Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Andrea Barbuti is active.

Publication


Featured researches published by Andrea Barbuti.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Deep bradycardia and heart block caused by inducible cardiac-specific knockout of the pacemaker channel gene Hcn4

Mirko Baruscotti; Annalisa Bucchi; Carlo Viscomi; Giacomo Mandelli; Giacomo Consalez; Tomaso Gnecchi-Rusconi; Nicola Montano; Karina Rabello Casali; Stefano Micheloni; Andrea Barbuti; Dario DiFrancesco

Cardiac pacemaking generation and modulation rely on the coordinated activity of several processes. Although a wealth of evidence indicates a relevant role of the If (“funny,” or pacemaker) current, whose molecular constituents are the hyperpolarization-activated, cyclic nucleotide-gated (HCN) channels and particularly HCN4, work with mice where Hcn genes were knocked out, or functionally modified, has challenged this view. However, no previous studies used a cardiac-specific promoter to induce HCN4 ablation in adult mice. We report here that, in an inducible and cardiac-specific HCN4 knockout (ciHCN4-KO) mouse model, ablation of HCN4 consistently leads to progressive development of severe bradycardia (∼50% reduction of original rate) and AV block, eventually leading to heart arrest and death in about 5 d. In vitro analysis of sinoatrial node (SAN) myocytes isolated from ciHCN4-KO mice at the mean time of death revealed a strong reduction of both the If current (by ∼70%) and of the spontaneous rate (by ∼60%). In agreement with functional results, immunofluorescence and Western blot analysis showed reduced expression of HCN4 protein in SAN tissue and cells. In ciHCN4-KO animals, the residual If was normally sensitive to β-adrenergic receptor (β-AR) modulation, and the permanence of rate response to β-AR stimulation was observed both in vivo and in vitro. Our data show that cardiac HCN4 channels are essential for normal heart impulse generation and conduction in adult mice and support the notion that dysfunctional HCN4 channels can be a direct cause of rhythm disorders. This work contributes to identifying the molecular mechanism responsible for cardiac pacemaking.


Circulation Research | 2004

Localization of Pacemaker Channels in Lipid Rafts Regulates Channel Kinetics

Andrea Barbuti; Biagio Gravante; Monica Riolfo; Raffaella Milanesi; Benedetta Terragni; Dario DiFrancesco

Lipid rafts are discrete membrane subdomains rich in sphingolipids and cholesterol. In ventricular myocytes a function of caveolae, a type of lipid rafts, is to concentrate in close proximity several proteins of the β-adrenergic transduction pathway. We have investigated the subcellular localization of HCN4 channels expressed in HEK cells and studied the effects of such localization on the properties of pacemaker channels in HEK and rabbit sinoatrial (SAN) cells. We used a discontinuous sucrose gradient and Western blot analysis to detect HCN4 proteins in HEK and in SAN cells, and found that HCN4 proteins localize to low-density membrane fractions together with flotillin (HEK) or caveolin-3 (SAN), structural proteins of caveolae. Lipid raft disruption by cell incubation with methyl-β-cyclodextrin (MβCD) impaired specific HCN4 localization. It also shifted the midpoint of activation of the HCN4 current in HEK cells and of If in SAN cells to the positive direction by 11.9 and 10.4 mV, respectively. These latter effects were not due to elevation of basal cyclic nucleotide levels because the cholesterol-depletion treatment did not alter the current response to cyclic nucleotides. In accordance with an increased If, MβCD-treated SAN cells showed large increases of diastolic depolarization slope (87%) and rate (58%). We also found that the kinetics of HCN4- and native f-channel deactivation were slower after lipid raft disorganization. In conclusion, our work indicates that pacemaker channels localize to lipid rafts and that disruption of lipid rafts causes channels to redistribute within the membrane and modifies their kinetic properties.


Journal of Molecular and Cellular Cardiology | 2010

The cardiac pacemaker current.

