Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Mol Cytogenet ; 11: 3, 2018.
Article in English | MEDLINE | ID: mdl-29344090

ABSTRACT

BACKGROUND: Translocations of the IGH locus on 14q32.3 are present in about 8% of patients with chronic lymphocytic leukemia (CLL) and contribute to leukemogenesis by deregulating the expression of the IGH-partner genes. Identification of these genes and investigation of the downstream effects of their deregulation can reveal disease-causing mechanisms. CASE PRESENTATION: We report on the molecular characterization of a novel t(12;14)(q23.2;q32.3) in CLL. As a consequence of the rearrangement ASCL1 was brought into proximity of the IGHJ-Cµ enhancer and was highly overexpressed in the aberrant B-cells of the patient, as shown by qPCR and immunohistochemistry. ASCL1 encodes for a transcription factor acting as a master regulator of neurogenesis, is overexpressed in neuroendocrine tumors and a promising therapeutic target in small cell lung cancer (SCLC). Its overexpression has also been recently reported in acute adult T-cell leukemia/lymphoma.To examine possible downstream effects of the ASCL1 upregulation in CLL, we compared the gene expression of sorted CD5+ cells of the translocation patient to that of CD19+ B-cells from seven healthy donors and detected 176 significantly deregulated genes (Fold Change ≥2, FDR p ≤ 0.01). Deregulation of 55 genes in our gene set was concordant with at least two studies comparing gene expression of normal and CLL B-lymphocytes. INSM1, a well-established ASCL1 target in the nervous system and SCLC, was the gene with the strongest upregulation (Fold Change = 209.4, FDR p = 1.37E-4).INSM1 encodes for a transcriptional repressor with extranuclear functions, implicated in neuroendocrine differentiation and overexpressed in the majority of neuroendocrine tumors. It was previously shown to be induced in CLL cells but not in normal B-cells upon treatment with IL-4 and to be overexpressed in CLL cells with unmutated versus mutated IGHV genes. Its role in CLL is still unexplored. CONCLUSION: We identified ASCL1 as a novel IGH-partner gene in CLL. The neural transcription factor was strongly overexpressed in the patient's CLL cells. Microarray gene expression analysis revealed the strong upregulation of INSM1, a prominent ASCL1 target, which was previously shown to be induced in CLL cells upon IL-4 treatment. We propose further investigation of the expression and potential role of INSM1 in CLL.

2.
Genes Chromosomes Cancer ; 55(1): 60-8, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26391436

ABSTRACT

Myeloid and lymphoid neoplasms with fibroblast growth factor receptor 1 (FGFR1) abnormalities, also known as 8p11 myeloproliferative syndrome (EMS), represent rare and aggressive disorders, associated with chromosomal aberrations that lead to the fusion of FGFR1 to different partner genes. We report on a third patient with a fusion of the translocated promoter region (TPR) gene, a component of the nuclear pore complex, to FGFR1 due to a novel ins(1;8)(q25;p11p23). The fact that this fusion is a rare but recurrent event in EMS prompted us to examine the localization and transforming potential of the chimeric protein. TPR-FGFR1 localizes in the cytoplasm, although the nuclear pore localization signal of TPR is retained in the fusion protein. Furthermore, TPR-FGFR1 enables cytokine-independent survival, proliferation, and granulocytic differentiation of the interleukin-3 dependent myeloid progenitor cell line 32Dcl3, reflecting the chronic phase of EMS characterized by myeloid hyperplasia. 32Dcl3 cells transformed with the TPR-FGFR1 fusion and treated with increasing concentrations of the tyrosine kinase inhibitors ponatinib (AP24534) and infigratinib (NVP-BGJ398) displayed reduced survival and proliferation with IC50 values of 49.8 and 7.7 nM, respectively. Ponatinib, a multitargeted tyrosine kinase inhibitor, is already shown to be effective against several FGFR1-fusion kinases. Infigratinib, tested only against FGFR1OP2-FGFR1 to date, is also efficient against TPR-FGFR1. Taking its high specificity for FGFRs into account, infigratinib could be beneficial for EMS patients and should be further investigated for the treatment of myeloproliferative neoplasms with FGFR1 abnormalities.


Subject(s)
Chromosomes, Human, Pair 8/genetics , Imidazoles/pharmacology , Myeloproliferative Disorders/genetics , Nuclear Pore Complex Proteins/genetics , Phenylurea Compounds/pharmacology , Proto-Oncogene Proteins/genetics , Pyridazines/pharmacology , Pyrimidines/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cytoplasm/metabolism , Humans , Imidazoles/therapeutic use , Male , Middle Aged , Mutagenesis, Insertional , Myeloproliferative Disorders/drug therapy , Nuclear Pore Complex Proteins/metabolism , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Phenylurea Compounds/therapeutic use , Proto-Oncogene Proteins/metabolism , Pyridazines/therapeutic use , Pyrimidines/therapeutic use , Receptor, Fibroblast Growth Factor, Type 1/metabolism
3.
Am J Med Genet A ; 164A(12): 3126-31, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25250687

ABSTRACT

Here, we report on a male patient with developmental delay, speech impairment, mild dysmorphic features, and borderline intellectual disability, bearing a de novo balanced t(5;6)(q11;q25.3). By combining FISH and long distance inverse PCR, we identified two genes, ADAMTS6 and ARID1B, which were disrupted at the translocation breakpoints. Due to the opposing transcriptional directions of the two genes, no fusion transcripts could be formed. ADAMTS6 on chromosome 5 encodes a zinc metalloprotease. To date, there has been no information about the substrates and the exact role of this enzyme protein. ARID1B on chromosome 6 is involved in chromatin remodeling and transcriptional activation and is known to play a role in neural development. To our knowledge, this is the fourth translocation involving ARID1B reported in association with intellectual disability. ARID1B haploinsufficiency has already been described in patients with intellectual disabilities with or without corpus callosum abnormalities, Coffin-Siris syndrome and autism (OMIM 614562 and OMIM 614556). A review of patients with ARID1B mutations reveals their broad phenotypic variability. The phenotype of the present patient is of the mildest described to date and further underscores this observation. We conclude that the most prominent and consistent clinical findings in patients with ARID1B haploinsufficiency are developmental delay, speech impairment and intellectual disability and propose that patients with unresolved genetic background and these clinical findings should be considered for ARID1B mutation screening.


Subject(s)
ADAM Proteins/genetics , Abnormalities, Multiple/genetics , Chromosomes, Human, Pair 5/genetics , Chromosomes, Human, Pair 6/genetics , DNA-Binding Proteins/genetics , Developmental Disabilities/genetics , Transcription Factors/genetics , Translocation, Genetic/genetics , ADAMTS Proteins , Humans , In Situ Hybridization, Fluorescence , Male , Polymerase Chain Reaction
4.
J Pediatr ; 163(4): 1174-8.e3, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23876976

ABSTRACT

OBJECTIVE: To study genotype-phenotype correlation of ring chromosome 18 [r(18)] in 9 patients with 46,XN karyotype. STUDY DESIGN: In 9 patients with a de novo 46,XN,r(18) karyotype (7 females, 2 males), we performed high-resolution single-nucleotide polymorphism array analysis (Illumina Human Omni1-QuadV1 array in 6 patients, Affymetrix 6.0 array in 3 patients), investigation of parental origin, and genotype-phenotype correlation. RESULTS: No breakpoint was recurrent. Single metaphases with loss of the ring, double rings, or secondarily rearranged rings were found in some cases, but true mosaicism was present in none of these cases. In 3 patients, additional duplications in 18p (of 1.4 Mb, 2 Mb, and 5.8 Mb) were detected. In 1 patient, an additional deletion of 472 kb in Xp22.33, including the SHOX gene, was found. Parental origin of r(18) was maternal in 2 patients and paternal in 4 patients, and formation was most likely meiotic. Karyotype was normal in all investigated parents (n = 15). At birth, mean maternal age was 30 years (n = 9) and mean paternal age was 34.4 years (n = 9). CONCLUSION: Genotype-phenotype correlation revealed extensive clinical variability but no characteristic r(18) phenotype. Severity of clinical signs were generally correlated with the size of the deletion. Patients with large deletions in 18p and small deletions in 18q exhibited mainly symptoms related to 18p-, whereas those with large deletions in 18q and small deletions in 18p had symptoms of 18q-.


