Imperial College London

ProfessorCesareTerracciano

Faculty of MedicineNational Heart & Lung Institute

Professor of Cardiac Electrophysiology
 
 
 
//

Contact

 

+44 (0)20 7594 2735c.terracciano Website CV

 
 
//

Location

 

430ICTEM buildingHammersmith Campus

//

Summary

 

Publications

Publication Type
Year
to

302 results found

Wang BX, Couch L, Kit-Anan W, Whittaker T, Nagelkerke A, Deidda G, Mitraki A, Harding SE, Stevens MM, MacLeod KT, Terracciano CMet al., 2018, Extracellular Vesicles From Human Cardiac Fibroblasts Increase Sarcoplasmic Reticulum-Dependency of Human Induced Pluripotent Stem Cell-Derived Cardiomyocyte Calcium Handling, Publisher: LIPPINCOTT WILLIAMS & WILKINS, ISSN: 0009-7322

Conference paper

Perbellini F, Watson SA, Bardi I, Terracciano CMet al., 2018, Heterocellularity and cellular cross-talk in the cardiovascular system, Frontiers in Cardiovascular Medicine, Vol: 5, ISSN: 2297-055X

Cellular specialization and interactions with other cell types are the essence of complex multicellular life. The orchestrated function of different cell populations in the heart, in combination with a complex network of intercellular circuits of communication, is essential to maintain a healthy heart and its disruption gives rise to pathological conditions. Over the past few years, the development of new biological research tools has facilitated more accurate identification of the cardiac cell populations and their specific roles. This review aims to provide an overview on the significance and contributions of the various cellular components: cardiomyocytes, fibroblasts, endothelial cells, vascular smooth muscle cells, pericytes, and inflammatory cells. It also aims to describe their role in cardiac development, physiology and pathology with a particular focus on the importance of heterocellularity and cellular interaction between these different cell types.

Journal article

Wang BX, Kit-Anan W, Terracciano CMN, 2018, Many cells make life work-multicellularity in stem cell-based cardiac disease modelling, International Journal of Molecular Sciences, Vol: 19, ISSN: 1422-0067

Cardiac disease causes 33% of deaths worldwide but our knowledge of disease progression is still very limited. In vitro models utilising and combining multiple, differentiated cell types have been used to recapitulate the range of myocardial microenvironments in an effort to delineate the mechanical, humoral, and electrical interactions that modulate the cardiac contractile function in health and the pathogenesis of human disease. However, due to limitations in isolating these cell types and changes in their structure and function in vitro, the field is now focused on the development and use of stem cell-derived cell types, most notably, human-induced pluripotent stem cell-derived CMs (hiPSC-CMs), in modelling the CM function in health and patient-specific diseases, allowing us to build on the findings from studies using animal and adult human CMs. It is becoming increasingly appreciated that communications between cardiomyocytes (CMs), the contractile cell of the heart, and the non-myocyte components of the heart not only regulate cardiac development and maintenance of health and adult CM functions, including the contractile state, but they also regulate remodelling in diseases, which may cause the chronic impairment of the contractile function of the myocardium, ultimately leading to heart failure. Within the myocardium, each CM is surrounded by an intricate network of cell types including endothelial cells, fibroblasts, vascular smooth muscle cells, sympathetic neurons, and resident macrophages, and the extracellular matrix (ECM), forming complex interactions, and models utilizing hiPSC-derived cell types offer a great opportunity to investigate these interactions further. In this review, we outline the historical and current state of disease modelling, focusing on the major milestones in the development of stem cell-derived cell types, and how this technology has contributed to our knowledge about the interactions between CMs and key non-myocyte components of th

Journal article

Armstrong J, Puetzer JL, Serio A, Guex AG, Kapnisi K, Breant A, Zong Y, Assal V, Skaalure S, King O, Murty T, Meinert C, Franklin AC, Bassindale PG, Nichols MK, Terracciano C, Hutmacher DW, Drinkwater BW, Klein TJ, Perriman AW, Stevens MMet al., 2018, Engineering anisotropic muscle tissue using acoustic cell patterning, Advanced Materials, Vol: 30, Pages: 1-7, ISSN: 0935-9648

