Network


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

Hotspot


Dive into the research topics where Sergio Signore is active.

Publication


Featured researches published by Sergio Signore.


American Journal of Pathology | 2011

Effects of Age and Heart Failure on Human Cardiac Stem Cell Function

Daniela Cesselli; Antonio Paolo Beltrami; Federica D'Aurizio; Patrizia Marcon; Natascha Bergamin; Barbara Toffoletto; Maura Pandolfi; Elisa Puppato; Laura Marino; Sergio Signore; Ugolino Livi; Roberto Verardo; Silvano Piazza; Luigi Marchionni; Claudia Fiorini; Claudio Schneider; Toru Hosoda; Marcello Rota; Jan Kajstura; Piero Anversa; Carlo Alberto Beltrami; Annarosa Leri

Currently, it is unknown whether defects in stem cell growth and differentiation contribute to myocardial aging and chronic heart failure (CHF), and whether a compartment of functional human cardiac stem cells (hCSCs) persists in the decompensated heart. To determine whether aging and CHF are critical determinants of the loss in growth reserve of the heart, the properties of hCSCs were evaluated in 18 control and 23 explanted hearts. Age and CHF showed a progressive decrease in functionally competent hCSCs. Chronological age was a major predictor of five biomarkers of hCSC senescence: telomeric shortening, attenuated telomerase activity, telomere dysfunction-induced foci, and p21(Cip1) and p16(INK4a) expression. CHF had similar consequences for hCSCs, suggesting that defects in the balance between cardiomyocyte mass and the pool of nonsenescent hCSCs may condition the evolution of the decompensated myopathy. A correlation was found previously between telomere length in circulating bone marrow cells and cardiovascular diseases, but that analysis was restricted to average telomere length in a cell population, neglecting the fact that telomere attrition does not occur uniformly in all cells. The present study provides the first demonstration that dysfunctional telomeres in hCSCs are biomarkers of aging and heart failure. The biomarkers of cellular senescence identified here can be used to define the birth date of hCSCs and to sort young cells with potential therapeutic efficacy.


Circulation | 2012

Cardiomyogenesis in the Aging and Failing Human Heart

Jan Kajstura; Marcello Rota; Donato Cappetta; Barbara Ogorek; Christian Arranto; Yingnan Bai; João Ferreira-Martins; Sergio Signore; Fumihiro Sanada; Alex Matsuda; James Kostyla; Maria Virginia Caballero; Claudia Fiorini; David A. D'Alessandro; Robert E. Michler; Federica del Monte; Toru Hosoda; Mark A. Perrella; Annarosa Leri; Bruce A. Buchholz; Joseph Loscalzo; Piero Anversa

Background— Two opposite views of cardiac growth are currently held; one views the heart as a static organ characterized by a large number of cardiomyocytes that are present at birth and live as long as the organism, and the other views the heart a highly plastic organ in which the myocyte compartment is restored several times during the course of life. Methods and Results— The average age of cardiomyocytes, vascular endothelial cells (ECs), and fibroblasts and their turnover rates were measured by retrospective 14C birth dating of cells in 19 normal hearts 2 to 78 years of age and in 17 explanted failing hearts 22 to 70 years of age. We report that the human heart is characterized by a significant turnover of ventricular myocytes, ECs, and fibroblasts, physiologically and pathologically. Myocyte, EC, and fibroblast renewal is very high shortly after birth, decreases during postnatal maturation, remains relatively constant in the adult organ, and increases dramatically with age. From 20 to 78 years of age, the adult human heart entirely replaces its myocyte, EC, and fibroblast compartment ≈8, ≈6, and ≈8 times, respectively. Myocyte, EC, and fibroblast regeneration is further enhanced with chronic heart failure. Conclusions— The human heart is a highly dynamic organ that retains a remarkable degree of plasticity throughout life and in the presence of chronic heart failure. However, the ability to regenerate cardiomyocytes, vascular ECs, and fibroblasts cannot prevent the manifestations of myocardial aging or oppose the negative effects of ischemic and idiopathic dilated cardiomyopathy.