Subject(s)
Chromosome Deletion , Polymorphism, Single Nucleotide , Adolescent , Adult , Body Size , Child , Child, Preschool , Chromosomes, Human, Pair 18/ultrastructure , Female , Genetic Association Studies , Head/physiology , Humans , Infant , Infant, Newborn , Karyotyping , Male , Maternal Age , Microsatellite Repeats/genetics , Middle Aged , Oligonucleotide Array Sequence Analysis , Ring Chromosomes , Young Adult
5.
Eur J Med Genet ; 56(7): 383-8, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23608969

ABSTRACT

De novo combined duplications/inversions are very rare chromosomal rearrangements. For chromosome 7 just some dozen cases of duplications of various parts of the long arm have been published. We report on a 12-year-old boy with muscular hypotonia, global developmental delay, short stature, and various facial dysmorphism including frontal bossing, temporal narrowing, slightly down-slanting palpebral fissures, a broad nasal root, a long philtrum, a thin and tented upper lip, a drooping lower lip, micrognathia, prominent ears, a short neck, and a low posterior hairline. Karyotype analysis and molecular investigations revealed a complex de novo chromosomal rearrangement on 7q. FISH analysis with locus specific YACs and BACs and SNP array with the Illumina(®) HumanOmni1-Quad v1.0 BeadChip disclosed a direct duplication in the long arm of chromosome 7 (q22.1→q32.2) and an inversion located at the breakpoint between the two copies of the duplication (q31.31→q31.33). In addition, breakpoint characterization at the molecular level revealed a 386 bp insertion carrying two Alu elements of chromosome 19p13.2 between the two copies of the duplication. By a comparison of the SNP haplotypes of the derivative chromosome of the patient and both parents a two-step formation during spermatogenesis was suggested as the most likely mechanism of formation.


Subject(s)
Abnormalities, Multiple/genetics , Chromosome Aberrations , Chromosomes, Human, Pair 19/genetics , Chromosomes, Human, Pair 7/genetics , Developmental Disabilities/genetics , Abnormalities, Multiple/diagnosis , Alu Elements , Child , Chromosome Breakpoints , Developmental Disabilities/diagnosis , Haplotypes , Humans , Male , Polymorphism, Single Nucleotide
6.
Am J Med Genet A ; 158A(9): 2239-44, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22821890

ABSTRACT

Exact breakpoint determination by DNA-array has dramatically improved the analysis of genotype-phenotype correlations in chromosome aberrations. It allows a more exact definition of the most relevant genes and particularly their isolated or combined impact on the phenotype in an unbalanced state. Here, we report on a 21-year-old female with severe growth retardation, severe intellectual disability, hypoplasia of the corpus callosum, unilateral sacral hypoplasia, tethered cord, various minor facial dysmorphisms, and a telomeric deletion of about 4.4 Mb in 7q36.2->qter combined with a telomeric duplication of about 8 Mb in 17pter->p13.1. Fine mapping was achieved with the Illumina® Infinium HumanOmni1-Quad v1.0 BeadChip. Most of the major clinical features correspond to the well-known effects of haploinsufficiency of the MNX1 and SHH genes. In addition, review of the literature suggests an association of the 17p duplication with specific facial dysmorphic features and skeletal anomalies, but also an aggravating effect of the duplication-deletion for severe growth retardation as well as sacral and corpus callosum hypoplasia by one or more genes located on the proximal half of the segmental 17p duplication could be elaborated by comparison with other patients from the literature carrying either the deletion or the duplication found in our patient.


Subject(s)
Abnormalities, Multiple/genetics , Chromosome Deletion , Chromosome Duplication , Adult , Female , Humans , Karyotyping
7.
Microvasc Res ; 84(1): 65-73, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22480576

ABSTRACT

The phenotypes and functions of endothelial cells (EC), a heterogeneous cell population, vary along the vascular tree and even in the same organ between different vessels. The placenta is an organ with abundant vessels. To enhance further knowledge concerning placenta derived EC, we develop a new method for isolation, purification and culture of these EC. Moreover, in order to investigate the peculiarity of placenta derived EC we compare their phenotypic and functional characteristics with human dermal lymphatic endothelial cells (HDLEC) and human umbilical vein endothelial cells (HUVEC). Freshly isolated placenta derived EC displayed an elongated shape with pale cytoplasm and showed the typical cobblestone pattern of EC but also a swirling pattern when confluent. FISH-analyses of the isolated EC from placentae of male fetus revealed an XY genotype strongly indicating their fetal origin. Characterisation of placenta derived fetal EC (fEC) underlined their blood vessel phenotype by the expression of vWF, Ulex europaeus lectin-1, HLA-class I molecules, CD31, CD34, CD36, CD51/61, CD54, CD62E, CD105, CD106, CD133, CD141, CD143, CD144, CD146, VEGFR-1, VEGFR-2, EN-4, PAL-E, BMA120, Tie-1, Tie-2 and α-Tubulin. In contrast to previous reports the expression of lymphatic markers, like VEGFR-3, LYVE-1, Prox-1 and Podoplanin was consistently negative. Haematopoietic surface markers like CD45 and CD14 were also always negative. Various functional tests (Dil-Ac-LDL uptake, Matrigel assay and TNF-α induced upregulation of CD62E and CD54) substantiated the endothelial nature of propagated fEC. At the ultrastructural level, fEC harboured numerous microvilli, micropinocytic vesicles at their basis, were rich in intermediate filaments and possessed typical Weibel - Palade bodies. In conclusion, the placenta is a plentiful source of fetal, microvascular, blood EC with an expression profile (CD34+, CD133+, VEGFR-2+, CD45-) suggestive of an endothelial progenitor phenotype.


Subject(s)
Antigens, CD/metabolism , Biomarkers/metabolism , Endothelial Cells/cytology , Placenta/blood supply , Vascular Endothelial Growth Factor Receptor-2/metabolism , AC133 Antigen , Adult , Antigens, CD34/metabolism , Cell Culture Techniques , Cells, Cultured , Cytoplasmic Structures/ultrastructure , Dermis/blood supply , Endothelial Cells/metabolism , Endothelial Cells/ultrastructure , Female , Glycoproteins/metabolism , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Leukocyte Common Antigens/metabolism , Microvilli/ultrastructure , Peptides/metabolism , Pregnancy , Term Birth
8.
Diagn Pathol ; 6: 19, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21401966

ABSTRACT

BACKGROUND: Characterization of novel fusion genes in acute leukemia is important for gaining information about leukemia genesis. We describe the characterization of a new ETV6 fusion gene in acute myeloid leukemia (AML) FAB M0 as a result of an uncommon translocation involving chromosomes 12 and 15. METHODS: The ETV6 locus at 12p13 was shown to be translocated and to constitute the 5' end of the fusion product by ETV6 break apart fluorescence in situ hybridisation (FISH). To identify a fusion partner 3' rapid amplification of cDNA-ends with polymerase chain reaction (RACE PCR) was performed followed by cloning and sequencing. RESULTS: The NTRK3 gene on chromosome 15 was found to constitute the 3' end of the fusion gene and the underlying ETV6-NTRK3 rearrangement was verified by reverse transcriptase PCR. No RNA of the reciprocal NTRK3-ETV6 fusion gene could be detected. CONCLUSION: We have characterized a novel ETV6-NTRK3 fusion transcript which has not been previously described in AML FAB M0 by FISH and RACE PCR. ETV6-NTRK3 rearrangements have been described in secretory breast carcinoma and congenital fibrosarcoma.


Subject(s)
Chromosomes, Human, Pair 12 , Chromosomes, Human, Pair 15 , Leukemia, Myeloid, Acute/genetics , Oncogene Proteins, Fusion/genetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Chromosome Aberrations , Cloning, Molecular , DNA Mutational Analysis , DNA, Neoplasm/analysis , Gene Rearrangement , Humans , In Situ Hybridization, Fluorescence , Leukemia, Myeloid, Acute/drug therapy , Male , Middle Aged
9.
Am J Med Genet A ; 152A(11): 2762-7, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20954245

ABSTRACT

Constitutional insertional translocations are rare findings in clinical cytogenetics. Here, we report on the unbalanced segregation of a balanced paternal insertional translocation ins(7;6)(p15;q16.1q21) to three children. Investigations by conventional karyotyping, FISH with locus-specific probes, microsatellite marker analysis, and SNP-array based copy number analysis revealed a direct orientation of the inserted segment, a size of 11.3 Mb, and breakpoints between rs4370337 and rs12660854 and rs12110990 and rs4946730 on 6q16.1 and 6q21, respectively, as well as within BAC clone RP11-182J2 on 7p15. A 17-year-old daughter inherited the der(6) chromosome and was affected by severe mental retardation, obesity, and minor anomalies. Two further children inherited the der(7) chromosome. A daughter shows an almost unremarkable phenotype and only minor features in neuropsychological testing at 19 years of age. Her 14-year-old half-brother demonstrates a mild delay in cognitive development most likely jointly caused by the chromosomal rearrangement and asphyxia during delivery. The patient with the deletion confirms the previously reported phenotype of severe mental retardation and obesity in patients with del(6)(q16.2), while both patients with partial trisomy for the same segment of chromosome 6 are further examples for a generally less severe phenotype associated with duplications than with deletions, and even for the recent insight that chromosomal aneusomies of several megabases may go without major clinical consequences.


Subject(s)
Chromosome Duplication/genetics , Chromosomes, Human, Pair 7/genetics , Mutagenesis, Insertional/genetics , Adolescent , Child , Child, Preschool , Chromosome Deletion , Chromosomes, Human, Pair 6/genetics , Female , Humans , Infant , Infant, Newborn , Male , Neuropsychological Tests , Pedigree , Phenotype , Pregnancy , Trisomy/genetics , Young Adult
10.
Cancer Res ; 70(11): 4613-23, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20460525

ABSTRACT

We describe for the first time a new integral molecular pathway, linking transcription factor E2F3a to epidermal growth factor receptor (EGFR) activation in ovarian cancer cells. Investigations on the role of E2F family members in EGFR-mediated mitogenic signaling revealed that E2F3a was selectively upregulated following EGFR activation, whereas all other E2F family members remained unaffected. In contrast, EGF treatment of healthy ovarian surface epithelial and mesothelial cells yielded a selective upregulation of proliferation-promoting E2F1 and E2F2 without influencing E2F3a expression. In ovarian cancer cell lines, the extent of EGF-induced proliferative stimulus was closely related to the magnitude of E2F3a increase, and proliferation inhibition by E2F3a knockdown was not overcome by EGF exposure. Furthermore, the EGFR-E2F3a axis was found to be signal transducer and activator of transcription 1/3 dependent and the ratio of IFN-regulatory factor (IRF)-1 to IRF-2 was shown to be determinative for E2F3a control. In a pilot study on 32 primary ovarian cancer specimens, a highly significant correlation between activated EGFR and E2F3a expression was disclosed. This new integral pathway in the EGFR-driven mitogenic cell response, which through its key player E2F3a was found to be essential in triggering proliferation in ovarian cancer cells, provides new insights into EGFR signaling and could represent the basis for appealing new therapeutic approaches in ovarian cancer.


Subject(s)
E2F3 Transcription Factor/metabolism , ErbB Receptors/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Cell Growth Processes/physiology , Cell Line, Tumor , E2F3 Transcription Factor/biosynthesis , E2F3 Transcription Factor/genetics , Epidermal Growth Factor/pharmacology , ErbB Receptors/antagonists & inhibitors , Female , Humans , Interferon Regulatory Factor-1/metabolism , Interferon Regulatory Factor-2/metabolism , Ovarian Neoplasms/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Signal Transduction , Transfection , Up-Regulation/drug effects
11.
Fertil Steril ; 94(1): 350.e12-5, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20117774

ABSTRACT

OBJECTIVE: To describe the parental origin and the mechanism of formation of a 46,X,der(X)(pter-->q21.1::p11.4-->pter)[23]/45,X[8] karyotype in a patient with mild Turner syndrome. DESIGN: Case report. SETTING: A university hospital. PATIENT(S): A 23-year-old woman with normal height, gonadal dysgenesis, and mild Turner stigmata. INTERVENTION(S): Genotype-phenotype correlation, array-based copy number analysis, fluorescence in situ hybridization with locus-specific probes, and microsatellite marker-mediated haplotype analysis subsequent to whole genome amplification of microdissected chromosomes. MAIN OUTCOME MEASURES: Genotype-phenotype correlation, mechanism of formation, and parental origin. RESULT(S): Formation in paternal meiosis by refolding in itself and unequal recombination between Xp and Xq were found as the most likely mechanism of formation. CONCLUSION(S): Formation of der(X) chromosomes in females can be more complex than previously thought. The nearly normal height of this patient could be explained by a combination of trisomy of the Xp-located SHOX gene and mosaicism with a 45,X cell line.


Subject(s)
Chromosomes, Human, X/genetics , Karyotyping/methods , Turner Syndrome/diagnosis , Turner Syndrome/genetics , Adult , Female , Humans , Male , Parents , Pregnancy , Young Adult
13.
Am J Med Genet A ; 149A(11): 2522-6, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19842202

