6 research outputs found

    Angiogenin is expressed in TH-positive neurons in the substantia nigra after viral injection.

    No full text
    <p>A. An AAV2-Ang injected mouse was sacrificed eight weeks after injection and stained with a primary antibody against the V5 tag fused to angiogenin and a CY3-conjugated secondary antibody, demonstrating overexpression of angiogenin in the substantia nigra. Scale bar = 100 µm. B. Same section of tissue stained with a primary antibody against tyrosine hydroxylase (TH) and an Alexa488-conjugated secondary antibody to identify dopaminergic nigral neurons. Scale bar = 100 µm. C. A merge of V5-angiogenin staining and TH staining demonstrating colocalization in dopaminergic cells in the substantia nigra. Scale bar = 100 µm. D. Higher magnification image demonstrating V5-angiogenin localized perinuclearly and accumulated in vesicular-like puncta within the cell. Scale bar = 10 µm. E. Control immunostaining of the substantia nigra from an AAV2-GFP injected mouse. No V5-angiogenin staining was observed. Scale bar = 100 µm. F. Same section stained for TH to identify dopaminergic neurons in the substantia nigra of an AAV2-GFP-injected mouse. Scale bar = 100 µm. G. GFP immunostaining of the substantia nigra from another section from the same AAV-GFP injected mouse, demonstrating GFP expression in the substantia nigra. H. Same section stained for TH to identify dopaminergic neurons in the substantia nigra of an AAV-GFP injected mouse. Scale bar = 100 µm.</p

    Dominant negative Akt does not inhibit angiogenin's protective effect against MPP+.

    No full text
    <p>A. Western blot demonstrating reduced Akt phosphorylation in SH-SY5Y cells transfected with the K179M DN-Akt construct. Twenty-four hours following transfection of empty pCMV5 vector (pC) or DN-Akt vector (DN), SH-SY5Y cells were treated with Insulin Growth Factor (50 ng/mL). Thirty seconds after Insulin Growth Factor treatment, cell lysates were collected for Western blotting. Actin was used as a loading control. B. Representative Western blot demonstrating reduced caspase-3 cleavage following angiogenin treatment in SH-SY5Y cells transfected with either empty vector or HA-tagged DN-Akt using Amaxa nucleoporation. Twenty-four hours after transfection, cells were pretreated with angiogenin (100 nM). MPP+ (0.75 mM) was then applied with fresh angiogenin for an additional 24 hours prior to collection. HA-tagged DN-Akt expression was comparable among all DN-Akt-transfected conditions. Actin was used as loading control. C. Densitometric quantification of caspase-3 cleavage bands normalized to actin. Results reflect three independent experiments. *p<0.05, ***p<0.001 (One-way ANOVA with Tukey's post-hoc test). Error bars reflect SEM.</p

    Angiogenin overexpression does not reduce MPTP-induced dopaminergic neuronal loss in the substantia nigra.

    No full text
    <p>Four weeks following AAV2 injection, MPTP was administered at 30 mg/kg/day for five days. Mice were sacrificed three weeks following the last MPTP injection and brain sections stained for tyrosine hydroxylase to identify DA neurons in the substantia nigra. Stereology counts of TH-positive nigral neurons in the AAV-injected side are shown for each of the four conditions: GFP-saline (n = 7), Ang-saline (n = 4), GFP-MPTP (n = 17), and Ang-MPTP (n = 16). MPTP induced a 28% reduction in TH-stained neurons in the substantia nigra of GFP-injected mice, and angiogenin did not ameliorate dopaminergic neuron loss induced by MPTP. *p<0.05, **p<0.01 (One-way ANOVA with Tukey's post-hoc test). Error bars reflect SEM.</p

    Angiogenin does not prevent MPTP-induced depletion of dopamine and its metabolites in the striatum.