Circulation Research | 2012

Cardiomyogenesis in the Developing Heart Is Regulated by C-Kit–Positive Cardiac Stem Cells

João Ferreira-Martins; Barbara Ogorek; Donato Cappetta; Alex Matsuda; Sergio Signore; Domenico D'Amario; James Kostyla; Elisabeth Steadman; Noriko Ide-Iwata; Fumihiro Sanada; Grazia Iaffaldano; Sergio Ottolenghi; Toru Hosoda; Annarosa Leri; Jan Kajstura; Piero Anversa; Marcello Rota

Rationale: Embryonic and fetal myocardial growth is characterized by a dramatic increase in myocyte number, but whether the expansion of the myocyte compartment is dictated by activation and commitment of resident cardiac stem cells (CSCs), division of immature myocytes or both is currently unknown. Objective: In this study, we tested whether prenatal cardiac development is controlled by activation and differentiation of CSCs and whether division of c-kit–positive CSCs in the mouse heart is triggered by spontaneous Ca2+ oscillations. Methods and Results: We report that embryonic-fetal c-kit–positive CSCs are self-renewing, clonogenic and multipotent in vitro and in vivo. The growth and commitment of c-kit–positive CSCs is responsible for the generation of the myocyte progeny of the developing heart. The close correspondence between values computed by mathematical modeling and direct measurements of myocyte number at E9, E14, E19 and 1 day after birth strongly suggests that the organogenesis of the embryonic heart is dependent on a hierarchical model of cell differentiation regulated by resident CSCs. The growth promoting effects of c-kit–positive CSCs are triggered by spontaneous oscillations in intracellular Ca2+, mediated by IP3 receptor activation, which condition asymmetrical stem cell division and myocyte lineage specification. Conclusions: Myocyte formation derived from CSC differentiation is the major determinant of cardiac growth during development. Division of c-kit–positive CSCs in the mouse is promoted by spontaneous Ca2+ spikes, which dictate the pattern of stem cell replication and the generation of a myocyte progeny at all phases of prenatal life and up to one day after birth.


Circulation Research | 2014

c-kit-Positive Cardiac Stem Cells Nested in Hypoxic Niches are Activated by Stem Cell Factor Reversing the Aging Myopathy

Fumihiro Sanada; Junghyun Kim; Noel Yan-Ki Chan; Sergio Signore; Barbara Ogorek; Kazuya Isobe; Ewa Wybieralska; Giulia Borghetti; Ada Pesapane; Andrea Sorrentino; Emily Mangano; Donato Cappetta; Chiara Mangiaracina; Mario Ricciardi; Maria Cimini; Emeka Ifedigbo; Mark A. Perrella; Polina Goichberg; Augustine M. K. Choi; Jan Kajstura; Toru Hosoda; Marcello Rota; Piero Anversa; Annarosa Leri

Rationale: Hypoxia favors stem cell quiescence, whereas normoxia is required for stem cell activation, but whether cardiac stem cell (CSC) function is regulated by the hypoxic/normoxic state of the cell is currently unknown. Objective: A balance between hypoxic and normoxic CSCs may be present in the young heart, although this homeostatic control may be disrupted with aging. Defects in tissue oxygenation occur in the old myocardium, and this phenomenon may expand the pool of hypoxic CSCs, which are no longer involved in myocyte renewal. Methods and Results: Here, we show that the senescent heart is characterized by an increased number of quiescent CSCs with intact telomeres that cannot re-enter the cell cycle and form a differentiated progeny. Conversely, myocyte replacement is controlled only by frequently dividing CSCs with shortened telomeres; these CSCs generate a myocyte population that is chronologically young but phenotypically old. Telomere dysfunction dictates their actual age and mechanical behavior. However, the residual subset of quiescent young CSCs can be stimulated in situ by stem cell factor reversing the aging myopathy. Conclusions: Our findings support the notion that strategies targeting CSC activation and growth interfere with the manifestations of myocardial aging in an animal model. Although caution has to be exercised in the translation of animal studies to human beings, our data strongly suggest that a pool of functionally competent CSCs persists in the senescent heart and that this stem cell compartment can promote myocyte regeneration effectively, partly correcting the aging myopathy.


