopy. Different apoptotic indices (AIs), based either on biochemical (DNA fragmentation) or morphological criteria (DNA condensation) alone or on a combination of both, were determined in different histological regions in and around the tumor. Using morphological criteria alone, 40-75% of all apoptotic cells were not detected. Based on previous observations this finding might be related to subsets of apoptotic cells which induce the process of DNA condensation without activation of processes responsible for DNA fragmentation. Moreover, the AIs of tumor cells and lymphocytes were highest in the tumor region, compared with regions around the tumor and distant from it; these findings are discussed in the context of the Fas/FasL system.


Subject(s)
Apoptosis , Carcinoma, Embryonal/physiopathology , Testicular Neoplasms/physiopathology , Carcinoma, Embryonal/pathology , Carcinoma, Embryonal/secondary , DNA/chemistry , DNA Fragmentation , Humans , Lymphocytes/physiology , Male , Methods , Testicular Neoplasms/pathology , Testis/pathology , Testis/physiopathology
15.
Dev Biol ; 213(2): 390-404, 1999 Sep 15.
Article in English | MEDLINE | ID: mdl-10479456

ABSTRACT

Besides nuclear retinoid receptors and cellular retinoid binding proteins also retinoic acid (RA)-synthesizing enzymes (using all-trans-retinal as substrate) and RA-catabolizing enzymes (producing hydroxylated products) may explain the specific effects of retinoids. In the past we have established an active role for 4-hydroxy-RA and 4-oxo-RA, which originally were considered to be inactive retinoids, but in fact are highly active modulators of positional specification in Xenopus development. Here we present evidence for a specific role of hydroxylated RA metabolites in the onset of neuronal differentiation. 4-Hydroxy- and 18-hydroxy-RA are products of the hydroxylation of RA by a novel cytochrome P450 (CYP)-type of enzyme, CYP26, expression of which is rapidly induced by RA. P19 embryonal carcinoma (EC) cell lines stably expressing hCYP26 undergo extensive and rapid neuronal differentiation in monolayer at already low concentrations of RA, while normally P19 cells under these conditions differentiate only in endoderm-like cells. Our results indicate that the effects on growth inhibition and RARbeta transactivation of P19 EC cells are mediated directly by RA, while the onset of neuronal differentiation and the subsequent expression of neuronal markers is mediated by hCYP26 via the conversion of RA to its hydroxylated products.


Subject(s)
Carcinoma, Embryonal/pathology , Carcinoma, Embryonal/physiopathology , Cytochrome P-450 Enzyme System/physiology , Neurons/pathology , Animals , Cell Differentiation/physiology , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Humans , Mice , Neurons/physiology , Retinoic Acid 4-Hydroxylase , Transfection , Tumor Cells, Cultured
16.
Int J Dev Biol ; 42(7): 917-25, 1998.
Article in English | MEDLINE | ID: mdl-9853822

ABSTRACT

Despite great technological advances in the study of mammalian development in the past two decades, certain problems in early development, such as how the extraembryonic lineages are established, have remained intractable. We suggest that teratocarcinoma (EC) and embryonic stem cells (ES) remain useful in vitro tools for studying some of these problems. We present a continuation of our studies on the role of IHH-based signaling in early development and demonstrate that the IHH N-peptide is expressed in the outer visceral endoderm cells of both the EC and ES-derived embryoid body. We also show that Bmp2 is upregulated and Bmp4 downregulated during the differentiation of F9 EC cells into embryoid bodies, whereas both genes are upregulated when J7 ES cells differentiate into embryoid bodies. We also examine the spatial localization of Ihh, Bmp2, and Bmp4 in day 6.5-7.0 and 7.5-8.0 embryos by in situ hybridization analysis. These data support the EC temporal expression data in that all 3 genes are expressed in visceral endoderm. Bmp4 expression appears to be limited to extraembryonic regions, where mesoderm as well as visceral endoderm are stained. Ihh and Bmp2 are expressed in extraembryonic tissues and the embryo proper. Functional roles for the observed expression patterns are discussed.


