10 research outputs found

    A novel approach to induce cell cycle reentry in terminally differentiated muscle cells.

    No full text
    During terminal differentiation, skeletal muscle cells permanently retract from the cell cycle. We and others have shown previously that this cell cycle withdrawal is an actively maintained state that can be reversed by transient expression of the SV40 large T antigen. In an attempt to avoid the hazards of gene transfer and the difficulties of regulating transgene expression, we have now used this cellular system as a model to test whether direct protein delivery could constitute a feasible alternative or complementing strategy to gene therapy-based approaches. Taking advantage of the recently described intercellular trafficking properties of the herpes simplex virus I VP22 protein, we have constructed a chimeric VP22-SV40 large T antigen fusion protein and shown that it can spread into terminally differentiated myotubes where it accumulates in the nucleus. This fusion protein retains the ability to override the cell cycle arrest as shown for SV40 large T antigen alone. Our results clearly show that the transduced fusion protein remains capable of inducing S-phase and mitosis in these otherwise terminally differentiated cells and opens now the way to exploit this novel strategy for tissue regeneration

    Effects of immunoadsorption on α<sub>1</sub>-AAB and blood pressure.

    No full text
    <p>(A) Ordinate shows neonatal cardiomyocyte spontaneous beating rate; abscissa shows response to immunoadorption performed on a representative patient. The α<sub>1</sub>-AAB activity decreased with every immunoadsorption with a total decrease in response over time. (B) Figure shows the initial spontaneous beating rate (closed circles). After five immunoadsorptions, this response is reduced to basal values (open circles) in all 5 patients. (C) Mean response of a representative patient in cardiomyocyte contraction assay over time to immunoadsorption is shown. (D) Mean arterial pressure (MAP) was measured 5 and 180 days after immunoadsorption. MAP was significantly reduced compared to before immunoadsorption.</p

    Epitope mapping for extracellular loop 1 and loop 2 of α<sub>1Α</sub>-AR is given.

    No full text
    <p>(A) α<sub>1</sub>-AAB were directed against the first extracellular loop (eL1) in 24% and against the second extracellular loop (eL2) in 27% of the hypertensive patients. (B) The peptide sequence P1 (YWAFGR) and P2 (GRVFCNI) (partially) for eL1 were able to diminish the spontaneous beating rate response completely. (C) Epitope mapping for loop 2 is given for the following amino acid sequences: P1: GWRQPA, P2: APEDET, P3: TICQIN, P4: INEEPG, P5: GYVLFS. The sequence P2 (APEDET) was able to diminish the spontaneous beating rate response completely.</p

    Activation of ERK 1/2 in response to treatment with PE, α<sub>1</sub>-AAB from patients or rabbit α<sub>1</sub>-AB is shown.

    No full text
    <p>(A) Cardiomyocytes were treated for 5 and 15 min with 10 µM PE, 2.5 µg/ml α<sub>1</sub>-AAB or rabbit α<sub>1</sub>-AB, respectively. Equivalent amounts of protein were analyzed by Western blotting with anti-pERK 1/2 antibody (44/42 kD) and the amount of ERK was analyzed with anti-ERK 1/2 antibody. PE, patient α<sub>1</sub>-AAB, and rabbit α<sub>1</sub>-AB caused ERK 1/2 phosphorylation. (B) CHO/α<sub>1Α</sub>-AR cells were incubated with patient α<sub>1</sub>-AAB for 5 min. Lane 1 and 2 represent untreated cells. Patients 1, 4–8 were α<sub>1</sub>-AAB-positive in the cardiomyocyte contraction assay. “Patient” 9 represents a pool of five α<sub>1</sub>-AAB positive patients. Patients 10–12 were α<sub>1</sub>-AAB negative in the cardiomyocyte contraction assay. Their serum samples were processed identically to those of α<sub>1</sub>-AAB-positive patients. ERK 1/2 phosphorylation was inhibited by peptide P2, but not by peptide P5. Eukaryotic initiation factor 4E (elF4E, 25 kD) antibody was used as translational control, the amount of ERK was analyzed with anti-ERK 1/2 antibody.</p

    Representative traces of responses in intracellular Ca<sup>2+</sup> of cultivated neonatal rat cardiomyocytes exposed to isolated patient α<sub>1</sub>-AAB and rabbit α<sub>1</sub>-AB are shown.

    No full text
    <p>(A) α<sub>1</sub>-AAB isolated from the serum of a patient with refractory hypertension elicited a Ca<sup>2+</sup> signal. (B) The rabbit α<sub>1</sub>-AB gave a similar Ca<sup>2+</sup> signal. (C) A human control IgG preparation was unable to affect intracellular Ca<sup>2+</sup>. Cardiomyocytes were electrically stimulated at 1 Hz, and the peak Ca<sup>2+</sup> was monitored.</p
    corecore