Circulation Research | 2011

The Ephrin A1–EphA2 System Promotes Cardiac Stem Cell Migration After Infarction

Polina Goichberg; Yingnan Bai; Domenico D'Amario; João Ferreira-Martins; Claudia Fiorini; Hanqiao Zheng; Sergio Signore; Federica del Monte; Sergio Ottolenghi; David A. D'Alessandro; Robert E. Michler; Toru Hosoda; Piero Anversa; Jan Kajstura; Marcello Rota; Annarosa Leri

Rationale: Understanding the mechanisms that regulate trafficking of human cardiac stem cells (hCSCs) may lead to development of new therapeutic approaches for the failing heart. Objective: We tested whether the motility of hCSCs in immunosuppressed infarcted animals is controlled by the guidance system that involves the interaction of Eph receptors with ephrin ligands. Methods and Results: Within the cardiac niches, cardiomyocytes expressed preferentially the ephrin A1 ligand, whereas hCSCs possessed the EphA2 receptor. Treatment of hCSCs with ephrin A1 resulted in the rapid internalization of the ephrin A1–EphA2 complex, posttranslational modifications of Src kinases, and morphological changes consistent with the acquisition of a motile cell phenotype. Ephrin A1 enhanced the motility of hCSCs in vitro, and their migration in vivo following acute myocardial infarction. At 2 weeks after infarction, the volume of the regenerated myocardium was 2-fold larger in animals injected with ephrin A1–activated hCSCs than in animals receiving control hCSCs; this difference was dictated by a greater number of newly formed cardiomyocytes and coronary vessels. The increased recovery in myocardial mass with ephrin A1–treated hCSCs was characterized by further restoration of cardiac function and by a reduction in arrhythmic events. Conclusions: Ephrin A1 promotes the motility of EphA2-positive hCSCs, facilitates their migration to the area of damage, and enhances cardiac repair. Thus, in situ stimulation of resident hCSCs with ephrin A1 or their ex vivo activation before myocardial delivery improves cell targeting to sites of injury, possibly providing a novel strategy for the management of the diseased heart.


Circulation | 2013

Inositol 1, 4, 5-Trisphosphate Receptors and Human Left Ventricular Myocytes

Sergio Signore; Andrea Sorrentino; João Ferreira-Martins; Ramaswamy Kannappan; Mehrdad Shafaie; Fabio Del Ben; Kazuya Isobe; Christian Arranto; Ewa Wybieralska; Andrew Webster; Fumihiro Sanada; Barbara Ogorek; Hanqiao Zheng; Xiaoxia Liu; Federica del Monte; David A. D’Alessandro; Oriyanhan Wunimenghe; Robert E. Michler; Toru Hosoda; Polina Goichberg; Annarosa Leri; Jan Kajstura; Piero Anversa; Marcello Rota

Background— Little is known about the function of inositol 1,4,5-trisphosphate receptors (IP3Rs) in the adult heart experimentally. Moreover, whether these Ca2+ release channels are present and play a critical role in human cardiomyocytes remains to be defined. IP3Rs may be activated after G&agr;q-protein–coupled receptor stimulation, affecting Ca2+ cycling, enhancing myocyte performance, and potentially favoring an increase in the incidence of arrhythmias. Methods and Results— IP3R function was determined in human left ventricular myocytes, and this analysis was integrated with assays in mouse myocytes to identify the mechanisms by which IP3Rs influence the electric and mechanical properties of the myocardium. We report that IP3Rs are expressed and operative in human left ventricular myocytes. After G&agr;q-protein–coupled receptor activation, Ca2+ mobilized from the sarcoplasmic reticulum via IP3Rs contributes to the decrease in resting membrane potential, prolongation of the action potential, and occurrence of early afterdepolarizations. Ca2+ transient amplitude and cell shortening are enhanced, and extrasystolic and dysregulated Ca2+ elevations and contractions become apparent. These alterations in the electromechanical behavior of human cardiomyocytes are coupled with increased isometric twitch of the myocardium and arrhythmic events, suggesting that G&agr;q-protein–coupled receptor activation provides inotropic reserve, which is hampered by electric instability and contractile abnormalities. Additionally, our findings support the notion that increases in Ca2+ load by IP3Rs promote Ca2+ extrusion by forward-mode Na+/Ca2+ exchange, an important mechanism of arrhythmic events. Conclusions— The G&agr;q-protein/coupled receptor/IP3R axis modulates the electromechanical properties of the human myocardium and its propensity to develop arrhythmias.