Subject(s)
Bone Morphogenetic Proteins/genetics , Carcinoma, Embryonal/genetics , Embryonic Induction , Proteins/genetics , Stem Cells/physiology , Trans-Activators , Animals , Animals, Genetically Modified , Carcinoma, Embryonal/physiopathology , Cell Differentiation , Cells, Cultured , Gene Expression Regulation, Developmental , Hedgehog Proteins , In Situ Hybridization , In Vitro Techniques , RNA, Messenger/metabolism , Tumor Cells, Cultured
17.
Int J Dev Biol ; 42(7): 943-50, 1998.
Article in English | MEDLINE | ID: mdl-9853825

ABSTRACT

In 1974, Dr. Ralph Brinster published a paper describing the consequences of injecting embryonal carcinoma cells, the predecessors of embryonic stem cells, into mouse blastocysts. Despite their early promise, embryonal carcinoma cells would not efficiently populate the germ line of mice. A decade later Elizabeth Robertson and I described the efficient generation of germline chimaeras from cultured embryonic stem cells and shortly afterwards the genetic manipulation of the mouse germline using ES cells. Our demonstration of the potency of Embryonic Stem cells gave birth to a new era in manipulative mouse genetics, one in which endogenous genes can now be mutated at will using gene targeting of retroviral mutagenesis. This review focuses on the development and testing of concepts and techniques during the thirteen years after we knew germline modification of endogenous genes in the mouse would be possible. This period is one in which more and more sophisticated tools for manipulating the mouse germline were developed and implemented. In this review I have taken the rare opportunity to reveal some of my thought processes, frustrations, successes and failures as we moved through this exciting period of rapid technological change. As I look forward to the next thirteen years, I feel that this will be an equally exciting period for manipulative genetics as we struggle to formulate concepts and design experiments that enable us to understand gene function in an era when the sequence of all genes will be known.


Subject(s)
Genetic Engineering/history , Mice/genetics , Animals , Carcinoma, Embryonal/genetics , Carcinoma, Embryonal/physiopathology , Cell Line , Gene Targeting , History, 20th Century , Mutagenesis , Retroviridae/genetics
18.
Oncogene ; 17(6): 761-7, 1998 Aug 13.
Article in English | MEDLINE | ID: mdl-9715278

ABSTRACT

A subset of male germ cell cancers presenting with advanced stage abundantly express the fibroblast growth factor-4 (FGF4). FGF4 expression is restricted in vitro to undifferentiated embryonal carcinomas (ECs). During induced differentiation, FGF4 expression is repressed in maturation sensitive but not resistant human ECs, suggesting FGF4 plays an important role in malignant growth or differentiation of ECs. To explore these FGF4 signals in male germ cell cancers, the multipotent human EC NTERA-2 clone D1 (NT2/D1) cell line was studied. All-trans-retinoic acid (RA)-treatment of these cells induces a neuronal phenotype and represses tumorigenicity and FGF4 expression. In contrast, RA-treatment of retinoid resistant lines derived from NT2/D1 cells failed to repress FGF4 expression. This implicated FGF4 directly in regulating human EC growth or differentiation. To evaluate further this FGF4 role, FGF4 was constitutively over-expressed in NT2/D1 cells using a CMV-driven expression vector containing the neomycin resistance gene. Three stable transfectants expressing exogenous FGF4 were studied as was a control transfectant only expressing the neomycin resistance gene. RA-treatment repressed endogenous but not exogenous FGF4 expression. RA-treatment of these transfectants induced morphologic and immunophenotypic maturation, changes in RA-regulated genes, and a G1 cell cycle arrest in a manner similar to parental NT2/D1 cells. This indicated FGF4 over-expression did not block RA-mediated differentiation. As expected, RA-treatment repressed tumorigenicity of the control transfectant after subcutaneous injection into athymic mice. Despite RA-treatment, this repressed tumorigenicity was overcome in all the transfectants over-expressing FGF4. The histopathology and neovascularization did not appreciably differ between xenograft tumors derived from FGF4 over-expressing versus control transfectants. FGF4 expression studies were extended to patient-derived germ cell tumors using total cellular RNA Northern analysis and an immunohistochemical assay developed to detect FGF4 protein expression. Germ cell tumors with EC components were significantly more likely to express FGF4 mRNA (P < or = 0.0179) than other examined germ cell tumors without EC components. Immunohistochemical results from 43 germ cell tumors demonstrated increased FGF4 expression especially in non-seminomas having EC components. Thus, FGF4 promotes directly malignant growth of cultured ECs, overcomes the antitumorigenic actions of RA, and is selectively expressed in specific histopathologic subsets of germ cell tumors. Taken together, these findings indicate how differentiation and anti-tumorigenic retinoic acid signals can be dissociated in germ cell cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Embryonal/physiopathology , Fibroblast Growth Factors/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Tretinoin/pharmacology , Cell Cycle , Cell Differentiation/drug effects , Drug Interactions , Fibroblast Growth Factor 4 , Fibroblast Growth Factors/genetics , Humans , Proto-Oncogene Proteins/genetics , Recombinant Proteins/biosynthesis , Signal Transduction , Transfection
19.
J Cell Sci ; 108 ( Pt 6): 2253-60, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7673345