Tissue engineering has offered unique opportunities for disease modeling and regenerative medicine; however, the success of these strategies is dependent on faithful reproduction of native cellular organization. Here, it is reported that ultrasound standing waves can be used to organize myoblast populations in material systems for the engineering of aligned muscle tissue constructs. Patterned muscle engineered using type I collagen hydrogels exhibits significant anisotropy in tensile strength, and under mechanical constraint, produced microscale alignment on a cell and fiber level. Moreover, acoustic patterning of myoblasts in gelatin methacryloyl hydrogels significantly enhances myofibrillogenesis and promotes the formation of muscle fibers containing aligned bundles of myotubes, with a width of 120–150 µm and a spacing of 180–220 µm. The ability to remotely pattern fibers of aligned myotubes without any material cues or complex fabrication procedures represents a significant advance in the field of muscle tissue engineering. In general, these results are the first instance of engineered cell fibers formed from the differentiation of acoustically patterned cells. It is anticipated that this versatile methodology can be applied to many complex tissue morphologies, with broader relevance for spatially organized cell cultures, organoid development, and bioelectronics.

Journal article

Masood E, Vesper I, Van Noorden R, Heard E, Bonacchi C, Terracciano C, Muscella S, Galsworthy Met al., 2018, SIX MONTHS UNTIL BREXIT: HOW SCIENTISTS ARE PREPARING FOR THE SPLIT, NATURE, Vol: 561, Pages: 452-454, ISSN: 0028-0836

Journal article

Amdursky N, Mazo M, Thomas MR, Humphrey E, Puetzer J, St-Pierre J, Skaalure S, Richardson RM, Terracciano C, Stevens MMet al., 2018, Elastic serum-albumin based hydrogels: mechanism of formation and application in cardiac tissue engineering, Journal of Materials Chemistry B, Vol: 6, Pages: 5604-5612, ISSN: 2050-750X

Hydrogels are promising materials for mimicking the extra-cellular environment. Here, we present a simple methodology for the formation of a free-standing viscoelastic hydrogel from the abundant and low cost protein serum albumin. We show that the mechanical properties of the hydrogel exhibit a complicated behaviour as a function of the weight fraction of the protein component. We further use X-ray scattering to shed light on the mechanism of gelation from the formation of a fibrillary network at low weight fractions to interconnected aggregates at higher fractions. Given the match between our hydrogel elasticity and that of the myocardium, we investigated its potential for supporting cardiac cells in vitro. Interestingly, the sehydrogels support the formation of several layers of myocytes and significantly promote the maintenance of a native-likegene expression profile compared to those cultured on glass. When confronted with a multicellular ventricular cell preparation, the hydrogels can support macroscopically contracting cardiac-like tissues with a distinct cell arrangement, and form mm-long vascular-like structures. We envisage that our simple approach for the formation of an elastic substrate from an abundant protein makes the hydrogel a compelling biomedical material candidate for a wide range of cell types.

Journal article

Wright PT, Sanchez-Alonso JL, Lucarelli C, Alvarez-Laviada A, Poulet CE, Bello SO, Faggian G, Terracciano CM, Gorelik Jet al., 2018, Partial mechanical unloading of the heart disrupts L-type calcium channel and beta-adrenoceptor signaling microdomains, Frontiers in Physiology, Vol: 9, ISSN: 1664-042X