Circulation | 2013

Age-Associated Defects in EphA2 Signaling Impair the Migration of Human Cardiac Progenitor Cells

Polina Goichberg; Ramaswamy Kannappan; Maria Cimini; Yingnan Bai; Fumihiro Sanada; Andrea Sorrentino; Sergio Signore; Jan Kajstura; Marcello Rota; Piero Anversa; Annarosa Leri

Background— Aging negatively impacts on the function of resident human cardiac progenitor cells (hCPCs). Effective regeneration of the injured heart requires mobilization of hCPCs to the sites of damage. In the young heart, signaling by the guidance receptor EphA2 in response to the ephrin A1 ligand promotes hCPC motility and improves cardiac recovery after infarction. Methods and Results— We report that old hCPCs are characterized by cell-autonomous inhibition of their migratory ability ex vivo and impaired translocation in vivo in the damaged heart. EphA2 expression was not decreased in old hCPCs; however, the elevated level of reactive oxygen species in aged cells induced post-translational modifications of the EphA2 protein. EphA2 oxidation interfered with ephrin A1-stimulated receptor auto-phosphorylation, activation of Src family kinases, and caveolin-1–mediated internalization of the receptor. Cellular aging altered the EphA2 endocytic route, affecting the maturation of EphA2-containing endosomes and causing premature signal termination. Overexpression of functionally intact EphA2 in old hCPCs corrected the defects in endocytosis and downstream signaling, enhancing cell motility. Based on the ability of phenotypically young hCPCs to respond efficiently to ephrin A1, we developed a novel methodology for the prospective isolation of live hCPCs with preserved migratory capacity and growth reserve. Conclusions— Our data demonstrate that the ephrin A1/EphA2 pathway may serve as a target to facilitate trafficking of hCPCs in the senescent myocardium. Importantly, EphA2 receptor function can be implemented for the selection of hCPCs with high therapeutic potential, a clinically relevant strategy that does not require genetic manipulation of stem cells.


Nature Communications | 2015

Late Na + current and protracted electrical recovery are critical determinants of the aging myopathy

Sergio Signore; Andrea Sorrentino; Giulia Borghetti; Antonio Cannata; Marianna Meo; Yu Zhou; Ramaswamy Kannappan; Francesco S. Pasqualini; Heather A. O'Malley; Mark Sundman; Nikolaos Tsigkas; Eric Zhang; Christian Arranto; Chiara Mangiaracina; Kazuya Isobe; Brena Sena; Junghyun Kim; Polina Goichberg; Matthias Nahrendorf; Lori L. Isom; Annarosa Leri; Piero Anversa; Marcello Rota

The aging myopathy manifests itself with diastolic dysfunction and preserved ejection fraction. We raised the possibility that, in a mouse model of physiological aging, defects in electromechanical properties of cardiomyocytes are important determinants of the diastolic characteristics of the myocardium, independently from changes in structural composition of the muscle and collagen framework. Here we show that an increase in the late Na+ current (INaL) in aging cardiomyocytes prolongs the action potential (AP) and influences temporal kinetics of Ca2+ cycling and contractility. These alterations increase force development and passive tension. Inhibition of INaL shortens the AP and corrects dynamics of Ca2+ transient, cell contraction and relaxation. Similarly, repolarization and diastolic tension of the senescent myocardium are partly restored. Thus, INaL offers inotropic support, but negatively interferes with cellular and ventricular compliance, providing a new perspective of the biology of myocardial aging and the aetiology of the defective cardiac performance in the elderly.


