GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
Filter
Material
Language
  • 1
    In: Journal of Molecular Medicine, Springer Science and Business Media LLC, Vol. 89, No. 12 ( 2011-12), p. 1219-1229
    Type of Medium: Online Resource
    ISSN: 0946-2716 , 1432-1440
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2011
    detail.hit.zdb_id: 1462132-0
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Hypertension, Ovid Technologies (Wolters Kluwer Health), Vol. 74, No. Suppl_1 ( 2019-09)
    Abstract: Hypertension is the greatest driver of cardiovascular disease, the most common cause of death. Finding novel model mechanisms for blood-pressure (BP) regulation is an important goal. Autosomal-dominant hypertension with brachydactyly (HTNB) clinically resembles salt-resistant essential hypertension and causes death by stroke, commonly before age 50 years. We identified mutated phosphodiesterase 3A as responsible earlier; however, definitive mechanistic proof and animal models were lacking. We now describe new kindreds, one substituted within the earlier-described regulatory region (G449S), and another with a mutation causing an amino acid substitution within the PDE3A enzymatic pocket (R864C). Both exhibit the HTNB phenotype. Mutant PDE3A (T445N) overexpression in mouse vascular smooth muscle cells causes hypertension (mean telemetry BP 130/88 mm Hg vs littermate control 115/84 mm Hg, p 〈 0.01). A 9 base-pair, three amino acid (3AA) deletion within the regulatory segment (using CRISPR-Cas9 technology), recapitulates HTNB in the rat, including the skeletal (with micro CT) phenotype. Three-weeks of radiotelemetry in homozygous 3AA+/+ rats at age 6 months revealed mean systolic BP 151, diastolic BP 119 mm Hg; heterozygous 3AA+/- (like the patients) showed 148/105 mm Hg; PDE3A knockout (frame-shift mutation) rats exhibited the lowest pressures at 115/87 mm Hg (all p 〈 0.001) vs. BP of wild-type Sprague-Dawley rats at mean 126/90 mm Hg. R864C causes increased oligomerization. Furthermore, R864C, T445N, and 3AA all cause increased interactions at the scaffolding protein, 14-3-3 theta, compared to the wild-type (P 〈 0.05). With two separate rodent models, our data not only direct mechanistic hypertension research along non-renal pathways, but also identify new areas of possible therapeutic intervention. We suggest that PDE3A could represent a generalizable novel therapeutic target in all instances of elevated blood pressure.
    Type of Medium: Online Resource
    ISSN: 0194-911X , 1524-4563
    Language: English
    Publisher: Ovid Technologies (Wolters Kluwer Health)
    Publication Date: 2019
    detail.hit.zdb_id: 2094210-2
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    Ovid Technologies (Wolters Kluwer Health) ; 2021
    In:  Circulation Vol. 144, No. Suppl_1 ( 2021-11-16)
    In: Circulation, Ovid Technologies (Wolters Kluwer Health), Vol. 144, No. Suppl_1 ( 2021-11-16)
    Abstract: Hypertension affects more than 1 billion people worldwide and can cause cardiac hypertrophy and ultimately heart failure, a condition with an unmet medical need. We have shown that missense mutations in a 15 bp regulatory domain of the phosphodiesterase 3A ( PDE3A ) gene cause hypertension with brachydactyly type E (HTNB). 1, 2 We recently discovered two HTNB families with new mutations in the region encoding the catalytic domain of PDE3A. CRISPR-Cas9 was used to generate rat models of HTNB by introducing mutations in the regulatory or catalytic domain of the PDE3A gene. Surprisingly, despite the hypertension the hearts of the HTNB rats, even in response to catecholamine challenge, were similar to those of the wild type controls, as indicated by echocardiography. mRNA and protein expression analyses did hardly detect significant differences between the left ventricles of the mutant and wild type animals. However, the mutations consistently caused aberrations of PDE3A phosphorylation, enhanced interaction with the adaptor protein, 14-3-3θ, and aberrant cellular localisation. Fluorescence resonance energy transfer (FRET) experiments showed that all PDE3A mutants were hyperactive. Calcium imaging of cardiac myocytes isolated from the rat models revealed differences in calcium cycling between the mutants and wild type animals. Taken together, the data indicate that mutant PDE3A causes aberrant signalling, and suggest that an enhanced local activity of PDE3A is cardioprotective and may provide a basis for new approaches towards heart failure treatment. 1.Maass PG et al. PDE3A mutations cause autosomal dominant hypertension with brachydactyly. Nat Genet . 2015;47:647-53.2.Ercu M, Marko L et al. Phosphodiesterase 3A and Arterial Hypertension. Circulation . 2020;142:133-149. HTNB Consortium: Ryan Walker-Gray, Anastasiia Sholokh, Claudia Loica, Tamara Pallien, Michael B. Mücke, Kerstin Zühlke, Andrea Geelhaar, Michael Russwurm, Lajos Markó, Theda U.P. Bartolomaeus, Hanna Napieczynska, Stefanie Schelenz, Martin Taube, Arnd Heuser, Tatiana Borodina, Daniele Yumi Sunaga-Franze, Jenny Eichhorst, Martin Lehmann, Fatimunnisa Qadri, Elena Popova, Reika Langanki, Sofia K. Forslund, Dominik N. Müller, Norbert Hübner, Sylvia Båhring, Michael Bader
    Type of Medium: Online Resource
    ISSN: 0009-7322 , 1524-4539
    Language: English
    Publisher: Ovid Technologies (Wolters Kluwer Health)
    Publication Date: 2021
    detail.hit.zdb_id: 1466401-X
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Circulation, Ovid Technologies (Wolters Kluwer Health), Vol. 144, No. Suppl_1 ( 2021-11-16)
    Abstract: Mutations causing hyperactivity of phosphodiesterase 3A (PDE3A) lead to autosomal dominant hypertension with brachydactyly type E. Affected patients suffer from profoundly increased blood pressure and, if untreated, die from stroke at the age of 50 years. Surprisingly, the hearts of affected individuals show little signs of hypertrophic responses to these conditions, indicating a beneficial effect of PDE3A hyperactivity on the damaging mechanisms of the heart. PDE3A inhibition was previously tested as a treatment for heart failure. While PDE3A inhibitors lead to short term positive inotropy, treatment with these drugs decreases overall survival of heart failure patients, underlining the importance of PDE3A in the heart. As arterial hypertension is a major risk factor for cardiovascular disease and proves to be a major burden on the healthcare system, dissecting the mechanisms underlying the protective effect of PDE3A can open new paths to preventing and treating heart failure. We hypothesize that PDE3A activation protects the heart from the detrimental effects of high blood pressure via spatially distinct regulation of cAMP levels through its different isoforms, leading to an altered calcium handling of cardiomyocytes. We are focusing on two mutations causing PDE3A hyperactivity, one in a regulatory hotspot the other in the catalytic domain. Using single nuclei sequencing of left ventricles of rats, genetically engineered to recapitulate the human mutations, we are dissecting the differences in the cell type-specific gene regulatory response to high blood pressure compared to the wildtype. To follow up in a human model system, we used genome editing to introduce the mutations of interest into human induced pluripotent stem cells (hiPSC). hiPSC lines containing PDE3A mutations as well as isogenic matched control hiPSCs were differentiated to generate hiPSC-derived cardiomyocytes (hiPSC-CMs). Preliminary data from a well-established 2D system show significantly altered calcium handling of PDE3A-mutant hiPSC-CMs. Performing RNA-sequencing, calcium imaging and cAMP measurements in isogenic hiPSC-CMs, we are functionally characterizing the effect of PDE3A activity on cardiomyocyte profile and physiology to a high resolution.
    Type of Medium: Online Resource
    ISSN: 0009-7322 , 1524-4539
    Language: English
    Publisher: Ovid Technologies (Wolters Kluwer Health)
    Publication Date: 2021
    detail.hit.zdb_id: 1466401-X
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    Wiley ; 2012
    In:  British Journal of Pharmacology Vol. 166, No. 2 ( 2012-05), p. 420-433
    In: British Journal of Pharmacology, Wiley, Vol. 166, No. 2 ( 2012-05), p. 420-433
    Abstract: A‐kinase anchoring proteins (AKAPs) crucially contribute to the spatial and temporal control of cellular signalling. They directly interact with a variety of protein binding partners and cellular constituents, thereby directing pools of signalling components to defined locales. In particular, AKAPs mediate compartmentalization of cAMP signalling. Alterations in AKAP expression and their interactions are associated with or cause diseases including chronic heart failure, various cancers and disorders of the immune system such as HIV. A number of cellular dysfunctions result from mutations of specific AKAPs. The link between malfunctions of single AKAP complexes and a disease makes AKAPs and their interactions interesting targets for the development of novel drugs. LINKED ARTICLES This article is part of a themed section on Novel cAMP Signalling Paradigms. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2012.166.issue‐2
    Type of Medium: Online Resource
    ISSN: 0007-1188 , 1476-5381
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2012
    detail.hit.zdb_id: 2029728-2
    SSG: 15,3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    The Company of Biologists ; 2003
    In:  Journal of Cell Science Vol. 116, No. 8 ( 2003-04-15), p. 1519-1525
    In: Journal of Cell Science, The Company of Biologists, Vol. 116, No. 8 ( 2003-04-15), p. 1519-1525
    Abstract: We have recently demonstrated that inhibition of Rho GTPase with Clostridium difficile toxin B, or with Clostridium botulinumC3 toxin, causes actin depolymerization and translocation of aquaporin 2(AQP2) in renal CD8 cells in the absence of hormonal stimulation. Here we demonstrate that Rho inhibition is part of the signal transduction cascade activated by vasopressin leading to AQP2 insertion into the apical membrane. Quantitation of active RhoA (GTP-bound) by selective pull down experiments demonstrated that the amount of active RhoA decreased upon stimulation of CD8 cells with the cAMP-elevating agent forskolin. Consistent with this observation, forskolin treatment resulted in a decreased expression of membrane-associated (active) Rho, as assessed by cell fractionation followed by western blotting analysis. In addition, the abundance of the endogenous Rho GDP dissociation inhibitor (Rho-GDI) was found to have decreased in the membrane fraction after forskolin stimulation. Co-immunoprecipitation experiments revealed that, after forskolin stimulation, the amount of Rho-GDI complexed with RhoA increased, suggesting that Rho GTPase inhibition occurs through association of RhoA with Rho-GDI. Finally, forskolin stimulation was associated with an increase in Rho phosphorylation on a serine residue, a protein modification known to stabilize the inactive form of RhoA and to increase its interaction with Rho-GDI. Taken together, these data demonstrate that RhoA inhibition through Rho phosphorylation and interaction with Rho-GDI is a key event for cytoskeletal dynamics controlling cAMP-induced AQP2 translocation.
    Type of Medium: Online Resource
    ISSN: 1477-9137 , 0021-9533
    Language: English
    Publisher: The Company of Biologists
    Publication Date: 2003
    detail.hit.zdb_id: 219171-4
    detail.hit.zdb_id: 1483099-1
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: EMBO reports, EMBO, Vol. 8, No. 11 ( 2007-11), p. 1061-1067
    Type of Medium: Online Resource
    ISSN: 1469-221X , 1469-3178
    Language: English
    Publisher: EMBO
    Publication Date: 2007
    detail.hit.zdb_id: 2025376-X
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Circulation Research, Ovid Technologies (Wolters Kluwer Health), Vol. 130, No. 1 ( 2022-01-07), p. 27-44
    Abstract: The sarcoplasmic reticulum (SR) Ca 2+ -ATPase 2 (SERCA2) mediates Ca 2+ reuptake into SR and thereby promotes cardiomyocyte relaxation, whereas the ryanodine receptor (RYR) mediates Ca 2+ release from SR and triggers contraction. Ca 2+ /CaMKII (CaM [calmodulin]-dependent protein kinase II) regulates activities of SERCA2 through phosphorylation of PLN (phospholamban) and RYR through direct phosphorylation. However, the mechanisms for CaMKIIδ anchoring to SERCA2-PLN and RYR and its regulation by local Ca 2+ signals remain elusive. The objective of this study was to investigate CaMKIIδ anchoring and regulation at SERCA2-PLN and RYR. Methods: A role for AKAP18δ (A-kinase anchoring protein 18δ) in CaMKIIδ anchoring and regulation was analyzed by bioinformatics, peptide arrays, cell-permeant peptide technology, immunoprecipitations, pull downs, transfections, immunoblotting, proximity ligation, FRET-based CaMKII activity and ELISA-based assays, whole cell and SR vesicle fluorescence imaging, high-resolution microscopy, adenovirus transduction, adenoassociated virus injection, structural modeling, surface plasmon resonance, and alpha screen technology. Results: Our results show that AKAP18δ anchors and directly regulates CaMKIIδ activity at SERCA2-PLN and RYR, via 2 distinct AKAP18δ regions. An N-terminal region (AKAP18δ-N) inhibited CaMKIIδ through binding of a region homologous to the natural CaMKII inhibitor peptide and the Thr17-PLN region. AKAP18δ-N also bound CaM, introducing a second level of control. Conversely, AKAP18δ-C, which shares homology to neuronal CaMKIIα activator peptide (N2B-s), activated CaMKIIδ by lowering the apparent Ca 2+ threshold for kinase activation and inducing CaM trapping. While AKAP18δ-C facilitated faster Ca 2+ reuptake by SERCA2 and Ca 2+ release through RYR, AKAP18δ-N had opposite effects. We propose a model where the 2 unique AKAP18δ regions fine-tune Ca 2+ -frequency-dependent activation of CaMKIIδ at SERCA2-PLN and RYR. Conclusions: AKAP18δ anchors and functionally regulates CaMKII activity at PLN-SERCA2 and RYR, indicating a crucial role of AKAP18δ in regulation of the heartbeat. To our knowledge, this is the first protein shown to enhance CaMKII activity in heart and also the first AKAP (A-kinase anchoring protein) reported to anchor a CaMKII isoform, defining AKAP18δ also as a CaM-KAP.
    Type of Medium: Online Resource
    ISSN: 0009-7330 , 1524-4571
    RVK:
    Language: English
    Publisher: Ovid Technologies (Wolters Kluwer Health)
    Publication Date: 2022
    detail.hit.zdb_id: 1467838-X
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Biochemical Journal, Portland Press Ltd., Vol. 398, No. 1 ( 2006-08-15), p. 23-36
    Abstract: The cAMP-specific phosphodiesterase PDE4D5 can interact with the signalling scaffold proteins RACK (receptors for activated C-kinase) 1 and β-arrestin. Two-hybrid and co-immunoprecipitation analyses showed that RACK1 and β-arrestin interact with PDE4D5 in a mutually exclusive manner. Overlay studies with PDE4D5 scanning peptide array libraries showed that RACK1 and β-arrestin interact at overlapping sites within the unique N-terminal region of PDE4D5 and at distinct sites within the conserved PDE4 catalytic domain. Screening scanning alanine substitution peptide arrays, coupled with mutagenesis and truncation studies, allowed definition of RACK1 and β-arrestin interaction sites. Modelled on the PDE4D catalytic domain, these form distinct well-defined surface-exposed patches on helices-15–16, for RACK1, and helix-17 for β-arrestin. siRNA (small interfering RNA)-mediated knockdown of RACK1 in HEK-293 (human embryonic kidney) B2 cells increased β-arrestin-scaffolded PDE4D5 approx. 5-fold, increased PDE4D5 recruited to the β2AR (β2-adrenergic receptor) upon isoproterenol challenge approx. 4-fold and severely attenuated (approx. 4–5 fold) both isoproterenol-stimulated PKA (protein kinase A) phosphorylation of the β2AR and activation of ERK (extracellular-signal-regulated kinase). The ability of a catalytically inactive form of PDE4D5 to exert a dominant negative effect in amplifying isoproterenol-stimulated ERK activation was ablated by a mutation that blocked the interaction of PDE4D5 with β-arrestin. In the present study, we show that the signalling scaffold proteins RACK1 and β-arrestin compete to sequester distinct ‘pools’ of PDE4D5. In this fashion, alterations in the level of RACK1 expression may act to modulate signal transduction mediated by the β2AR.
    Type of Medium: Online Resource
    ISSN: 0264-6021 , 1470-8728
    RVK:
    Language: English
    Publisher: Portland Press Ltd.
    Publication Date: 2006
    detail.hit.zdb_id: 1473095-9
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Cellular Signalling, Elsevier BV, Vol. 27, No. 4 ( 2015-04), p. 756-769
    Type of Medium: Online Resource
    ISSN: 0898-6568
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2015
    detail.hit.zdb_id: 1496718-2
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...