ABSTRACT

Partial duplication 3q is a well defined clinical entity characterized by growth retardation, cryptorchism, microcephaly, and characteristic dysmorphisms. Most patients present with large duplications or are associated with a second chromosomal imbalance, which makes the definition of the phenotype difficult. Here, we report on a 4-year and 8-month-old girl with pre- and postnatal measurements in the high normal range, developmental delay, minor dysmorphic features, and a de novo unbalanced 3/4 translocation with trisomy 3q27 --> qter and monosomy of the subtelomeric region of 4p. Conventional karyotyping, FISH with probes from the Wolf-Hirschhorn syndrome critical region and chromosome 4p locus-specific probes, microsatellite marker-based haplotyping, and SNP microarray copy number analysis revealed a terminal 4p deletion of less than 500 kb with a breakpoint distal to the Wolf-Hirschhorn syndrome critical region, a chromosome 3q duplication of around 15.3 Mb, with origin of the rearrangement in paternal meiosis. Thus, our case clearly characterizes the phenotype of pure partial duplication 3q more exactly, and moreover, indicates that small chromosome rearrangements might lead to growth in the upper normal range or even cause overgrowth.


Subject(s)
Chromosomes, Human, Pair 3/genetics , Trisomy/genetics , Adult , Child, Preschool , Chromosome Banding , Female , Gene Duplication , Humans , In Situ Hybridization, Fluorescence , Infant, Newborn , Phenotype
14.
Blood ; 113(3): 755-64, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-18824596

ABSTRACT

Allograft vasculopathy is the leading cause of death in patients with heart transplantation. Accelerated endothelial regeneration mediated by enhanced endothelial progenitor cell (EPC) incorporation may attenuate the development of allograft vasculopathy. We investigated the hypothesis that modulation of EPC biology and attenuation of allograft vasculopathy by increased high-density lipoprotein cholesterol after human apo A-I (AdA-I) transfer requires scavenger receptor (SR)-BI expression in bone marrow-derived EPCs. After AdA-I transfer, the number of circulating EPCs increased 2.0-fold (P < .001) at different time points in C57BL/6 mice transplanted with SR-BI(+/+) bone marrow but remained unaltered in mice with SR-BI(-/-) bone marrow. The effect of high-density lipoprotein on EPC migration in vitro requires signaling via SR-BI and extracellular signal-regulated kinases and is dependent on increased nitric oxide (NO) production in EPCs. Human apo A-I transfer 2 weeks before paratopic artery transplantation reduced intimal area at day 21 3.7-fold (P < .001) in mice with SR-BI(+/+) bone marrow but had no effect in mice with SR-BI(-/-) bone marrow. AdA-I transfer potently stimulated EPC incorporation and accelerated endothelial regeneration in chimeric SR-BI(+/+) mice but not in chimeric SR-BI(-/-) mice. In conclusion, human apo A-I transfer accelerates endothelial regeneration mediated via SR-BI expressing bone marrow-derived EPCs, thereby preventing allograft vasculopathy.


Subject(s)
Apolipoprotein A-I/metabolism , Blood Vessels/pathology , Endothelial Cells/metabolism , Scavenger Receptors, Class B/metabolism , Stem Cells/metabolism , Animals , Apolipoprotein A-I/genetics , Blotting, Western , Bone Marrow Cells/metabolism , Bone Marrow Transplantation , Carotid Arteries/transplantation , Cell Movement , Cholesterol, HDL/blood , Enzyme-Linked Immunosorbent Assay , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Flow Cytometry , Humans , Immunohistochemistry , In Situ Hybridization , Male , Mice , Nitric Oxide/metabolism , Phosphorylation , Regeneration , Scavenger Receptors, Class B/genetics , Signal Transduction/physiology , Transplantation, Homologous
15.
Wien Klin Wochenschr ; 120(13-14): 435-9, 2008.
Article in English | MEDLINE | ID: mdl-18726671

ABSTRACT

Mayer-Rokitansky-Kuster-Hauser (MRKH) syndrome is characterized by the congenital absence of uterus and upper part of the vagina as a result of Mullerian duct agenesis. The combination of MRKH syndrome with renal anomalies and cervicothoracic dysplasia is known as MURCS association (Mullerian aplasia, Renal anomalies, and Cervicothoracic Somite dysplasia). The etiology remains poorly understood. We delineate this disease by reporting on a 16-year-old patient showing the cardinal features of MURCS association accompanied by a persistent left superior vena cava and atrial septal defect, orofacial clefting, and mild reduction deformities of the left hand. Fluorescence in situ hybridization did not show major deletions or duplications of the DiGeorge/VCFS (velocardiofacial syndrome) region at chromosome 22q11.1 as well as the TBX5/TBX3 region at 12q24.1. In addition, sequencing of the TP63L (p63) gene, residing at 3q27, remained normal in the presented patient. Thus, we provide further evidence for the genetic heterogeneity of MURCS association.


Subject(s)
Abnormalities, Multiple/genetics , Cervical Vertebrae/abnormalities , Genetic Variation/genetics , Genitalia, Female/abnormalities , Kidney/abnormalities , Mullerian Ducts/abnormalities , Thoracic Vertebrae/abnormalities , Abnormalities, Multiple/diagnosis , Adolescent , Chromosome Deletion , Chromosomes, Human, Pair 12 , Chromosomes, Human, Pair 22 , Chromosomes, Human, Pair 3 , DNA Mutational Analysis , DiGeorge Syndrome/diagnosis , DiGeorge Syndrome/genetics , Female , Fingers/abnormalities , Genetic Heterogeneity , Heart Defects, Congenital/diagnosis , Heart Defects, Congenital/genetics , Humans , In Situ Hybridization, Fluorescence , Syndrome
16.
Fertil Steril ; 90(5): 2009.e13-7, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18687426

ABSTRACT

OBJECTIVE: To describe the parental origin and the mechanism of formation of an 47,XY,idic(X)(p11.2) karyotype in a patient with Klinefelter syndrome. DESIGN: Case report. SETTING: A university hospital. PATIENT(S): A 36-year-old man with primary infertility. INTERVENTION(S): Genotype-phenotype correlation and microsatellite marker-mediated haplotype analysis subsequent to whole genome amplification of microdissected chromosomes. MAIN OUTCOME MEASURE(S): Genotype-phenotype correlation, mechanism of formation, and parental origin. RESULT(S): Maternal origin of the isochromosome and the normal X chromosome and loss of maternal heterozygosity for all informative Xq markers on the isochromosome and in each case, the presence of the other maternal allele on the normal homologue was shown. Comparative analysis of the clinical features of 17 additional cases and of 1 case with a 46,XY/47,XY,i(X)(q10) karyotype reported in the literature revealed a phenotype very similar to the clinical findings in patients with a 47,XXY karyotype. CONCLUSION(S): The molecular results in our patient indicate a maternal origin of a true dicentric isochromosome and most likely postzygotic formation subsequent to a nondisjunction in maternal meiosis II. With the exception of the final height the phenotype of Klinefelter syndrome appears not to be the consequence of trisomy of the pseudoautosomal region on Xp.


Subject(s)
Chromosomes, Human, X , Chromosomes, Human, Y , Infertility, Male/genetics , Isochromosomes , Klinefelter Syndrome/genetics , Adult , Chromosome Banding , Chromosome Painting , Genotype , Humans , Male , Pedigree , Phenotype
17.
Int J Hematol ; 87(4): 382-386, 2008 May.
Article in English | MEDLINE | ID: mdl-18418698

ABSTRACT

Disease recurrence has been and remains the leading cause of treatment failure in patients with high-risk leukemia. We retrospectively analyzed outcome in 61 patients with high-risk leukemia receiving a combination of fludarabine and intermediate-dose cytarabine as induction (n = 11) or salvage therapy (n = 35). Thirty-six patients having a suitable stem cell donor proceeded to allogeneic hematopoietic stem cell transplantation (HSCT). Ten patients received fludarabine-based salvage therapy without consecutive allogeneic transplantation and 15 patients received fludarabine/intermediate-dose cytarabine because of disease relapse following allogeneic stem cell transplantation. In patients without prior allogeneic HSCT (n = 46) the complete remission rate (CR) was 41% with a CR rate of 46 and 14% in patients with acute myeloid leukemia (AML) and with acute lymphoblastic leukemia (ALL), respectively. Overall survival for patients achieving a CR was 41 versus 0% for patients not achieving CR (P < 0.0001). The best outcome was observed in patients receiving an allogeneic HSCT in CR following fludarabine/ intermediate-dose cytarabine (47 vs. 0% for patients not in CR at the time of allografting, P = 0.01). All 10 patients receiving fludarabine/intermediate-dose cytarabine without subsequent allogeneic HSCT died within 3 years either of disease relapse/progression or infection. Only 1/15 (7%) patients receiving fludarabine/intermediate-dose cytarabine because of relapse following allogeneic HSCT became a long-term survivor. By multivariate analysis achieving CR, receiving an allogeneic HSCT, and being in first relapse or untreated were the only parameters that significantly determine the outcome. Although preliminary only high-risk AML patients having a stem cell donor are candidates for fludarabine/intermediate-dose cytarabine and only those achieving a CR should be referred to subsequent allogeneic HSCT. All other patients with high-risk leukemia are candidates for experimental therapies within controlled trials.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cytarabine/therapeutic use , Hematopoietic Stem Cell Transplantation , Leukemia/drug therapy , Vidarabine/analogs & derivatives , Adult , Aged , Dose-Response Relationship, Drug , Female , Humans , Leukemia/surgery , Male , Middle Aged , Recurrence , Risk Factors , Survival Rate , Transplantation, Homologous , Vidarabine/therapeutic use
18.
Am J Med Genet A ; 146A(7): 925-9, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18302251

ABSTRACT

The phenotype of patients with a ring chromosome 6 can be highly variable ranging from almost normal to severe malformations and mental retardation. Size and structure of the ring chromosome as well as the level of mosaicism are important factors for the clinical phenotype. Here, we report on a 25-year-old woman with short stature, minor scoliosis, normal fertility, appropriate psychomotor development, minor dysmorphisms, and a de novo ring chromosome 6. Conventional karyotyping as well as molecular cytogenetic and molecular investigations of DUSP22 on 6p and RP1-191N21.4 on 6q by a new technical approach indicated breakpoints less than 240 kb and less than 190 kb proximal to the telomeres of 6p and 6q, respectively. In addition, formation of the ring chromosome from the paternal chromosome was demonstrated. Thus this case clearly shows that in patients with ring chromosomes without loss of euchromatic material mitotic instability of the ring chromosome is the most important reason for growth retardation and minor congenital anomalies.


Subject(s)
Chromosomes, Human, Pair 6 , Growth Disorders/genetics , Ring Chromosomes , Adult , Base Sequence , DNA Primers , Genomic Imprinting , Humans , Male
19.
FASEB J ; 20(14): 2600-2, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17077285

ABSTRACT

Glucocorticoids (GCs) specifically induce apoptosis in malignant lymphoblasts and are thus pivotal in the treatment of acute lymphoblastic leukemia (ALL). However, GC-resistance is a therapeutic problem with an unclear molecular mechanism. We generated approximately 70 GC-resistant sublines from a GC-sensitive B- and a T-ALL cell line and investigated their mechanisms of resistance. In response to GCs, all GC-resistant subclones analyzed by real-time polymerase chain reaction (PCR) showed a deficient up-regulation of the GC-receptor (GR) and its downstream target, GC-induced leucine zipper. This deficiency in GR up-regulation was confirmed by Western blotting and on retroviral overexpression of GR in resistant subclones GC-sensitivity was restored. All GC-resistant subclones were screened for GR mutations using denaturing high-pressure liquid chromatography (DHPLC), DNA-fingerprinting, and fluorescence in situ hybridization (FISH). Among the identified mutations were some previously not associated with GC resistance: A484D, P515H, L756N, Y663H, L680P, and R714W. This approach revealed three genotypes, complete loss of functional GR in the mismatch repair deficient T-ALL model, apparently normal GR genes in B-ALLs, and heterozygosity in both. In the first genotype, deficiency in GR up-regulation was fully explained by mutational events, in the second by a putative regulatory defect, and in the third by a combination thereof. In all instances, GC-resistance occurred at the level of the GR in both models.


Subject(s)
Drug Resistance, Neoplasm , Glucocorticoids/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Receptors, Glucocorticoid/metabolism , Cell Line, Tumor , DNA Mismatch Repair , Drug Resistance, Neoplasm/genetics , Glucocorticoids/metabolism , Humans , Mutation , Receptors, Glucocorticoid/genetics , Transcription Factors/metabolism
20.
Haematologica ; 91(9): 1291-3, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16956841

ABSTRACT

We investigated tumor cell apoptosis in vivo in 14 heavily pretreated patients with B-cell chronic lymphocytic leukemia undergoing rituximab monotherapy. Apoptosis induction was more pronounced in patients with mutated IgVH genes than in those with unmutated IgVH genes, independently of the levels of expression of CD20, CD38, and ZAP-70 and of the presence of 17p deletion. Our results suggest an association between IgVH gene mutational status and rituximab-induced apoptosis.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Apoptosis/drug effects , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Variable Region/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Aged , Antibodies, Monoclonal, Murine-Derived , Apoptosis/immunology , Female , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Male , Middle Aged , Rituximab , Somatic Hypermutation, Immunoglobulin
SELECTION OF CITATIONS
SEARCH DETAIL
...