Mirko Baruscotti; Andrea Barbuti; Annalisa Bucchi

In mammals cardiac rate is determined by the duration of the diastolic depolarization of sinoatrial node (SAN) cells which is mainly determined by the pacemaker I(f) current. f-channels are encoded by four members of the hyperpolarization-activated cyclic nucleotide-gated gene (HCN1-4) family. HCN4 is the most abundant isoform in the SAN, and its relevance to pacemaking has been further supported by the discovery of four loss-of-function mutations in patients with mild or severe forms of cardiac rate disturbances. Due to its selective contribution to pacemaking, the I(f) current is also the pharmacological target of a selective heart rate-reducing agent (ivabradine) currently used in the clinical practice. Albeit to a minor extent, the I(f) current is also present in other spontaneously active myocytes of the cardiac conduction system (atrioventricular node and Purkinje fibres). In working atrial and ventricular myocytes f-channels are expressed at a very low level and do not play any physiological role; however in certain pathological conditions over-expression of HCN proteins may represent an arrhythmogenic mechanism. In this review some of the most recent findings on f/HCN channels contribution to pacemaking are described.


Cell Death & Differentiation | 2008

Cardiac mesoangioblasts are committed, self-renewable progenitors, associated with small vessels of juvenile mouse ventricle

Beatriz G. Gálvez; Maurilio Sampaolesi; Andrea Barbuti; Alessia Crespi; Diego Covarello; Silvia Brunelli; Arianna Dellavalle; Stefania Crippa; Giovanna Balconi; Ivan Cuccovillo; Fabiola Molla; Lidia Staszewsky; Roberto Latini; Dario DiFrancesco; Giulio Cossu

Different cardiac stem/progenitor cells have been recently identified in the post-natal heart. We describe here the identification, clonal expansion and characterization of self-renewing progenitors that differ from those previously described for high spontaneous cardiac differentiation. Unique coexpression of endothelial and pericyte markers identify these cells as cardiac mesoangioblasts and allow prospective isolation and clonal expansion from the juvenile mouse ventricle. Cardiac mesoangioblasts express many cardiac transcription factors and spontaneously differentiate into beating cardiomyocytes that assemble mature sarcomeres and express typical cardiac ion channels. Cells similarly isolated from the atrium do not spontaneously differentiate. When injected into the ventricle after coronary artery ligation, cardiac mesoangioblasts efficiently generate new myocardium in the peripheral area of the necrotic zone, as they do when grafted in the embryonic chick heart. These data identify cardiac mesoangioblasts as committed progenitors, downstream of earlier stem/progenitor cells and suitable for the cell therapy of a subset of juvenile cardiac diseases.


Journal of Biological Chemistry | 2004

Interaction of the Pacemaker Channel HCN1 with Filamin A

Biagio Gravante; Andrea Barbuti; Raffaella Milanesi; Ivan Zappi; Carlo Viscomi; Dario DiFrancesco

Pacemaker channels are encoded by the HCN gene family and are responsible for a variety of cellular functions including control of spontaneous activity in cardiac myocytes and control of excitability in different types of neurons. Some of these functions require specific membrane localization. Although several voltage-gated channels are known to interact with intracellular proteins exerting auxiliary functions, no cytoplasmic proteins have been found so far to modulate HCN channels. Through the use of a yeast two-hybrid technique, here we showed that filamin A interacts with HCN1, an HCN isoform widely expressed in the brain, but not with HCN2 or HCN4. Filamin A is a cytoplasmic scaffold protein with actin-binding domains whose main function is to link transmembrane proteins to the actin cytoskeleton. Using several HCN1 C-terminal constructs, we identified a filamin A-interacting region of 22 amino acids located downstream from the cyclic nucleotide-binding domain; this region is not conserved in HCN2, HCN3, or HCN4. We also verified by immunoprecipitation from bovine brain that the filamin A-HCN1 interaction is functional in vivo. In filamin A-expressing cells (filamin+), HCN1 (but not HCN4) channels were expressed in hot spots, whereas they were evenly distributed on the membrane of cells lacking filamin A (filamin–) indicating that interaction with filamin A affects membrane localization. Also, in filamin– cells the gating kinetics of HCN1 were strongly accelerated relative to filamin+ cells. The interaction with filamin A may contribute to localizing HCN1 channels to specific neuronal areas and to modulating channel activity.


Cardiovascular Research | 2011

Human cardiac and bone marrow stromal cells exhibit distinctive properties related to their origin

Alessandra Rossini; Caterina Frati; Costanza Lagrasta; Gallia Graiani; Stefano Cavalli; Ezio Musso; Marco Baccarin; Marina Di Segni; Francesco Fagnoni; Antonia Germani; Eugenio Quaini; Manuel Mayr; Qingbo Xu; Andrea Barbuti; Dario DiFrancesco; Giulio Pompilio; Federico Quaini; Carlo Gaetano; Maurizio C. Capogrossi

AIMS Bone marrow mesenchymal stromal cell (BMStC) transplantation into the infarcted heart improves left ventricular function and cardiac remodelling. However, it has been suggested that tissue-specific cells may be better for cardiac repair than cells from other sources. The objective of the present work has been the comparison of in vitro and in vivo properties of adult human cardiac stromal cells (CStC) to those of syngeneic BMStC. METHODS AND RESULTS Although CStC and BMStC exhibited a similar immunophenotype, their gene, microRNA, and protein expression profiles were remarkably different. Biologically, CStC, compared with BMStC, were less competent in acquiring the adipogenic and osteogenic phenotype but more efficiently expressed cardiovascular markers. When injected into the heart, in rat a model of chronic myocardial infarction, CStC persisted longer within the tissue, migrated into the scar, and differentiated into adult cardiomyocytes better than BMStC. CONCLUSION Our findings demonstrate that although CStC and BMStC share a common stromal phenotype, CStC present cardiovascular-associated features and may represent an important cell source for more efficient cardiac repair.


Journal of Cardiovascular Electrophysiology | 2007

The pacemaker current: from basics to the clinics.

Andrea Barbuti; Mirko Baruscotti; Dario DiFrancesco

Activation of the pacemaker (“funny,” If) current during diastole is the main process underlying generation of the diastolic depolarization and spontaneous activity of cardiac pacemaker cells. If modulation by autonomic transmitters is responsible for the chronotropic regulation of heart rate. Given its role in pacemaking, If has been a major target of investigation aimed to exploit its rate‐controlling function in a clinical perspective. In this short review, we describe some of the most recent clinically relevant applications of the concept of If‐based pacemaking.


Journal of Molecular and Cellular Cardiology | 2009

Distribution of the pacemaker HCN4 channel mRNA and protein in the rabbit sinoatrial node.

Chiara Brioschi; Stefano Micheloni; James O. Tellez; G. Pisoni; Renato Longhi; P. Moroni; Rudi Billeter; Andrea Barbuti; Halina Dobrzynski; Mark R. Boyett; Dario DiFrancesco; Mirko Baruscotti

Several studies of the pacemaker mechanisms in mammalian cells, most of which were carried out in cells isolated from the rabbit sinoatrial node (SAN), have highlighted the role of the I(f) current. While the distribution of Hyperpolarization-activated Cyclic Nucleotide-gated (HCN) channels, the molecular correlates of f-channels, is known at the mRNA level, the identification of f-channel proteins in this tissue is still undetermined. Here we investigate HCN protein expression in the rabbit pacemaker region. We found that HCN4 is the main isoform, and set therefore to analyze its distribution within the SAN and surrounding areas with the aim of correlating protein expression and pacemaking function. The analysis was carried out in tissue slices and single cells of the intercaval area, which includes the crista terminalis (CT), the SAN, and the septum interatrialis (SI). Immunolabeling, in situ hybridization, qRT-PCR analysis, and electrophysiological recordings identified the SAN as a region characterized by high HCN4 signal and current levels, while the expression in the CT and in the SI was either negligible or absent. Detailed analysis of the central SAN area showed that cells are predominantly distributed in islets interconnected by cell prolongations, and single-cell HCN4 labeling suggested sites of channel clustering. Our data indicate that in the rabbit SAN, HCN4 proteins are major constituents of native f-channels, and their distribution matches closely the SAN as defined morphologically and electrophysiologically. Until recently, the SAN was identified as the region where Cx43 and atrial natriuretic peptide are not expressed; we propose here that expression of HCN4 is an appropriate tool to map and identify the cardiac SAN pacemaker region.


The Journal of Neuroscience | 2011

Recessive Loss-of-Function Mutation in the Pacemaker HCN2 Channel Causing Increased Neuronal Excitability in a Patient with Idiopathic Generalized Epilepsy

Jacopo C. DiFrancesco; Andrea Barbuti; Raffaella Milanesi; Stefania Coco; Annalisa Bucchi; Georgia Bottelli; Carlo Ferrarese; Silvana Franceschetti; Benedetta Terragni; Mirko Baruscotti; Dario DiFrancesco

The hyperpolarization-activated Ih current, coded for by hyperpolarization-activated, cyclic nucleotide-gated (HCN) channels, controls synaptic integration and intrinsic excitability in many brain areas. Because of their role in pacemaker function, defective HCN channels are natural candidates for contributing to epileptogenesis. Indeed, Ih is pathologically altered after experimentally induced seizures, and several independent data indicate a link between dysfunctional HCN channels and different forms of epilepsy. However, direct evidence for functional changes of defective HCN channels correlating with the disease in human patients is still elusive. By screening families with epilepsy for mutations in Hcn1 and Hcn2 genes, we found a recessive loss-of-function point mutation in the gene coding for the HCN2 channel in a patient with sporadic idiopathic generalized epilepsy. Of 17 screened members of the same family, the proband was the only one affected and homozygous for the mutation. The mutation (E515K) is located in the C-linker, a region known to affect channel gating. Functional analysis revealed that homomeric mutant, but not heteromeric wild-type/mutant channels, have a strongly inhibited function caused by a large negative shift of activation range and slowed activation kinetics, effectively abolishing the HCN2 contribution to activity. After transfection into acutely isolated newborn rat cortical neurons, homomeric mutant, but not heteromeric wild type/mutant channels, lowered the threshold of action potential firing and strongly increased cell excitability and firing frequency when compared with wild-type channels. This is the first evidence in humans for a single-point, homozygous loss-of-function mutation in HCN2 potentially associated with generalized epilepsy with recessive inheritance.


Annals of the New York Academy of Sciences | 2008

Control of Cardiac Rate by "Funny" Channels in Health and Disease

Andrea Barbuti; Dario DiFrancesco

Activation of the “funny” (pacemaker, If) current during the diastolic depolarization phase of an action potential is the main mechanism underlying spontaneous, rhythmic activity of cardiac pacemaker cells. In the past three decades, a wealth of evidence elucidating the function of the funny current in the generation and modulation of cardiac pacemaker activity has been gathered. The slope of early diastolic depolarization, and thus the heart rate, is controlled precisely by the degree of If activation during diastole. If is also accurately and rapidly modulated by changes of the cytosolic concentration of the second messenger cAMP, operated by the autonomous nervous system through β‐adrenergic, mainly β2, and in the opposite way by muscarinic receptor, stimulation. Recently, novel in vivo data, both in animal models and humans, have been collected that confirm the key role of If in pacemaking. In particular, an inheritable point mutation in the cyclic nucleotide‐binding domain of human HCN4, the main hyperpolarization‐activated cyclic nucleotide (HCN) isoform contributing to native funny channels of the sinoatrial node, was shown to be associated with sinus bradycardia in a large family. Because of their properties, funny channels have long been a major target of classical pharmacological research and are now target of innovative gene/cell‐based therapeutic approaches aimed to exploit their function in cardiac rate control.

Collaboration


Dive into the Andrea Barbuti's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Carlo Gaetano

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge