Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Cell ; 176(6): 1461-1476.e23, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30849374

ABSTRACT

Maintaining the optimal performance of cell processes and organelles is the task of auto-regulatory systems. Here we describe an auto-regulatory device that helps to maintain homeostasis of the endoplasmic reticulum (ER) by adjusting the secretory flux to the cargo load. The cargo-recruiting subunit of the coatomer protein II (COPII) coat, Sec24, doubles as a sensor of folded cargo and, upon cargo binding, acts as a guanine nucleotide exchange factor to activate the signaling protein Gα12 at the ER exit sites (ERESs). This step, in turn, activates a complex signaling network that activates and coordinates the ER export machinery and attenuates proteins synthesis, thus preventing large fluctuations of folded and potentially active cargo that could be harmful to the cell or the organism. We call this mechanism AREX (autoregulation of ER export) and expect that its identification will aid our understanding of human physiology and diseases that develop from secretory dysfunction.


Subject(s)
Endoplasmic Reticulum/metabolism , Vesicular Transport Proteins/metabolism , Biological Transport , COP-Coated Vesicles/metabolism , COP-Coated Vesicles/physiology , Cell Line , Coatomer Protein/metabolism , Endoplasmic Reticulum/physiology , Endoplasmic Reticulum Stress/physiology , Female , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Golgi Apparatus/metabolism , Guanine Nucleotide Exchange Factors/physiology , HeLa Cells , Humans , Male , Protein Folding , Protein Transport , Proteostasis/physiology , Signal Transduction
2.
Nano Lett ; 20(12): 8725-8732, 2020 12 09.
Article in English | MEDLINE | ID: mdl-33231075

ABSTRACT

Whereas most of the reports on the nonlinear properties of micro- and nanostructures address the generation of distinct signals, such as second or third harmonic, here we demonstrate that the novel generation of dual output lasers recently developed for microscopy can readily increase the accessible parameter space and enable the simultaneous excitation and detection of multiple emission orders such as several harmonics and signals stemming from various sum and difference frequency mixing processes. This rich response, which in our case features 10 distinct emissions and encompasses the whole spectral range from the deep ultraviolet to the short-wave infrared region, is demonstrated using various nonlinear oxide nanomaterials while being characterized and simulated temporally and spectrally. Notably, we show that the response is conserved when the particles are embedded in biological media opening the way to novel biolabeling and phototriggering strategies.


Subject(s)
Metal Nanoparticles , Nanostructures , Lasers , Oxides
3.
Biochem Soc Trans ; 47(5): 1341-1353, 2019 10 31.
Article in English | MEDLINE | ID: mdl-31671182

ABSTRACT

Cardiac stress initiates a pathological remodeling process that is associated with cardiomyocyte loss and fibrosis that ultimately leads to heart failure. In the injured heart, a pathologically elevated synthesis of reactive oxygen species (ROS) is the main driver of oxidative stress and consequent cardiomyocyte dysfunction and death. In this context, the cAMP-dependent protein kinase (PKA) plays a central role in regulating signaling pathways that protect the heart against ROS-induced cardiac damage. In cardiac cells, spatiotemporal regulation of PKA activity is controlled by A-kinase anchoring proteins (AKAPs). This family of scaffolding proteins tether PKA and other transduction enzymes at subcellular microdomains where they can co-ordinate cellular responses regulating oxidative stress. In this review, we will discuss recent literature illustrating the role of PKA and AKAPs in modulating the detrimental impact of ROS production on cardiac function.


Subject(s)
A Kinase Anchor Proteins/metabolism , Myocardium/metabolism , Oxidative Stress , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Humans , Reactive Oxygen Species/metabolism , Signal Transduction
4.
Biochim Biophys Acta Mol Cell Res ; 1864(12): 2336-2346, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28923249