Introduction: We investigated the effect of partial mechanical unloading (PMU) of the heart on the physiology of calcium and beta-adrenoceptor-cAMP (βAR-cAMP) microdomains. Previous studies have investigated PMU using a model of heterotopic-heart and lung transplantation (HTHAL). These studies have demonstrated that PMU disrupts the structure of cardiomyocytes and calcium handling. We sought to understand these processes by studying L-Type Calcium Channel (LTCC) activity and sub-type-specific βAR-cAMP signaling within cardiomyocyte membrane microdomains.Method: We utilized an 8-week model of HTHAL, whereby the hearts of syngeneic Lewis rats were transplanted into the abdomens of randomly assigned cage mates. A pronounced atrophy was observed in hearts after HTHAL. Cardiomyocytes were isolated via enzymatic perfusion. We utilized Förster Resonance Energy Transfer (FRET) based cAMP-biosensors and scanning ion conductance microscopy (SICM) based methodologies to study localization of LTCC and βAR-cAMP signaling.Results: β2AR-cAMP responses measured by FRET in the cardiomyocyte cytosol were reduced by PMU (loaded 28.51 ± 7.18% vs. unloaded 10.84 ± 3.27% N,n 4/10-13 mean ± SEM ∗p < 0.05). There was no effect of PMU on β2AR-cAMP signaling in RII_Protein Kinase A domains. β1AR-cAMP was unaffected by PMU in either microdomain. Consistent with this SICM/FRET analysis demonstrated that β2AR-cAMP was specifically reduced in t-tubules (TTs) after PMU (loaded TT 0.721 ± 0.106% vs. loaded crest 0.104 ± 0.062%, unloaded TT 0.112 ± 0.072% vs. unloaded crest 0.219 ± 0.084% N,n 5/6-9 mean ± SEM ∗∗p < 0.01, ∗∗∗p < 0.001 vs. loaded TT). By comparison β1AR-cAMP responses in either TT or sarcolemmal crests were unaffected by the PMU. LTCC occurrence and open probability (Po) were reduced by PMU (loaded TT Po 0.073 ± 0.011% vs. load

Journal article

Kane C, Terracciano CMN, 2018, Human cardiac fibroblasts engage the sarcoplasmic reticulum in induced pluripotent stem cell-derived cardiomyocyte excitation-contraction coupling, Journal of the American College of Cardiology, Vol: 72, Pages: 1061-1063, ISSN: 0735-1097

Journal article

Sanzari I, Humphrey EJ, Dinelli F, Terracciano CM, Prodromakis Tet al., 2018, Effect of patterned polyacrylamide hydrogel on morphology and orientation of cultured NRVMs, Scientific Reports, Vol: 8, ISSN: 2045-2322

We recently demonstrated that patterned Parylene C films could be effectively used as a mask for directly copolymerizing proteins on polyacrylamide hydrogel (PAm). In this work, we have proved the applicability of this technique for studying the effect such platforms render on neonatal rat ventricular myocytes (NRVMs). Firstly, we have characterised topographically and mechanically the scaffolds in liquid at the nano-scale level. We thus establish that such platforms have physical properties that closely mimics the in vivo extracellular environment of cells. We have then studied the cell morphology and physiology by comparing cultures on flat uniformly-covered and collagen-patterned scaffolds. We show that micro-patterns promote the elongation of cells along the principal axis of the ridges coated with collagen. In several cases, cells also tend to create bridges across the grooves. We have finally studied cell contraction, monitoring Ca2+ cycling at a certain stimulation. Cells seeded on patterned scaffolds present significant responses in comparison to the isotropic ones.

Journal article

Watson SA, Duff JD, Bardi IB, Zabielska M, Atanur SS, Jabbour RJ, Smolenski RT, Harding SE, Perbellini F, Terracciano CMet al., 2018, A novel platform to maintain adult cardiac tissue in vitro: myocardial slices and electromechanical stimulation with physiological preload, European-Society-of-Cardiology Congress, Publisher: OXFORD UNIV PRESS, Pages: 1090-1090, ISSN: 0195-668X

Conference paper

Wang B, Deidda G, Mitraki A, MacLeod KT, Terracciano CMet al., 2018, Self-assembling arginine-glycine-aspartic acid-containing peptides abbreviate human cardiomyocyte calcium transients and increase sarcoplasmic reticulum contribution to excitation-contraction coupling, European-Society-of-Cardiology Congress, Publisher: OXFORD UNIV PRESS, Pages: 1209-1209, ISSN: 0195-668X

Conference paper

Jabbour R, Kapnisi K, Mawad D, Handa B, Couch L, Mansfield C, Perbillini F, Terracciano C, Stevens M, Lyon A, Smith G, Peters N, Ng FS, Harding Set al., 2018, Conductive polymers affect myocardial conduction velocity but are not pro-arrhythmic, European-Society-of-Cardiology Congress, Publisher: OXFORD UNIV PRESS, Pages: 1205-1205, ISSN: 0195-668X

Conference paper

Mustroph J, Wagemann O, Luecht CM, Trum M, Hammer KP, Sag CM, Lebek S, Tarnowski D, Reinders J, Perbellini F, Terracciano C, Schmid C, Schopka S, Hilker M, Zausig Y, Pabel S, Sossalla ST, Schweda F, Maier LS, Wagner Set al., 2018, Empagliflozin reduces Ca/calmodulin-dependent kinase II activity in isolated ventricular cardiomyocytes, ESC Heart Failure, Vol: 5, Pages: 642-648, ISSN: 2055-5822

AimsThe EMPA‐REG OUTCOME study showed reduced mortality and hospitalization due to heart failure (HF) in diabetic patients treated with empagliflozin. Overexpression and Ca2+‐dependent activation of Ca2+/calmodulin‐dependent kinase II (CaMKII) are hallmarks of HF, leading to contractile dysfunction and arrhythmias. We tested whether empagliflozin reduces CaMKII‐ activity and improves Ca2+‐handling in human and murine ventricular myocytes.Methods and resultsMyocytes from wild‐type mice, mice with transverse aortic constriction (TAC) as a model of HF, and human failing ventricular myocytes were exposed to empagliflozin (1 μmol/L) or vehicle. CaMKII activity was assessed by CaMKII–histone deacetylase pulldown assay. Ca2+ spark frequency (CaSpF) as a measure of sarcoplasmic reticulum (SR) Ca2+ leak was investigated by confocal microscopy. [Na+]i was measured using Na+/Ca2+‐exchanger (NCX) currents (whole‐cell patch clamp). Compared with vehicle, 24 h empagliflozin exposure of murine myocytes reduced CaMKII activity (1.6 ± 0.7 vs. 4.2 ± 0.9, P < 0.05, n = 10 mice), and also CaMKII‐dependent ryanodine receptor phosphorylation (0.8 ± 0.1 vs. 1.0 ± 0.1, P < 0.05, n = 11 mice), with similar results upon TAC. In murine myocytes, empagliflozin reduced CaSpF (TAC: 1.7 ± 0.3 vs. 2.5 ± 0.4 1/100 μm−1 s−1, P < 0.05, n = 4 mice) but increased SR Ca2+ load and Ca2+ transient amplitude. Importantly, empagliflozin also significantly reduced CaSpF in human failing ventricular myocytes (1 ± 0.2 vs. 3.3 ± 0.9, P < 0.05, n = 4 patients), while Ca2+ transient amplitude was increased (F/F0: 0.53 ± 0.05 vs. 0.36 ± 0.02, P < 0.05, n = 3 patients). In contrast, 30 min exposure with empagliflozin did not affect CaMKII activity nor Ca2+‐handling but significantly reduced [Na+]i.ConclusionsWe show for the first time that empagliflozin reduces CaMKII activity and CaMKII‐dependent SR Ca2+ le

Journal article

Wright P, Lucarelli C, Sanchez-Alonso J, Bello S, Faggian G, Terracciano C, Gorelik Jet al., 2018, Mechanical Unloading Suppresses Localized Beta-2 Adrenoceptor and L-type Calcium Channel Function in Healthy and Failing Cardiomyocytes, Circulation 136(Suppl_1.18365):14 Nov 2017

Journal article

Zwi-Dantsis L, Stuckey D, Marijon C, Wang B, Terracciano C, Stevens Met al., 2018, ENGINEERING MAGNETIC, PATTERNED HUMAN HEART PATCHES FOR CARDIAC REGENERATION, Annual Conference of the British-Cardiovascular-Society on High Performing Teams, Publisher: BMJ PUBLISHING GROUP, Pages: A82-A82, ISSN: 1355-6037

Conference paper

Kapnisi M, Mansfield C, Marijon C, Guex AG, Perbellini F, Bardi I, Humphrey EJ, Puetzer J, Mawad D, Koutsogeorgis DC, Stuckey DJ, Terracciano CM, Harding SE, Stevens MMet al., 2018, Auxetic cardiac patches with tunable mechanical and conductive properties toward treating myocardial infarction, Advanced Functional Materials, Vol: 28, ISSN: 1616-301X

An auxetic conductive cardiac patch (AuxCP) for the treatment of myocardial infarction (MI) is introduced. The auxetic design gives the patch a negative Poisson's ratio, providing it with the ability to conform to the demanding mechanics of the heart. The conductivity allows the patch to interface with electroresponsive tissues such as the heart. Excimer laser microablation is used to micropattern a re‐entrant honeycomb (bow‐tie) design into a chitosan‐polyaniline composite. It is shown that the bow‐tie design can produce patches with a wide range in mechanical strength and anisotropy, which can be tuned to match native heart tissue. Further, the auxetic patches are conductive and cytocompatible with murine neonatal cardiomyocytes in vitro. Ex vivo studies demonstrate that the auxetic patches have no detrimental effect on the electrophysiology of both healthy and MI rat hearts and conform better to native heart movements than unpatterned patches of the same material. Finally, the AuxCP applied in a rat MI model results in no detrimental effect on cardiac function and negligible fibrotic response after two weeks in vivo. This approach represents a versatile and robust platform for cardiac biomaterial design and could therefore lead to a promising treatment for MI.

Journal article

Chowdhury RA, Tzortzis KN, Dupont E, Selvadurai S, Perbellini F, Cantwell C, Ng FS, Simon A, Terracciano C, Peters NSet al., 2018, Concurrent micro- to macro-cardiac electrophysiology in myocyte cultures and human heart slices, Scientific Reports, Vol: 8, ISSN: 2045-2322

The contact cardiac electrogram is derived from the extracellular manifestation of cellular action potentials and cell-to-cell communication. It is used to guide catheter based clinical procedures. Theoretically, the contact electrogram and the cellular action potential are directly related, and should change in conjunction with each other during arrhythmogenesis, however there is currently no methodology by which to concurrently record both electrograms and action potentials in the same preparation for direct validation of their relationships and their direct mechanistic links. We report a novel dual modality apparatus for concurrent electrogram and cellular action potential recording at a single cell level within multicellular preparations. We further demonstrate the capabilities of this system to validate the direct link between these two modalities of voltage recordings.

Journal article

Bello S, Singh C, Jayakumar S, Punjabi P, Terracciano Cet al., 2018, Role of Cardiomyocyte Regeneration in Mechanical Unloading Induced Reverse Cardiac Remodeling, Circulation 134(Suppl_1.20205):11 Nov 2016, ISSN: 0009-7322

Journal article

Cui C, Faraji N, Lauto A, Travaglini L, Tonkin J, Mahns D, Humphrey E, Terracciano C, Gooding JJ, Seidel J, Mawad Det al., 2018, A flexible polyaniline-based bioelectronic patch, BIOMATERIALS SCIENCE, Vol: 6, Pages: 493-500, ISSN: 2047-4830

Journal article

Ricardo CP, Hellen N, Foldes G, Kodagoda T, Terracciano CM, Duchen MR, Harding SEet al., 2018, Low levels of the A3243G MTDNA mutation in human induced pluripotent stem cellcardiomyocytes do not cause functional or metabolic disturbances but increase with further passaging, Autumn Meeting of the British-Society-for-Cardiovascular-Research on Cardiac Metabolic Disorders and Mitochondrial Dysfunction, Publisher: BMJ Publishing Group, Pages: A8-A9, ISSN: 1355-6037

Conference paper

Vicinanza C, Aquila I, Scalise M, Cristiano F, Marino F, Cianflone E, Mancuso T, Marotta P, Sacco W, Lewis FC, Couch L, Shone V, Gritti G, Torella A, Smith AJ, Terracciano CMN, Britti D, Veltri P, Indolfi C, Nadal-Ginard B, Ellison-Hughes GM, Torella Det al., 2017, Adult cardiac stem cells are multipotent and robustly myogenic: c-kit expression is necessary but not sufficient for their identification, Cell Death and Differentiation, Vol: 24, Pages: 2101-2116, ISSN: 1350-9047

Multipotent adult resident cardiac stem cells (CSCs) were first identified by the expression of c-kit, the stem cell factor receptor. However, in the adult myocardium c-kit alone cannot distinguish CSCs from other c-kit-expressing (c-kitpos) cells. The adult heart indeed contains a heterogeneous mixture of c-kitpos cells, mainly composed of mast and endothelial/progenitor cells. This heterogeneity of cardiac c-kitpos cells has generated confusion and controversy about the existence and role of CSCs in the adult heart. Here, to unravel CSC identity within the heterogeneous c-kit-expressing cardiac cell population, c-kitpos cardiac cells were separated through CD45-positive or -negative sorting followed by c-kitpos sorting. The blood/endothelial lineage-committed (Lineagepos) CD45posc-kitpos cardiac cells were compared to CD45neg(Lineageneg/Linneg) c-kitpos cardiac cells for stemness and myogenic properties in vitro and in vivo. The majority (~90%) of the resident c-kitpos cardiac cells are blood/endothelial lineage-committed CD45posCD31posc-kitpos cells. In contrast, the LinnegCD45negc-kitpos cardiac cell cohort, which represents 10% of the total c-kitpos cells, contain all the cardiac cells with the properties of adult multipotent CSCs. These characteristics are absent from the c-kitneg and the blood/endothelial lineage-committed c-kitpos cardiac cells. Single Linnegc-kitpos cell-derived clones, which represent only 1–2% of total c-kitpos myocardial cells, when stimulated with TGF-β/Wnt molecules, acquire full transcriptome and protein expression, sarcomere organisation, spontaneous contraction and electrophysiological properties of differentiated cardiomyocytes (CMs). Genetically tagged cloned progeny of one Linnegc-kitpos cell when injected into the infarcted myocardium, results in significant regeneration of new CMs, arterioles and capillaries, derived from the injected cells. The CSC’s myogenic regenerative capacity is dependent on commitment t

Journal article

Watson SA, Scigliano M, Bardi I, Ascione R, Terracciano CM, Perbellini Fet al., 2017, Preparation of viable adult ventricular myocardial slices from large and small mammals., Nature Protocols, Vol: 12, Pages: 2623-2639, ISSN: 1750-2799

This protocol describes the preparation of highly viable adult ventricular myocardial slices from the hearts of small and large mammals, including rodents, pigs, dogs and humans. Adult ventricular myocardial slices are 100- to 400-μm-thick slices of living myocardium that retain the native multicellularity, architecture and physiology of the heart. This protocol provides a list of the equipment and reagents required alongside a detailed description of the methodology for heart explantation, tissue preparation, slicing with a vibratome and handling of myocardial slices. Supplementary videos are included to visually demonstrate these steps. A number of critical steps are addressed that must be followed in order to prepare highly viable myocardial slices. These include identification of myocardial fiber direction and fiber alignment within the tissue block, careful temperature control, use of an excitation-contraction uncoupler, optimal vibratome settings and correct handling of myocardial slices. Many aspects of cardiac structure and function can be studied using myocardial slices in vitro. Typical results obtained with hearts from a small mammal (rat) and a large mammal (human) with heart failure are shown, demonstrating myocardial slice viability, maximum contractility, Ca2+ handling and structure. This protocol can be completed in ∼4 h.

Journal article

Watson SA, Scigliano M, Bardi I, Ascione R, Terracciano CMN, Perbellini Fet al., 2017, Preparation of viable adult ventricular myocardial slices from large and small mammals, Nature Protocols, Vol: 12, Pages: 2623-2639, ISSN: 1750-2799

This protocol describes a robust and reproducible method for the preparation of highly viable adult ventricular myocardial slices from small and large mammalian hearts. Adult ventricular myocardial slices are 100-400μm-thick slices of living myocardium that retain the native multicellularity, architecture and physiology of the heart. This protocol provides a list of the equipment and reagents required alongside a detailed description of methodology for heart explantation, tissue preparation, slicing with a vibratome and handling of myocardial slices. Supplementary videos are included to visually demonstrate these steps. A number of critical steps are addressed that must be followed in order to prepare highly viable myocardial slices. These include: identification of myocardial fiber direction and fiber alignment within tissue block, careful temperature control, use of an excitation-contraction uncoupler and optimal vibratome settings and correctly handling myocardial slices. Many aspects of cardiac structure and function can be studied using myocardial slices in vitro. Typical results obtained from rat (small mammal) & human heart failure (large mammal) samples are shown, demonstrating myocardial slice viability, maximum contractility, Ca2+ handling & structure. This protocol can be completed in ~4 hours.