    No full text
    <p>Three weeks after MPTP or saline injections, AAV2-GFP and AAV2-Ang-injected mice were sacrificed and striatal tissue immediately removed and frozen for HPLC. DA and its metabolites, DOPAC and HVA, were analyzed by HPLC. GFP-Saline n = 10, Ang-Saline n = 10, GFP-MPTP n = 9, Ang-MPTP n = 6. Error bars reflect SEM. A. Striatal dopamine levels as determined by HPLC. AAV2-Ang mice treated with MPTP showed a comparable loss in striatal DA. ***p<0.001 (One-way ANOVA with Tukey's post-hoc test). B. Striatal DOPAC levels as determined by HPLC. A similar reduction in DOPAC was seen in MPTP-treated AAV2-Ang injected mice as compared to AAV2-Ang MPTP. ***p<0.001 (One-way ANOVA with Tukey's post-hoc test). C. Striatal HVA levels as determined by HPLC. AAV2-Ang-injected mice treated with MPTP had a similar decrease in HVA as compared to AAV2-GFP mice treated with MPTP. ***p<0.001 (One-way ANOVA with Tukey's post-hoc test).</p

    K40I fails to activate Akt phosphorylation but protects against MPP+.

    No full text
    <p>A. Immunostaining of SH-SY5Y cells treated with exogenous angiogenin (100 nM) or K40I (100 nM) for 12 hours. Control is immunostaining of SH-SY5Y cells not treated with recombinant angiogenin and reflects low endogenous angiogenin levels in SH-SY5Y cells. Both wildtype and K40I recombinant angiogenin are taken up by SH-SY5Y cells. As the intensity in staining is stronger for wildtype angiogenin compared to K40I, either less K40I is taken up compared to wildtype or the antibody used better recognizes the wildtype form. Scale bar = 25 µm. B. Quantification of angiogenin immunostaining of SH-SY5Y cells treated with exogenous wildtype or K40I angiogenin. 10 random fields per well were imaged by confocal microscopy, and intensity was quantified using Image J. Average of 30 cells was analyzed per field, and two separate immunostaining runs were quantified. ***p<0.001 (students' t-test). C. Western blot demonstrating increased Akt phosphorylation after 30 sec treatment with recombinant angiogenin (100 nM) but not with the K40I angiogenin mutant (100 nM) in SH-SY5Y cells. Actin was used as the loading control. D. LDH assay of SH-SY5Y cells treated with MPP+ with and without wildtype or K40I angiogenin. Cells were pretreated with wildtype or K40I angiogenin (50–200 nM) for 12 hrs and then co-incubated with MPP+ (0.75 µM) and fresh angiogenin or K40I for another twenty-four hours. Angiogenin demonstrated a protective effect that was not significantly different from K40I. n = 4–6, **p<0.01, ***p<0.001 compared to MPP+ treatment (One-way ANOVA with Tukey's post-hoc test). There is no significant difference between wildtype and K40I-treated conditions. Error bars reflect SEM.</p

    Pharmacological studies on the NOP and opioid receptor agonist PWT2-[Dmt1]N/OFQ(1-13)

    Full text link
    An innovative chemical strategy named peptide welding technology (PWT) has been developed for the facile synthesis of tetrabranched peptides. [Dmt1]N/OFQ(1–13)-NH2 acts as a universal agonist for nociceptin/orphanin FQ (N/OFQ) and classical opioid receptors. The present study investigated the pharmacological profile of the PWT derivative of [Dmt1]N/OFQ(1–13)NH2 (PWT2-[Dmt1]) in several assays in vitro and in vivo after spinal administration in monkeys subjected to the tail withdrawal assay. PWT2-[Dmt1] mimicked the effects of [Dmt1]N/OFQ(1–13)-NH2 displaying full agonist activity, similar affinity/potency and selectivity at human recombinant N/OFQ (NOP) and opioid receptors in receptor binding, stimulation of [35S]GTPγS binding, calcium mobilization in cells expressing chimeric G proteins, and BRET studies for measuring receptor/G-protein and receptor/β-arrestin 2 interaction. In vivo in monkeys PWT2-[Dmt1] elicited dose-dependent and robust antinociceptive effects being more potent and longer lasting than [Dmt1]N/OFQ(1–13)-NH2. The analgesic action of PWT2-[Dmt1] was sensitive to the NOP receptor antagonist J-113397, but not naltrexone. Thus, the present study demonstrated that the tetrabranched derivative of [Dmt1]N/OFQ(1–13)-NH2 obtained with the PWT technology maintains the in vitro pharmacological profile of the parent peptide but displays higher potency and longer lasting action in vivo
    corecore