ABSTRACT

Doxorubicin (DOX) is a chemotherapic agent that is widely used to treat hematological and solid tumors. Despite its efficacy, DOX displays significant cardiac toxicity associated with cardiomyocytes death and heart failure. Cardiac toxicity is mainly associated with the ability of DOX to alter mitochondrial function. The current lack of treatments to efficiently prevent DOX cardiotoxicity underscores the need of new therapeutic approaches. Our current findings show that stimulation of cardiomyocytes with the α1-adrenergic receptor (AR) agonist phenylephrine (PE) significantly inhibits the apoptotic effect of DOX. Importantly, our results indicate that AKAP-Lbc is critical for transducing protective signals downstream of α1-ARs. In particular, we could show that suppression of AKAP-Lbc expression by infecting primary cultures of ventricular myocytes with lentiviruses encoding AKAP-Lbc specific short hairpin (sh) RNAs strongly impairs the ability of PE to reduce DOX-induced apoptosis. AKAP-Lbc-mediated cardiomyocyte protection requires the activation of anchored protein kinase D1 (PKD1)-dependent prosurvival pathways that promote the expression of the anti-apoptotic protein Bcl2 and inhibit the translocation of the pro-apoptotic protein Bax to mitochondria. In conclusion, AKAP-Lbc emerges as a coordinator of signals that protect cardiomyocytes against the toxic effects of DOX.


Subject(s)
A Kinase Anchor Proteins/genetics , Apoptosis/drug effects , Doxorubicin/adverse effects , Minor Histocompatibility Antigens/genetics , Myocytes, Cardiac/drug effects , Proto-Oncogene Proteins/genetics , A Kinase Anchor Proteins/metabolism , Adrenergic alpha-1 Receptor Agonists/administration & dosage , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Lentivirus/genetics , Minor Histocompatibility Antigens/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Neoplasms/complications , Neoplasms/drug therapy , Phenylephrine/administration & dosage , Proto-Oncogene Proteins/metabolism , Signal Transduction/drug effects
5.
Biochim Biophys Acta ; 1863(7 Pt B): 1926-36, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26643253

ABSTRACT

Heart and blood vessels ensure adequate perfusion of peripheral organs with blood and nutrients. Alteration of the homeostatic functions of the cardiovascular system can cause hypertension, atherosclerosis, and coronary artery disease leading to heart injury and failure. A-kinase anchoring proteins (AKAPs) constitute a family of scaffolding proteins that are crucially involved in modulating the function of the cardiovascular system both under physiological and pathological conditions. AKAPs assemble multifunctional signaling complexes that ensure correct targeting of the cAMP-dependent protein kinase (PKA) as well as other signaling enzymes to precise subcellular compartments. This allows local regulation of specific effector proteins that control the function of vascular and cardiac cells. This review will focus on recent advances illustrating the role of AKAPs in cardiovascular pathophysiology. The accent will be mainly placed on the molecular events linked to the control of vascular integrity and blood pressure as well as on the cardiac remodeling process associated with heart failure. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Integration of Developmental and Environmental Cues in the Heart edited by Marcus Schaub and Hughes Abriel.


Subject(s)
A Kinase Anchor Proteins/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Heart Failure/enzymology , Muscle, Smooth, Vascular/enzymology , Myocytes, Cardiac/enzymology , Myocytes, Smooth Muscle/enzymology , Animals , Blood Pressure , Heart Failure/pathology , Heart Failure/physiopathology , Humans , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/physiopathology , Myocytes, Cardiac/pathology , Myocytes, Smooth Muscle/pathology , Signal Transduction , Vascular Remodeling , Ventricular Remodeling
6.
Mol Cell ; 32(2): 169-79, 2008 Oct 24.
Article in English | MEDLINE | ID: mdl-18951085

ABSTRACT

Elevated catecholamines in the heart evoke transcriptional activation of the Myocyte Enhancer Factor (MEF) pathway to induce a cellular response known as pathological myocardial hypertrophy. We have discovered that the A-Kinase Anchoring Protein (AKAP)-Lbc is upregulated in hypertrophic cardiomyocytes. It coordinates activation and movement of signaling proteins that initiate MEF2-mediated transcriptional reprogramming events. Live-cell imaging, fluorescent kinase activity reporters, and RNA interference techniques show that AKAP-Lbc couples activation of protein kinase D (PKD) with the phosphorylation-dependent nuclear export of the class II histone deacetylase HDAC5. These studies uncover a role for AKAP-Lbc in which increased expression of the anchoring protein selectively amplifies a signaling pathway that drives cardiac myocytes toward a pathophysiological outcome.


Subject(s)
A Kinase Anchor Proteins/physiology , Cardiomegaly/metabolism , Guanine Nucleotide Exchange Factors/physiology , Proto-Oncogene Proteins/physiology , Signal Transduction , 14-3-3 Proteins/metabolism , A Kinase Anchor Proteins/genetics , A Kinase Anchor Proteins/metabolism , Active Transport, Cell Nucleus , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression Regulation , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Heart Ventricles/drug effects , Histone Deacetylases/metabolism , Humans , MEF2 Transcription Factors , Minor Histocompatibility Antigens , Models, Biological , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myogenic Regulatory Factors/metabolism , Phenylephrine/pharmacology , Phosphorylation , Protein Kinase C/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA Interference , Rats
7.
Biochim Biophys Acta ; 1843(2): 335-45, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24269843

ABSTRACT

In response to stress or injury the heart undergoes an adverse remodeling process associated with cardiomyocyte hypertrophy and fibrosis. Transformation of cardiac fibroblasts to myofibroblasts is a crucial event initiating the fibrotic process. Cardiac myofibroblasts invade the myocardium and secrete excess amounts of extracellular matrix proteins, which cause myocardial stiffening, cardiac dysfunctions and progression to heart failure. While several studies indicate that the small GTPase RhoA can promote profibrotic responses, the exchange factors that modulate its activity in cardiac fibroblasts are yet to be identified. In the present study, we show that AKAP-Lbc, an A-kinase anchoring protein (AKAP) with an intrinsic Rho-specific guanine nucleotide exchange factor (GEF) activity, is critical for activating RhoA and transducing profibrotic signals downstream of type I angiotensin II receptors (AT1Rs) in cardiac fibroblasts. In particular, our results indicate that suppression of AKAP-Lbc expression by infecting adult rat ventricular fibroblasts with lentiviruses encoding AKAP-Lbc specific short hairpin (sh) RNAs strongly reduces the ability of angiotensin II to promote RhoA activation, differentiation of cardiac fibroblasts to myofibroblasts, collagen deposition as well as myofibroblast migration. Interestingly, AT1Rs promote AKAP-Lbc activation via a pathway that requires the α subunit of the heterotrimeric G protein G12. These findings identify AKAP-Lbc as a key Rho-guanine nucleotide exchange factor modulating profibrotic responses in cardiac fibroblasts.


Subject(s)
A Kinase Anchor Proteins/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Heart Ventricles/pathology , Signal Transduction , Actins/metabolism , Angiotensin II/pharmacology , Animals , Cell Differentiation/drug effects , Cell Movement/drug effects , Collagen/biosynthesis , Enzyme Activation/drug effects , Fibroblasts/drug effects , Fibrosis , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Gene Silencing/drug effects , Minor Histocompatibility Antigens , Models, Biological , Myofibroblasts/drug effects , Myofibroblasts/pathology , Phenotype , Rats , Signal Transduction/drug effects , Transforming Growth Factor beta1/metabolism , Up-Regulation/drug effects , rhoA GTP-Binding Protein/metabolism
8.
Proc Natl Acad Sci U S A ; 109(43): E2929-38, 2012 Oct 23.
Article in English | MEDLINE | ID: mdl-23027962

ABSTRACT

Spatial regulation of tyrosine phosphorylation is important for many aspects of cell biology. However, phosphotyrosine accounts for less than 1% of all phosphorylated substrates, and it is typically a very transient event in vivo. These factors complicate the identification of key tyrosine kinase substrates, especially in the context of their extraordinary spatial organization. Here, we describe an approach to identify tyrosine kinase substrates based on their subcellular distribution from within cells. This method uses an unnatural amino acid-modified Src homology 2 (SH2) domain that is expressed within cells and can covalently trap phosphotyrosine proteins on exposure to light. This SH2 domain-based photoprobe was targeted to cellular structures, such as the actin cytoskeleton, mitochondria, and cellular membranes, to capture tyrosine kinase substrates unique to each cellular region. We demonstrate that RhoA, one of the proteins associated with actin, can be phosphorylated on two tyrosine residues within the switch regions, suggesting that phosphorylation of these residues might modulate RhoA signaling to the actin cytoskeleton. We conclude that expression of SH2 domains within cellular compartments that are capable of covalent phototrapping can reveal the spatial organization of tyrosine kinase substrates that are likely to be important for the regulation of subcellular structures.


Subject(s)
Phosphoproteins/metabolism , Phosphotyrosine/metabolism , Subcellular Fractions/metabolism , src Homology Domains , Cell Compartmentation , HEK293 Cells , Humans , Mass Spectrometry , Phosphorylation
9.
Biochim Biophys Acta ; 1833(4): 901-8, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22889610

ABSTRACT

In response to stress or injury the heart undergoes a pathological remodeling process, associated with hypertrophy, cardiomyocyte death and fibrosis, that ultimately causes cardiac dysfunction and heart failure. It has become increasingly clear that signaling events associated with these pathological cardiac remodeling events are regulated by scaffolding and anchoring proteins, which allow coordination of pathological signals in space and time. A-kinase anchoring proteins (AKAPs) constitute a family of functionally related proteins that organize multiprotein signaling complexes that tether the cAMP-dependent protein kinase (PKA) as well as other signaling enzymes to ensure integration and processing of multiple signaling pathways. This review will discuss the role of AKAPs in the cardiac response to stress. Particular emphasis will be given to the adaptative process associated with cardiac hypoxia as well as the remodeling events linked to cardiac hypertrophy and heart failure. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Cardiac Pathways of Differentiation, Metabolism and Contraction.


Subject(s)
A Kinase Anchor Proteins/genetics , Cardiomegaly/metabolism , Hypoxia/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , A Kinase Anchor Proteins/metabolism , Adaptation, Physiological , Cardiomegaly/genetics , Cardiomegaly/pathology , Cyclic AMP/metabolism , Gene Expression Regulation , Humans , Hypoxia/genetics , Hypoxia/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Myocardium/pathology , Myocytes, Cardiac/pathology , Oxygen/metabolism , Protein Binding , Signal Transduction , Stress, Physiological
10.
Biochim Biophys Acta Mol Cell Res ; 1871(3): 119674, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38242328

ABSTRACT

Cardiac fibrosis is a major cause of dysfunctions and arrhythmias in failing hearts. At the cellular level fibrosis is mediated by cardiac myofibroblasts, which display an increased migratory capacity and secrete large amounts of extracellular matrix. These properties allow myofibroblasts to invade, remodel and stiffen the myocardium and eventually alter cardiac function. While the enhanced ability of cardiac myofibroblasts to migrate has been proposed to contribute to the initiation of the fibrotic process, the molecular mechanisms controlling their motile function have been poorly defined. In this context, our current findings indicate that A-kinase anchoring protein 2 (AKAP2) associates with actin at the leading edge of migrating cardiac myofibroblasts. Proteomic analysis of the AKAP2 interactome revealed that this anchoring protein assembles a signaling complex composed of the extracellular regulated kinase 1 (ERK1) and its upstream activator Grb2 that mediates the activation of ERK in cardiac myofibroblasts. Silencing AKAP2 expression results in a significant reduction in the phosphorylation of ERK1 and its downstream effector WAVE2, a protein involved in actin polymerization, and impairs the ability of cardiac myofibroblasts to migrate. Importantly, disruption of the interaction between AKAP2 and F-actin using cell-permeant competitor peptides, inhibits the activation of the ERK-WAVE2 signaling axis, resulting in a reduction of the translocation of Arp2 to the leading-edge membrane and in inhibition of cardiac myofibroblast migration. Collectively, these findings suggest that AKAP2 functions as an F-actin bound molecular scaffold mediating the activation of an ERK1-dependent promigratory transduction pathway in cardiac myofibroblasts.


Subject(s)
Actins , Myofibroblasts , Mitogen-Activated Protein Kinase 3 , Proteomics , Heart
11.
Virchows Arch ; 483(5): 635-643, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37726450

ABSTRACT

Extra-pituitary ACTH secretion is associated with a variety of neoplastic conditions and may cause the so-called ectopic ACTH-dependent Cushing syndrome (CS). The clarification of the mechanisms of extra-pituitary ACTH expression would provide potential therapeutic targets for this complex and severe disease. In the adenohypophysis, the transcription factor TPIT, co-operating with other molecules, induces POMC expression and ACTH production. However, no data are currently available on the presence and role of TPIT expression in extra-pituitary ACTH-producing neoplasms. This study was designed to explore TPIT expression in a series of pulmonary and pancreatic ACTH-producing tumors, either CS-associated or not. Forty-one extra-pituitary ACTH-producing neuroendocrine tumors (NETs) were included in the study, encompassing 32 NETs of the lung (LuNETs), 7 of the pancreas (PanNETs), and 2 pheochromocytomas. Of these, 9 LuNETs, all PanNETs, and the two pheochromocytomas were CS-associated. For comparison, 6 NETs of the pituitary gland (PitNETs; 3 ACTH-secreting and 3 ACTH-negative) and 35 ACTH-negative extra-pituitary NETs (15 Lu-NETs and 20 PanNETs) were analyzed. Immunohistochemistry with specific anti-TPIT antibodies and quantitative real-time PCR (qRT-PCR) were performed using standard protocols. TPIT expression was completely absent (protein and mRNA) in PanNETs, pheochromocytomas, and all ACTH-negative NETs. In contrast, it was expressed in 16/32 LuNETs, although with lower levels than in PitNETs. No definite relationship was found between immunohistochemistry TPIT expression and NET grade or the presence of Cushing syndrome. This study further highlights the clinical and biological heterogeneity of extra-pituitary ACTH secretion and suggests that the differences between ACTH-secreting PanNETs and LuNETs may mirror distinct molecular mechanisms underlying POMC expression. Our results point towards the recognition of a real corticotroph-like phenotype of ACTH-producing LuNETs, that is not a feature of ACTH-producing PanNETs.