Journal article

Wright PT, Lucarelli C, Sanchez-Alonso J, Bello S, Faggian G, Terracciano C, Gorelik Jet al., 2017, Mechanical Unloading Suppresses Localized Beta-2 Adrenoceptor and L-type Calcium Channel Function in Healthy and Failing Cardiomyocytes, Scientific Sessions of the American-Heart-Association / Resuscitation Science Symposium, Publisher: LIPPINCOTT WILLIAMS & WILKINS, ISSN: 0009-7322

Conference paper

Bello SO, Singh C, Punjabi P, Perbellini F, Terracciano Cet al., 2017, Coronary Reperfusion Ameliorates the Deleterious Effect of Mechanical Unloading on Infarct Size and Interstitial Fibrosis After Acute Myocardial Infarction, Scientific Sessions of the American-Heart-Association / Resuscitation Science Symposium, Publisher: LIPPINCOTT WILLIAMS & WILKINS, ISSN: 0009-7322

Conference paper

Bello S, Singh C, Punjabi P, Perbellini F, Terracciano Cet al., 2017, Coronary Reperfusion Ameliorates the Deleterious Effect of Mechanical Unloading on Infarct Size and Interstitial Fibrosis After Acute Myocardial Infarction, Circulation, Vol: 136

Journal article

Couch L, Clayton R, Wienecke L, Derda AA, Terracciano C, Thum T, Harding SEet al., 2017, Takotsubo Syndrome Associated miR-16 and miR-26a Reduce Contractility of Apical, but Not Basal, Cardiomyocytes in vitro, Scientific Sessions of the American-Heart-Association / Resuscitation Science Symposium, Publisher: LIPPINCOTT WILLIAMS & WILKINS, ISSN: 0009-7322

Conference paper

Perbellini F, Liu AK, Watson SA, Bardi I, Terracciano CMet al., 2017, Free-of-acrylamide SDS-based Tissue Clearing (FASTClear) for Three Dimensional Imaging of Collagen Content and Vascular Organization in Adult Ventricular Tissue, Scientific Sessions of the American-Heart-Association / Resuscitation Science Symposium, Publisher: LIPPINCOTT WILLIAMS & WILKINS, ISSN: 0009-7322

Conference paper

Lucarelli C, Wright P, Diakonov I, Bello S, Faggian G, Terracciano C, Gorelik Jet al., 2017, Partial Mechanical Unloading Distorts Cardiomyocyte Membrane Organization and Regional ß2AR-cAMP Compartmentation, Scientific Sessions of the American-Heart-Association / Resuscitation Science Symposium, Publisher: LIPPINCOTT WILLIAMS & WILKINS, ISSN: 0009-7322

Conference paper

Wang BX, Couch L, MacLeod KT, Harding SE, Terracciano CMet al., 2017, Extracellular Vesicles Secreted From Human Fibroblasts Modulate Human Induced Pluripotent Stem Cell- Cardiomyocyte Calcium Cycling, Scientific Sessions of the American-Heart-Association / Resuscitation Science Symposium, Publisher: LIPPINCOTT WILLIAMS & WILKINS, ISSN: 0009-7322

Conference paper

This data is extracted from the Web of Science and reproduced under a licence from Thomson Reuters. You may not copy or re-distribute this data in whole or in part without the written consent of the Science business of Thomson Reuters.

Request URL: http://wlsprd.imperial.ac.uk:80/respub/WEB-INF/jsp/search-html.jsp Request URI: /respub/WEB-INF/jsp/search-html.jsp Query String: limit=30&id=00155523&person=true&page=3&respub-action=search.html