131 results on '"Gabriele G. Schiattarella"'
Search Results
2. Genetics-diet crossroads: Unveiling new insights into hypertrophic cardiomyopathy
- Author
-
Steffen P. Häseli and Gabriele G. Schiattarella
- Subjects
Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Published
- 2023
- Full Text
- View/download PDF
3. High‐Resolution Transthoracic Echocardiography Accurately Detects Pulmonary Arterial Pressure and Decreased Right Ventricular Contractility in a Mouse Model of Pulmonary Fibrosis and Secondary Pulmonary Hypertension
- Author
-
Thomas S. Hansen, Kristen J. Bubb, Gabriele G. Schiattarella, Martin Ugander, Timothy C. Tan, and Gemma A. Figtree
- Subjects
bleomycin ,echocardiography ,mouse model ,pulmonary hypertension ,right ventricle ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
Background To date, assessment of right ventricular (RV) function in mice has relied extensively on invasive measurements. Echocardiographic advances have allowed adaptation of measures used in humans for serial, noninvasive RV functional assessment in mice. We evaluated the diagnostic performance of tricuspid annular plane systolic excursion (TAPSE), RV peak systolic myocardial velocity (s'), RV myocardial performance index (MPI), and RV fractional area change (FAC) in a mouse model of pulmonary hypertension. Methods and Results Echocardiography was performed on mice at baseline and 3 weeks after induction of pulmonary hypertension using inhaled bleomycin or saline, including adapted measures of TAPSE, s', MPI, and FAC. RV systolic pressure was measured by invasive catheterization, and RV contractility was measured as the peak slope of the RV systolic pressure recording (maximum change pressure/change time). Postmortem morphological assessment of RV hypertrophy was performed. RV systolic pressure was elevated and maximum change pressure/change time was reduced in bleomycin versus control (n=8; P=0.002). Compared with controls, bleomycin mice had reduced TAPSE (0.79±0.05 versus 1.06±0.04 mm; P=0.003), s' (21.3±1.2 versus 29.2±1.3 mm/s; P
- Published
- 2022
- Full Text
- View/download PDF
4. Xbp1s-FoxO1 axis governs lipid accumulation and contractile performance in heart failure with preserved ejection fraction
- Author
-
Gabriele G. Schiattarella, Francisco Altamirano, Soo Young Kim, Dan Tong, Anwarul Ferdous, Hande Piristine, Subhajit Dasgupta, Xuliang Wang, Kristin M. French, Elisa Villalobos, Stephen B. Spurgin, Maayan Waldman, Nan Jiang, Herman I. May, Theodore M. Hill, Yuxuan Luo, Heesoo Yoo, Vlad G. Zaha, Sergio Lavandero, Thomas G. Gillette, and Joseph A. Hill
- Subjects
Science - Abstract
Heart failure with preserved ejection fraction (HFpEF) is a global, major health issue for which no effective therapies are available. Here, the authors discover that the interplay between two transcription factors, Xbp1s and FoxO1, is critical for metabolic adaptation and lipid handling in HFpEF-stressed cardiomyocytes.
- Published
- 2021
- Full Text
- View/download PDF
5. FoxO1–Dio2 signaling axis governs cardiomyocyte thyroid hormone metabolism and hypertrophic growth
- Author
-
Anwarul Ferdous, Zhao V. Wang, Yuxuan Luo, Dan L. Li, Xiang Luo, Gabriele G. Schiattarella, Francisco Altamirano, Herman I. May, Pavan K. Battiprolu, Annie Nguyen, Beverly A. Rothermel, Sergio Lavandero, Thomas G. Gillette, and Joseph A. Hill
- Subjects
Science - Abstract
Disease stress-induced cardiac hypertrophy is a major mechanism of pathological cardiac remodeling. Here, the authors unveil a previously unrecognized role of a FoxO1–Dio2 signaling axis in maladaptive, afterload-induced cardiac hypertrophy and intracellular thyroid hormone homeostasis.
- Published
- 2020
- Full Text
- View/download PDF
6. Loss of Akap1 Exacerbates Pressure Overload-Induced Cardiac Hypertrophy and Heart Failure
- Author
-
Gabriele G. Schiattarella, Nicola Boccella, Roberta Paolillo, Fabio Cattaneo, Valentina Trimarco, Anna Franzone, Stefania D’Apice, Giuseppe Giugliano, Laura Rinaldi, Domenica Borzacchiello, Alessandra Gentile, Assunta Lombardi, Antonio Feliciello, Giovanni Esposito, and Cinzia Perrino
- Subjects
heart failure ,Akt ,cardiac hypertrophy ,cardiomyocytes ,pressure overload ,Physiology ,QP1-981 - Abstract
Left ventricular hypertrophy (LVH) is a major contributor to the development of heart failure (HF). Alterations in cyclic adenosine monophosphate (cAMP)-dependent signaling pathways participate in cardiomyocyte hypertrophy and mitochondrial dysfunction occurring in LVH and HF. cAMP signals are received and integrated by a family of cAMP-dependent protein kinase A (PKA) anchor proteins (AKAPs), tethering PKA to discrete cellular locations. AKAPs encoded by the Akap1 gene (mitoAKAPs) promote PKA mitochondrial targeting, regulating mitochondrial structure and function, reactive oxygen species production, and cell survival. To determine the role of mitoAKAPs in LVH development, in the present investigation, mice with global genetic deletion of Akap1 (Akap1-/-), Akap1 heterozygous (Akap1+/-), and their wild-type (wt) littermates underwent transverse aortic constriction (TAC) or SHAM procedure for 1 week. In wt mice, pressure overload induced the downregulation of AKAP121, the major cardiac mitoAKAP. Compared to wt, Akap1-/- mice did not display basal alterations in cardiac structure or function and cardiomyocyte size or fibrosis. However, loss of Akap1 exacerbated LVH and cardiomyocyte hypertrophy induced by pressure overload and accelerated the progression toward HF in TAC mice, and these changes were not observed upon prevention of AKAP121 degradation in seven in absentia homolog 2 (Siah2) knockout mice (Siah2-/-). Loss of Akap1 was also associated to a significant increase in cardiac apoptosis as well as lack of activation of Akt signaling after pressure overload. Taken together, these results demonstrate that in vivo genetic deletion of Akap1 enhances LVH development and accelerates pressure overload-induced cardiac dysfunction, pointing at Akap1 as a novel repressor of pathological LVH. These results confirm and extend the important role of mitoAKAPs in cardiac response to stress.
- Published
- 2018
- Full Text
- View/download PDF
7. Molecular mechanisms underlying the beneficial effects of exercise and dietary interventions in the prevention of cardiometabolic diseases
- Author
-
Maurizio Forte, Daniele Rodolico, Pietro Ameri, Daniele Catalucci, Cristina Chimenti, Lia Crotti, Leonardo Schirone, Annachiara Pingitore, Daniele Torella, Giuliano Iacovone, Valentina Valenti, Gabriele G. Schiattarella, Cinzia Perrino, and Sebastiano Sciarretta
- Subjects
cardiometabolic disease ,mitochondrial dysfunction ,oxidative stress ,dietary restriction ,exercise ,General Medicine ,Cardiology and Cardiovascular Medicine - Published
- 2022
8. Cardiac metabolism in HFpEF: from fuel to signalling
- Author
-
Federico Capone, Cristian Sotomayor-Flores, David Bode, Rongling Wang, Daniele Rodolico, Stefano Strocchi, Gabriele G Schiattarella, Capone, Federico, Sotomayor-Flores, Cristian, Bode, David, Wang, Rongling, Rodolico, Daniele, Strocchi, Stefano, and Schiattarella, Gabriele G
- Subjects
Metabolism ,Physiology ,Physiology (medical) ,Heart failure ,HFpEF ,Cardiology and Cardiovascular Medicine ,Post-translational modifications - Abstract
Heart failure (HF) is marked by distinctive changes in myocardial uptake and utilization of energy substrates. Among the different types of HF, HF with preserved ejection fraction (HFpEF) is a highly prevalent, complex, and heterogeneous condition for which metabolic derangements seem to dictate disease progression. Changes in intermediate metabolism in cardiometabolic HFpEF—among the most prevalent forms of HFpEF—have a large impact both on energy provision and on a number of signalling pathways in the heart. This dual, metabolic vs. signalling, role is played in particular by long-chain fatty acids (LCFAs) and short-chain carbon sources [namely, short-chain fatty acids (SCFAs) and ketone bodies (KBs)]. LCFAs are key fuels for the heart, but their excess can be harmful, as in the case of toxic accumulation of lipid by-products (i.e. lipotoxicity). SCFAs and KBs have been proposed as a potential major, alternative source of energy in HFpEF. At the same time, both LCFAs and short-chain carbon sources are substrate for protein post-translational modifications and other forms of direct and indirect signalling of pivotal importance in HFpEF pathogenesis. An in-depth molecular understanding of the biological functions of energy substrates and their signalling role will be instrumental in the development of novel therapeutic approaches to HFpEF. Here, we summarize the current evidence on changes in energy metabolism in HFpEF, discuss the signalling role of intermediate metabolites through, at least in part, their fate as substrates for post-translational modifications, and highlight clinical and translational challenges around metabolic therapy in HFpEF.
- Published
- 2022
9. Cardiometabolic HFpEF: NASH of the Heart
- Author
-
Federico Capone, Roberto Vettor, and Gabriele G. Schiattarella
- Subjects
Physiology (medical) ,Cardiology and Cardiovascular Medicine - Published
- 2023
10. The Lure of Cardiac Metabolism in the Diagnosis, Prevention, and Treatment of Heart Failure
- Author
-
Daniele Rodolico, Gabriele G. Schiattarella, and Heinrich Taegtmeyer
- Subjects
Cardiology and Cardiovascular Medicine - Published
- 2023
11. Long COVID and the cardiovascular system—elucidating causes and cellular mechanisms in order to develop targeted diagnostic and therapeutic strategies : A joint Scientific Statement of the ESC Working Groups on Cellular Biology of the Heart and Myocardial and Pericardial Diseases
- Author
-
Mariann Gyöngyösi, Pilar Alcaide, Folkert W Asselbergs, Bianca J J M Brundel, Giovanni G Camici, Paula da Costa Martins, Péter Ferdinandy, Marianna Fontana, Henrique Girao, Massimiliano Gnecchi, Can Gollmann-Tepeköylü, Petra Kleinbongard, Thomas Krieg, Rosalinda Madonna, Melanie Paillard, Antonis Pantazis, Cinzia Perrino, Maurizio Pesce, Gabriele G Schiattarella, Joost P G Sluijter, Sabine Steffens, Carsten Tschöpe, Sophie Van Linthout, Sean M Davidson, Martins, Paula da Costa [0000-0002-0695-1187], Fontana, Marianna [0000-0002-9233-9831], Girao, Henrique [0000-0002-5786-8447], Gnecchi, Massimiliano [0000-0001-7435-4328], Kleinbongard, Petra [0000-0003-3576-3772], Krieg, Thomas [0000-0002-5192-580X], Perrino, Cinzia [0000-0002-2274-0048], Pesce, Maurizio [0000-0002-3097-8961], Sluijter, Joost PG [0000-0003-2088-9102], Steffens, Sabine [0000-0002-6892-9751], Van Linthout, Sophie [0000-0002-7581-7870], Apollo - University of Cambridge Repository, Medizinische Universität Wien = Medical University of Vienna, Tufts University School of Medicine [Boston], University Medical Center [Utrecht], Utrecht University [Utrecht], University College of London [London] (UCL), Amsterdam UMC - Amsterdam University Medical Center, Vrije Universiteit Amsterdam [Amsterdam] (VU), Universität Zürich [Zürich] = University of Zurich (UZH), University hospital of Zurich [Zurich], Maastricht University [Maastricht], Semmelweis University [Budapest], Pharmahungary Group [Szeged, Hungary] (PG), University of Coimbra [Portugal] (UC), Coimbra Institute for Clinical and Biomedical Research [Coimbra, Portugal] (iCBR - Faculty of Medicine), Università degli Studi di Pavia = University of Pavia (UNIPV), Fondazione IRCCS Policlinico San Matteo [Pavia], Innsbruck Medical University = Medizinische Universität Innsbruck (IMU), Universitätsklinikum Essen [Universität Duisburg-Essen] (Uniklinik Essen), Addenbrooke's Hospital, Cambridge University NHS Trust, University of Pisa - Università di Pisa, Jefferson (Philadelphia University + Thomas Jefferson University), Cardiovasculaire, métabolisme, diabétologie et nutrition (CarMeN), Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Hospices Civils de Lyon (HCL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Imperial College London, Royal Brompton and Harefield NHS Foundation Trust, University of Naples Federico II = Università degli studi di Napoli Federico II, Centro Cardiologico Monzino [Milano], Dpt di Scienze Cliniche e di Comunità [Milano] (DISCCO), Università degli Studi di Milano = University of Milan (UNIMI)-Università degli Studi di Milano = University of Milan (UNIMI)-Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Charité - UniversitätsMedizin = Charité - University Hospital [Berlin], German Center for Cardiovascular Research (DZHK), Berlin Institute of Health (BIH), Max Delbrück Center for Molecular Medicine [Berlin] (MDC), Helmholtz-Gemeinschaft = Helmholtz Association, Utrecht Brain Center [UMC], Ludwig-Maximilians-Universität München (LMU), Charité Campus Virchow-Klinikum (CVK), The Hatter Cardiovascular Institute (THCI), CarMeN, laboratoire, Gyöngyösi, Mariann, Alcaide, Pilar, Asselbergs, Folkert W, Brundel, Bianca J J M, Camici, Giovanni G, da Costa Martins, Paula, Ferdinandy, Péter, Fontana, Marianna, Girao, Henrique, Gnecchi, Massimiliano, Gollmann-Tepeköylü, Can, Kleinbongard, Petra, Krieg, Thoma, Madonna, Rosalinda, Paillard, Melanie, Pantazis, Antoni, Perrino, Cinzia, Pesce, Maurizio, Schiattarella, Gabriele G, Sluijter, Joost P G, Steffens, Sabine, Tschöpe, Carsten, Van Linthout, Sophie, and Davidson, Sean M
- Subjects
Long COVID ,Heart Diseases ,Physiology ,SARS-CoV-2 ,[SDV]Life Sciences [q-bio] ,Myocardium ,Medizin ,COVID-19 ,Heart ,virus ,Post COVID ,Cardiovascular ,[SDV] Life Sciences [q-bio] ,Post-Acute COVID-19 Syndrome ,COVID-19 Testing ,Cardiovascular and Metabolic Diseases ,Physiology (medical) ,Humans ,Cardiology and Cardiovascular Medicine ,Cardiac - Abstract
Long COVID has become a world-wide, non-communicable epidemic, caused by long-lasting multiorgan symptoms that endure for weeks or months after SARS-CoV-2 infection has already subsided. This scientific document aims to provide insight into the possible causes and therapeutic options available for the cardiovascular manifestations of long COVID. In addition to chronic fatigue, which is a common symptom of long COVID, patients may present with chest pain, ECG abnormalities, postural orthostatic tachycardia, or newly developed supraventricular or ventricular arrhythmias. Imaging of the heart and vessels has provided evidence of chronic, post-infectious perimyocarditis with consequent left or right ventricular failure, arterial wall inflammation, or microthrombosis in certain patient populations. Better understanding of the underlying cellular and molecular mechanisms of long COVID will aid in the development of effective treatment strategies for its cardiovascular manifestations. A number of mechanisms have been proposed, including those involving direct effects on the myocardium, microthrombotic damage to vessels or endothelium, or persistent inflammation. Unfortunately, existing circulating biomarkers, coagulation, and inflammatory markers, are not highly predictive for either the presence or outcome of long COVID when measured 3 months after SARS-CoV-2 infection. Further studies are needed to understand underlying mechanisms, identify specific biomarkers, and guide future preventive strategies or treatments to address long COVID and its cardiovascular sequelae.
- Published
- 2023
12. The DWORF micropeptide enhances contractility and prevents heart failure in a mouse model of dilated cardiomyopathy
- Author
-
Catherine A Makarewich, Amir Z Munir, Gabriele G Schiattarella, Svetlana Bezprozvannaya, Olga N Raguimova, Ellen E Cho, Alexander H Vidal, Seth L Robia, Rhonda Bassel-Duby, and Eric N Olson
- Subjects
calcium ,contractility ,cardiac ,heart failure ,cardiomyopathy ,SERCA ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Calcium (Ca2+) dysregulation is a hallmark of heart failure and is characterized by impaired Ca2+ sequestration into the sarcoplasmic reticulum (SR) by the SR-Ca2+-ATPase (SERCA). We recently discovered a micropeptide named DWORF (DWarf Open Reading Frame) that enhances SERCA activity by displacing phospholamban (PLN), a potent SERCA inhibitor. Here we show that DWORF has a higher apparent binding affinity for SERCA than PLN and that DWORF overexpression mitigates the contractile dysfunction associated with PLN overexpression, substantiating its role as a potent activator of SERCA. Additionally, using a well-characterized mouse model of dilated cardiomyopathy (DCM) due to genetic deletion of the muscle-specific LIM domain protein (MLP), we show that DWORF overexpression restores cardiac function and prevents the pathological remodeling and Ca2+ dysregulation classically exhibited by MLP knockout mice. Our results establish DWORF as a potent activator of SERCA within the heart and as an attractive candidate for a heart failure therapeutic.
- Published
- 2018
- Full Text
- View/download PDF
13. Heart failure with preserved ejection fraction in humans and mice
- Author
-
Gabriele G. Schiattarella, Rudolf A. de Boer, Laura M G Meems, Carolyn S.P. Lam, Coenraad Withaar, Withaar, C., Lam, C. S. P., Schiattarella, G. G., De Boer, R. A., and Meems, L. M. G.
- Subjects
medicine.medical_specialty ,Consensus ,Mouse ,Context (language use) ,Signs and symptoms ,Consensu ,030204 cardiovascular system & hematology ,H2FPEF ,Age and sex ,HFA-PEFF ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Natriuretic Peptide ,Internal medicine ,medicine ,State of the Art Review ,Animals ,Humans ,AcademicSubjects/MED00200 ,Natriuretic Peptides ,Clinical syndrome ,Heart Failure and Cardiomyopathies ,030304 developmental biology ,Heart Failure ,0303 health sciences ,Ejection fraction ,business.industry ,Animal ,Multifactorial disease ,Translational ,Stroke Volume ,medicine.disease ,HFpEF ,3. Good health ,Algorithm ,Heart failure ,Cardiology ,Cardiology and Cardiovascular Medicine ,business ,Heart failure with preserved ejection fraction ,Algorithms ,Human - Abstract
Heart failure (HF) with preserved ejection fraction (HFpEF) is a multifactorial disease accounting for a large and increasing proportion of all clinical HF presentations. As a clinical syndrome, HFpEF is characterized by typical signs and symptoms of HF, a distinct cardiac phenotype and raised natriuretic peptides. Non-cardiac comorbidities frequently co-exist and contribute to the pathophysiology of HFpEF. To date, no therapy has proven to improve outcomes in HFpEF, with drug development hampered, at least partly, by lack of consensus on appropriate standards for pre-clinical HFpEF models. Recently, two clinical algorithms (HFA-PEFF and H2FPEF scores) have been developed to improve and standardize the diagnosis of HFpEF. In this review, we evaluate the translational utility of HFpEF mouse models in the context of these HFpEF scores. We systematically recorded evidence of symptoms and signs of HF or clinical HFpEF features and included several cardiac and extra-cardiac parameters as well as age and sex for each HFpEF mouse model. We found that most of the pre-clinical HFpEF models do not meet the HFpEF clinical criteria, although some multifactorial models resemble human HFpEF to a reasonable extent. We therefore conclude that to optimize the translational value of mouse models to human HFpEF, a novel approach for the development of pre-clinical HFpEF models is needed, taking into account the complex HFpEF pathophysiology in humans., Graphical Abstract An in-depth review of existing pre-clinical HFpEF mouse models with validation of their translational value using the HFA-PEFF and H2FPEF scores.
- Published
- 2021
14. Cooperative Binding of ETS2 and NFAT Links Erk1/2 and Calcineurin Signaling in the Pathogenesis of Cardiac Hypertrophy
- Author
-
Anwarul Ferdous, Xiang Luo, Nan Jiang, Ding Sheng Jiang, Guihao Chen, Qinfeng Li, Joseph A. Hill, Yuxuan Luo, Sergio Lavandero, Beverly A. Rothermel, Herman I. May, Thomas G. Gillette, Gabriele G. Schiattarella, Chao Li, Luo, Yuxuan, Jiang, Nan, May, Herman I, Luo, Xiang, Ferdous, Anwarul, Schiattarella, Gabriele G, Chen, Guihao, Li, Qinfeng, Li, Chao, Rothermel, Beverly A, Jiang, Dingsheng, Lavandero, Sergio, Gillette, Thomas G, and Hill, Joseph A
- Subjects
Male ,MAPK/ERK pathway ,MAP Kinase Signaling System ,ETS2 ,Mice, Transgenic ,030204 cardiovascular system & hematology ,Proto-Oncogene Protein c-ets-2 ,Rats, Sprague-Dawley ,Pathogenesis ,Mice ,03 medical and health sciences ,0302 clinical medicine ,mir-223 ,Original Research Articles ,Physiology (medical) ,medicine ,Animals ,Humans ,calcineurin-NFAT pathway ,Risk factor ,Cells, Cultured ,030304 developmental biology ,Mice, Knockout ,0303 health sciences ,NFATC Transcription Factors ,business.industry ,Calcineurin ,cardiac hypertrophy ,Cooperative binding ,NFAT ,medicine.disease ,Rats ,HEK293 Cells ,Cardiovascular and Metabolic Diseases ,cardiomegaly ,Heart failure ,MAPK/Erk pathway ,ComputingMethodologies_DOCUMENTANDTEXTPROCESSING ,Cancer research ,microRNA-223 ,Cardiology and Cardiovascular Medicine ,business ,Protein Binding - Abstract
Supplemental Digital Content is available in the text., Background: Cardiac hypertrophy is an independent risk factor for heart failure, a leading cause of morbidity and mortality globally. The calcineurin/NFAT (nuclear factor of activated T cells) pathway and the MAPK (mitogen-activated protein kinase)/Erk (extracellular signal-regulated kinase) pathway contribute to the pathogenesis of cardiac hypertrophy as an interdependent network of signaling cascades. How these pathways interact remains unclear and few direct targets responsible for the prohypertrophic role of NFAT have been described. Methods: By engineering cardiomyocyte-specific ETS2 (a member of the E26 transformation-specific sequence [ETS] domain family) knockout mice, we investigated the role of ETS2 in cardiac hypertrophy. Primary cardiomyocytes were used to evaluate ETS2 function in cell growth. Results: ETS2 is phosphorylated and activated by Erk1/2 on hypertrophic stimulation in both mouse (n=3) and human heart samples (n=8 to 19). Conditional deletion of ETS2 in mouse cardiomyocytes protects against pressure overload–induced cardiac hypertrophy (n=6 to 11). Silencing of ETS2 in the hearts of calcineurin transgenic mice significantly attenuates hypertrophic growth and contractile dysfunction (n=8). As a transcription factor, ETS2 is capable of binding to the promoters of hypertrophic marker genes, such as ANP, BNP, and Rcan1.4 (n=4). We report that ETS2 forms a complex with NFAT to stimulate transcriptional activity through increased NFAT binding to the promoters of at least 2 hypertrophy-stimulated genes: Rcan1.4 and microRNA-223 (=n4 to 6). Suppression of microRNA-223 in cardiomyocytes inhibits calcineurin-mediated cardiac hypertrophy (n=6), revealing microRNA-223 as a novel prohypertrophic target of the calcineurin/NFAT and Erk1/2-ETS2 pathways. Conclusions: Our findings point to a critical role for ETS2 in calcineurin/NFAT pathway-driven cardiac hypertrophy and unveil a previously unknown molecular connection between the Erk1/2 activation of ETS2 and expression of NFAT/ETS2 target genes.
- Published
- 2021
15. Abstract P3016: Role Of AMPK/Sirtuin3 Signaling In HFpEF-associated Atrial Myopathy
- Author
-
Dan Tong, Gabriele G Schiattarella, Nan Jiang, Daniel Daou, Yuxuan Luo, Mark S Link, Sergio Lavandero, Thomas G Gillette, and Joseph A Hill
- Subjects
Physiology ,Cardiology and Cardiovascular Medicine - Abstract
Background: Patients with heart failure with preserved ejection fraction (HFpEF) are uniquely predisposed to atrial fibrillation (AF), which significantly worsens clinical outcomes. Pathological atrial remodeling, i.e. atrial myopathy, usually precedes clinical AF and is deemed to be an underlying cause of AF. However, little is known about its molecular features, and no effective treatments have been identified. Method: Wild type C57Bl6 mice were fed a high fat diet (HFD) and received L-NAME via drinking water (5-8 weeks). Atrial morphology and function were assessed by echocardiography. AF was induced by transesophageal pacing. Results: We discovered that HFpEF mice manifest prominent sinoatrial node (SAN) dysfunction and are highly susceptible to pacing-induced AF, suggesting that this is an ideal model for studying HFpEF-associated atrial changes. Our findings revealed that atrial hypertrophy, contractile dysfunction, and conduction abnormalities are key features of HFpEF-associated atrial myopathy. Interestingly, we did not observe significant atrial fibrosis in this model, a prominent feature of other AF models. Importantly, we discovered impaired AMPK/Sirt3 signaling in the atria of HFpEF mice, and similar atrial defects were observed in mice with cardiomyocyte-specific loss of sirtuin3 (Sirt3) or AMPK, indicating a critical role of AMPK/Sirt3 signaling in HFpEF-associated atrial myopathy. Lastly, we found that metformin, a widely used clinical AMPK agonist, significantly attenuated AF inducibility and SAN dysfunction in HFpEF mice. Conclusion: Our “two-hit” HFpEF model successfully recapitulates HFpEF-associated atrial phenotypes. Using this model, we unveiled molecular features of a distinct atrial myopathy. We identified a critical role of AMPK/Sirt3 signaling in HFpEF-associated atrial remodeling and demonstrated the therapeutic effect of metformin. As next steps, confirming this benefit in a clinical trial is warranted.
- Published
- 2022
16. Up next: the dawn of systems biology in HFpEF research
- Author
-
David Bode, Cristian Sotomayor-Flores, and Gabriele G. Schiattarella
- Subjects
Heart Failure ,Cardiovascular and Metabolic Diseases ,Systems Biology ,Humans ,Stroke Volume ,Cardiology and Cardiovascular Medicine ,Molecular Biology ,Biomarkers - Published
- 2022
17. Transverse aortic constriction induces gut barrier alterations, microbiota remodeling and systemic inflammation
- Author
-
Lorena Coretti, Mariella Cuomo, Orlando Paciello, S D'Apice, Francesca Lembo, Giuseppina Mattace Raso, Roberta Paolillo, Gina Cavaliere, Cinzia Perrino, Ilaria d'Aquino, Maria Pina Mollica, Giuseppe Giugliano, Gabriele G. Schiattarella, Giovanni Esposito, Adriano Lama, and Nicola Boccella
- Subjects
0301 basic medicine ,Lipopolysaccharide ,medicine.medical_treatment ,030204 cardiovascular system & hematology ,Gut flora ,Systemic inflammation ,Mice ,Feces ,chemistry.chemical_compound ,0302 clinical medicine ,Intestinal Mucosa ,Multidisciplinary ,Ventricular Remodeling ,biology ,Pathophysiology ,Cardiac hypertrophy ,Cytokine ,Echocardiography ,Heart Function Tests ,Medicine ,Disease Susceptibility ,medicine.symptom ,Science ,Article ,Permeability ,Proinflammatory cytokine ,03 medical and health sciences ,medicine ,Animals, Aortic Valve Stenosis, Biomarkers, Disease Models, Disease Susceptibility, Echocardiography, Feces, Heart Failure, Inflammation, Intestinal Mucosa, Permeability, Ventricular Remodeling, Gastrointestinal Microbiome ,Animals ,Heart Failure ,Inflammation ,Pressure overload ,business.industry ,Aortic Valve Stenosis ,biology.organism_classification ,medicine.disease ,Gastrointestinal Microbiome ,Disease Models, Animal ,stomatognathic diseases ,030104 developmental biology ,chemistry ,Cardiovascular and Metabolic Diseases ,Heart failure ,Disease Models ,Immunology ,Dysbiosis ,Metagenome ,Microbiome ,Metagenomics ,business ,Biomarkers - Abstract
Accumulating evidence suggests that modifications of gut function and microbiota composition might play a pivotal role in the pathophysiology of several cardiovascular diseases, including heart failure (HF). In this study we systematically analysed gut microbiota composition, intestinal barrier integrity, intestinal and serum cytokines and serum endotoxin levels in C57BL/6 mice undergoing pressure overload by transverse aortic constriction (TAC) for 1 and 4 weeks. Compared to sham-operated animals, TAC induced prompt and strong weakening of intestinal barrier integrity, long-lasting decrease of colon anti-inflammatory cytokine levels, significant increases of serum levels of bacterial lipopolysaccharide and proinflammatory cytokines. TAC also exerted effects on microbiota composition, inducing significant differences in bacterial genera inside Actinobacteria, Firmicutes, Proteobacteria and TM7 phyla as shown by 16S rDNA sequencing of fecal samples from TAC or sham mice. These results suggest that gut modifications represent an important element to be considered in the development and progression of cardiac dysfunction in response to TAC and support this animal model as a valuable tool to establish the role and mechanisms of gut-heart crosstalk in HF. Evidence arising in this field might identify new treatment options targeting gut integrity and microbiota components to face adverse cardiac events.
- Published
- 2021
18. Xbp1s-FoxO1 axis governs lipid accumulation and contractile performance in heart failure with preserved ejection fraction
- Author
-
Joseph A. Hill, Xuliang Wang, Nan Jiang, Soo Young Kim, Stephen B. Spurgin, Hande Piristine, Sergio Lavandero, Kristin M. French, Vlad G. Zaha, Francisco Altamirano, Subhajit Dasgupta, Anwarul Ferdous, Theodore M. Hill, Herman I. May, Thomas G. Gillette, Maayan Waldman, Dan Tong, Gabriele G. Schiattarella, Heesoo Yoo, Yuxuan Luo, Elisa Villalobos, Schiattarella, G. G., Altamirano, F., Kim, S. Y., Tong, D., Ferdous, A., Piristine, H., Dasgupta, S., Wang, X., French, K. M., Villalobos, E., Spurgin, S. B., Waldman, M., Jiang, N., May, H. I., Hill, T. M., Luo, Y., Yoo, H., Zaha, V. G., Lavandero, S., Gillette, T. G., and Hill, J. A.
- Subjects
0301 basic medicine ,X-Box Binding Protein 1 ,Ubiquitin-Protein Ligase ,Transcription, Genetic ,General Physics and Astronomy ,FOXO1 ,030204 cardiovascular system & hematology ,Heart Ventricle ,Mice ,0302 clinical medicine ,Ubiquitin ,HEK293 Cell ,Myocytes, Cardiac ,Proteolysi ,Conserved Sequence ,Multidisciplinary ,biology ,Chemistry ,Forkhead Box Protein O1 ,Protein Stability ,Cell biology ,Ubiquitin ligase ,Phenotype ,Cell signalling ,Human ,Heart Ventricles ,Ubiquitin-Protein Ligases ,Science ,Models, Biological ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,medicine ,Animals ,Humans ,Transcription factor ,STUB1 ,Heart Failure ,Binding Sites ,Base Sequence ,Animal ,Myocardium ,Binding Site ,Stroke Volume ,General Chemistry ,medicine.disease ,Lipid Metabolism ,Myocardial Contraction ,Mice, Inbred C57BL ,HEK293 Cells ,030104 developmental biology ,Cardiovascular and Metabolic Diseases ,Heart failure ,Proteolysis ,biology.protein ,Unfolded protein response ,Heart failure with preserved ejection fraction ,Gene Deletion - Abstract
Heart failure with preserved ejection fraction (HFpEF) is now the dominant form of heart failure and one for which no efficacious therapies exist. Obesity and lipid mishandling greatly contribute to HFpEF. However, molecular mechanism(s) governing metabolic alterations and perturbations in lipid homeostasis in HFpEF are largely unknown. Here, we report that cardiomyocyte steatosis in HFpEF is coupled with increases in the activity of the transcription factor FoxO1 (Forkhead box protein O1). FoxO1 depletion, as well as over-expression of the Xbp1s (spliced form of the X-box-binding protein 1) arm of the UPR (unfolded protein response) in cardiomyocytes each ameliorates the HFpEF phenotype in mice and reduces myocardial lipid accumulation. Mechanistically, forced expression of Xbp1s in cardiomyocytes triggers ubiquitination and proteasomal degradation of FoxO1 which occurs, in large part, through activation of the E3 ubiquitin ligase STUB1 (STIP1 homology and U-box-containing protein 1) a novel and direct transcriptional target of Xbp1s. Our findings uncover the Xbp1s-FoxO1 axis as a pivotal mechanism in the pathogenesis of cardiometabolic HFpEF and unveil previously unrecognized mechanisms whereby the UPR governs metabolic alterations in cardiomyocytes., Heart failure with preserved ejection fraction (HFpEF) is a global, major health issue for which no effective therapies are available. Here, the authors discover that the interplay between two transcription factors, Xbp1s and FoxO1, is critical for metabolic adaptation and lipid handling in HFpEF-stressed cardiomyocytes.
- Published
- 2021
19. Immunometabolic Mechanisms of Heart Failure with Preserved Ejection Fraction
- Author
-
Gabriele G. Schiattarella, Pilar Alcaide, Gianluigi Condorelli, Thomas G. Gillette, Stephane Heymans, Elizabeth A. V. Jones, Marinos Kallikourdis, Andrew Lichtman, Federica Marelli-Berg, Sanjiv J. Shah, Edward B. Thorp, Joseph A. Hill, Schiattarella, Gabriele G, Alcaide, Pilar, Condorelli, Gianluigi, Gillette, Thomas G, Heymans, Stephane, Jones, Elizabeth A V, Kallikourdis, Marino, Lichtman, Andrew, Marelli-Berg, Federica, Shah, Sanjiv, Thorp, Edward B, and Hill, Joseph A
- Subjects
immune system ,HFpEF ,metabolism ,Article - Abstract
Heart failure with preserved ejection fraction (HFpEF) is increasing in prevalence worldwide, already accounting for at least half of all heart failure (HF). As most patients with HFpEF are obese with metabolic syndrome, metabolic stress has been implicated in syndrome pathogenesis. Recently, compelling evidence for bidirectional crosstalk between metabolic stress and chronic inflammation has emerged, and alterations in systemic and cardiac immune responses are held to participate in HFpEF pathophysiology. Indeed, based on both preclinical and clinical evidence, comorbidity-driven systemic inflammation, coupled with metabolic stress, have been implicated together in HFpEF pathogenesis. As metabolic alterations impact immune function(s) in HFpEF, major changes in immune cell metabolism are also recognized in HFpEF and in HFpEF-predisposing conditions. Both arms of immunity - innate and adaptive - are implicated in the cardiomyocyte response in HFpEF. Indeed, we submit that crosstalk among adipose tissue, the immune system, and the heart represents a critical component of HFpEF pathobiology. Here, we review recent evidence in support of immunometabolic mechanisms as drivers of HFpEF pathogenesis, discuss pivotal biological mechanisms underlying the syndrome, and highlight questions requiring additional inquiry.
- Published
- 2022
20. Immunoglobulins G modulate endothelial function and affect insulin sensitivity in humans
- Author
-
Raffaele Napoli, Antonio Ruvolo, Fabio Magliulo, Cecilia Nigro, Claudia Miele, Giuseppe Spadaro, Antonio Pecoraro, Antonio Cittadini, Giovanni Esposito, Paola Triggianese, Simona Grassi, Gabriele G. Schiattarella, Amato de Paulis, Nella Prevete, Napoli, R., Ruvolo, A., Triggianese, P., Prevete, N., Schiattarella, G. G., Nigro, C., Miele, C., Magliulo, F., Grassi, S., Pecoraro, A., Cittadini, A., Esposito, G., de Paulis, A., and Spadaro, G.
- Subjects
Blood Glucose ,Male ,Time Factors ,Brachial Artery ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Medicine (miscellaneous) ,chemistry.chemical_compound ,Insulin ,Medicine ,Infusions, Intravenou ,Brachial artery ,Endothelial dysfunction ,Infusions, Intravenous ,Cells, Cultured ,Nutrition and Dietetics ,biology ,Flow mediated dilation ,Immunoglobulins, Intravenous ,Vasodilation ,Treatment Outcome ,Atherosclerosi ,Female ,Antibody ,Case-Control Studie ,Cardiology and Cardiovascular Medicine ,Human ,medicine.medical_specialty ,Time Factor ,Adolescent ,Nitric Oxide ,Nitric oxide ,Young Adult ,Insulin resistance ,Internal medicine ,Diabetes mellitus ,medicine.artery ,Immunoglobulin ,Humans ,business.industry ,Common variable immunodeficiency ,Endothelial function ,Biomarker ,medicine.disease ,Common Variable Immunodeficiency ,Endocrinology ,chemistry ,Immunoglobulins, Intravenou ,Case-Control Studies ,Immunoglobulin G ,biology.protein ,Endothelium, Vascular ,Insulin Resistance ,business ,Biomarkers - Abstract
Background and aims: Data from animals suggest that immunoglobulins G (IgG) play a mechanistic role in atherosclerosis and diabetes through endothelial dysfunction and insulin resistance. Patients with common variable immunodeficiency (CVID), who have low circulating levels of IgG and are treated with intravenous polyclonal IgG (IVIgG), may provide an ideal model to clarify whether circulating IgG modulate endothelial function and affect insulin sensitivity in humans. Methods and results: We studied 24 patients with CVID and 17 matched healthy controls (HC). Endothelial function was evaluated as flow mediated dilation (FMD) of the brachial artery at baseline and 1, 7, 14, and 21 days after IVIgG infusion in the CVID patients. We measured also plasma glucose, insulin, and calculated the HOMA-IR index. We also investigated the role of human IgG on the production of Nitric Oxide (NO) in vitro in Human Coronary Artery Endothelial Cells (HCAEC). Compared to HC, FMD of CVID patients was significantly impaired at baseline (9.4 ± 0.9 and 7.6 ± 0.6% respectively, p < 0.05) but rose above normal levels 1 and 7 days after IVIgG infusion to return at baseline at 14 and 21 days. Serum insulin concentration and HOMA-IR index dropped by 50% in CVID patients after IVIgG (p < 0.002 vs. baseline). In vitro IgG stimulated NO production in HCAEC. Conclusions: Reduced IgG levels are associated with endothelial dysfunction and IVIgG stimulates endothelial function directly while improving insulin sensitivity. The current findings may suggest an anti-atherogenic role of human IgG.
- Published
- 2020
21. Impaired AMP-Activated Protein Kinase Signaling in Heart Failure With Preserved Ejection Fraction-Associated Atrial Fibrillation
- Author
-
Dan Tong, Gabriele G. Schiattarella, Nan Jiang, Daniel Daou, Yuxuan Luo, Mark S. Link, Sergio Lavandero, Thomas G. Gillette, Joseph A. Hill, Tong, Dan, Schiattarella, Gabriele G, Jiang, Nan, Daou, Daniel, Luo, Yuxuan, Link, Mark S, Lavandero, Sergio, Gillette, Thomas G, and Hill, Joseph A
- Subjects
Heart Failure ,Physiology (medical) ,Humans ,Stroke Volume ,atrial fibrillation ,AMP-Activated Protein Kinases ,Cardiology and Cardiovascular Medicine ,Prognosis ,heart failure, diastolic ,Ventricular Function, Left ,Article - Published
- 2022
22. Small nucleolar RNA SNORD3A: a potential new biomarker and molecular player in heart failure
- Author
-
C L Holley, Cinzia Perrino, A. Della Corte, Gabriele G. Schiattarella, S D'Apice, Roberta Paolillo, Giovanni Esposito, and Ciro Bancone
- Subjects
business.industry ,Heart failure ,medicine ,Cancer research ,Biomarker (medicine) ,Small nucleolar RNA ,Cardiology and Cardiovascular Medicine ,medicine.disease ,business - Abstract
Background Despite optimal therapy, heart failure (HF) remains a relentless and deadly disease. Given the relative inaccessibility of myocardial human tissues, identification of circulating biomarkers mirroring myocardial pathological signaling pathways, especially in peripheral blood mononuclear cells (PBMC) is expected to be extremely relevant. Small Nucleolar RNAs (snoRNAs) have been shown to play important roles in various cellular physiological processes. However, the connection between snoRNAs and pathological dysfunction in the heart or peripheral blood mononuclear cells (PBMC) is still poorly understood. Purpose To identify novel circulating PBMC biomarkers linked to myocardial dysfunction and HF. Methods Myocardial left ventricle (LV) samples and PBMC were obtained from patients affected by ischemic HF (HF, n=13) undergoing heart transplantation and control donors (CD, n=7) and analyzed by RNA sequencing analysis (RNASeq). SNORD3A expression levels in the different groups were evaluated by quantitative real-time PCR. HF was induced in 8-week-old wild type C57BL/6 mice by transverse aortic constriction (TAC). Sham-operated mice (sham) were used as controls. After twelve-week-TAC (12w) or sham operation, mice were anesthetized, cardiac function was analyzed by echocardiography, and cardiac/PBMC samples were collected after sacrifice. In order to test the role of SNORD3A in cardiomyocyte hypoxia, H9C2 cardiomyoblasts were transfected with SNORD3A-targeted antisense oligonucleotides (ASO) and cell survival was analyzed by cleaved caspase-3 and PARP1 immunoblotting. Results RnaSeq analysis identified a small set of genes differentially expressed in the heart and PBMC from HF patients. Among these, SNORD3A was up-regulated in cardiac and PBMC samples from HF patients compared to CD (Figure 1A-B). Similarly, in murine HF induced by 12w TAC, SNORD3A levels were increased by rtPCR, both in the heart and PBMC (Figure 1C-D). SNORD3A expression levels were also significantly increased in H9C2 cells exposed to in vitro hypoxia (Figure 1E). Interestingly, H9C2 transfection with SNORD3A-specific ASO significantly reduced hypoxia-induced SNORD3A upregulation and reduced hypoxia-induced cell death (Figure 1F-G). Conclusions In this study, we identify SNORD3A as a novel possible biomarker in human HF, similarly up-regulated in the heart and PBMC, induced by hypoxia in vitro and modulating cell survival. Funding Acknowledgement Type of funding sources: Public grant(s) – National budget only. Main funding source(s): STAR GRANT Figure 1
- Published
- 2021
23. Abstract P464: Cardiac Dysfunction In A Mouse Model For Autosomal Dominant Polycystic Kidney Disease
- Author
-
William Perez, Gabriele G. Schiattarella, Francisco Altamirano, Troy Hendrickson, and Joseph A. Hill
- Subjects
Pathology ,medicine.medical_specialty ,Physiology ,business.industry ,Autosomal dominant polycystic kidney disease ,medicine ,Cardiology and Cardiovascular Medicine ,medicine.disease ,business ,Cardiac dysfunction - Abstract
Mutations in polycystin-1 (PC1) cause autosomal dominant polycystic kidney disease (ADPKD), a disorder that manifests with cardiac hypertrophy and dysfunction. We recently showed that cardiomyocyte-specific PC1 KO mice exhibit both systolic and diastolic dysfunction without signs of cardiac hypertrophy. The purpose of this study was to determine the effects of ADPKD-causing PC1 mutations on cardiac function using a mouse model for ADPKD harboring a mutation in PC1 R3277C (RC/RC). We used echocardiography to determine cardiac function and Western blot analysis to explore signaling pathways in WT and RC/RC mice (2-4 months of age).We observed a slight but significant decrease in ejection fraction (77.2±0.7 vs 86.2±2.4 %, N=5, 4, P=0.015) without signs of left ventricular hypertrophy (LV mass 78.4±5.5 vs 93.5±4.2 mg, N=5, 4, P>0.05) in RC/RC compared to WT mice. Western blot analysis from total heart lysates (WT and RC/RC; N=5) revealed no changes in protein levels of hypertrophic markers: beta myosin heavy chain (β-MHC) and regulator of calcineurin 1 (RCAN1). In addition, we studied multiple signaling pathways involved in cardiac hypertrophy by analyzing their phosphorylation status by Western blot (phosphorylated/total protein). We observed no changes in mTOR, S6K1 and S6 phosphorylation. However, a decrease in p-4EBP1 and p-eIF4B was observed in RC/RC compared to WT. Moreover, we observed a significant increase in p-ERK and p-CaMKII. Our data suggest that alterations in PC1 signaling promote cardiac dysfunction but do not promote hypertrophy in young mice (2-4 months of age). Published evidence (PMID: 32730856) suggest that RC/RC hearts become hypertrophic at 6 months of age. However, our data suggest there may be dysfunction prior to cardiac hypertrophy. This warrants further investigation into the more primary role of ADPKD-associated co-morbidities. More studies, with a larger animal cohort, are necessary to unveil the effects of mutant PC1 on cardiac function.
- Published
- 2021
24. Activation of Autophagic Flux Blunts Cardiac Ischemia/Reperfusion Injury
- Author
-
Min Xie, John M. Shelton, Nan Jiang, Jessica Medina, Francisco Altamirano, Dan L. Li, Joseph A. Hill, Cyndi R. Morales, Anwarul Ferdous, Gabriele G. Schiattarella, Geoffrey W Cho, Xiang Luo, Herman I. May, Thomas G. Gillette, Yongli Kong, Xie, Min, Cho, Geoffrey W, Kong, Yongli, Li, Dan L, Altamirano, Francisco, Luo, Xiang, Morales, Cyndi R, Jiang, Nan, Schiattarella, Gabriele G, May, Herman I, Medina, Jessica, Shelton, John M, Ferdous, Anwarul, Gillette, Thomas G, and Hill, Joseph A
- Subjects
medicine.medical_specialty ,Physiology ,Ischemia ,Myocardial Reperfusion Injury ,ischemia ,Autophagy-Related Protein 7 ,Article ,Rats, Sprague-Dawley ,Mice ,Internal medicine ,medicine ,Autophagy ,Animals ,Myocytes, Cardiac ,Cells, Cultured ,cardiomyopathie ,business.industry ,Cardiac ischemia ,medicine.disease ,Myocardial Contraction ,Recombinant Proteins ,reperfusion ,Rats ,Mice, Inbred C57BL ,histone deacetylase ,Cardiology ,Beclin-1 ,Cardiology and Cardiovascular Medicine ,business ,Reactive Oxygen Species ,Flux (metabolism) ,Reperfusion injury - Abstract
Rationale: Reperfusion injury accounts for up to half of myocardial infarct size, and meaningful clinical therapies targeting it do not exist. We have reported previously that autophagy is reduced during reperfusion and that HDAC (histone deacetylase) inhibition enhances cardiomyocyte autophagy and blunts ischemia/reperfusion (I/R) injury when administered at the time of reperfusion. However, whether inducing autophagy per se, as opposed to other effects triggered by HDAC inhibition, is sufficient to protect against reperfusion injury is not clear. Objective: We set out to test whether augmentation of autophagy using a specific autophagy-inducing peptide, TB (Tat-Beclin), protects the myocardium through reduction of reactive oxygen species (ROS) during reperfusion injury. Methods and Results: Eight- to 12-week-old, WT (wild type) C57BL6 mice and tamoxifen-inducible cardiomyocyte-specific ATG7 KO (ATG7 knockout) mice (to test the dependency on autophagy) were randomized into 2 groups: exposed to a control TS (Tat-scrambled) peptide or a TB peptide. Each group was subjected to I/R surgery (45-minute coronary ligation, 24-hour reperfusion). Infarct size, systolic function, autophagic flux, and ROS were assayed. Cultured neonatal rat ventricular myocytes were exposed to TB during simulated I/R injury. ATG7 knockdown by small interfering RNA in neonatal rat ventricular myocytes was used to evaluate the role of autophagy. TB treatment at reperfusion reduced infarct size by 20% (absolute reduction; 50% relative reduction) and improved contractile function. Improvement correlated with increased autophagic flux in the border zone with less oxidative stress. ATG7 KO mice did not manifest TB-promoted cardioprotection during I/R. In neonatal rat ventricular myocytes subjected to I/R, TB reduced cell death by 41% and reduced I/R-induced ROS generation. Conversely, ATG7 knockdown in neonatal rat ventricular myocytes abolished these beneficial effects of TB on cell death and ROS reduction. Conclusions: Induction of autophagy at the time of reperfusion is sufficient to mitigate myocardial reperfusion injury by reducing ROS and cell death. Maintenance of appropriate autophagic flux may emerge as a viable clinical therapy to reduce reperfusion injury in acute myocardial infarction.
- Published
- 2021
25. Remodeling of substrate consumption in the murine sTAC model of heart failure
- Author
-
Matthew E. Merritt, Aslan T. Turer, Francisco Altamirano, Craig R. Malloy, Herman I. May, Thomas G. Gillette, Gabriele G. Schiattarella, Turer, A., Altamirano, F., Schiattarella, G. G., May, H., Gillette, T. G., Malloy, C. R., and Merritt, M. E.
- Subjects
Male ,0301 basic medicine ,Pyruvate decarboxylation ,Citric Acid Cycle ,Metabolomic ,Oxidative phosphorylation ,030204 cardiovascular system & hematology ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Acetyl Coenzyme A ,Pyruvic Acid ,medicine ,Animals ,Metabolomics ,Glycolysis ,Molecular Biology ,Aorta ,Heart Failure ,chemistry.chemical_classification ,Animal ,Myocardium ,Fatty acid ,Substrate selection ,Heart ,medicine.disease ,Ketone ,Constriction ,Hypertensive heart disease ,Mice, Inbred C57BL ,Citric acid cycle ,Disease Models, Animal ,Glucose ,030104 developmental biology ,chemistry ,Biochemistry ,Anaplerosi ,Heart failure ,Metabolome ,Energy Metabolism ,Cardiology and Cardiovascular Medicine ,Energy source ,Oxidation-Reduction - Abstract
Background Energy metabolism and substrate selection are key aspects of correct myocardial mechanical function. Myocardial preference for oxidizable substrates changes in both hypertrophy and in overt failure. Previous work has shown that glucose oxidation is upregulated in overpressure hypertrophy, but its fate in overt failure is less clear. Anaplerotic flux of pyruvate into the tricarboxylic acid cycle (TCA) has been posited as a secondary fate of glycolysis, aside from pyruvate oxidation or lactate production. Methods and results A model of heart failure that emulates both valvular and hypertensive heart disease, the severe transaortic constriction (sTAC) mouse, was assayed for changes in substrate preference using metabolomic and carbon-13 flux measurements. Quantitative measures of O2 consumption in the Langendorff perfused mouse heart were paired with 13C isotopomer analysis to assess TCA cycle turnover. Since the heart accommodates oxidation of all physiological energy sources, the utilization of carbohydrates, fatty acids, and ketones were measured simultaneously using a triple-tracer NMR method. The fractional contribution of glucose to acetyl-CoA production was upregulated in heart failure, while other sources were not significantly different. A model that includes both pyruvate carboxylation and anaplerosis through succinyl-CoA produced superior fits to the data compared to a model using only pyruvate carboxylation. In the sTAC heart, anaplerosis through succinyl-CoA is elevated, while pyruvate carboxylation was not. Metabolomic data showed depleted TCA cycle intermediate pool sizes versus the control, in agreement with previous results. Conclusion In the sTAC heart failure model, the glucose contribution to acetyl-CoA production was significantly higher, with compensatory changes in fatty acid and ketone oxidation not reaching a significant level. Anaplerosis through succinyl-CoA is also upregulated, and is likely used to preserve TCA cycle intermediate pool sizes. The triple tracer method used here is new, and can be used to assess sources of acetyl-CoA production in any oxidative tissue.
- Published
- 2019
26. Nitrosative stress drives heart failure with preserved ejection fraction
- Author
-
Nan Jiang, Sergio Lavandero, Kristin M. French, Thomas G. Gillette, Kavita Sharma, Virginia S. Hahn, Gabriele G. Schiattarella, David A. Kass, Dong I. Lee, Dan Tong, Soo Young Kim, Zhao V. Wang, Elisa Villalobos, Xiang Luo, Joseph A. Hill, Jian Huang, Francisco Altamirano, Pradeep P.A. Mammen, Theodore M. Hill, Herman I. May, Schiattarella, G. G., Altamirano, F., Tong, D., French, K. M., Villalobos, E., Kim, S. Y., Luo, X., Jiang, N., May, H. I., Wang, Z. V., Hill, T. M., Mammen, P. P. A., Huang, J., Lee, D. I., Hahn, V. S., Sharma, K., Kass, D. A., Lavandero, S., Gillette, T. G., and Hill, J. A.
- Subjects
Male ,X-Box Binding Protein 1 ,0301 basic medicine ,medicine.medical_specialty ,XBP1 ,Protein Serine-Threonine Kinase ,Nitrosative Stre ,Nitric Oxide Synthase Type II ,030204 cardiovascular system & hematology ,Diet, High-Fat ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Myocyte ,Myocytes, Cardiac ,Endoribonuclease ,Heart Failure ,Multidisciplinary ,Ejection fraction ,biology ,Animal ,business.industry ,Stroke Volume ,Stroke volume ,Mice, Inbred C57BL ,Nitric oxide synthase ,Disease Models, Animal ,NG-Nitroarginine Methyl Ester ,Phenotype ,030104 developmental biology ,Endocrinology ,biology.protein ,Unfolded protein response ,Signal transduction ,business ,Heart failure with preserved ejection fraction ,Human ,Signal Transduction - Abstract
Heart failure with preserved ejection fraction (HFpEF) is a common syndrome with high morbidity and mortality for which there are no evidence-based therapies. Here we report that concomitant metabolic and hypertensive stress in mice—elicited by a combination of high-fat diet and inhibition of constitutive nitric oxide synthase using Nω-nitro-l-arginine methyl ester (l-NAME)—recapitulates the numerous systemic and cardiovascular features of HFpEF in humans. Expression of one of the unfolded protein response effectors, the spliced form of X-box-binding protein 1 (XBP1s), was reduced in the myocardium of our rodent model and in humans with HFpEF. Mechanistically, the decrease in XBP1s resulted from increased activity of inducible nitric oxide synthase (iNOS) and S-nitrosylation of the endonuclease inositol-requiring protein 1α (IRE1α), culminating in defective XBP1 splicing. Pharmacological or genetic suppression of iNOS, or cardiomyocyte-restricted overexpression of XBP1s, each ameliorated the HFpEF phenotype. We report that iNOS-driven dysregulation of the IRE1α–XBP1 pathway is a crucial mechanism of cardiomyocyte dysfunction in HFpEF. iNOS-driven dysregulation of the IRE1α–XBP1 pathway leads to cardiomyocyte dysfunction in mice and recapitulates the systemic and cardiovascular features of human heart failure with preserved ejection fraction.
- Published
- 2019
27. Diagnostics and therapeutic implications of gut microbiota alterations in cardiometabolic diseases
- Author
-
Giovanni Esposito, Gabriele G. Schiattarella, Cinzia Perrino, Anna Sannino, Schiattarella, Gabriele G., Sannino, Anna, Esposito, Giovanni, and Perrino, Cinzia
- Subjects
Heart failure ,Disease ,030204 cardiovascular system & hematology ,Gut flora ,Bioinformatics ,digestive system ,Metagenomic ,03 medical and health sciences ,0302 clinical medicine ,Metabolic Diseases ,Trimethylamine n-oxide ,Animals ,Humans ,Medicine ,030212 general & internal medicine ,Microbiome ,Enzyme Inhibitors ,Exercise ,Bacteria ,biology ,business.industry ,Probiotics ,Cardiovascular risk ,biology.organism_classification ,Anti-Bacterial Agents ,Gastrointestinal Microbiome ,Diet ,Prebiotics ,Cardiovascular Diseases ,Host-Pathogen Interactions ,Dysbiosis ,Digestive tract ,Diet, Healthy ,Cardiology and Cardiovascular Medicine ,business ,Metabolic activity ,Risk Reduction Behavior - Abstract
Alterations in gut microbiota composition and its metabolic activity are emerging as one of the most powerful determinants of cardiovascular disease. Although our knowledge of the precise molecular mechanisms by which gut microbiota influences cardiometabolic homeostasis is still limited, a growing body of knowledge has recently been uncovered about the potential modulation of microbiome for cardiovascular diagnostic and therapeutic aspects. The multitude of interactions between the microorganisms inhabiting the digestive tract and the host has been recognized crucial in the development and progression of atherosclerosis, obesity, diabetes and hypertension. Here, we summarize the role of gut microbiota in host physiology as well as in the pathophysiology of the most common cardio-metabolic disorders, discussing the potential therapeutic opportunities offered by interventions aimed at modifying microbiome composition and activity.
- Published
- 2019
28. NAD
- Author
-
Dan, Tong, Gabriele G, Schiattarella, Nan, Jiang, Francisco, Altamirano, Pamela A, Szweda, Abdallah, Elnwasany, Dong I, Lee, Heesoo, Yoo, David A, Kass, Luke I, Szweda, Sergio, Lavandero, Eric, Verdin, Thomas G, Gillette, and Joseph A, Hill
- Subjects
Male ,Niacinamide ,Heart Failure, Diastolic ,Fatty Acids ,Down-Regulation ,Mitochondrial Myopathies ,Acetylation ,Ketone Oxidoreductases ,Pyridinium Compounds ,NAD ,Acyl-CoA Dehydrogenase ,Mitochondria, Heart ,Article ,Mice, Inbred C57BL ,Disease Models, Animal ,Mice ,Oxygen Consumption ,Sirtuin 3 ,Animals ,Humans ,Oxidation-Reduction - Abstract
[Figure: see text].
- Published
- 2021
29. Metabolism and Inflammation in Cardiovascular Health and Diseases: Mechanisms to Therapies
- Author
-
Gabriele G. Schiattarella, Joseph A. Hill, Rong Tian, Yibin Wang, Schiattarella, G. G., Wang, Y., Tian, R., and Hill, J. A.
- Subjects
business.industry ,Animal ,Cardiovascular health ,MEDLINE ,Disease Management ,Inflammation ,Bioinformatics ,Cardiovascular System ,Article ,Cardiovascular Physiological Phenomena ,Cardiovascular Diseases ,Cardiovascular Disease ,medicine ,Animals ,Humans ,Disease Susceptibility ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,business ,Energy Metabolism ,Molecular Biology ,Human - Published
- 2021
30. Early In Vivo Detection of Radiation-Induced Cardiotoxicity With Hyperpolarized C-13 Pyruvate Magnetic Resonance Spectroscopy
- Author
-
Junjie Ma, Jae Mo Park, E.R. Zhang-Velten, Prasanna G. Alluri, Xuliang Wang, Thomas G. Gillette, Joseph A. Hill, Jun Chen, J. Sharma, Gabriele G. Schiattarella, Craig R. Malloy, and Vlad G. Zaha
- Subjects
Cancer Research ,medicine.medical_specialty ,Cardiotoxicity ,Radiation ,Ejection fraction ,Heart disease ,business.industry ,Cellular respiration ,Metabolism ,Mitochondrion ,medicine.disease ,Citric acid cycle ,Oncology ,In vivo ,Internal medicine ,Cardiology ,medicine ,Radiology, Nuclear Medicine and imaging ,business - Abstract
Purpose/Objective(s) Radiation therapy is commonly used for treatment of patients with intrathoracic malignancies, such as lung cancer, esophageal cancer, and lymphoma. Radiation-Induced Heart Disease (RIHD) is a major source of morbidity and mortality in patients receiving thoracic radiation. Therefore, there is an acute need for development of non-invasive approaches for detection of RIHD at a stage that offers potential for early intervention and reversibility. Cardiac mitochondrial dysfunction is a hallmark of radiation-induced cardiac injury. During aerobic respiration, pyruvate enters the tricarboxylic acid cycle and is metabolized into bicarbonate in mitochondria. We hypothesized that radiation-induced mitochondrial dysfunction results in decreased conversion of pyruvate to bicarbonate in the mitochondria and increased conversion to lactate in cytosol. We sought to non-invasively assess radiation-induced changes in mitochondrial myocardial metabolism by tracking the fate of hyperpolarized (HP) C-13 pyruvate utilization using Magnetic Resonance Spectroscopy (MRS). Materials/Methods Sprague-Dawley rats (n = 10) underwent baseline echocardiography and HP C-13 pyruvate MRS. Rats in the cardiac radiation group (n = 5) underwent image-guided cardiac radiation with cone-beam CT, to a total dose of 40 Gy in 5 fractions. All rats underwent repeat echocardiography and hyperpolarized C-13 pyruvate MRS one week later. Results For the first time, we have demonstrated feasibility of employing HP C-13 pyruvate MRS for detecting radiation-induced myocardial mitochondrial metabolic changes in a pre-clinical rat model. In the cardiac radiation group, C-13 pyruvate MRS demonstrated a statistically significant decrease in cardiac bicarbonate-to-lactate ratio compared to pre-radiation baseline (P = 0.02, one-tailed paired t-test), suggesting increased metabolism of pyruvate into lactate in the cytoplasm (at the expense of metabolism into bicarbonate in the mitochondria) due to mitochondrial dysfunction. No significant decrease in this ratio was observed in the non-irradiated, age matched controls (P = 0.90). No significant changes in left ventricular ejection fraction or global longitudinal strain were observed in either the cardiac irradiation or control group of rats at this time point. Conclusion Radiation-induced myocardial mitochondrial dysfunction is an early event and can be detected in vivo by hyperpolarized C-13 pyruvate MRS within 1 week after radiation, and prior to onset of echocardiographic changes. Due to its non-invasive nature, this technology has the potential to serve as a platform for building radiation-focused cardio-oncology programs for early detection and mitigation of radiation-induced cardiac injury in hundreds of thousands of patients receiving thoracic radiation annually.
- Published
- 2021
31. NAD+Repletion Reverses Heart Failure with Preserved Ejection Fraction
- Author
-
Sergio Lavandero, Pamela A. Szweda, Joseph A. Hill, Luke I. Szweda, Heesoo Yoo, Abdallah Elnwasany, Francisco Altamirano, Gabriele G. Schiattarella, Nan Jiang, Dong I. Lee, Dan Tong, David A. Kass, Eric Verdin, Thomas G. Gillette, Tong, D., Schiattarella, G. G., Jiang, N., Altamirano, F., Szweda, P. A., Elnwasany, A., Lee, D. I., Yoo, H., Kass, D. A., Szweda, L. I., Lavandero, S., Verdin, E., Gillette, T. G., and Hill, J. A.
- Subjects
0301 basic medicine ,Male ,Niacinamide ,medicine.medical_specialty ,Physiology ,Cardiomyopathy ,heart failure ,Down-Regulation ,030204 cardiovascular system & hematology ,Ketone Oxidoreductase ,Acyl-CoA Dehydrogenase ,Mitochondria, Heart ,03 medical and health sciences ,Mice ,Pyridinium Compound ,0302 clinical medicine ,Mitochondrial Myopathie ,Oxygen Consumption ,Internal medicine ,Sirtuin 3 ,medicine ,Heart Failure, Diastolic ,Chemistry ,Animal ,Acetylation ,medicine.disease ,NAD ,mitochondria ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Heart failure ,Cardiology ,NAD+ kinase ,Cardiology and Cardiovascular Medicine ,Heart failure with preserved ejection fraction ,cardiomyopathy ,Oxidation-Reduction ,Fatty Acid ,Human - Abstract
Rationale: Heart failure with preserved ejection fraction (HFpEF) is a mortal clinical syndrome without effective therapies. We recently demonstrated in mice that a combination of metabolic and hypertensive stress recapitulates key features of human HFpEF. Objective: Using this novel preclinical HFpEF model, we set out to define and manipulate metabolic dysregulations occurring in HFpEF myocardium. Methods and Results: We observed impairment in mitochondrial fatty acid oxidation associated with hyperacetylation of key enzymes in the pathway. Downregulation of sirtuin 3 and deficiency of NAD + secondary to an impaired NAD + salvage pathway contribute to this mitochondrial protein hyperacetylation. Impaired expression of genes involved in NAD + biosynthesis was confirmed in cardiac tissue from patients with HFpEF. Supplementing HFpEF mice with nicotinamide riboside or a direct activator of NAD + biosynthesis led to improvement in mitochondrial function and amelioration of the HFpEF phenotype. Conclusions: Collectively, these studies demonstrate that HFpEF is associated with myocardial mitochondrial dysfunction and unveil NAD + repletion as a promising therapeutic approach in the syndrome.
- Published
- 2021
32. Canagliflozin and myocardial oxidative stress: SGLT1 inhibition takes centre stage
- Author
-
Gabriele G. Schiattarella, David Bode, Schiattarella, Gabriele G, and Bode, David
- Subjects
Canagliflozin ,medicine.medical_specialty ,business.industry ,Urology ,Medicine ,Stage (cooking) ,Cardiology and Cardiovascular Medicine ,business ,medicine.disease_cause ,Oxidative stress ,medicine.drug - Published
- 2021
33. Metabolic inflammation in heart failure with preserved ejection fraction
- Author
-
Joseph A. Hill, Daniele Rodolico, Gabriele G. Schiattarella, Schiattarella, G. G., Rodolico, D., and Hill, J. A.
- Subjects
Adult ,Male ,Metabolic inflammation ,Physiology ,Anti-Inflammatory Agents ,Reviews ,Cardiac metabolism ,Inflammation ,Disease ,Comorbidity ,Bioinformatics ,Ventricular Function, Left ,Pathogenesis ,Immunity ,Physiology (medical) ,medicine ,Humans ,Obesity ,Inflammation Mediator ,Heart Failure ,business.industry ,Myocardium ,Stroke Volume ,HFpEF ,Pathophysiology ,Anti-Inflammatory Agent ,Metabolism ,Female ,Inflammation Mediators ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,Heart failure with preserved ejection fraction ,business ,Energy Metabolism ,Human ,Signal Transduction - Abstract
One in 10 persons in the world aged 40 years and older will develop the syndrome of HFpEF (heart failure with preserved ejection fraction), the most common form of chronic cardiovascular disease for which no effective therapies are currently available. Metabolic disturbance and inflammatory burden contribute importantly to HFpEF pathogenesis. The interplay within these two biological processes is complex; indeed, it is now becoming clear that the notion of metabolic inflammation—metainflammation—must be considered central to HFpEF pathophysiology. Inflammation and metabolism interact over the course of syndrome progression, and likely impact HFpEF treatment and prevention. Here, we discuss evidence in support of a causal, mechanistic role of metainflammation in shaping HFpEF, proposing a framework in which metabolic comorbidities profoundly impact cardiac metabolism and inflammatory pathways in the syndrome.
- Published
- 2021
34. Abstract 13192: Xbp1s - Foxo1 Axis Governs Lipid Accumulation and Cardiac Performance in Heart Failure With Preserved Ejection Fraction
- Author
-
Theodore M. Hill, Stephen B. Spurgin, Herman I. May, Thomas G. Gillette, Esther Kim, Nan Jiang, Xuliang Wang, Mayaan Hotiner Waldman, Sergio Lavandero, Kristin M. French, Joseph A. Hill, Vlad G. Zaha, Gabriele G. Schiattarella, Dan Tong, Subhajit Dasgupta, Francisco Altamirano, Elisa Villalobos, Anwarul Ferdous, and Hande Piristine
- Subjects
medicine.medical_specialty ,Lipid accumulation ,business.industry ,Physiology (medical) ,Heart failure ,Internal medicine ,medicine ,Cardiology ,FOXO1 ,Cardiology and Cardiovascular Medicine ,medicine.disease ,business ,Heart failure with preserved ejection fraction - Abstract
Introduction: Heart failure with preserved ejection fraction (HFpEF) is now the dominant form of heart failure (HF). Limited insight into underlying mechanisms has culminated in the longstanding absence of evidence-based therapies capable of mitigating the substantial morbidity and mortality associated with the syndrome. Existing clinical and epidemiological evidence suggests that excessive body fat and lipid mishandling contribute to HFpEF. However, molecular mechanism(s) governing metabolic alterations and perturbations in lipid homeostasis in HFpEF are unknown. We recently developed a novel, clinically relevant, murine model of HFpEF, uncovering suppression of the Xbp1s (spliced form of the X-box-binding protein 1) arm of the UPR (unfolded protein response) signaling pathway as a critical driver of HFpEF pathogenesis. Objectives: To define and manipulate mechanisms downstream of Xbp1s in HFpEF and decipher its cardioprotective actions. Methods and Results: In the myocardium of experimental HFpEF, we detected cardiomyocyte steatosis coupled with increases in the abundance and activity of FoxO1 (Forkhead box protein O1), a conserved transcription factor involved in cell metabolism. FoxO1 depletion, as well as Xbp1s over-expression, in cardiomyocytes each ameliorated the HFpEF phenotype and reduced myocardial lipid accumulation. Strikingly, forced expression of Xbp1s in cardiomyocytes triggered proteasomal degradation of FoxO1. Furthermore, we discovered that FoxO1 is ubiquitinated upon Xbp1s over-expression, and Xbp1s-induced proteasomal degradation of FoxO1 occurs, in large part, through activation of the E3 ubiquitin ligase STUB1 (STIP1 homology and U-Box-containing protein 1), a protein we identified as a novel and direct transcriptional target of Xbp1s. Conclusions: Our findings uncover the Xbp1s-FoxO1 axis as a pivotal mechanism in the pathogenesis of HFpEF and unveil previously unrecognized mechanisms whereby the UPR governs metabolic alterations in cardiomyocytes.
- Published
- 2020
35. Transverse aortic constriction impairs intestinal barrier integrity, promotes inflammation and alterations in gut microbiota composition
- Author
-
G. Mattace Raso, Adriano Lama, Giovanni Esposito, Cinzia Perrino, S D'Apice, Lorena Coretti, Francesca Lembo, Maria Pina Mollica, Gabriele G. Schiattarella, Roberta Paolillo, and Nicola Boccella
- Subjects
biology ,business.industry ,Aortic constriction ,medicine ,Barrier integrity ,Inflammation ,medicine.symptom ,Gut flora ,Cardiology and Cardiovascular Medicine ,biology.organism_classification ,business ,Cell biology - Abstract
Background Microbiota composition plays an important role in the pathophysiology of heart failure (HF). Reduced cardiac output can disrupt intestinal barrier function and promote systemic inflammation through bacterial translocation. Several gut and cardiac pathological conditions are reciprocally linked at multiple levels and through different, still largely undefined mechanisms. Purpose We analyzed the effects of Transverse aortic constriction (TAC) on aortic pressures, gut barrier integrity, systemic inflammation and gut microbiota composition. Methods TAC was induced in C57BL6 mice of either sex. Sham-operated (Sham) mice were used as controls. After one-week (1w) and four weeks (4w), mice have been anesthetized, cardiac function and abdominal aortic blood flow were analyzed. Colon, serum and feces samples were collected after sacrifice. Intestinal barrier integrity was evaluated in colon samples by Tight junction protein ZO-1 (Tjp1) and Occludin (Ocln) mRNA analysis. Circulating levels of Tumor Necrosis Factor-alpha (TNF-alpha), Lipopolysaccharide (LPS), Interleukin-10 and Interleukin-1 were measured. Microbial DNA was extracted from feces samples and gut microbiota composition was evaluated by Illumina Mi-Seq analysis. Results TAC induced left ventricular hypertrophy and systolic dysfunction. Abdominal aortic blood flow was significantly reduced in TAC mice compared to sham (Figure 1A). Decreased intestinal perfusion in TAC mice was associated to a prompt and strong weakening of intestinal barrier integrity and long-lasting decrease of colonic anti-inflammatory cytokine levels, as shown by reduced mRNA expression of interleukin-10 (IL-10) and Occludin (Ocln) (Figure 1B). Serum levels of lipopolysaccharide (LPS) were increased after TAC surgery and significant increases of circulating proinflammatory cytokines tumor necrosis factor-a (TNF-a) were detected in TAC mice (Figure 1C). High-resolution approach was used to obtain bacterial species assignment of key genera with significant differences among groups. After TAC, significant increases of Bifidobacterium, Lactobacillus and Turicibacter, whereas the genus Oscillospira was significantly less (Figure 1D). Butyrate-producing bacteria are considered relevant colonizers of the gastrointestinal tract being butyrate important in anti-inflammation and maintaining intestinal barrier integrity. Oscillospira genus members have been described as butyrate producers. Notably, in old patients with heart failure and in animal models of hypertension, increase in lactate-producing Lactobacillus was found. Conclusions These data indicate a remodeling of specific bacterial species abundance within identified key genera starting soon after TAC, designating a clear effect of the treatment on microbiota profiles and, possibly, on microbiota functionality. Gut dysbiosis may represent an element to be considered in the development or progression of cardiac dysfunction. Figure 1 Funding Acknowledgement Type of funding source: Other. Main funding source(s): CP was supported by Ministero dell'Istruzione, Università e Ricerca Scientifica grant (2015583WMX) and Programma STAR grant by Federico II University and Compagnia di San Paolo. RP was supported by a research grant provided by the Cardiopath PhD program. LC was supported by 2018-2019 Postdoctoral Fellowship Grants provided by Fondazione Umberto Veronesi.
- Published
- 2020
36. Epigenetic Reader BRD4 (Bromodomain-Containing Protein 4) Governs Nucleus-Encoded Mitochondrial Transcriptome to Regulate Cardiac Function
- Author
-
Gabriele G. Schiattarella, Thaís A. R. Ramos, Soo Young Kim, Sergio Lavandero, Kristin M. French, Xin Zhang, Luke I. Szweda, Hongliang Li, Vinicius Maracaja-Coutinho, Herman I. May, Thomas G. Gillette, Xiang Luo, Pamela A. Szweda, Francisco Altamirano, Bret M. Evers, Joseph A. Hill, Nan Jiang, Kim, S. Y., Zhang, X., Schiattarella, G. G., Altamirano, F., Ramos, T. A. R., French, K. M., Jiang, N., Szweda, P. A., Evers, B. M., May, H. I., Luo, X., Li, H., Szweda, L. I., Maracaja-Coutinho, V., Lavandero, S., Gillette, T. G., and Hill, J. A.
- Subjects
Cardiac function curve ,Cardiomyopathy, Dilated ,BRD4 ,transcription, genetic ,Transcription Factor ,Mitochondrion ,Mitochondria, Heart ,Ventricular Function, Left ,Article ,Epigenesis, Genetic ,Transcriptome ,03 medical and health sciences ,Ventricular Dysfunction, Left ,0302 clinical medicine ,Transcription (biology) ,Physiology (medical) ,Medicine ,Animals ,Myocytes, Cardiac ,electron transport ,Epigenetics ,Cell Nucleu ,Nuclear Protein ,030304 developmental biology ,Cell Nucleus ,Heart Failure ,Mice, Knockout ,0303 health sciences ,Electron Transport Chain Complex Protein ,Animal ,business.industry ,Gene Expression Profiling ,Estrogen Receptor alpha ,Nuclear Proteins ,BRD4 protein, human ,Cell biology ,Bromodomain ,mitochondria ,medicine.anatomical_structure ,Electron Transport Chain Complex Proteins ,Cardiology and Cardiovascular Medicine ,business ,Energy Metabolism ,Nucleus ,epigenetic ,030217 neurology & neurosurgery ,Transcription Factors - Abstract
Background: BET (bromodomain and extraterminal) epigenetic reader proteins, in particular BRD4 (bromodomain-containing protein 4), have emerged as potential therapeutic targets in a number of pathological conditions, including cancer and cardiovascular disease. Small-molecule BET protein inhibitors such as JQ1 have demonstrated efficacy in reversing cardiac hypertrophy and heart failure in preclinical models. Yet, genetic studies elucidating the biology of BET proteins in the heart have not been conducted to validate pharmacological findings and to unveil potential pharmacological side effects. Methods: By engineering a cardiomyocyte-specific BRD4 knockout mouse, we investigated the role of BRD4 in cardiac pathophysiology. We performed functional, transcriptomic, and mitochondrial analyses to evaluate BRD4 function in developing and mature hearts. Results: Unlike pharmacological inhibition, loss of BRD4 protein triggered progressive declines in myocardial function, culminating in dilated cardiomyopathy. Transcriptome analysis of BRD4 knockout mouse heart tissue identified early and specific disruption of genes essential to mitochondrial energy production and homeostasis. Functional analysis of isolated mitochondria from these hearts confirmed that BRD4 ablation triggered significant changes in mitochondrial electron transport chain protein expression and activity. Computational analysis identified candidate transcription factors participating in the BRD4-regulated transcriptome. In particular, estrogen-related receptor α, a key nuclear receptor in metabolic gene regulation, was enriched in promoters of BRD4-regulated mitochondrial genes. Conclusions: In aggregate, we describe a previously unrecognized role for BRD4 in regulating cardiomyocyte mitochondrial homeostasis, observing that its function is indispensable to the maintenance of normal cardiac function.
- Published
- 2020
37. Left ventricular dysfunction in heart failure with preserved ejection fraction—molecular mechanisms and impact on right ventricular function
- Author
-
Gabriele G. Schiattarella, Florian Blaschke, Niklas Hegemann, Rudolf A. de Boer, Felix Hohendanner, Jana Grune, Uwe Primessnig, Frank R. Heinzel, Burkert Pieske, and Wolfgang M. Kuebler
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Ejection fraction ,business.industry ,Diastole ,Context (language use) ,030204 cardiovascular system & hematology ,medicine.disease ,Pulmonary hypertension ,Review Article on Right Ventricular Dysfunction ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Ion homeostasis ,Cardiovascular and Metabolic Diseases ,Internal medicine ,Heart failure ,medicine ,Cardiology ,Cardiology and Cardiovascular Medicine ,Heart failure with preserved ejection fraction ,business ,Kidney disease - Abstract
The current classification of heart failure (HF) based on left ventricular (LV) ejection fraction (EF) identifies a large group of patients with preserved ejection fraction (HFpEF) with significant morbidity and mortality but without prognostic benefit from current HF therapy. Co-morbidities and conditions such as arterial hypertension, diabetes mellitus, chronic kidney disease, adiposity and aging shape the clinical phenotype and contribute to mortality. LV diastolic dysfunction and LV structural remodeling are hallmarks of HFpEF, and are linked to remodeling of the cardiomyocyte and extracellular matrix. Pulmonary hypertension (PH) and right ventricular dysfunction (RVD) are particularly common in HFpEF, and mortality is up to 10-fold higher in HFpEF patients with vs. without RV dysfunction. Here, we review alterations in cardiomyocyte function (i.e., ion homeostasis, sarcomere function and cellular metabolism) associated with diastolic dysfunction and summarize the main underlying cellular pathways. The contribution and interaction of systemic and regional upstream signaling such as chronic inflammation, neurohumoral activation, and NO-cGMP-related pathways are outlined in detail, and their diagnostic and therapeutic potential is discussed in the context of preclinical and clinical studies. In addition, we summarize prevalence and pathomechanisms of RV dysfunction in the context of HFpEF and discuss mechanisms connecting LV and RV dysfunction in HFpEF. Dissecting the molecular mechanisms of LV and RV dysfunction in HFpEF may provide a basis for an improved classification of HFpEF and for therapeutic approaches tailored to the molecular phenotype.
- Published
- 2020
38. Impact of chronic kidney disease on platelet aggregation in patients with acute coronary syndrome
- Author
-
Gabriele G. Schiattarella, Sara Cimino, Carmine Morisco, Giuseppe Giugliano, Nicola Verde, Giuseppe Gargiulo, Enrico Coscioni, Giovanni Esposito, Eugenio Stabile, Roberta Paolillo, Marco Ferrone, Cinzia Perrino, Plinio Cirillo, Federica Ilardi, Ilardi, F., Gargiulo, G., Paolillo, R., Ferrone, M., Cimino, S., Giugliano, G., Schiattarella, G. G., Verde, N., Stabile, E., Perrino, C., Cirillo, P., Coscioni, E., Morisco, C., and Esposito, G.
- Subjects
Male ,medicine.medical_specialty ,Acute coronary syndrome ,Ticagrelor ,Platelet Aggregation ,Platelet Function Tests ,Drug Resistance ,Renal function ,Kidney ,Gastroenterology ,Risk Assessment ,Severity of Illness Index ,Coronary artery disease ,P2Y12 ,Percutaneous Coronary Intervention ,Risk Factors ,Internal medicine ,medicine ,Humans ,Platelet ,Acute Coronary Syndrome ,Renal Insufficiency, Chronic ,Aged ,Aspirin ,business.industry ,Dual Anti-Platelet Therapy ,General Medicine ,Odds ratio ,Middle Aged ,medicine.disease ,Clopidogrel ,Treatment Outcome ,Purinergic P2Y Receptor Antagonists ,Female ,Cardiology and Cardiovascular Medicine ,business ,Prasugrel Hydrochloride ,Platelet Aggregation Inhibitors ,medicine.drug ,Kidney disease ,Glomerular Filtration Rate - Abstract
AIMS: Chronic kidney disease (CKD) is associated with increased thrombotic events and seems to influence platelet reactivity. Conflicting results have been published on platelet response in CKD patients with stable coronary artery disease. The aim of our study was to investigate the impact of CKD on platelet aggregation in acute coronary syndrome (ACS) patients receiving dual antiplatelet therapy, included the more potent P2Y12 inhibitors. METHODS: We enrolled 206 patients with ACS, divided in two groups, according to the presence or the absence of moderate/severe CKD. Platelet aggregation was performed with light transmission aggregometry and results are expressed as percentage of maximum platelet aggregation. High residual platelet reactivity (HRPR) was defined as maximum platelet aggregation more than 59%. RESULTS: Patients with CKD [estimate glomerular filtration rate (eGFR)
- Published
- 2020
39. The role of mitochondrial dynamics in cardiovascular diseases
- Author
-
Sebastiano Sciarretta, Gabriele G. Schiattarella, Leonardo Schirone, Lia Crotti, Cinzia Perrino, Daniele Catalucci, Ciro Indolfi, Jessica Modica, Maurizio Forte, Daniele Torella, Speranza Rubattu, Cristina Basso, Cristina Chimenti, Giacomo Frati, Pietro Ameri, Forte, M, Schirone, L, Ameri, P, Basso, C, Catalucci, D, Modica, J, Chimenti, C, Crotti, L, Frati, G, Rubattu, S, Schiattarella, G, Torella, D, Perrino, C, Indolfi, C, Sciarretta, S, Forte, M., Schirone, L., Ameri, P., Basso, C., Catalucci, D., Modica, J., Chimenti, C., Crotti, L., Frati, G., Rubattu, S., Schiattarella, G. G., Torella, D., Perrino, C., Indolfi, C., and Sciarretta, S.
- Subjects
0301 basic medicine ,Ischemia ,Disease ,Mitochondrion ,Mitochondrial Dynamics ,Brain Ischemia ,03 medical and health sciences ,0302 clinical medicine ,cardiovascular disease ,medicine ,Humans ,Pharmacology ,Pressure overload ,BIO/15 - BIOLOGIA FARMACEUTICA ,mitochondria ,mitochondrial dynamics ,fission ,fusion ,cardiovascular ,heart ,myocardium ,myocardial ,drp1 ,mfn1 ,mfn2 ,opa1 ,business.industry ,Myocardium ,MED/11 - MALATTIE DELL'APPARATO CARDIOVASCOLARE ,medicine.disease ,Pathophysiology ,Stroke ,030104 developmental biology ,Cardiovascular Diseases ,Heart failure ,business ,Reperfusion injury ,Neuroscience ,030217 neurology & neurosurgery ,Biogenesis - Abstract
The process of mitochondrial dynamics is emerging as a core player in cardiovascular homeostasis. This process refers to the co-ordinated cycles of biogenesis, fusion, fission and degradation to which mitochondria constantly undergo to maintain their integrity, distribution and size. These mechanisms represent an early response to mitochondrial stress, confining organelle portions that are irreversibly damaged and preserving mitochondrial function. Accumulating evidence demonstrates that impairment in mitochondrial dynamics leads to myocardial damage and cardiac disease progression in a variety of disease models, including pressure overload, ischaemia/reperfusion and metabolic disturbance. These findings suggest that modulation of mitochondrial dynamics may be considered as a valid therapeutic strategy in cardiovascular diseases. In this review, we discuss the current evidence about the role of mitochondrial dynamics in cardiac pathophysiology, with a particular focus on the mechanisms underlying the development of cardiac hypertrophy and heart failure, metabolic and genetic cardiomyopathies, ischaemia/reperfusion injury, atherosclerosis and ischaemic stroke. LINKED ARTICLES: This article is part of a themed issue on Cellular metabolism and diseases. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.10/issuetoc.
- Published
- 2020
40. Distinctive patterns of inflammation across the heart failure syndrome
- Author
-
Pietro Ameri, Gabriele G. Schiattarella, Vasco Sequeira, Schiattarella, G. G., Sequeira, V., and Ameri, P.
- Subjects
Prognosi ,Inflammation ,Pathogenesis ,030204 cardiovascular system & hematology ,Bioinformatics ,Ventricular Function, Left ,Comorbidities ,03 medical and health sciences ,Ventricular Dysfunction, Left ,0302 clinical medicine ,Pathogenesi ,medicine ,Humans ,In patient ,030212 general & internal medicine ,Myocardial infarction ,Cytokines ,Heart failure ,Cytokine ,Heart Failure ,Ejection fraction ,business.industry ,Cancer ,Stroke Volume ,medicine.disease ,Prognosis ,Comorbiditie ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,business ,Human ,Kidney disease - Abstract
Inflammation has long been known to play a role in heart failure (HF). Earlier studies demonstrated that inflammation contributes to the pathogenesis of HF with reduced ejection fraction (HFrEF), and the knowledge about molecules and cell types specifically involved in inflammatory events has been constantly increased ever since. However, conflicting results of several trials with anti-inflammatory treatments led to the conclusions that inflammation does participate in the progression of HFrEF, but more likely it is not the primary event. Conversely, it has been suggested that inflammation drives the development of HF with preserved ejection fraction (HFpEF). Recently the pharmacological blockade of interleukin-1 has been shown to prevent HF hospitalization and mortality in patients with prior myocardial infarction, lending renewed support to the hypothesis that inflammation is a promising therapeutic target in HF. Inflammation has also been proposed to underlie both HF and commonly associated conditions, such as chronic kidney disease or cancer. Within this last paradigm, an emergent role has been ascribed to clonal hematopoiesis of indeterminate potential. Here, we summarize the recent evidence about the role of inflammation in HF, highlighting the similarities and differences in HFrEF vs. HFpEF, and discuss the diagnostic and therapeutic opportunities raised by antinflammatory-based approaches.
- Published
- 2020
41. FoxO1–Dio2 signaling axis governs cardiomyocyte thyroid hormone metabolism and hypertrophic growth
- Author
-
Sergio Lavandero, Anwarul Ferdous, Annie Nguyen, Gabriele G. Schiattarella, Xiang Luo, Yuxuan Luo, Zhao V. Wang, Herman I. May, Thomas G. Gillette, Francisco Altamirano, Beverly A. Rothermel, Pavan K. Battiprolu, Joseph A. Hill, Dan L. Li, Ferdous, A., Wang, Z. V., Luo, Y., Li, D. L., Luo, X., Schiattarella, G. G., Altamirano, F., May, H. I., Battiprolu, P. K., Nguyen, A., Rothermel, B. A., Lavandero, S., Gillette, T. G., and Hill, J. A.
- Subjects
0301 basic medicine ,Thyroid Hormones ,endocrine system ,Science ,General Physics and Astronomy ,DIO2 ,Cardiomegaly ,FOXO1 ,030204 cardiovascular system & hematology ,Biology ,Iodide Peroxidase ,Article ,General Biochemistry, Genetics and Molecular Biology ,Mice ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Myocytes, Cardiac ,lcsh:Science ,Ventricular remodeling ,Cells, Cultured ,Mice, Knockout ,Multidisciplinary ,Ventricular Remodeling ,Forkhead Box Protein O1 ,FOXO Family ,General Chemistry ,medicine.disease ,Rats ,Cell biology ,Thyroid diseases ,Cardiac hypertrophy ,030104 developmental biology ,Animals, Newborn ,Gene Expression Regulation ,Cardiovascular and Metabolic Diseases ,cardiovascular system ,FOXO3 ,lcsh:Q ,Signal transduction ,hormones, hormone substitutes, and hormone antagonists ,Homeostasis ,Signal Transduction ,Hormone - Abstract
Forkhead box O (FoxO) proteins and thyroid hormone (TH) have well established roles in cardiovascular morphogenesis and remodeling. However, specific role(s) of individual FoxO family members in stress-induced growth and remodeling of cardiomyocytes remains unknown. Here, we report that FoxO1, but not FoxO3, activity is essential for reciprocal regulation of types II and III iodothyronine deiodinases (Dio2 and Dio3, respectively), key enzymes involved in intracellular TH metabolism. We further show that Dio2 is a direct transcriptional target of FoxO1, and the FoxO1–Dio2 axis governs TH-induced hypertrophic growth of neonatal cardiomyocytes in vitro and in vivo. Utilizing transverse aortic constriction as a model of hemodynamic stress in wild-type and cardiomyocyte-restricted FoxO1 knockout mice, we unveil an essential role for the FoxO1–Dio2 axis in afterload-induced pathological cardiac remodeling and activation of TRα1. These findings demonstrate a previously unrecognized FoxO1–Dio2 signaling axis in stress-induced cardiomyocyte growth and remodeling and intracellular TH homeostasis., Disease stress-induced cardiac hypertrophy is a major mechanism of pathological cardiac remodeling. Here, the authors unveil a previously unrecognized role of a FoxO1–Dio2 signaling axis in maladaptive, afterload-induced cardiac hypertrophy and intracellular thyroid hormone homeostasis.
- Published
- 2020
42. Can hfpef and hfref coexist?
- Author
-
Gabriele G. Schiattarella, Dan Tong, Joseph A. Hill, Schiattarella, G. G., Tong, D., and Hill, J. A.
- Subjects
medicine.medical_specialty ,Ejection fraction ,Treatment outcome ,Protease Inhibitor ,MEDLINE ,Angiotensin-Converting Enzyme Inhibitors ,Article ,Ventricular Function, Left ,Paradigm hf ,Angiotensin Receptor Antagonists ,Risk Factors ,Physiology (medical) ,Internal medicine ,Medicine ,Humans ,Hfref ,Protease Inhibitors ,Hfpef ,Paragon-hf ,Heart Failure ,Ventricular function ,Extramural ,business.industry ,Arni ,Risk Factor ,Angiotensin Receptor Antagonist ,Hfmref ,Angiotensin-Converting Enzyme Inhibitor ,Stroke Volume ,Recovery of Function ,Paradigm-hf ,Phenotype ,Treatment Outcome ,Cardiology ,Neprilysin ,Cardiology and Cardiovascular Medicine ,business ,Human - Published
- 2020
43. Feeding Diastolic Dysfunction: Is It a Bug?
- Author
-
Gabriele G. Schiattarella, Giovanni Esposito, Esposito, G., and Schiattarella, G. G.
- Subjects
medicine.medical_specialty ,Heart Diseases ,Fibrosi ,Diastole ,Inflammation ,TMAO ,Choline ,Mice ,Internal medicine ,medicine ,microbiota ,Animals ,Heart Failure ,business.industry ,Animal ,Stroke Volume ,medicine.disease ,HFpEF ,Fibrosis ,Diet ,Heart Disease ,Heart failure ,mouse models of cardiovascular disease ,Cardiology ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,business - Published
- 2020
44. Cytosolic DNA Sensing Promotes Macrophage Transformation and Governs Myocardial Ischemic Injury
- Author
-
Zhijian J. Chen, Elisa Villalobos, Tuo Li, Gabriele G. Schiattarella, Dian J. Cao, Herman I. May, Thomas G. Gillette, Joseph A. Hill, and Nan Jiang
- Subjects
0301 basic medicine ,Angiogenesis ,Myocardial Infarction ,Article ,Mice ,03 medical and health sciences ,Cytosol ,Immune system ,Physiology (medical) ,medicine ,Animals ,Macrophage ,Ventricular remodeling ,Receptor ,Innate immune system ,Ventricular Remodeling ,biology ,business.industry ,Macrophages ,Myocardium ,Pattern recognition receptor ,DNA ,medicine.disease ,Nucleotidyltransferases ,Cell biology ,Nitric oxide synthase ,030104 developmental biology ,biology.protein ,Cardiology and Cardiovascular Medicine ,business ,Signal Transduction - Abstract
Background: Myocardium irreversibly injured by ischemic stress must be efficiently repaired to maintain tissue integrity and contractile performance. Macrophages play critical roles in this process. These cells transform across a spectrum of phenotypes to accomplish diverse functions ranging from mediating the initial inflammatory responses that clear damaged tissue to subsequent reparative functions that help rebuild replacement tissue. Although macrophage transformation is crucial to myocardial repair, events governing this transformation are poorly understood. Methods: Here, we set out to determine whether innate immune responses triggered by cytoplasmic DNA play a role. Results: We report that ischemic myocardial injury, along with the resulting release of nucleic acids, activates the recently described cyclic GMP-AMP synthase–stimulator of interferon genes pathway. Animals lacking cyclic GMP-AMP synthase display significantly improved early survival after myocardial infarction and diminished pathological remodeling, including ventricular rupture, enhanced angiogenesis, and preserved ventricular contractile function. Furthermore, cyclic GMP-AMP synthase loss of function abolishes the induction of key inflammatory programs such as inducible nitric oxide synthase and promotes the transformation of macrophages to a reparative phenotype, which results in enhanced repair and improved hemodynamic performance. Conclusions: These results reveal, for the first time, that the cytosolic DNA receptor cyclic GMP-AMP synthase functions during cardiac ischemia as a pattern recognition receptor in the sterile immune response. Furthermore, we report that this pathway governs macrophage transformation, thereby regulating postinjury cardiac repair. Because modulators of this pathway are currently in clinical use, our findings raise the prospect of new treatment options to combat ischemic heart disease and its progression to heart failure.
- Published
- 2018
45. Microbial metabolites as predictive biomarkers: a paradigm shift for cardiovascular risk stratification
- Author
-
Bruno Trimarco, Gabriele G. Schiattarella, Schiattarella, G. G., and Trimarco, B.
- Subjects
Oncology ,medicine.medical_specialty ,Prognosi ,business.industry ,Lysine ,Risk Factor ,Biomarker ,Stratification (mathematics) ,Methylamine ,Cardiovascular Diseases ,Paradigm shift ,Internal medicine ,Risk stratification ,medicine ,Acute Coronary Syndrome ,Cardiology and Cardiovascular Medicine ,business ,Human ,Predictive biomarker - Published
- 2019
46. Double layered stents for carotid angioplasty: A meta-analysis of available clinical data
- Author
-
Tullio Tesorio, Evelina Toscano, Gabriele G. Schiattarella, Bruno Trimarco, Anna Franzone, Anna Sannino, Giovanni Esposito, Eugenio Stabile, Giuseppe Giugliano, Sannino, Anna, Giugliano, Giuseppe, Toscano, Evelina, Schiattarella, Gabriele G., Franzone, Anna, Tesorio, Tullio, Trimarco, Bruno, Esposito, Giovanni, and Stabile, Eugenio
- Subjects
Male ,medicine.medical_specialty ,Time Factors ,medicine.medical_treatment ,Intervention ,Subgroup analysis ,030204 cardiovascular system & hematology ,Prosthesis Design ,03 medical and health sciences ,0302 clinical medicine ,Risk Factors ,Clinical endpoint ,Humans ,Medicine ,Carotid Stenosis ,Radiology, Nuclear Medicine and imaging ,cardiovascular diseases ,030212 general & internal medicine ,Adverse effect ,Stroke ,Aged ,Evidence-Based Medicine ,Carotid artery disease ,business.industry ,Incidence ,Stent design/structure/coating ,Mortality rate ,Angioplasty ,Stent ,General Medicine ,medicine.disease ,Stenosis ,Treatment Outcome ,Meta-analysis ,Female ,Stents ,Radiology ,Cardiology and Cardiovascular Medicine ,business - Abstract
OBJECTIVES The objective of this meta-analysis is to evaluate clinical efficacy of double layered mesh covered carotid stent systems in the clinical practice. BACKGROUND The need for an increase plaque coverage to decrease the risk of debris dislodgement through the stent struts, following carotid artery stenting (CAS), has brought to the design of a new generation of double layered carotid stents. Several small sized clinical studies evaluating two different devices have been recently published, unfortunately these are not sufficiently powered to test for device related and clinical endpoints and no comparison, between the two available devices, has been reported yet. METHODS Ten studies, enrolling 635 patients, were included in the present meta-analysis. Our study analyzed a composite endpoint of 30-day stroke and death and the occurrence of procedural unsuccess after CAS with the use of two different double layered carotid stent systems. RESULTS Thirty-day stroke and death rate was quite low (patients 635, event rate 0.02, 95% CI: 0.01-0.04, P
- Published
- 2017
47. Epigenetic control of lipid metabolism: implications for lifespan and healthspan
- Author
-
Gabriele G. Schiattarella and Joseph A. Hill
- Subjects
0301 basic medicine ,biology ,Physiology ,Extramural ,business.industry ,media_common.quotation_subject ,Longevity ,Lipid metabolism ,Computational biology ,biology.organism_classification ,03 medical and health sciences ,030104 developmental biology ,Physiology (medical) ,Medicine ,Epigenetics ,Cardiology and Cardiovascular Medicine ,business ,Caenorhabditis elegans ,Epigenesis ,media_common - Published
- 2018
48. Obesity induces cardiac hypertrophy without functional or fibrotic alterations
- Author
-
Sergio Lavandero, Alfredo Criollo, Joseph A. Hill, Francisco Altamirano, Gabriele G. Schiattarella, Elisa Villalobos, and Thomas G. Gillette
- Subjects
medicine.medical_specialty ,business.industry ,Cardiac hypertrophy ,Internal medicine ,medicine ,Cardiology ,medicine.disease ,business ,Obesity - Published
- 2019
49. Abstract 867: NAD + Repletion Reverses HFpEF by Attenuating Myocardial Metabolic Dysfunction
- Author
-
David A. Kass, Pamela A. Szweda, Abdallah Elnwasany, Nan Jiang, Francisco Altamirano, Luke I. Szweda, Gabriele G. Schiattarella, Thomas G. Gillette, Joseph A. Hill, Dong I. Lee, and Dan Tong
- Subjects
medicine.medical_specialty ,Physiology ,business.industry ,Internal medicine ,Heart failure ,Cardiology ,Medicine ,NAD+ kinase ,Cardiology and Cardiovascular Medicine ,business ,Heart failure with preserved ejection fraction ,medicine.disease - Abstract
Background: Heart failure with preserved ejection fraction (HFpEF) is a highly prevalent clinical condition associated with significant morbidity, mortality and health care expenses. Yet, no effective treatment has been identified. We recently demonstrated that concomitant metabolic and hypertensive stress in mice elicited by a combination of high fat diet (HFD) and constitutive nitric oxide synthase inhibition by N [w] -nitro-l-arginine methyl ester (L-NAME) faithfully recapitulates the numerous and myriad features of human HFpEF (paper in press at Nature ). Methods: Wild type C57Bl6 mice were fed with HFD and L-NAME via drinking water for 5-8 weeks. Myocardial mitochondrial morphology was assessed by electron microscopy. Mitochondrial function was assessed by measuring oxygen consumption rates using an oxygen electrode. Protein expression and modification were assessed by Western blotting and immunoprecipitation. Results: Using this novel mouse model, we discovered significant impairment of mitochondrial fatty acid oxidation (FAO) associated with hyperacetylation of key FAO enzymes in HFpEF myocardium. Mechanistically, downregulation of Sirtuin3, the major mitochondrial deacetylase, and deficiency of its co-substrate nicotinamide adenine dinucleotide (NAD + ), culminated in mitochondrial protein hyperacetylation. Strikingly, supplementation with nicotinamide riboside (NR), a NAD + precursor, led to dramatic improvement of mitochondrial function, and importantly, amelioration of the HFpEF phenotype. Conclusion: In summary, we have unveiled that protein hyperacetylation-mediated mitochondrial dysfunction is a crucial mechanism of HFpEF pathogenesis. This is, to our knowledge, the first study identifying a specific signature of metabolic remodeling in HFpEF heart. We also demonstrated the therapeutic effect of NAD + repletion in a preclinical HFpEF model. In next steps, confirming this benefit in a clinical trial is warranted.
- Published
- 2019
50. Abstract 190: Polycystin-1 Assembles With Kv Channels to Govern Cardiomyocyte Repolarization and Contractility
- Author
-
Sergio Lavandero, Kristin M. French, Gabriele G. Schiattarella, César A. Ramírez-Sarmiento, Felipe Engelberger, Jay W. Schneider, Soo Young Kim, Thomas G. Gillette, Sergii Kyrychenko, Francisco Altamirano, Elisa Villalobos, Dan Tong, and Joseph A. Hill
- Subjects
Polycystin-1 ,endocrine system ,Physiology ,Autosomal dominant polycystic kidney disease ,chemistry.chemical_element ,Biology ,Calcium ,medicine.disease ,Cell biology ,Kv channel ,Contractility ,Electrophysiology ,chemistry ,medicine ,Repolarization ,Cardiology and Cardiovascular Medicine ,Gene - Abstract
Mutations in the gene encoding polycystin-1 (PC1) underlie autosomal dominant polycystic kidney disease (ADPKD). ADPKD patients present with multiple cardiovascular co-morbidities believed to be caused by renal dysfunction. LV hypertrophy and diastolic dysfunction can manifest during childhood or in young adults prior to a formal diagnosis of hypertension, and evidence suggests that LV function is impaired in ADPKD patients with normal or moderately reduced kidney function. These facts suggest that cardiomyocyte-autonomous effects may contribute to the cardiovascular abnormalities seen in ADPKD. Contractile function (systolic and diastolic) measured by echo was significantly reduced in PC1 cKO ( Pkd1 F/F ;αMHC-Cre) mice compared with controls ( Pkd1 F/F ). PC1 cKO cardiomyocytes manifest impaired contractility and smaller and slower Ca 2+ transients. Using a multidimensional approach, we discovered that cardiomyocytes lacking PC1 have shorter action potentials (APD50/90) and decreased SERCA activity. These alterations impair EC-coupling and decrease SR Ca 2+ loading during pacing. Remarkably, square pulses under voltage clamp (-80 to +10 mV) produced Ca 2+ transients with similar amplitude between genotypes, which highlights that alterations in action potential (AP) duration drive most of the EC-coupling changes. PC1-deficient cardiomyocytes manifested an increase in outward K + currents (I to , I Kslow1/2 and I ss ) but not in inward currents (I K1 ). PC1 over-expression in HEK293T cells reduced the currents of heterologously expressed Kv4.3/2.1/1.5 channels. The inhibitory effects of PC1 on Kv4.3 currents were mediated by PC1-CT (C-terminus) through its coiled-coil domain (CCD). Interestingly, a naturally occurring human mutant PC1 R4228X , located in the CCD, manifested no suppressive effects on Kv4.3 channel. Finally, to begin to test for relevance to human pathology, we found that PC1 ablation reduces AP duration, and PC1-CT over-expression had the opposite effect in human stem cell-derived cardiomyocytes. Our findings uncover a novel role for PC1 controlling action potential duration and SERCA. PC1-deficient cardiomyocytes manifest impaired contractility, likely contributing to contractile dysfunction in ADPKD patients.
- Published
- 2019
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.