Subject(s)
Adrenal Gland Neoplasms , Carcinoma, Neuroendocrine , Cushing Syndrome , Lung Neoplasms , Neuroendocrine Tumors , Pheochromocytoma , Pituitary Diseases , Pituitary Neoplasms , Humans , Adrenocorticotropic Hormone/metabolism , Lung Neoplasms/metabolism , Pancreas/pathology , Pituitary Gland/pathology , Pituitary Neoplasms/pathology , Pro-Opiomelanocortin/genetics , Pro-Opiomelanocortin/metabolism
12.
J Biol Chem ; 286(10): 7925-7937, 2011 Mar 11.
Article in English | MEDLINE | ID: mdl-21224381

ABSTRACT

The mitogen-activated protein kinases (MAPKs) pathways are highly organized signaling systems that transduce extracellular signals into a variety of intracellular responses. In this context, it is currently poorly understood how kinases constituting these signaling cascades are assembled and activated in response to receptor stimulation to generate specific cellular responses. Here, we show that AKAP-Lbc, an A-kinase anchoring protein (AKAP) with an intrinsic Rho-specific guanine nucleotide exchange factor activity, is critically involved in the activation of the p38α MAPK downstream of α(1b)-adrenergic receptors (α(1b)-ARs). Our results indicate that AKAP-Lbc can assemble a novel transduction complex containing the RhoA effector PKNα, MLTK, MKK3, and p38α, which integrates signals from α(1b)-ARs to promote RhoA-dependent activation of p38α. In particular, silencing of AKAP-Lbc expression or disrupting the formation of the AKAP-Lbc·p38α signaling complex specifically reduces α(1)-AR-mediated p38α activation without affecting receptor-mediated activation of other MAPK pathways. These findings provide a novel mechanistic hypothesis explaining how assembly of macromolecular complexes can specify MAPK signaling downstream of α(1)-ARs.


Subject(s)
A Kinase Anchor Proteins/metabolism , MAP Kinase Signaling System/physiology , Multienzyme Complexes/metabolism , Protein Kinase C/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, Adrenergic, alpha-1/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , A Kinase Anchor Proteins/genetics , Enzyme Activation/physiology , HEK293 Cells , Humans , Minor Histocompatibility Antigens , Multienzyme Complexes/genetics , Protein Kinase C/genetics , Proto-Oncogene Proteins/genetics , Receptors, Adrenergic, alpha-1/genetics , p38 Mitogen-Activated Protein Kinases/genetics , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
13.
Cardiovasc Res ; 118(2): 573-584, 2022 01 29.
Article in English | MEDLINE | ID: mdl-33576779

ABSTRACT

AIMS: Angiotensin (Ang) II signalling has been suggested to promote cardiac fibrosis in inflammatory heart diseases; however, the underlying mechanisms remain obscure. Using Agtr1a-/- mice with genetic deletion of angiotensin receptor type 1 (ATR1) and the experimental autoimmune myocarditis (EAM) model, we aimed to elucidate the role of Ang II-ATR1 pathway in development of heart-specific autoimmunity and post-inflammatory fibrosis. METHODS AND RESULTS: EAM was induced in wild-type (WT) and Agtr1a-/- mice by subcutaneous injections with alpha myosin heavy chain peptide emulsified in complete Freund's adjuvant. Agtr1a-/- mice developed myocarditis to a similar extent as WT controls at day 21 but showed reduced fibrosis and better systolic function at day 40. Crisscross bone marrow chimaera experiments proved that ATR1 signalling in the bone marrow compartment was critical for cardiac fibrosis. Heart infiltrating, bone-marrow-derived cells produced Ang II, but lack of ATR1 in these cells reduced transforming growth factor beta (TGF-ß)-mediated fibrotic responses. At the molecular level, Agtr1a-/- heart-inflammatory cells showed impaired TGF-ß-mediated phosphorylation of Smad2 and TAK1. In WT cells, TGF-ß induced formation of RhoA-GTP and RhoA-A-kinase anchoring protein-Lbc (AKAP-Lbc) complex. In Agtr1a-/- cells, stabilization of RhoA-GTP and interaction of RhoA with AKAP-Lbc were largely impaired. Furthermore, in contrast to WT cells, Agtr1a-/- cells stimulated with TGF-ß failed to activate canonical Wnt pathway indicated by suppressed activity of glycogen synthase kinase-3 (GSK-3)ß and nuclear ß-catenin translocation and showed reduced expression of Wnts. In line with these in vitro findings, ß-catenin was detected in inflammatory regions of hearts of WT, but not Agtr1a-/- mice and expression of canonical Wnt1 and Wnt10b were lower in Agtr1a-/- hearts. CONCLUSION: Ang II-ATR1 signalling is critical for development of post-inflammatory fibrotic remodelling and dilated cardiomyopathy. Our data underpin the importance of Ang II-ATR1 in effective TGF-ß downstream signalling response including activation of profibrotic Wnt/ß-catenin pathway.


Subject(s)
Angiotensin II/metabolism , Autoimmune Diseases/metabolism , Autoimmunity , CD4-Positive T-Lymphocytes/metabolism , Myocarditis/metabolism , Myocytes, Cardiac/metabolism , Receptor, Angiotensin, Type 1/metabolism , Wnt Signaling Pathway , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , CD4-Positive T-Lymphocytes/immunology , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Fibrosis , Inflammation Mediators/metabolism , Lymphocyte Activation , Mice, Inbred BALB C , Mice, Knockout , Myocarditis/genetics , Myocarditis/immunology , Myocarditis/pathology , Myocytes, Cardiac/immunology , Myocytes, Cardiac/pathology , Receptor, Angiotensin, Type 1/genetics , Wnt Proteins/genetics , Wnt Proteins/metabolism , Wnt1 Protein/genetics , Wnt1 Protein/metabolism , beta Catenin/genetics , beta Catenin/metabolism
14.
ACS Nanosci Au ; 2(4): 355-366, 2022 Aug 17.
Article in English | MEDLINE | ID: mdl-35996436

ABSTRACT

Nanoparticle-based drug delivery systems have the potential for increasing the efficiency of chemotherapeutics by enhancing the drug accumulation at specific target sites, thereby reducing adverse side effects and mitigating patient acquired resistance. In particular, photo-responsive nanomaterials have attracted much interest due to their ability to release molecular cargos on demand upon light irradiation. In some settings, they can also provide complementary information by optical imaging on the (sub)cellular scale. We herein present a system based on lithium niobate harmonic nanoparticles (LNO HNPs) for the decoupled multi-harmonic cell imaging and near-infrared light-triggered delivery of an erlotinib derivative (ELA) for the treatment of epidermal growth factor receptor (EGFR)-overexpressing carcinomas. The ELA cargo was covalently conjugated to the surface of silica-coated LNO HNPs through a coumarinyl photo-cleavable linker, achieving a surface loading of the active molecule of 27 nmol/mg NPs. The resulting nanoconjugates (LNO-CM-ELA NPs) were successfully imaged upon pulsed laser excitation at 1250 nm in EGFR-overexpressing human prostate cancer cells DU145 by detecting the second harmonic emission at 625 nm, in the tissue transparency window. Tuning the laser at 790 nm resulted in the uncaging of the ELA cargo as a result of the second harmonic emission of the inorganic HNP core at 395 nm. This protocol induced a significant growth inhibition in DU145 cells, which was only observed upon specific irradiation at 790 nm, highlighting the promising capabilities of LNO-CM-ELA NPs for theranostic applications.

15.
Am J Physiol Heart Circ Physiol ; 301(5): H1742-53, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21856912

ABSTRACT

The pleiotropic cyclic nucleotide cAMP is the primary second messenger responsible for autonomic regulation of cardiac inotropy, chronotropy, and lusitropy. Under conditions of prolonged catecholaminergic stimulation, cAMP also contributes to the induction of both cardiac myocyte hypertrophy and apoptosis. The formation of localized, multiprotein complexes that contain different combinations of cAMP effectors and regulatory enzymes provides the architectural infrastructure for the specialization of the cAMP signaling network. Scaffolds that bind protein kinase A are called "A-kinase anchoring proteins" (AKAPs). In this review, we discuss recent advances in our understanding of how PKA is compartmentalized within the cardiac myocyte by AKAPs and how AKAP complexes modulate cardiac function in both health and disease.


Subject(s)
A Kinase Anchor Proteins/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Heart Diseases/enzymology , Myocardium/enzymology , Second Messenger Systems , Animals , Cardiovascular Agents/therapeutic use , Heart Diseases/drug therapy , Heart Diseases/physiopathology , Humans , Second Messenger Systems/drug effects
16.
Cells ; 10(11)2021 10 23.
Article in English | MEDLINE | ID: mdl-34831084

ABSTRACT

Myocardial infarction (MI) is a leading cause of maladaptive cardiac remodeling and heart failure. In the damaged heart, loss of function is mainly due to cardiomyocyte death and remodeling of the cardiac tissue. The current study shows that A-kinase anchoring protein 2 (AKAP2) orchestrates cellular processes favoring cardioprotection in infarcted hearts. Induction of AKAP2 knockout (KO) in cardiomyocytes of adult mice increases infarct size and exacerbates cardiac dysfunction after MI, as visualized by increased left ventricular dilation and reduced fractional shortening and ejection fraction. In cardiomyocytes, AKAP2 forms a signaling complex with PKA and the steroid receptor co-activator 3 (Src3). Upon activation of cAMP signaling, the AKAP2/PKA/Src3 complex favors PKA-mediated phosphorylation and activation of estrogen receptor α (ERα). This results in the upregulation of ER-dependent genes involved in protection against apoptosis and angiogenesis, including Bcl2 and the vascular endothelial growth factor a (VEGFa). In line with these findings, cardiomyocyte-specific AKAP2 KO reduces Bcl2 and VEGFa expression, increases myocardial apoptosis and impairs the formation of new blood vessels in infarcted hearts. Collectively, our findings suggest that AKAP2 organizes a transcriptional complex that mediates pro-angiogenic and anti-apoptotic responses that protect infarcted hearts.


Subject(s)
A Kinase Anchor Proteins/metabolism , Cardiotonic Agents/metabolism , Membrane Proteins/metabolism , Myocardial Infarction/metabolism , A Kinase Anchor Proteins/genetics , Animals , Animals, Newborn , Apoptosis , Cyclic AMP-Dependent Protein Kinases/metabolism , Electrocardiography , Fibrosis , Gene Deletion , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/genetics , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Nuclear Receptor Coactivator 3/metabolism , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Up-Regulation/genetics , Vascular Endothelial Growth Factor A/metabolism
17.
J Biol Chem ; 284(41): 28232-28242, 2009 Oct 09.
Article in English | MEDLINE | ID: mdl-19696020

ABSTRACT

AKAP-Lbc is a member of the A-kinase anchoring protein (AKAP) family that has been recently associated with the development of pathologies, such as cardiac hypertrophy and cancer. We have previously demonstrated that, at the molecular level, AKAP-Lbc functions as a guanine nucleotide exchange factor (GEF) that promotes the specific activation of RhoA. In the present study, we identified the ubiquitin-like protein LC3 as a novel regulatory protein interacting with AKAP-Lbc. Mutagenesis studies revealed that LC3, through its NH(2)-terminal alpha-helical domain, interacts with two binding sites located within the NH(2)-terminal regulatory region of AKAP-Lbc. Interestingly, LC3 overexpression strongly reduced the ability of AKAP-Lbc to interact with RhoA, profoundly impairing the Rho-GEF activity of the anchoring protein and, as a consequence, its ability to promote cytoskeletal rearrangements associated with the formation of actin stress fibers. Moreover, AKAP-Lbc mutants that fail to interact with LC3 show a higher basal Rho-GEF activity as compared with the wild type protein and become refractory to the inhibitory effect of LC3. This suggests that LC3 binding maintains AKAP-Lbc in an inactive state that displays a reduced ability to promote downstream signaling. Collectively, these findings provide evidence for a previously uncharacterized role of LC3 in the regulation of Rho signaling and in the reorganization of the actin cytoskeleton.


Subject(s)
A Kinase Anchor Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Microtubule-Associated Proteins/metabolism , Proto-Oncogene Proteins/metabolism , A Kinase Anchor Proteins/genetics , Animals , Cell Line , Guanine Nucleotide Exchange Factors/genetics , Humans , Mice , Microtubule-Associated Proteins/genetics , Minor Histocompatibility Antigens , Models, Molecular , NIH 3T3 Cells , Proto-Oncogene Proteins/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Rho Guanine Nucleotide Exchange Factors , Signal Transduction/physiology , Two-Hybrid System Techniques
18.
Curr Opin Pharmacol ; 8(2): 166-73, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18165153

ABSTRACT

The cAMP-dependent kinase (PKA) is a broad specificity kinase that controls several fundamental processes in the heart including the strength and the frequency of contraction, the duration of the cardiac action potential as well as the activation of signaling pathways associated with the onset of cardiac hypertrophy and heart failure. It is now appreciated that to perform these functions, PKA must be precisely targeted in proximity to its cellular substrates. Evidence collected over the last years demonstrates that compartmentalization of the kinase is achieved through the association with A-kinase anchoring proteins (AKAPs). This family of functionally related proteins organize multivalent signaling complexes that target PKA and other signaling enzymes at precise subcellular sites within cardiomyocytes where they can be accessed by activators and, in turn, phosphorylate and modulate particular substrates.


Subject(s)
A Kinase Anchor Proteins/physiology , Cyclic AMP-Dependent Protein Kinases/physiology , Myocardial Contraction/physiology , Action Potentials/physiology , Animals , Calcium/metabolism , Cardiomegaly/drug therapy , Cardiomegaly/physiopathology , Drug Delivery Systems , Heart Failure/drug therapy , Heart Failure/physiopathology , Humans , Signal Transduction
19.
Cells ; 9(1)2019 Dec 26.
Article in English | MEDLINE | ID: mdl-31888098

ABSTRACT

Myocardial stress and injury invariably promote remodeling of the cardiac tissue, which is associated with cardiomyocyte death and development of fibrosis. The fibrotic process is initially triggered by the differentiation of resident cardiac fibroblasts into myofibroblasts. These activated fibroblasts display increased proliferative capacity and secrete large amounts of extracellular matrix. Uncontrolled myofibroblast activation can thus promote heart stiffness, cardiac dysfunction, arrhythmias, and progression to heart failure. Despite the well-established role of myofibroblasts in mediating cardiac disease, our current knowledge on how signaling pathways promoting fibrosis are regulated and coordinated in this cell type is largely incomplete. In this respect, cyclic adenosine monophosphate (cAMP) signaling acts as a major modulator of fibrotic responses activated in fibroblasts of injured or stressed hearts. In particular, accumulating evidence now suggests that upstream cAMP modulators including G protein-coupled receptors, adenylyl cyclases (ACs), and phosphodiesterases (PDEs); downstream cAMP effectors such as protein kinase A (PKA) and the guanine nucleotide exchange factor Epac; and cAMP signaling organizers such as A-kinase anchoring proteins (AKAPs) modulate a variety of fundamental cellular processes involved in myocardial fibrosis including myofibroblast differentiation, proliferation, collagen secretion, and invasiveness. The current review will discuss recent advances highlighting the role of cAMP and AKAP-mediated signaling in regulating pathophysiological responses controlling cardiac fibrosis.


Subject(s)
Cardiomyopathies/etiology , Cardiomyopathies/metabolism , Cyclic AMP/metabolism , Disease Susceptibility , Myocytes, Cardiac/metabolism , Signal Transduction , Animals , Biomarkers , Cardiomyopathies/pathology , Fibroblasts/metabolism , Fibrosis , Gene Expression Regulation , Humans
20.
J Cardiovasc Dev Dis ; 5(1)2018 Feb 08.
Article in English | MEDLINE | ID: mdl-29419761

ABSTRACT

Heart failure is a lethal disease that can develop after myocardial infarction, hypertension, or anticancer therapy. In the damaged heart, loss of function is mainly due to cardiomyocyte death and associated cardiac remodeling and fibrosis. In this context, A-kinase anchoring proteins (AKAPs) constitute a family of scaffolding proteins that facilitate the spatiotemporal activation of the cyclic adenosine monophosphate (AMP)-dependent protein kinase (PKA) and other transduction enzymes involved in cardiac remodeling. AKAP-Lbc, a cardiac enriched anchoring protein, has been shown to act as a key coordinator of the activity of signaling pathways involved in cardiac protection and remodeling. This review will summarize and discuss recent advances highlighting the role of the AKAP-Lbc signalosome in orchestrating adaptive responses in the stressed heart.

SELECTION OF CITATIONS
SEARCH DETAIL