ABSTRACT

The assembly of focal adhesions was investigated in F9 embryonal carcinoma cells in which the expression of vinculin was eliminated by a targeted disruption of the vinculin gene. Vinculin-deficient F9 cells were capable of adhering to fibronectin-coated surfaces, though they displayed a reduced spreading compared to the parental cells. Transmission electron microscopy as well as interference reflection microscopy of live cells showed that vinculin-null F9 cells formed focal adhesions that were indistinguishable from those of the control cells. Fluorescent labeling for actin, talin, alpha-actinin, paxillin and phosphotyrosinated components indicated that the organization of all these focal contact-associated components was essentially identical in the vinculin-containing and vinculin-null cells. However, quantitative, digitized microscopy indicated that the intensity of fluorescence labeling in focal adhesions for alpha-actinin, talin and paxillin was significantly higher in cells lacking vinculin. The results suggest that there are multiple molecular mechanisms for the formation of focal adhesions in the absence of vinculin.


Subject(s)
Carcinoma, Embryonal/physiopathology , Ovarian Neoplasms/physiopathology , Vinculin/genetics , Animals , Carcinoma, Embryonal/genetics , Cell Adhesion , Female , Fibronectins , Mice , Mice, Knockout , Ovarian Neoplasms/genetics , Tumor Cells, Cultured , Vinculin/physiology
20.
Bioessays ; 16(5): 343-8, 1994 May.
Article in English | MEDLINE | ID: mdl-8024542

ABSTRACT

The differentiation of mammalian neurons during development is a highly complex process involving regulation and coordination of gene expression at multiple steps. The P19 mouse embryonal carcinoma cell line is a suitable model system with which to analyze regulation of neuronal differentiation. These multipotential cells can be maintained and propagated in tissue culture in an undifferentiated state. Exposure of aggregated P19 cells to retinoic acid results in the differentiation of cells with many fundamental phenotypes of mammalian neurons. Undifferentiated P19 cells are amenable to genetic manipulations such as transfection and establishment of stable clonal cell lines expressing introduced genes. Proteins that play a key role in the neuronal differentiation of P19 cells are beginning to be identified. These include retinoic acid receptors, the epidermal growth factor receptor and the transcription factors Oct-3 and Brn-2. The biological and technical advantages of this system should facilitate deeper analysis of the activities of proteins that play a role in neuronal differentiation.


Subject(s)
Carcinoma, Embryonal/physiopathology , Neurons/cytology , Animals , Carcinoma, Embryonal/genetics , Carcinoma, Embryonal/pathology , Cell Differentiation , Cell Line , Gene Expression Regulation, Neoplastic , Mice , Models, Neurological , Transcription Factors/metabolism , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...