American Journal of Physiology-heart and Circulatory Physiology | 2017

Hyperglycemia induces defective Ca2+ homeostasis in cardiomyocytes

Andrea Sorrentino; Giulia Borghetti; Yu Zhou; Antonio Cannata; Marianna Meo; Sergio Signore; Piero Anversa; Annarosa Leri; Polina Goichberg; Khaled Qanud; Jason T. Jacobson; Thomas H. Hintze; Marcello Rota

Diabetes and other metabolic conditions characterized by elevated blood glucose constitute important risk factors for cardiovascular disease. Hyperglycemia targets myocardial cells rendering ineffective mechanical properties of the heart, but cellular alterations dictating the progressive deterioration of cardiac function with metabolic disorders remain to be clarified. In the current study, we examined the effects of hyperglycemia on cardiac function and myocyte physiology by employing mice with high blood glucose induced by administration of streptozotocin, a compound toxic to insulin-producing β-cells. We found that hyperglycemia initially delayed the electrical recovery of the heart, whereas cardiac function became defective only after ~2 mo with this condition and gradually worsened with time. Prolonged hyperglycemia was associated with increased chamber dilation, thinning of the left ventricle (LV), and myocyte loss. Cardiomyocytes from hyperglycemic mice exhibited defective Ca2+ transients before the appearance of LV systolic defects. Alterations in Ca2+ transients involved enhanced spontaneous Ca2+ releases from the sarcoplasmic reticulum (SR), reduced cytoplasmic Ca2+ clearance, and declined SR Ca2+ load. These defects have important consequences on myocyte contraction, relaxation, and mechanisms of rate adaptation. Collectively, our data indicate that hyperglycemia alters intracellular Ca2+ homeostasis in cardiomyocytes, hindering contractile activity and contributing to the manifestation of the diabetic cardiomyopathy. NEW & NOTEWORTHY We have investigated the effects of hyperglycemia on cardiomyocyte physiology and ventricular function. Our results indicate that defective Ca2+ handling is a critical component of the progressive deterioration of cardiac performance of the diabetic heart.


Journal of the American Heart Association | 2016

Reduction in Kv Current Enhances the Temporal Dispersion of the Action Potential in Diabetic Myocytes: Insights From a Novel Repolarization Algorithm

Marianna Meo; Olivier Meste; Sergio Signore; Andrea Sorrentino; Antonio Cannata; Yu Zhou; Alex Matsuda; Marco Luciani; Ramaswamy Kannappan; Polina Goichberg; Annarosa Leri; Piero Anversa; Marcello Rota

Background Diabetes is associated with prolongation of the QT interval of the electrocardiogram and enhanced dispersion of ventricular repolarization, factors that, together with atherosclerosis and myocardial ischemia, may promote the occurrence of electrical disorders. Thus, we tested the possibility that alterations in transmembrane ionic currents reduce the repolarization reserve of myocytes, leading to action potential (AP) prolongation and enhanced beat‐to‐beat variability of repolarization. Methods and Results Diabetes was induced in mice with streptozotocin (STZ), and effects of hyperglycemia on electrical properties of whole heart and myocytes were studied with respect to an untreated control group (Ctrl) using electrocardiographic recordings in vivo, ex vivo perfused hearts, and single‐cell patch‐clamp analysis. Additionally, a newly developed algorithm was introduced to obtain detailed information of the impact of high glucose on AP profile. Compared to Ctrl, hyperglycemia in STZ‐treated animals was coupled with prolongation of the QT interval, enhanced temporal dispersion of electrical recovery, and susceptibility to ventricular arrhythmias, defects observed, in part, in the Akita mutant mouse model of type I diabetes. AP was prolonged and beat‐to‐beat variability of repolarization was enhanced in diabetic myocytes, with respect to Ctrl cells. Density of Kv K+ and L‐type Ca2+ currents were decreased in STZ myocytes, in comparison to cells from normoglycemic mice. Pharmacological reduction of Kv currents in Ctrl cells lengthened AP duration and increased temporal dispersion of repolarization, reiterating features identified in diabetic myocytes. Conclusions Reductions in the repolarizing K+ currents may contribute to electrical disturbances of the diabetic heart.

Collaboration


Dive into the Sergio Signore's collaboration.

Top Co-Authors

Avatar

Marcello Rota

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Annarosa Leri

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Piero Anversa

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Jan Kajstura

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Polina Goichberg

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Andrea Sorrentino

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ramaswamy Kannappan

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Barbara Ogorek

Brigham and Women's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge