83 results on '"Youker KA"'
Search Results
2. Reciprocal regulation of myocardial microRNAs and messenger RNA in human cardiomyopathy and reversal of the microRNA signature by biomechanical support.
- Author
-
Matkovich SJ, Van Booven DJ, Youker KA, Torre-Amione G, Diwan A, Eschenbacher WH, Dorn LE, Watson MA, Margulies KB, Dorn GW 2nd, Matkovich, Scot J, Van Booven, Derek J, Youker, Keith A, Torre-Amione, Guillermo, Diwan, Abhinav, Eschenbacher, William H, Dorn, Lisa E, Watson, Mark A, Margulies, Kenneth B, and Dorn, Gerald W 2nd
- Published
- 2009
- Full Text
- View/download PDF
3. Rad GTPase deficiency leads to cardiac hypertrophy.
- Author
-
Chang L, Zhang J, Tseng YH, Xie CQ, Ilany J, Brüning JC, Sun Z, Zhu X, Cui T, Youker KA, Yang Q, Day SM, Kahn CR, Chen YE, Chang, Lin, Zhang, Jifeng, Tseng, Yu-Hua, Xie, Chang-Qing, Ilany, Jacob, and Brüning, Jens C
- Published
- 2007
- Full Text
- View/download PDF
4. Inhibitory cardiac transcription factor, SRF-N, is generated by caspase 3 cleavage in human heart failure and attenuated by ventricular unloading.
- Author
-
Chang J, Wei L, Otani T, Youker KA, Entman ML, Schwartz RJ, Kitsis RN, Chang, Jiang, Wei, Lei, Otani, Takayuki, Youker, Keith A, Entman, Mark L, and Schwartz, Robert J
- Published
- 2003
5. Myocardial Recovery versus Myocardial Regeneration: Mechanisms and Therapeutic Modulation.
- Author
-
Cooke JP, Youker KA, and Lai L
- Subjects
- Humans, Animals, Heart Failure physiopathology, Heart Failure therapy, Myocytes, Cardiac pathology, Myocytes, Cardiac metabolism, Mitochondria, Heart metabolism, Mitochondria, Heart pathology, Regeneration, Recovery of Function, Myocardium pathology, Myocardium metabolism
- Abstract
Myocardial recovery is characterized by a return toward normal structure and function of the heart after an injury. Mechanisms of myocardial recovery include restoration and/or adaptation of myocyte structure and function, mitochondrial activity and number, metabolic homeostasis, electrophysiological stability, extracellular matrix remodeling, and myocardial perfusion. Myocardial regeneration is an element of myocardial recovery that involves the generation of new myocardial tissue, a process which is limited in adult humans but may be therapeutically augmented. Understanding the mechanisms of myocardial recovery and myocardial regeneration will lead to novel therapies for heart failure., Competing Interests: Drs. Cooke and Lai have invention disclosures related to vascular regeneration that are assigned to Houston Methodist Hospital., (Copyright: © 2024 The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
6. Future Impact of mRNA Therapy on Cardiovascular Diseases.
- Author
-
Cooke JP and Youker KA
- Subjects
- Humans, RNA, Messenger genetics, Clinical Trials, Phase II as Topic, Cardiovascular Diseases diagnosis, Cardiovascular Diseases genetics, Cardiovascular Diseases therapy, Coronary Artery Disease, Heart Failure, Myocardial Ischemia
- Abstract
The silver lining of the recent pandemic was that it accelerated the emergence of messenger ribonucleic acid (mRNA) therapeutics. The great promise of mRNA therapeutics was highlighted by the speed at which the vaccines were created, tested, and proven to be relatively safe and highly effective. There are a wide variety of mRNA therapeutics now under development, and dozens of these are in clinical trials. These therapeutics are generating a major paradigm shift in medical therapy, including the treatment of cardiovascular disease. Most of the cardiovascular mRNA therapies are still in preclinical development, although a phase 2a trial of mRNA therapy for myocardial ischemia has been completed with promising results.
1 The application of mRNA therapies to cardiovascular diseases is virtually limitless, and ongoing work includes mRNA therapies for myocardial ischemia, heart failure, arrhythmias, hypercholesterolemia, and arterial occlusive diseases. In addition, mRNA may be used to enhance cell therapies. In the future, mRNA therapies for cardiovascular disease are destined to supplant some of our current biologics and pharmacotherapies and will be used to treat previously untreatable cardiovascular diseases. Furthermore, mRNA therapies can be personalized, and they can be rapidly generated in current Good Manufacturing Practice facilities with a modest footprint, facilitating the rise of hospital-based regional centers of RNA therapeutics., Competing Interests: Dr. Cooke is a member of the Scientific Advisory Board for Humann, the founder of Cooke Consulting, and has a sponsored research agreement with VGXI Inc. He is the inventor of related mRNA technologies assigned to Stanford University and Houston Methodist Hospital, some of which are licensed to ChromeX Bio, founded by Dr. Cooke. Dr. Youker has no competing interests to declare., (Copyright: © 2022 The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
7. An In Vitro Platform to Study Reversible Endothelial-to-Mesenchymal Transition.
- Author
-
Krishnamoorthi MK, Thandavarayan RA, Youker KA, and Bhimaraj A
- Abstract
Endothelial cells can acquire a mesenchymal phenotype in response to external stimuli through both mechanical and biological factors, using a process known as endothelial-to-mesenchymal (EndoMT) transition. EndoMT is characterized by the decrease in endothelial characteristics, increase in mesenchymal markers, and morphological changes. It has been recognized not only during development but also in different pathological conditions including organ/tissue fibrosis in adults. The ability to modulate the EndoMT process could have a therapeutic potential in many fibrotic diseases. An in vitro method is presented here to induce EndoMT with Nω-nitro-L-arginine methyl ester hydrochloride (L-NAME) and angiotensin II (Ang II) followed by a protocol to study the reversibility of EndoMT. Using this method, we furnish evidence that the combination of L-NAME and Ang II can stimulate EndoMT in Human umbilical vascular endothelial cells (HUVECs) and this process can be reversed as observed using endothelial functionality assays. This method may serve as a model to screen and identify potential pharmacological molecules to target and regulate the EndoMT process, with applications in drug discovery for human diseases., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Krishnamoorthi, Thandavarayan, Youker and Bhimaraj.)
- Published
- 2022
- Full Text
- View/download PDF
8. STK35 Gene Therapy Attenuates Endothelial Dysfunction and Improves Cardiac Function in Diabetes.
- Author
-
Joladarashi D, Zhu Y, Willman M, Nash K, Cimini M, Thandavarayan RA, Youker KA, Song X, Ren D, Li J, Kishore R, Krishnamurthy P, and Wang L
- Abstract
Diabetic cardiomyopathy (DCM) is characterized by microvascular pathology and interstitial fibrosis that leads to progressive heart failure. The mechanisms underlying DCM pathogenesis remain obscure, and no effective treatments for the disease have been available. In the present study, we observed that STK35, a novel kinase, is decreased in the diabetic human heart. High glucose treatment, mimicking hyperglycemia in diabetes, downregulated STK35 expression in mouse cardiac endothelial cells (MCEC). Knockdown of STK35 attenuated MCEC proliferation, migration, and tube formation, whereas STK35 overexpression restored the high glucose-suppressed MCEC migration and tube formation. Angiogenesis gene PCR array analysis revealed that HG downregulated the expression of several angiogenic genes, and this suppression was fully restored by STK35 overexpression. Intravenous injection of AAV9-STK35 viral particles successfully overexpressed STK35 in diabetic mouse hearts, leading to increased vascular density, suppression of fibrosis in the heart, and amelioration of left ventricular function. Altogether, our results suggest that hyperglycemia downregulates endothelial STK35 expression, leading to microvascular dysfunction in diabetic hearts, representing a novel mechanism underlying DCM pathogenesis. Our study also emerges STK35 is a novel gene therapeutic target for preventing and treating DCM., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Joladarashi, Zhu, Willman, Nash, Cimini, Thandavarayan, Youker, Song, Ren, Li, Kishore, Krishnamurthy and Wang.)
- Published
- 2022
- Full Text
- View/download PDF
9. Endothelial Dysfunction-related Neurological Bleeds with Continuous Flow-Left Ventricular Assist Devices Measured by Digital Thermal Monitor.
- Author
-
Ali A, Uribe C, Araujo-Gutierrez R, Cruz-Solbes AS, Marcos-Abdala HG, Youker KA, Guha A, Torre-Amione G, Nguyen DT, Graviss EA, Cooke JP, and Bhimaraj A
- Subjects
- Aged, Cross-Sectional Studies, Female, Heart Failure physiopathology, Humans, Male, Middle Aged, Vasodilation physiology, Endothelium, Vascular physiology, Heart-Assist Devices adverse effects, Hemorrhage etiology
- Abstract
Endothelial dysfunction has been demonstrated in patients with Continuous Flow-Left Ventricular Assist Devices (CF-LVADs) but association with adverse events has not been shown. We used a noninvasive, operator-independent device called VENDYS® to assess vasodilatory function based on digital thermal measurements postrelease of a brachial artery occlusion in ambulatory patients with CF-LVAD (n = 56). Aortic valve opening and pulse perception were also documented before the test. Median duration of CF-LVAD support was 438 days. The VENDYS® test generates a vascular reactivity index (VRI). Outcomes for the CF-LVAD patients were compared between VRI < 1 and VRI ≥ 1. The bleeding events were driven primarily by a difference in neurologic bleeds. Multivariate analysis showed that VRI < 1 correlated with future bleeding events (HR: 5.56; P = 0.01). The C-statistic with the VRI dichotomized as above was 0.82. There was a trend toward a worse survival in patients with poor endothelial function. Endothelial vasodilatory dysfunction measured by a simple test utilizing digital thermal monitoring can predict adverse bleeding events in patients with CF-LVADs., Competing Interests: Disclosure: Drs. Guha and Bhimaraj have consulting agreements with Abbott, the current maker of Heartmate-II LVADs. The other authors have no conflicts of interest to report., (Copyright © ASAIO 2020.)
- Published
- 2021
- Full Text
- View/download PDF
10. Erratum for Kirkpatrick et al., "Inducible Lung Epithelial Resistance Requires Multisource Reactive Oxygen Species Generation To Protect against Viral Infections".
- Author
-
Kirkpatrick CT, Wang Y, Leiva Juarez MM, Shivshankar P, Pantaleón García J, Plumer AK, Kulkarni VV, Ware HH, Gulraiz F, Chavez Cavasos MA, Martinez Zayas G, Wali S, Rice AP, Liu H, Tour JM, Sikkema WKA, Cruz Solbes AS, Youker KA, Tuvim MJ, Dickey BF, and Evans SE
- Published
- 2019
- Full Text
- View/download PDF
11. Small molecule disruption of G protein βγ subunit signaling reprograms human macrophage phenotype and prevents autoimmune myocarditis in rats.
- Author
-
Karuppagounder V, Bajpai A, Meng S, Arumugam S, Sreedhar R, Giridharan VV, Guha A, Bhimaraj A, Youker KA, Palaniyandi SS, Karmouty-Quintana H, Kamal F, Spiller KL, Watanabe K, and Thandavarayan RA
- Subjects
- Animals, Autoimmune Diseases metabolism, G-Protein-Coupled Receptor Kinase 2 metabolism, HMGB1 Protein metabolism, Heart Failure metabolism, Heart Failure prevention & control, Humans, Macrophage Activation drug effects, Macrophages classification, Macrophages metabolism, Male, Myocarditis metabolism, Rats, Inbred Lew, Autoimmune Diseases prevention & control, GTP-Binding Protein beta Subunits metabolism, GTP-Binding Protein gamma Subunits metabolism, Macrophages drug effects, Myocarditis prevention & control, Signal Transduction drug effects, Xanthenes pharmacology
- Abstract
The purpose of this study was to determine whether blocking of G protein βγ (Gβγ) signaling halts heart failure (HF) progression by macrophage phenotype manipulation. Cardiac Gβγ signaling plays a crucial role in HF pathogenesis. Previous data suggested that inhibiting Gβγ signaling reprograms T helper cell 1 (Th1) and Th2 cytokines, suggesting that Gβγ might be a useful drug target for treating HF. We investigated the efficacy of a small molecule Gβγ inhibitor, gallein, in a clinically relevant, experimental autoimmune myocarditis (EAM) model of HF as well as in human macrophage phenotypes in vitro. In the myocardium of HF patients, we observed that G protein coupled receptor kinase (GRK)2 levels were down-regulated compared with healthy controls. In rat EAM, treatment with gallein effectively improved survival and cardiac function, suppressed cardiac remodeling, and further attenuated myocardial protein expression of GRK2 as well as high mobility group box (HMGB)1 and its cascade signaling proteins. Furthermore, gallein effectively inhibited M1 polarization and promoted M2 polarization in vivo in the EAM heart and in vitro in human monocyte-derived macrophages. Taken together, these data suggest that the small molecule Gβγ inhibitor, gallein, could be an important pharmacologic therapy for HF as it can switch the phenotypic reprogramming from M1 to M2 phenotype in a rat model of EAM heart and in human macrophages., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2018
- Full Text
- View/download PDF
12. Efficacy of sustained delivery of GC-1 from a Nanofluidic system in a spontaneously obese non-human primate: a case study.
- Author
-
Filgueira CS, Bruno G, Smith ZW, Chua CYX, Ballerini A, Folci M, Gilbert AL, Jain P, Sastry JK, Nehete PN, Shelton KA, Hill LR, Ali A, Youker KA, and Grattoni A
- Subjects
- Animals, Macaca mulatta, Acetates metabolism, Drug Delivery Systems instrumentation, Nanotechnology instrumentation, Obesity metabolism, Phenols metabolism
- Abstract
With nearly 40% of U.S. adults obese, and childhood and adolescent rates rising, obesity and associated comorbidities are serious public health concerns with massive societal costs. Often, lifestyle interventions do not offer sufficient weight loss to improve health, requiring surgery and medications as adjunct management strategies. Here, we present a 4-month case study in which the sustained, low-dose, and constant administration of the thyroid receptor β selective agonist GC-1 (sobetirome) from a novel nanochannel membrane implant was assessed in an obese, pre-diabetic rhesus macaque. Dramatic loss of white adipose tissue in the abdomen from 36 to 18% was observed via magnetic resonance imaging in conjunction with normalized serum insulin and glycemia, with no signs of cardiotoxicity shown. The non-human primate study highlights sustained low-dose delivery of GC-1 from our minimally invasive subcutaneous implant as a valuable approach to induce weight loss and manage obesity and comorbidities, including type 2 diabetes.
- Published
- 2018
- Full Text
- View/download PDF
13. Inducible Lung Epithelial Resistance Requires Multisource Reactive Oxygen Species Generation To Protect against Viral Infections.
- Author
-
Kirkpatrick CT, Wang Y, Leiva Juarez MM, Shivshankar P, Pantaleón García J, Plumer AK, Kulkarni VV, Ware HH, Gulraiz F, Chavez Cavasos MA, Martinez Zayas G, Wali S, Rice AP, Liu H, Tour JM, Sikkema WKA, Cruz Solbes AS, Youker KA, Tuvim MJ, Dickey BF, and Evans SE
- Subjects
- Animals, Epithelial Cells virology, Female, Humans, Influenza A Virus, H3N2 Subtype genetics, Influenza, Human genetics, Influenza, Human virology, Interferon Type I genetics, Interferon Type I immunology, Lung cytology, Lung immunology, Lung virology, Male, Mice, Mice, Inbred C57BL, Toll-Like Receptors genetics, Toll-Like Receptors immunology, Epithelial Cells immunology, Influenza A Virus, H3N2 Subtype physiology, Influenza, Human immunology, Reactive Oxygen Species immunology
- Abstract
Viral pneumonias cause profound worldwide morbidity, necessitating novel strategies to prevent and treat these potentially lethal infections. Stimulation of intrinsic lung defenses via inhalation of synergistically acting Toll-like receptor (TLR) agonists protects mice broadly against pneumonia, including otherwise-lethal viral infections, providing a potential opportunity to mitigate infectious threats. As intact lung epithelial TLR signaling is required for the inducible resistance and as these cells are the principal targets of many respiratory viruses, the capacity of lung epithelial cells to be therapeutically manipulated to function as autonomous antiviral effectors was investigated. Our work revealed that mouse and human lung epithelial cells could be stimulated to generate robust antiviral responses that both reduce viral burden and enhance survival of isolated cells and intact animals. The antiviral protection required concurrent induction of epithelial reactive oxygen species (ROS) from both mitochondrial and dual oxidase sources, although neither type I interferon enrichment nor type I interferon signaling was required for the inducible protection. Taken together, these findings establish the sufficiency of lung epithelial cells to generate therapeutically inducible antiviral responses, reveal novel antiviral roles for ROS, provide mechanistic insights into inducible resistance, and may provide an opportunity to protect patients from viral pneumonia during periods of peak vulnerability. IMPORTANCE Viruses are the most commonly identified causes of pneumonia and inflict unacceptable morbidity, despite currently available therapies. While lung epithelial cells are principal targets of respiratory viruses, they have also been recently shown to contribute importantly to therapeutically inducible antimicrobial responses. This work finds that lung cells can be stimulated to protect themselves against viral challenges, even in the absence of leukocytes, both reducing viral burden and improving survival. Further, it was found that the protection occurs via unexpected induction of reactive oxygen species (ROS) from spatially segregated sources without reliance on type I interferon signaling. Coordinated multisource ROS generation has not previously been described against viruses, nor has ROS generation been reported for epithelial cells against any pathogen. Thus, these findings extend the potential clinical applications for the strategy of inducible resistance to protect vulnerable people against viral infections and also provide new insights into the capacity of lung cells to protect against infections via novel ROS-dependent mechanisms., (Copyright © 2018 Kirkpatrick et al.)
- Published
- 2018
- Full Text
- View/download PDF
14. Functionally redundant control of cardiac hypertrophic signaling by inositol 1,4,5-trisphosphate receptors.
- Author
-
Garcia MI, Karlstaedt A, Chen JJ, Amione-Guerra J, Youker KA, Taegtmeyer H, and Boehning D
- Subjects
- Animals, Animals, Newborn, Cardiomegaly complications, Cardiomegaly pathology, Cell Nucleus drug effects, Cell Nucleus metabolism, Cells, Cultured, Cytosol drug effects, Cytosol metabolism, Endothelin-1 pharmacology, Heart Failure complications, Heart Failure pathology, Heart Ventricles pathology, Hyperglycemia pathology, Myocardial Contraction drug effects, Myocytes, Cardiac drug effects, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology, NFATC Transcription Factors metabolism, Protein Isoforms metabolism, Rats, Sprague-Dawley, Ryanodine Receptor Calcium Release Channel metabolism, Sarcoplasmic Reticulum Calcium-Transporting ATPases metabolism, Signal Transduction drug effects, Cardiomegaly metabolism, Hyperglycemia metabolism, Inositol 1,4,5-Trisphosphate Receptors metabolism
- Abstract
Calcium plays an integral role to many cellular processes including contraction, energy metabolism, gene expression, and cell death. The inositol 1, 4, 5-trisphosphate receptor (IP
3 R) is a calcium channel expressed in cardiac tissue. There are three IP3 R isoforms encoded by separate genes. In the heart, the IP3 R-2 isoform is reported to being most predominant with regards to expression levels and functional significance. The functional roles of IP3 R-1 and IP3 R-3 in the heart are essentially unexplored despite measureable expression levels. Here we show that all three IP3 Rs isoforms are expressed in both neonatal and adult rat ventricular cardiomyocytes, and in human heart tissue. The three IP3 R proteins are expressed throughout the cardiomyocyte sarcoplasmic reticulum. Using isoform specific siRNA, we found that expression of all three IP3 R isoforms are required for hypertrophic signaling downstream of endothelin-1 stimulation. Mechanistically, IP3 Rs specifically contribute to activation of the hypertrophic program by mediating the positive inotropic effects of endothelin-1 and leading to downstream activation of nuclear factor of activated T-cells. Our findings highlight previously unidentified functions for IP3 R isoforms in the heart with specific implications for hypertrophic signaling in animal models and in human disease., (Copyright © 2017 Elsevier Ltd. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
15. Rapamycin nanoparticles localize in diseased lung vasculature and prevent pulmonary arterial hypertension.
- Author
-
Segura-Ibarra V, Amione-Guerra J, Cruz-Solbes AS, Cara FE, Iruegas-Nunez DA, Wu S, Youker KA, Bhimaraj A, Torre-Amione G, Ferrari M, Karmouty-Quintana H, Guha A, and Blanco E
- Subjects
- Administration, Intravenous, Animals, Disease Models, Animal, Lung pathology, Rats, Rats, Sprague-Dawley, Sirolimus administration & dosage, Hypertension, Pulmonary drug therapy, Lung drug effects, Nanoparticles administration & dosage, Sirolimus pharmacology
- Abstract
Vascular remodeling resulting from pulmonary arterial hypertension (PAH) leads to endothelial fenestrations. This feature can be exploited by nanoparticles (NP), allowing them to extravasate from circulation and accumulate in remodeled pulmonary vessels. Hyperactivation of the mTOR pathway in PAH drives pulmonary arterial smooth muscle cell proliferation. We hypothesized that rapamycin (RAP)-loaded NPs, an mTOR inhibitor, would accumulate in diseased lungs, selectively targeting vascular mTOR and preventing PAH progression. RAP poly(ethylene glycol)-block-poly(ε-caprolactone) (PEG-PCL) NPs were fabricated. NP accumulation and efficacy were examined in a rat monocrotaline model of PAH. Following intravenous (IV) administration, NP accumulation in diseased lungs was verified via LC/MS analysis and confocal imaging. Pulmonary arteriole thickness, right ventricular systolic pressures, and ventricular remodeling were determined to assess the therapeutic potential of RAP NPs. Monocrotaline-exposed rats showed increased NP accumulation within lungs compared to healthy controls, with NPs present to a high extent within pulmonary perivascular regions. RAP, in both free and NP form, attenuated PAH development, with histological analysis revealing minimal changes in pulmonary arteriole thickness and no ventricular remodeling. Importantly, NP-treated rats showed reduced systemic side effects compared to free RAP. This study demonstrates the potential for nanoparticles to significantly impact PAH through site-specific delivery of therapeutics., (Copyright © 2017 Elsevier B.V. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
16. AIBP Limits Angiogenesis Through γ-Secretase-Mediated Upregulation of Notch Signaling.
- Author
-
Mao R, Meng S, Gu Q, Araujo-Gutierrez R, Kumar S, Yan Q, Almazan F, Youker KA, Fu Y, Pownall HJ, Cooke JP, Miller YI, and Fang L
- Subjects
- Animals, Hindlimb blood supply, Hindlimb metabolism, Hindlimb pathology, Humans, Ischemia metabolism, Ischemia pathology, Mice, Mice, Knockout, Racemases and Epimerases, Retina metabolism, Retina pathology, Zebrafish, Amyloid Precursor Protein Secretases physiology, Carrier Proteins biosynthesis, Neovascularization, Physiologic physiology, Phosphoproteins biosynthesis, Receptors, Notch biosynthesis, Signal Transduction physiology, Up-Regulation physiology
- Abstract
Rationale: Angiogenesis improves perfusion to the ischemic tissue after acute vascular obstruction. Angiogenesis in pathophysiological settings reactivates signaling pathways involved in developmental angiogenesis. We showed previously that AIBP (apolipoprotein A-I [apoA-I]-binding protein)-regulated cholesterol efflux in endothelial cells controls zebra fish embryonic angiogenesis., Objective: This study is to determine whether loss of AIBP affects angiogenesis in mice during development and under pathological conditions and to explore the underlying molecular mechanism., Methods and Results: In this article, we report the generation of AIBP knockout ( Apoa1bp
-/- ) mice, which are characterized of accelerated postnatal retinal angiogenesis. Mechanistically, AIBP triggered relocalization of γ-secretase from lipid rafts to nonlipid rafts where it cleaved Notch. Consistently, AIBP treatment enhanced DLL4 (delta-like ligand 4)-stimulated Notch activation in human retinal endothelial cells. Increasing high-density lipoprotein levels in Apoa1bp-/- mice by crossing them with apoA-I transgenic mice rescued Notch activation and corrected dysregulated retinal angiogenesis. Notably, the retinal vessels in Apoa1bp-/- mice manifested normal pericyte coverage and vascular integrity. Similarly, in the subcutaneous Matrigel plug assay, which mimics ischemic/inflammatory neovascularization, angiogenesis was dramatically upregulated in Apoa1bp-/- mice and associated with a profound inhibition of Notch activation and reduced expression of downstream targets. Furthermore, loss of AIBP increased vascular density and facilitated the recovery of blood vessel perfusion function in a murine hindlimb ischemia model. In addition, AIBP expression was significantly increased in human patients with ischemic cardiomyopathy., Conclusions: Our data reveal a novel mechanistic connection between AIBP-mediated cholesterol metabolism and Notch signaling, implicating AIBP as a possible druggable target to modulate angiogenesis under pathological conditions., (© 2017 American Heart Association, Inc.)- Published
- 2017
- Full Text
- View/download PDF
17. MicroRNA-126 overexpression rescues diabetes-induced impairment in efferocytosis of apoptotic cardiomyocytes.
- Author
-
Suresh Babu S, Thandavarayan RA, Joladarashi D, Jeyabal P, Krishnamurthy S, Bhimaraj A, Youker KA, and Krishnamurthy P
- Subjects
- 3' Untranslated Regions, Animals, Apoptosis, Gene Expression Regulation drug effects, Glucose pharmacology, Humans, Macrophages drug effects, Mice, Phagocytosis, Proto-Oncogene Mas, RAW 264.7 Cells, THP-1 Cells, c-Mer Tyrosine Kinase metabolism, ADAM Proteins genetics, Diabetes Mellitus, Experimental genetics, Macrophages cytology, Membrane Proteins genetics, MicroRNAs genetics, Myocytes, Cardiac cytology
- Abstract
Efferocytosis, a process of clearance of apoptotic cells by phagocytes, is essential for successful resolution of inflammation and maintenance of tissue homeostasis. Diabetes compromises the function of macrophages leading to adverse inflammatory response during wound healing, myocardial injury, atherosclerosis and autoimmune disorders. However, the effect of diabetes on macrophage-mediated efferocytosis of apoptotic cardiomyocytes (ACM) and the molecular mechanisms involved are not understood so far. In the present study we found that invitro efferocytosis of ACM was impaired in macrophages from db/db (diabetic) mice. Macrophages exposed to high glucose (HG) decreases microRNA-126 (miR-126) expression with a corresponding increase in ADAM9 expression. Dual-luciferase reporter assay confirms that ADAM9 3'UTR contains miR-126 target site. ADAM9 inhibition reduces HG-induced proteolytic cleavage of Mer tyrosine receptor kinase (MerTK, a proto-oncogene that plays a critical role in phagocytosis), resulting in shedding of soluble-Mer (sMER) and loss of MERTK function. Over-expression of miR-126 attenuates HG-induced impairment of efferocytosis. Furthermore, human diabetic hearts show lower miR-126 expression with a corresponding increase in ADAM9 expression vs. normal counterparts. These data suggests that diabetes impairs efferocytosis of ACM and that strategies to enhance efferocytosis might attenuate diabetes-induced impairment in inflammation resolution and cardiac repair after injury.
- Published
- 2016
- Full Text
- View/download PDF
18. MicroRNA-9 inhibits hyperglycemia-induced pyroptosis in human ventricular cardiomyocytes by targeting ELAVL1.
- Author
-
Jeyabal P, Thandavarayan RA, Joladarashi D, Suresh Babu S, Krishnamurthy S, Bhimaraj A, Youker KA, Kishore R, and Krishnamurthy P
- Subjects
- Animals, Cell Line, Cells, Cultured, Diabetic Cardiomyopathies pathology, ELAV-Like Protein 1 metabolism, Gene Expression Regulation, Gene Knockdown Techniques, Heart Ventricles pathology, Humans, Hyperglycemia metabolism, Mice, MicroRNAs metabolism, Myocytes, Cardiac physiology, ELAV-Like Protein 1 genetics, Hyperglycemia genetics, MicroRNAs genetics, Myocytes, Cardiac pathology, Pyroptosis genetics
- Abstract
Diabetic cardiomyopathy is a common complication in patients with diabetes and is associated with underlying chronic inflammation and cardiac cell death, subsequently leading to heart failure (HF). ELAV-like protein 1 (ELAVL1) plays a critical role in the progression of inflammation and HF. However the role of ELAVL-1 in inflammation induced cardiac cell death (pyroptosis) under hyperglycemic condition remains elusive. Our data demonstrates that ELAVL1 expression augmented with a concomitant increase in caspase-1 and IL-1 beta expression in human hearts and human ventricular cardiomyocytes under hyperglycemic condition. Furthermore, ELAVL1 knockdown abrogates TNF-α induced canonical pyroptosis via NLRP3, caspase-1 and IL-1beta suppression. Bioinformatics analysis and target validation assays showed that miR-9 directly targets ELAVL1. Interestingly, miRNA-9 expression significantly reduced in high glucose treated cardiomyocytes and in human diabetic hearts. Inhibition of miR-9 upregulates ELAVL1 expression and activates caspase-1. Alternatively, treatment with miR-9 mimics attenuates hyperglycemia-induced ELAVL1 and inhibits cardiomyocyte pyroptosis. Taken together our study highlights the potential therapeutic implications of targeting miR-9/ELAVL1 in preventing cardiomyocyte cell loss during HF in diabetics., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
19. Combination of angiotensin II and l-NG-nitroarginine methyl ester exacerbates mitochondrial dysfunction and oxidative stress to cause heart failure.
- Author
-
Hamilton DJ, Zhang A, Li S, Cao TN, Smith JA, Vedula I, Cordero-Reyes AM, Youker KA, Torre-Amione G, and Gupte AA
- Subjects
- Animals, Atrial Natriuretic Factor, Calcium-Calmodulin-Dependent Protein Kinase Type 2 drug effects, Calcium-Calmodulin-Dependent Protein Kinase Type 2 genetics, Cardiomegaly, DNA, Mitochondrial drug effects, DNA, Mitochondrial metabolism, Electron Transport Complex I drug effects, Electron Transport Complex I metabolism, Electron Transport Complex II drug effects, Electron Transport Complex II metabolism, Gene Expression drug effects, Heart drug effects, Hydrogen Peroxide metabolism, Mice, Mitochondria, Heart metabolism, Myocardium metabolism, Myocardium pathology, Natriuretic Peptide, Brain drug effects, Natriuretic Peptide, Brain genetics, Natriuretic Peptide, C-Type drug effects, Natriuretic Peptide, C-Type genetics, Protein Precursors drug effects, Protein Precursors genetics, Pyruvate Dehydrogenase Complex drug effects, Pyruvate Dehydrogenase Complex metabolism, Reverse Transcriptase Polymerase Chain Reaction, Sarcoplasmic Reticulum Calcium-Transporting ATPases drug effects, Sarcoplasmic Reticulum Calcium-Transporting ATPases genetics, Angiotensin II pharmacology, Enzyme Inhibitors pharmacology, Heart Failure etiology, Mitochondria, Heart drug effects, NG-Nitroarginine Methyl Ester pharmacology, Nitric Oxide metabolism, Oxidative Stress drug effects, Vasoconstrictor Agents pharmacology
- Abstract
Mitochondrial dysfunction has been implicated as a cause of energy deprivation in heart failure (HF). Herein, we tested individual and combined effects of two pathogenic factors of nonischemic HF, inhibition of nitric oxide synthesis [with l-N(G)-nitroarginine methyl ester (l-NAME)] and hypertension [with angiotensin II (AngII)], on myocardial mitochondrial function, oxidative stress, and metabolic gene expression. l-NAME and AngII were administered individually and in combination to mice for 5 wk. Although all treatments increased blood pressure and reduced cardiac contractile function, the l-NAME + AngII group was associated with the most severe HF, as characterized by edema, hypertrophy, oxidative stress, increased expression of Nppa and Nppb, and decreased expression of Atp2a2 and Camk2b. l-NAME + AngII-treated mice exhibited robust deterioration of cardiac mitochondrial function, as observed by reduced respiratory control ratios in subsarcolemmal mitochondria and reduced state 3 levels in interfibrillar mitochondria for complex I but not for complex II substrates. Cardiac myofibrils showed reduced ADP-supported and oligomycin-inhibited oxygen consumption. Mitochondrial functional impairment was accompanied by reduced mitochondrial DNA content and activities of pyruvate dehydrogenase and complex I but increased H2O2 production and tissue protein carbonyls in hearts from AngII and l-NAME + AngII groups. Microarray analyses revealed the majority of the gene changes attributed to the l-NAME + AngII group. Pathway analyses indicated significant changes in metabolic pathways, such as oxidative phosphorylation, mitochondrial function, cardiac hypertrophy, and fatty acid metabolism in l-NAME + AngII hearts. We conclude that l-NAME + AngII is associated with impaired mitochondrial respiratory function and increased oxidative stress compared with either l-NAME or AngII alone, resulting in nonischemic HF., (Copyright © 2016 the American Physiological Society.)
- Published
- 2016
- Full Text
- View/download PDF
20. Rnd3/RhoE Modulates Hypoxia-Inducible Factor 1α/Vascular Endothelial Growth Factor Signaling by Stabilizing Hypoxia-Inducible Factor 1α and Regulates Responsive Cardiac Angiogenesis.
- Author
-
Yue X, Lin X, Yang T, Yang X, Yi X, Jiang X, Li X, Li T, Guo J, Dai Y, Shi J, Wei L, Youker KA, Torre-Amione G, Yu Y, Andrade KC, and Chang J
- Subjects
- Animals, Blotting, Western, Coronary Vessels metabolism, Disease Models, Animal, Endothelium, Vascular metabolism, Endothelium, Vascular pathology, Humans, Hypertension metabolism, Hypertension pathology, Hypoxia-Inducible Factor 1, alpha Subunit biosynthesis, Mice, Mice, Knockout, Mice, Transgenic, Neovascularization, Pathologic metabolism, RNA genetics, Signal Transduction, Vascular Endothelial Growth Factor A biosynthesis, rho GTP-Binding Proteins biosynthesis, Coronary Vessels pathology, Gene Expression Regulation, Hypertension genetics, Hypoxia-Inducible Factor 1, alpha Subunit genetics, Neovascularization, Pathologic genetics, Vascular Endothelial Growth Factor A genetics, rho GTP-Binding Proteins genetics
- Abstract
The insufficiency of compensatory angiogenesis in the heart of patients with hypertension contributes to heart failure transition. The hypoxia-inducible factor 1α-vascular endothelial growth factor (HIF1α-VEGF) signaling cascade controls responsive angiogenesis. One of the challenges in reprograming the insufficient angiogenesis is to achieve a sustainable tissue exposure to the proangiogenic factors, such as HIF1α stabilization. In this study, we identified Rnd3, a small Rho GTPase, as a proangiogenic factor participating in the regulation of the HIF1α-VEGF signaling cascade. Rnd3 physically interacted with and stabilized HIF1α, and consequently promoted VEGFA expression and endothelial cell tube formation. To demonstrate this proangiogenic role of Rnd3 in vivo, we generated Rnd3 knockout mice. Rnd3 haploinsufficient (Rnd3(+/-)) mice were viable, yet developed dilated cardiomyopathy with heart failure after transverse aortic constriction stress. The poststress Rnd3(+/-) hearts showed significantly impaired angiogenesis and decreased HIF1α and VEGFA expression. The angiogenesis defect and heart failure phenotype were partially rescued by cobalt chloride treatment, a HIF1α stabilizer, confirming a critical role of Rnd3 in stress-responsive angiogenesis. Furthermore, we generated Rnd3 transgenic mice and demonstrated that Rnd3 overexpression in heart had a cardioprotective effect through reserved cardiac function and preserved responsive angiogenesis after pressure overload. Finally, we assessed the expression levels of Rnd3 in the human heart and detected significant downregulation of Rnd3 in patients with end-stage heart failure. We concluded that Rnd3 acted as a novel proangiogenic factor involved in cardiac responsive angiogenesis through HIF1α-VEGFA signaling promotion. Rnd3 downregulation observed in patients with heart failure may explain the insufficient compensatory angiogenesis involved in the transition to heart failure., (© 2016 American Heart Association, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
21. A specifically designed nanoconstruct associates, internalizes, traffics in cardiovascular cells, and accumulates in failing myocardium: a new strategy for heart failure diagnostics and therapeutics.
- Author
-
Ruiz-Esparza GU, Segura-Ibarra V, Cordero-Reyes AM, Youker KA, Serda RE, Cruz-Solbes AS, Amione-Guerra J, Yokoi K, Kirui DK, Cara FE, Paez-Mayorga J, Flores-Arredondo JH, Guerrero-Beltrán CE, Garcia-Rivas G, Ferrari M, Blanco E, and Torre-Amione G
- Subjects
- Animals, Biocompatible Materials, Disease Models, Animal, Heart Failure physiopathology, Humans, Injections, Intravenous, Male, Mice, Mice, Inbred C57BL, Myocardium, Polymers, Silicon, Heart physiology, Heart physiopathology, Heart Failure diagnosis, Heart Failure therapy, Myocytes, Cardiac physiology, Nanostructures therapeutic use
- Abstract
Aims: Ongoing inflammation and endothelial dysfunction occurs within the local microenvironment of heart failure, creating an appropriate scenario for successful use and delivery of nanovectors. This study sought to investigate whether cardiovascular cells associate, internalize, and traffic a nanoplatform called mesoporous silicon vector (MSV), and determine its intravenous accumulation in cardiac tissue in a murine model of heart failure., Methods and Results: In vitro cellular uptake and intracellular trafficking of MSVs was examined by scanning electron microscopy, confocal microscopy, time-lapse microscopy, and flow cytometry in cardiac myocytes, fibroblasts, smooth muscle cells, and endothelial cells. The MSVs were internalized within the first hours, and trafficked to perinuclear regions in all the cell lines. Cytotoxicity was investigated by annexin V and cell cycle assays. No significant evidence of toxicity was found. In vivo intravenous cardiac accumulation of MSVs was examined by high content fluorescence and confocal microscopy, with results showing increased accumulation of particles in failing hearts compared with normal hearts. Similar to observations in vitro, MSVs were able to associate, internalize, and traffic to the perinuclear region of cardiomyocytes in vivo., Conclusions: Results show that MSVs associate, internalize, and traffic in cardiovascular cells without any significant toxicity. Furthermore, MSVs accumulate in failing myocardium after intravenous administration, reaching intracellular regions of the cardiomyocytes. These findings represent a novel avenue to develop nanotechnology-based therapeutics and diagnostics in heart failure., Competing Interests: Conflicts of Interest: None declared., (© 2016 The Authors European Journal of Heart Failure © 2016 European Society of Cardiology.)
- Published
- 2016
- Full Text
- View/download PDF
22. Full Expression of Cardiomyopathy Is Partly Dependent on B-Cells: A Pathway That Involves Cytokine Activation, Immunoglobulin Deposition, and Activation of Apoptosis.
- Author
-
Cordero-Reyes AM, Youker KA, Trevino AR, Celis R, Hamilton DJ, Flores-Arredondo JH, Orrego CM, Bhimaraj A, Estep JD, and Torre-Amione G
- Subjects
- Angiotensin II, Animals, B-Lymphocytes immunology, Cardiomyopathies chemically induced, Cardiomyopathies genetics, Cardiomyopathies immunology, Cardiomyopathies pathology, Cardiomyopathies physiopathology, Collagen metabolism, Cytokines immunology, Disease Models, Animal, Fibroblasts metabolism, Fibroblasts pathology, Fibrosis, Genetic Predisposition to Disease, Heart Failure chemically induced, Heart Failure genetics, Heart Failure immunology, Heart Failure pathology, Heart Failure physiopathology, Hypertrophy, Left Ventricular immunology, Hypertrophy, Left Ventricular metabolism, Hypertrophy, Left Ventricular pathology, Hypertrophy, Left Ventricular prevention & control, Immunoglobulin G immunology, Magnetic Resonance Imaging, Male, Mice, Inbred C57BL, Mice, Knockout, Mice, Nude, Mice, SCID, Myocardium immunology, Myocardium pathology, NG-Nitroarginine Methyl Ester, Phenotype, Sialic Acid Binding Ig-like Lectin 2 deficiency, Sialic Acid Binding Ig-like Lectin 2 genetics, Signal Transduction, Sodium Chloride, Stroke Volume, Time Factors, Ventricular Dysfunction, Left immunology, Ventricular Dysfunction, Left metabolism, Ventricular Dysfunction, Left physiopathology, Ventricular Dysfunction, Left prevention & control, Ventricular Function, Left, Ventricular Remodeling, Apoptosis, B-Lymphocytes metabolism, Cardiomyopathies metabolism, Cytokines metabolism, Heart Failure metabolism, Immunoglobulin G metabolism, Myocardium metabolism
- Abstract
Background: Limited information exists on the role of B-cell-dependent mechanisms in the progression of heart failure (HF). However, in failing human myocardium, there is evidence of deposition of activated complement components as well as anticardiac antibodies. We aimed to determine the contribution of B-cells in HF progression using a nonsurgical mouse model of nonischemic cardiomyopathy (CMP)., Methods and Results: CMP protocol involved the use of l-NAME and NaCl in the drinking water and angiotensin-II infusion for 35 days. At day 35, mice were analyzed by cardiac magnetic resonance imaging, gene expression, and histology. Mice (12 weeks old) were divided into 4 groups, all in C57BL/6 background: wild-type (WT) CMP; severe combined immunodeficiency (SCID) CMP (T- and B-cell deficient); CD22(-) CMP (B-cell depleted); and Nude CMP (T-cell deficient), with their respective controls. We performed B-cell depletion and reconstitution protocols. The protective effect of B-cell depletion was demonstrated by a significant reduction of cell hypertrophy and collagen deposition and a preserved ejection fraction in the CD22(-) CMP group compared to WT CMP. Once SCID mice underwent B-cell reconstitution with isolated CMP B-cells, the CMP phenotype was restored. Furthermore, deposition of IgG3 and apoptosis in the myocardium follows the development of CMP; in addition, in vitro studies demonstrated that activated B-cells stimulate collagen production by cardiac fibroblasts., Conclusions: The absence of B-cells in this model of HF resulted in less hypertrophy and collagen deposition, preservation of left ventricular function, and, in association with these changes, a reduction in expression of proinflammatory cytokines, immunoglobulin G deposition, and apoptosis in the myocardium. Taken together, these data suggest that B-cells play a contributory role in an angiotensin-II-induced HF model., (© 2016 The Authors. Published on behalf of the American Heart Association, Inc., by Wiley Blackwell.)
- Published
- 2016
- Full Text
- View/download PDF
23. Differential Mitochondrial Function in Remodeled Right and Nonremodeled Left Ventricles in Pulmonary Hypertension.
- Author
-
Gupte AA, Cordero-Reyes AM, Youker KA, Matsunami RK, Engler DA, Li S, Loebe M, Ashrith G, Torre-Amione G, and Hamilton DJ
- Subjects
- Adolescent, Aged, Echocardiography, Electron Transport Complex I metabolism, Female, Heart Failure etiology, Humans, Middle Aged, Mitochondria, Heart enzymology, Mitochondrial ADP, ATP Translocases metabolism, Proteomics, Ventricular Dysfunction, Right etiology, Heart Failure physiopathology, Heart Ventricles physiopathology, Hypertension, Pulmonary complications, Mitochondria, Heart metabolism, Ventricular Dysfunction, Right physiopathology, Ventricular Remodeling
- Abstract
Objectives: Right ventricular failure is the primary reason for mortality in pulmonary hypertension (PH), but little is understood about the energetics of the failing right myocardium. Our aim was to examine mitochondrial function and proteomic signatures in paired remodeled right (RM-RV) and non-remodeled left (NRM-LV) ventricular tissue samples procured during heart-lung transplantation., Methods and Results: Contractile dysfunction in RM-RV and preserved contractile function in NRM-LV were determined clinically and by echocardiography. Mitochondria were isolated from fresh paired RV and LV wall specimens of explanted hearts. Respiratory states in response to 4 substrates and an uncoupler were analyzed. Proteomic analysis on the mitochondrial isolates was performed with the use of liquid chromatography-mass spectrometry. The RM-RV mitochondria exhibited higher succinate state 4 levels with lower respiratory control ratio (RCR) compared with state 4 levels for pyruvate-malate (PM) and glutamate-malate (GM). RM-RV mitochondria also exhibited lower state 3 for palmitoyl-carnitine (PC) and state 4 for all complex I substrates compared with NRM-LV. The mean RCR were greater in RM-RVs than in NRM-LVs for PM and GM, which is consistent with tight coupling (low state 4 rates, higher RCRs); however, low RM-RV state 3 rates suggest concurrent substrate-dependent impairment in respiratory capacity. Mitochondrial proteomics revealed greater levels of mitochondrial ADP-ATP translocase and proteins of ATP synthesis, mitochondrial pyruvate and short branched chain acyl-CoA metabolism in RM-RV., Conclusions: The mitochondrial respiration and proteomics in RM-RV are different from NRM-LV. These results have important implications in expanding our understanding of RV metabolism and future management of RV failure., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
24. Enhanced Cardiac Regenerative Ability of Stem Cells After Ischemia-Reperfusion Injury: Role of Human CD34+ Cells Deficient in MicroRNA-377.
- Author
-
Joladarashi D, Garikipati VNS, Thandavarayan RA, Verma SK, Mackie AR, Khan M, Gumpert AM, Bhimaraj A, Youker KA, Uribe C, Suresh Babu S, Jeyabal P, Kishore R, and Krishnamurthy P
- Subjects
- Adult, Animals, Antigens, CD34, Female, Heart, Humans, Inflammation pathology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Myocardium cytology, Myocardium pathology, Neovascularization, Physiologic physiology, Reperfusion Injury pathology, Reverse Transcriptase Polymerase Chain Reaction, Endothelial Progenitor Cells metabolism, Heart Failure metabolism, MicroRNAs metabolism, Myocardium metabolism, Reperfusion Injury metabolism
- Abstract
Background: MicroRNA (miR) dysregulation in the myocardium has been implicated in cardiac remodeling after injury or stress., Objectives: The aim of this study was to explore the role of miR in human CD34(+) cell (hCD34(+)) dysfunction in vivo after transplantation into the myocardium under ischemia-reperfusion (I-R) conditions., Methods: In response to inflammatory stimuli, the miR array profile of endothelial progenitor cells was analyzed using a polymerase chain reaction-based miR microarray. miR-377 expression was assessed in myocardial tissue from human patients with heart failure (HF). We investigated the effect of miR-377 inhibition on an hCD34(+) cell angiogenic proteome profile in vitro and on cardiac repair and function after I-R injury in immunodeficient mice., Results: The miR array data from endothelial progenitor cells in response to inflammatory stimuli indicated changes in numerous miR, with a robust decrease in the levels of miR-377. Human cardiac biopsies from patients with HF showed significant increases in miR-377 expression compared with nonfailing control hearts. The proteome profile of hCD34(+) cells transfected with miR-377 mimics showed significant decrease in the levels of proangiogenic proteins versus nonspecific control-transfected cells. We also validated that serine/threonine kinase 35 is a target of miR-377 using a dual luciferase reporter assay. In a mouse model of myocardial I-R, intramyocardial transplantation of miR-377 silenced hCD34(+) cells in immunodeficient mice, promoting neovascularization (at 28 days, post-I-R) and lower interstitial fibrosis, leading to improved left ventricular function., Conclusions: These findings indicate that HF increased miR-377 expression in the myocardium, which is detrimental to stem cell function, and transplantation of miR-377 knockdown hCD34(+) cells into ischemic myocardium promoted their angiogenic ability, attenuating left ventricular remodeling and cardiac fibrosis., (Copyright © 2015 American College of Cardiology Foundation. Published by Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
25. Mechanical unloading promotes myocardial energy recovery in human heart failure.
- Author
-
Gupte AA, Hamilton DJ, Cordero-Reyes AM, Youker KA, Yin Z, Estep JD, Stevens RD, Wenner B, Ilkayeva O, Loebe M, Peterson LE, Lyon CJ, Wong ST, Newgard CB, Torre-Amione G, Taegtmeyer H, and Hsueh WA
- Subjects
- Adult, Aged, Female, Gene Expression Profiling, Heart Failure genetics, Heart Failure therapy, Heart-Assist Devices, Humans, Male, Middle Aged, Myocytes, Cardiac metabolism, Transcription Factors genetics, Energy Metabolism, Heart Failure metabolism, Heart Ventricles metabolism, Myocardium metabolism
- Abstract
Background: Impaired bioenergetics is a prominent feature of the failing heart, but the underlying metabolic perturbations are poorly understood., Methods and Results: We compared metabolomic, gene transcript, and protein data from 6 paired samples of failing human left ventricular tissue obtained during left ventricular assist device insertion (heart failure samples) and at heart transplant (post-left ventricular assist device samples). Nonfailing left ventricular wall samples procured from explanted hearts of patients with right heart failure served as novel comparison samples. Metabolomic analyses uncovered a distinct pattern in heart failure tissue: 2.6-fold increased pyruvate concentrations coupled with reduced Krebs cycle intermediates and short-chain acylcarnitines, suggesting a global reduction in substrate oxidation. These findings were associated with decreased transcript levels for enzymes that catalyze fatty acid oxidation and pyruvate metabolism and for key transcriptional regulators of mitochondrial metabolism and biogenesis, peroxisome proliferator-activated receptor γ coactivator 1α (PGC1A, 1.3-fold) and estrogen-related receptor α (ERRA, 1.2-fold) and γ (ERRG, 2.2-fold). Thus, parallel decreases in key transcription factors and their target metabolic enzyme genes can explain the decreases in associated metabolic intermediates. Mechanical support with left ventricular assist device improved all of these metabolic and transcriptional defects., Conclusions: These observations underscore an important pathophysiologic role for severely defective metabolism in heart failure, while the reversibility of these defects by left ventricular assist device suggests metabolic resilience of the human heart., (© 2014 American Heart Association, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
26. High proportion of patients with end-stage heart failure regardless of aetiology demonstrates anti-cardiac antibody deposition in failing myocardium: humoral activation, a potential contributor of disease progression.
- Author
-
Youker KA, Assad-Kottner C, Cordero-Reyes AM, Trevino AR, Flores-Arredondo JH, Barrios R, Fernandez-Sada E, Estep JD, Bhimaraj A, and Torre-Amione G
- Subjects
- Adenosine Triphosphatases immunology, Antigens metabolism, Case-Control Studies, Disease Progression, Female, Humans, Immunity, Humoral immunology, Immunoglobulin G metabolism, Male, Middle Aged, Antibodies metabolism, Heart Failure immunology, Myocardium immunology
- Abstract
Aims: Various reports have raised the possibility of humoral immune responses as contributors for the progression of heart failure. Previous studies, however, have focused on the analysis of serum and documented circulating antibodies against a variety of cardiac proteins. However, there is little evidence on whether anti-cardiac antibodies are deposited in end-stage failing myocardium. Our objective was to determine whether or not there was evidence of deposition of anti-cardiac antibodies and/or activated complement components in end-stage failing human myocardium., Methods and Results: Myocardial samples were obtained from 100 end-stage heart failure patients and 40 donor control biopsies. Sections were cut and stained using standard fluorescent immunohistochemistry techniques with anti-human immunoglobulin G (IgG), IgG3, and C3c. Gel electrophoresis and protein identification by mass spectrometry were used to confirm the presence of IgG and its antigen. Immunoglobulin G was localized to the sarcolemma in 71% of patients, 48% of those being positive for the subtype IgG3. The proportion of patients with ischaemic heart disease that was positive for IgG was 65% and among those with non-ischaemic aetiologies was 76%. In a subgroup analysis, the presence of IgG and its subunits were confirmed by mass spectrometry and adenosine triphosphate synthase β subunit identified as an antigen. Complement was activated in 31% of all patients. The presence of IgG, IgG3, and C3c was directly correlated with the length of disease (r = 0.451, P = 0.006)., Conclusion: Evidence of anti-cardiac antibodies and complement activation was found in a large number of patients with end-stage cardiomyopathy regardless of the aetiology. Adenosine triphosphate synthase appears to be a new prominent antigenic stimulus; but more interestingly, the simultaneous co-existence of activated complement components suggests that this humoral mechanism may participate in disease progression.
- Published
- 2014
- Full Text
- View/download PDF
27. Freshly isolated mitochondria from failing human hearts exhibit preserved respiratory function.
- Author
-
Cordero-Reyes AM, Gupte AA, Youker KA, Loebe M, Hsueh WA, Torre-Amione G, Taegtmeyer H, and Hamilton DJ
- Subjects
- Adult, CD36 Antigens genetics, CD36 Antigens metabolism, Case-Control Studies, Cell Respiration, Electron Transport Complex I metabolism, Electron Transport Complex II metabolism, Female, Heart Failure pathology, Heart Ventricles metabolism, Heart Ventricles pathology, Humans, Male, Middle Aged, Oxidation-Reduction, Oxygen metabolism, Transcriptome, Young Adult, Heart Failure metabolism, Mitochondria, Heart metabolism
- Abstract
In heart failure mitochondrial dysfunction is thought to be responsible for energy depletion and contractile dysfunction. The difficulties in procuring fresh left ventricular (LV) myocardium from humans for assessment of mitochondrial function have resulted in the reliance on surrogate markers of mitochondrial function and limited our understanding of cardiac energetics. We isolated mitochondria from fresh LV wall tissue of patients with heart failure and reduced systolic function undergoing heart transplant or left ventricular assist device placement, and compared their function to mitochondria isolated from the non-failing LV (NFLV) wall tissue with normal systolic function from patients with pulmonary hypertension undergoing heart-lung transplant. We performed detailed mitochondrial functional analyses using 4 substrates: glutamate-malate (GM), pyruvate-malate (PM) palmitoyl carnitine-malate (PC) and succinate. NFLV mitochondria showed preserved respiratory control ratios and electron chain integrity with only few differences for the 4 substrates. In contrast, HF mitochondria had greater respiration with GM, PM and PC substrates and higher electron chain capacity for PM than for PC. Surprisingly, HF mitochondria had greater respiratory control ratios and lower ADP-independent state 4 rates than NFLV mitochondria for GM, PM and PC substrates demonstrating that HF mitochondria are capable of coupled respiration ex vivo. Gene expression studies revealed decreased expression of key genes in pathways for oxidation of both fatty acids and glucose. Our results suggest that mitochondria from the failing LV myocardium are capable of tightly coupled respiration when isolated and supplied with ample substrates. Thus energy starvation in the failing heart may be the result of dysregulation of metabolic pathways, impaired substrate supply or reduced mitochondrial number but not the result of reduced mitochondrial electron transport capacity., (Copyright © 2014 Elsevier Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
28. The role of B-cells in heart failure.
- Author
-
Cordero-Reyes AM, Youker KA, and Torre-Amione G
- Subjects
- Animals, Heart Failure physiopathology, Heart Failure therapy, Humans, Immunotherapy methods, Lymphocyte Activation, Signal Transduction, B-Lymphocytes immunology, Heart Failure immunology
- Abstract
Heart failure is a complex disease that has great impact on morbidity and mortality in the general population. No recent therapies have proven to be effective; however, the discovery of new potential pathophysiological mechanisms involved in heart failure expression and progression could offer novel therapeutic strategies. A number of studies have shown that the immune system may be a central mediator in the development and progression of heart failure, and here we describe how the B-cell and B-cell-mediated pathways play specific roles in the heart failure state. Therapies aimed at B-cells, either blocking antibody production or inactivating B-cell function, may suggest potential new treatment strategies.
- Published
- 2013
- Full Text
- View/download PDF
29. Cellular evidence of reverse cardiac remodeling induced by cardiac resynchronization therapy.
- Author
-
Orrego CM, Nasir N, Oliveira GH, Flores-Arredondo JH, Cordero-Reyes AM, Loebe M, Youker KA, and Torre-Amione G
- Subjects
- Aged, Biopsy, Chronic Disease, Echocardiography, Female, Heart Failure metabolism, Humans, Male, Middle Aged, Myocardium metabolism, Myocardium pathology, Myocytes, Cardiac pathology, Prospective Studies, Tumor Necrosis Factor-alpha metabolism, Cardiac Resynchronization Therapy, Heart Failure physiopathology, Heart Failure therapy, Heart-Assist Devices, Ventricular Remodeling physiology
- Abstract
Left ventricular assist devices (LVADs) induce reverse cardiac remodeling by reducing myocyte size and collagen deposition. On the other hand, cardiac resynchronization therapy (CRT) induces reverse cardiac remodeling by improving electromechanical synchronization. The clinical and structural changes produced by CRT in failing myocardium are known, but whether these changes are accompanied by reverse cellular remodeling is unknown. A total of 12 patients with chronic heart failure (CHF) who underwent CRT and 15 patients who had LVAD therapy as clinically indicated and 8 healthy controls were compared. Demographics, echocardiographic data, and histologic samples from myocardial biopsies were analyzed and compared among groups. The authors found significant increases in myocyte size, myocardial fibrosis, and inflammation in both CHF groups who underwent CRT or LVAD, compared with healthy controls. After CRT or LVAD therapy, a significant decrease in myocyte size and tumor necrosis factor α (TNF-α) expression compared with healthy controls (P < .05) was found. In the CRT group, 6 of 8 patients demonstrated reduction in myocyte size and interstitial fibrosis. In addition, there was a decrease in myocyte size by 13%, total collagen by 27% and TNF-α by 49% in the CRT group vs 28%, 45%, and 45% in the LVAD group. CRT produces cellular reverse remodeling in failing human hearts that are comparable with those produced by LVAD therapy., (© 2011 Wiley Periodicals, Inc.)
- Published
- 2011
- Full Text
- View/download PDF
30. Reversal of secondary pulmonary hypertension by axial and pulsatile mechanical circulatory support.
- Author
-
Torre-Amione G, Southard RE, Loebe MM, Youker KA, Bruckner B, Estep JD, Tierney M, and Noon GP
- Subjects
- Adult, Aged, Female, Heart Failure complications, Heart Failure mortality, Hemodynamics, Humans, Hypertension, Pulmonary complications, Hypertension, Pulmonary mortality, Male, Middle Aged, Retrospective Studies, Risk Factors, Treatment Outcome, Heart Failure surgery, Heart-Assist Devices, Hypertension, Pulmonary surgery
- Abstract
Background: Pulmonary hypertension associated with chronic congestive heart failure posses a significant risk of morbidity and death after heart transplantation. Isolated observations suggest that chronic ventricular unloading may lead to normalization of pulmonary pressures and thus render a patient likely to be a heart transplant candidate., Methods: This study is a retrospective analysis of 9 heart failure patients with secondary pulmonary hypertension (transpulmonary gradient [TPG] > 15 mm/Hg). Two were treated with a pulsatile left ventricular assist device (LVAD) and 7 with an axial-flow LVAD., Results: After LVAD support, mean pulmonary artery pressure decreased from 39 +/- 7 to 31 +/- 5 mm Hg, and the TPG decreased from 19 +/- 3 to 13 +/- 4 mm Hg (p < 0.01). The 1-year Kaplan-Meier survival curve for patients with pre-LVAD TPG > 15 mm Hg vs those with TPG < 15 mm Hg showed no difference in survival (p = 0.6). This finding was supported by analysis of a large multi-institutional cohort obtained from the Organ Procurement and Transplantation Network database, where no differences in survival were found in the same groups., Conclusions: Pulmonary hypertension that is secondary to congestive heart failure, as defined by a TPG > 15 mm Hg can be reversed by the use of pulsatile and axial-flow LVADs; furthermore, post-transplant survival for patients with secondary pulmonary hypertension treated with an LVAD was no different than for those without pulmonary hypertension who received LVAD support.
- Published
- 2010
- Full Text
- View/download PDF
31. Therapeutic plasma exchange a potential strategy for patients with advanced heart failure.
- Author
-
Torre-Amione G, Orrego CM, Khalil N, Kottner-Assad C, Leveque C, Celis R, Youker KA, and Estep JD
- Subjects
- Adult, B-Lymphocytes immunology, Chronic Disease, Female, Heart Failure immunology, Humans, Immunity, Humoral, Immunoglobulin G blood, Lymphocyte Activation, Male, Middle Aged, Myocardium immunology, Heart Failure therapy, Plasma Exchange
- Abstract
Background: Previous reports had emphasized the importance of humoral immunity in heart failure in humans, primarily determined by the presence of circulating antibodies. However, there is little or no information about the frequency of anticardiac antibodies present in failing human myocardium., Methods: Clinical data and myocardial tissue samples were analyzed to determine the role of humoral immunity in patients with chronic heart failure (CHF) in different SETTINGS., Results: Anticardiac antibodies were found present in failing hearts but not in normal control hearts. Further, the level of expression of these anticardiac antibodies changed with the severity of the disease state; and in patients with acute heart failure, we found selective activation of B cells. Finally, treatment of CHF patients with therapeutic plasma exchange, a strategy that removes circulating antibodies, resulted in a reduction in anticardiac antibody deposition and improvements in cardiac function., Conclusion: These data collectively suggest a role of humoral immunity in the progression of heart failure., (Copyright © 2010 Wiley-Liss, Inc.)
- Published
- 2010
- Full Text
- View/download PDF
32. Mast cell burden and reticulin fibrosis in the myeloproliferative neoplasms: a computer-assisted image analysis study.
- Author
-
Ahmed A, Powers MP, Youker KA, Rice L, Ewton A, Dunphy CH, and Chang CC
- Subjects
- Fibrosis, Humans, Image Interpretation, Computer-Assisted, Immunohistochemistry, Mast Cells pathology, Myeloproliferative Disorders immunology, Myeloproliferative Disorders pathology, Reticulin metabolism
- Abstract
The mast cell has been associated with fibrosis in many different tissues, organs, and different disease processes including hematopoietic malignancies. Mast cells are often increased in the bone marrow of patients with primary bone marrow disorders, and patients with systemic mastocytosis often have a second concomitant neoplastic disease of the bone marrow. The goals of the current study were to determine the role the mast cell has in the pathogenesis of myeloproliferative neoplasms (MPN) and to correlate the mast cell burden with the degree of reticulin fibrosis. We used computer-assisted image analysis of bone marrow core biopsies stained for mast cell tryptase from patients with myeloproliferative neoplasms [31 cases: 12 chronic myelogenous leukemia (CML), 6 primary myelofibrosis (PMF), 4 essential thrombocythemia (ET), 4 polycythemia vera (PV), and 5 chronic myeloproliferative disorder, unclassifiable (CMPD-U)]. Although the number of cases of some subtypes of MPN was small, the results suggested that PMF and ET each had significantly more mast cells than both CML and control cases (P<0.01 and 0.05, respectively, Mann-Whitney test). CMPD-U and PV showed no significant differences from the control cases, but the CML cases had significantly fewer mast cells than our control cases (P=0.02, Mann-Whitney test). In addition, the quantity of mast cells seen in the bone marrows of MPN patients correlated with reticulin fibrosis (P=0.04, Mann-Whitney test). Our studies highlight the different mast cell quantities in different myeloproliferative neoplasms and suggest a direct role for the mast cell in intramedullary fibrosis. Further studies are warranted to confirm our observation and to study the mechanisms by which mast cells contribute to fibrosis in the MPN setting.
- Published
- 2009
- Full Text
- View/download PDF
33. Increased expression of stem cell factor and its receptor after left ventricular assist device support: a potential novel target for therapeutic interventions in heart failure.
- Author
-
Jahanyar J, Youker KA, Torre-Amione G, Koerner MM, Bruckner B, Noon GP, and Loebe M
- Subjects
- Adult, Aged, Case-Control Studies, Female, Gene Expression, Heart Failure therapy, Heart Transplantation, Heart-Assist Devices, Humans, Immunohistochemistry, In Situ Hybridization, Male, Middle Aged, Reverse Transcriptase Polymerase Chain Reaction, Heart Failure metabolism, Mast Cells metabolism, Proto-Oncogene Proteins c-kit biosynthesis, Stem Cell Factor biosynthesis
- Abstract
Background: Left ventricular assist devices (LVADs) cause an influx of mast cells into the failing heart, but the underlying mechanism is unknown. This study investigates the potential role of stem cell factor (SCF) and its receptor (c-Kit) in promoting the recruitment of mast cells during heart failure and after LVAD support., Methods: Myocardial samples were collected from 10 end-stage heart failure patients undergoing LVAD implantation (pre-LVAD) and paired with samples taken at the time of orthotopic heart transplantation (post-LVAD). Biopsies of normal hearts served as controls. We assessed gene expression of SCF and c-Kit. In addition, we stained for SCF, c-Kit, tryptase and chymase, and utilized in situ hybridization to determine the origin of SCF., Results: SCF mRNA and overall mast cell numbers were significantly increased (p < 0.01/p < 0.001) after LVAD support as compared with paired heart failure tissues. c-Kit mRNA was significantly increased post-LVAD compared with normal tissues (p < 0.05). The c-Kit protein was expressed only in cardiac mast cells. SCF mRNA was found in endothelial cells, myocytes and interstitial cells, as confirmed by antibody staining., Conclusions: LVADs cause an increase of SCF and c-Kit gene expression, which coincides with a surge of mast cells after ventricular unloading. This suggests that SCF functions as an important mediator for the recruitment of mast cells to the mechanically unloaded human heart.
- Published
- 2008
- Full Text
- View/download PDF
34. The role of mast cells after solid organ transplantation.
- Author
-
Jahanyar J, Koerner MM, Loebe M, Youker KA, Torre-Amione G, and Noon GP
- Subjects
- Acute Disease, Chronic Disease, Graft Rejection, Hematopoietic Stem Cells physiology, Humans, Immunosuppressive Agents therapeutic use, Transplantation Immunology, Transplantation, Homologous physiology, Mast Cells physiology, Organ Transplantation physiology
- Abstract
Mast cells are best known as primary responders in allergic reactions, including anaphylaxis and asthma. However, recent studies have shown that mast cells are functionally diverse cells with immunoregulatory properties that influence both the innate and adaptive immunities. Mast cells are capable of producing an array of both proinflammatory and anti-inflammatory mediators, acting as antigen-presenting cells, and expressing a spectrum of costimulatory molecules. Moreover, mast cells seem to confer a certain degree of immune privilege to tissues in concert with T-regulatory cells and are essential players in fibrotic conditions. The following review of the literature serves to further define the role of mast cells in the immunologic reactions affecting transplanted solid organ grafts.
- Published
- 2008
- Full Text
- View/download PDF
35. Mast cell-derived cathepsin g: a possible role in the adverse remodeling of the failing human heart.
- Author
-
Jahanyar J, Youker KA, Loebe M, Assad-Kottner C, Koerner MM, Torre-Amione G, and Noon GP
- Subjects
- Adult, Aged, Angiotensin II metabolism, Cardiac Output, Low pathology, Cardiac Output, Low surgery, Case-Control Studies, Cathepsin G, Cathepsins antagonists & inhibitors, Chymases antagonists & inhibitors, Chymases metabolism, Disease Progression, Female, Heart-Assist Devices, Humans, Male, Mast Cells pathology, Middle Aged, Myocardium metabolism, Necrosis prevention & control, Tryptases physiology, Cardiac Output, Low physiopathology, Cathepsins physiology, Mast Cells physiology, Serine Endopeptidases physiology, Ventricular Remodeling physiology
- Abstract
Background: The role of cardiac mast cells (MCs) in the progression to heart failure has recently become increasingly evident. Cathepsin g is a neutrophil- and mast cell-derived protease, which can convert angiotensin I to angiotensin II and thereby activate the TGF-beta pathway, resulting in myocyte necrosis, hypertrophy, and increased fibrosis. This study focuses on mast cell-derived cathepsin g in the human heart during heart failure and following mechanical unloading by means of heart-assist devices (LVADs)., Materials and Methods: Myocardial tissue was obtained from 10 patients with end-stage cardiomyopathy at the time of LVAD implantation (pre-LVAD) and following orthotopic heart transplantation (post-LAVD). In addition, biopsies of four normal hearts served as a control group. Paraffin-embedded sections were dual stained for cathepsin g and tryptase, a known marker for mast cells, using standard immunohistochemistry protocols. Total cathepsin g positive mast cells were counted., Results: No cathepsin g positive MCs were found in normal hearts. However, we found evidence for cathepsin g in cardiac MCs in heart failure tissues (pre-LVAD). During heart failure, 46% of total MCs were cathepsin g positive as compared to after mechanical unloading, where only 11% of total MCs were cathepsin g positive (P<0.001)., Conclusion: Heart failure causes an increase of myocardial MCs. We have provided evidence that cathepsin g positive MCs accumulate during heart failure and their total percentage decreases after ventricular unloading. This coincides with the decrease in myocyte necrosis, hypertrophy, and fibrosis. Thus, cathepsin g may play a role in the progression to heart failure by activating angiotensin II, leading to detrimental effects on the heart.
- Published
- 2007
- Full Text
- View/download PDF
36. Lack of NF-kappaB1 (p105/p50) attenuates unloading-induced downregulation of PPARalpha and PPARalpha-regulated gene expression in rodent heart.
- Author
-
Razeghi P, Wang ME, Youker KA, Golfman L, Stepkowski S, and Taegtmeyer H
- Subjects
- Animals, Biomechanical Phenomena, Cell Size, DNA metabolism, Gene Expression Regulation, Genes, Developmental, I-kappa B Kinase metabolism, I-kappa B Proteins metabolism, Male, Mice, Mice, Knockout, Myocytes, Cardiac pathology, NF-kappa B metabolism, NF-kappa B p50 Subunit genetics, NF-kappa B p50 Subunit metabolism, NF-kappa B p52 Subunit genetics, NF-kappa B p52 Subunit metabolism, Protein Binding, Rats, Rats, Wistar, Down-Regulation, Heart Transplantation, Myocardium metabolism, NF-kappa B genetics, PPAR alpha metabolism
- Abstract
Objective: Unloading of the rodent heart activates the fetal gene program, decreases peroxisome proliferator-activated receptor alpha (PPARalpha) and PPARalpha-regulated gene expression (MCAD), and induces cardiomyocyte atrophy. NF-kappaB regulates the fetal gene program and PPARalpha-regulated gene expression during cardiac hypertrophy and induces atrophy in skeletal muscle. Our objective was to test the hypothesis that NF-kappaB is the regulator for activation of the fetal gene program, for downregulation of PPARalpha and PPARalpha-regulated gene expression, and for cardiomyocyte atrophy in the heart subjected to mechanical unloading., Methods: Activation of the inhibitory kappa B kinase beta (IKKbeta)/NF-kappaB pathways were measured in the heterotopically transplanted rat heart using Western blotting of total and phospho-IKKbeta and using transcription factor ELISA's for the five members of the NF-kappaB family (p65 (Rel A), p105/p50, c-Rel, RelB, and p100/p52). In loss of function experiments, we transplanted hearts of p105/p50 knockout mice into wildtype mice and compared changes in gene expression and cardiomyocyte size with wildtype hearts transplanted into wildtype mice., Results: Total and phospho-IKKbeta levels significantly increased in the transplanted heart seven days after surgery. The activation of IKKbeta was paralleled by increased DNA binding activity of p65 and p105/p50. Mechanical unloading induced myosin heavy chain beta expression and decreased cardiomyocyte size in hearts of both wildtype and p105/p050 knockout animals. In contrast, the downregulation of PPARalpha and MCAD was significantly attenuated or prevented in the hearts of p105/p50 knockout mice., Conclusions: The IKKbeta/p65/p50 pathway is activated in the unloaded rodent heart and a likely regulator for the downregulation of PPARalpha and PPARalpha-regulated gene expression.
- Published
- 2007
- Full Text
- View/download PDF
37. Mechanical unloading of the heart activates the calpain system.
- Author
-
Razeghi P, Volpini KC, Wang ME, Youker KA, Stepkowski S, and Taegtmeyer H
- Subjects
- Animals, Calcium-Binding Proteins biosynthesis, Cell Size, Disease Models, Animal, Gene Expression Regulation, Heart Failure pathology, Heart Failure surgery, Humans, Male, Mice, Middle Aged, Myocardium pathology, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology, Rats, Time Factors, Calpain biosynthesis, Heart Failure metabolism, Heart Transplantation, Heart-Assist Devices, Myocardium metabolism, Transplantation, Heterotopic
- Abstract
The mechanism for the decrease in cardiomyocyte size with mechanical unloading is unknown. The calpain system regulates cardiomyocyte atrophy. We obtained samples from failing human hearts at the time of implantation and explantation of a left ventricular assist device. For mechanical unloading, we also heterotopically transplanted rat or mouse hearts for 1 week. The effect of calpain inhibition on cardiac atrophy was assessed in transplanted hearts overexpressing calpastatin. We measured transcript levels of calpain 1 and 2 in the human and the rodent model, as well as calpain activity, a calpain-specific degradation product and cardiomyocyte size in the two rodent models. Mechanical unloading of the failing human heart significantly increased calpain 2 gene expression. Transcript levels of calpain 1 and 2, calpain activity and a calpain-specific degradation product all significantly increased in the unloaded rat heart. Unexpectedly, in hearts of animals overexpressing calpastatin, cardiomyocyte size also decreased. Mechanical unloading of the mammalian heart activates the calpain system, although other proteolytic systems may compensate for decreased calpain activity when calpastatin is overexpressed.
- Published
- 2007
- Full Text
- View/download PDF
38. Recurrent device thrombi during mechanical circulatory support with an axial-flow pump is a treatable condition and does not preclude successful long-term support.
- Author
-
Jahanyar J, Noon GP, Koerner MM, Youker KA, Malaisrie SC, Ngo UQ, Torre-Amione G, and Loebe M
- Subjects
- Humans, Male, Middle Aged, Recurrence, Remission Induction, Heart Diseases drug therapy, Heart Diseases etiology, Heart-Assist Devices adverse effects, Thrombolytic Therapy, Thrombosis drug therapy, Thrombosis etiology
- Abstract
The formation of pump thrombus after implantation of axial-flow ventricular assist devices has been described previously. We report a case of recurrent pump thrombus formation in a 63-year-old man who was bridged to heart transplantation with a MicroMed DeBakey ventricular assist device. He was treated with a low, dose-adjusted thrombolytic treatment protocol, and was subsequently successfully bridged to transplantation. Transient pump thrombus formation does not preclude safe long-term support with an axial-flow pump when diagnosed and treated appropriately.
- Published
- 2007
- Full Text
- View/download PDF
39. Decorin-mediated transforming growth factor-beta inhibition ameliorates adverse cardiac remodeling.
- Author
-
Jahanyar J, Joyce DL, Southard RE, Loebe M, Noon GP, Koerner MM, Torre-Amione G, and Youker KA
- Subjects
- Adolescent, Adult, Aged, Cells, Cultured, Collagen biosynthesis, Collagen genetics, Decorin, Endomyocardial Fibrosis etiology, Endomyocardial Fibrosis metabolism, Female, Fibroblasts drug effects, Fibroblasts metabolism, Fibroblasts pathology, Gene Expression Regulation drug effects, Heart Failure metabolism, Heart Failure pathology, Heart Ventricles metabolism, Heart Ventricles pathology, Humans, Male, Microscopy, Fluorescence, Middle Aged, Polymerase Chain Reaction, RNA, Messenger genetics, Transforming Growth Factor beta metabolism, Treatment Outcome, Endomyocardial Fibrosis prevention & control, Extracellular Matrix Proteins pharmacology, Heart Failure therapy, Heart Ventricles physiopathology, Heart-Assist Devices adverse effects, Proteoglycans pharmacology, Transforming Growth Factor beta antagonists & inhibitors, Ventricular Remodeling drug effects
- Abstract
Background: Implantation of a left ventricular assist device (LVAD) has been shown to induce regression of fibrosis in patients with congestive heart failure (CHF) and improve myocardial function. The mechanism of reverse remodeling after mechanical circulatory support (MCS), however, has not been fully characterized. In this study we examined the anti-fibrotic effects of decorin, an extracellular matrix (ECM) proteoglycan, on the transforming growth factor-beta (TGF-beta) pathway., Methods: Human myocardial tissue samples were obtained from patients undergoing LVAD implantation and again following subsequent transplantation after a sustained period of MCS. The specimens were examined by utilizing different molecular and histologic techniques, including human cardiac fibroblast in vitro studies. We assessed gene expression, mRNA and protein levels., Results: We found a significant decrease in interstitial fibrosis after MCS, with a decrease in collagen mRNA transcription rates, serving as an indirect measurement of collagen synthesis. Both the mRNA and protein levels of decorin were significantly increased after a period of MCS. Decorin mRNA was up-regulated by 44% after MCS (p < 0.01), which paralleled the increase in interstitial decorin deposition (p < 0.001). In addition, p-SMAD2, a molecular marker downstream of the TGF-beta pathway, was found to be inactivated after MCS (p < 0.02). Moreover, cultured human cardiac fibroblasts exposed to TGF-beta demonstrated decreased collagen production when exogenous decorin was added (p < 0.03)., Conclusions: The decorin molecule is potentially involved in reverse cardiac remodeling, by directly inhibiting the TGF-beta pathway and its pro-fibrotic effects on the failing human heart.
- Published
- 2007
- Full Text
- View/download PDF
40. Myocardial contrast echocardiography of radiofrequency ablation lesions.
- Author
-
Panescu D, Rao L, Ding C, Sun H, Youker KA, Nagueh SF, and Khoury DS
- Subjects
- Animals, Dogs, Female, Heart Conduction System physiology, Male, Myocardium, Catheter Ablation methods, Echocardiography methods, Heart physiopathology, Heart Ventricles diagnostic imaging
- Abstract
Introduction: The study tested the feasibility of differentiating radiofrequency ablation lesions from normal myocardium and quantifying their dimensions by myocardial contrast echocardiography (MCE)., Methods and Results: In 11 normal dogs, we created 14 focal and 4 linear lesions at different left ventricular sites.MCE was performed both before and after ablation by using an intracardiac echocardiography catheter (9 MHz)and infusing contrast microbubbles through the left coronary artery. An independent observer examined the lesion pathology. We found that intracardiac echocardiography alone could not delineate lesion dimensions. However, after ablation, MCE localized the lesions as well-defined, low-contrast areas within the normally opacified myocardium. Lesion dimensions byMCE immediately after ablation and 30 minutes later were similar. In 12 focal lesions, the average maximum depth (5.55 +/- 1.38 mm) and average maximum diameter(10.38 +/- 2.09 mm) by MCE were in excellent agreement with the pathologic depth (5.20 +/- 1.45 mm) and diameter(10.61 +/- 1.67 mm). Two focal lesions could not be detected by MCE and later were found to be superficial. Three-dimensional MCE correctly reconstructed the extent and shape of linear lesions compared to pathology (length: 18.7+/- 5.7 vs 18.5 +/- 5.6 mm; maximum longitudinal cross-sectional area: 81.2 +/- 9.6 vs 76.0 +/-10.3 mm(2))., Conclusion: MCE accurately localized and quantified radiofrequency ablation lesions in the normal leftv entricle. This new application of MCE may advance'ablation for managing ventricular arrhythmias that involve intramural or epicardial regions by providing instantaneous anatomic feedback on the effects of ablation during catheterization.
- Published
- 2006
- Full Text
- View/download PDF
41. Quantitative changes in mast cell populations after left ventricular assist device implantation.
- Author
-
Akgul A, Youker KA, Noon GP, and Loebe M
- Subjects
- Adult, Aged, Cell Count, Female, Heart Failure pathology, Humans, Male, Middle Aged, Myocardium pathology, Heart-Assist Devices, Mast Cells pathology
- Abstract
Mast cells have been implicated as important in tissue remodeling and fibrosis. We investigated the effect of mechanical ventricular unloading upon myocardial fibrosis and cardiac mast cell density in patients undergoing left ventricular assist device (LVAD) implantation. Paired myocardial tissue samples were obtained from 30 patients with end-stage cardiomyopathy at the time of LVAD implantation and at the time of removal and were compared with samples taken from donor hearts. Tissue sections were stained and quantitated for mast cells and myocardial fibrosis. Mast cell density (tryptase positive cells) in cardiomyopathy was higher than that in donor hearts (33.5 +/- 3.6 SEM cells/10 fields vs.15.2 +/- 2.0 SEM cells/10 fields respectively, p = 0.04) and was lower than LVAD supported hearts (33.5 +/- 3.6 SEM cells/10 fields vs. 49.8 +/- 5.7 SEM cells/10 fields respectively, p = 0.01). Mast cells are primarily localized in areas of increased interstitial fibrosis adjacent to myocardial cells and not vessels. There was statistically significant correlation between mast cells and interstitial collagen (p = 0.03) in patients before LVAD implantation that did not persist after mechanical support (p = 0.18). These results suggest that mechanical support with left ventricular assist devices induces an increase in mast cell number in the myocardium and an associated decrease in myocardial fibrosis. We believe these data demonstrate a dual role for cardiac mast cells in the increase in fibrosis in heart failure and the decrease after LVAD and its associated cardiac improvement.
- Published
- 2005
- Full Text
- View/download PDF
42. Fluorescence imaging microscopy of cellular markers in ischemic vs non-ischemic cardiomyopathy after left ventricular unloading.
- Author
-
Bick RJ, Bagwell SH, Jones CE, Poindexter BJ, Buja LM, Youker KA, Grigore A, Clubb F, Radovancevic B, and Frazier OH
- Subjects
- Atrial Natriuretic Factor metabolism, Biomarkers metabolism, Biopsy, Blotting, Western, Cardiomyopathy, Dilated pathology, Cardiomyopathy, Dilated therapy, Device Removal, Electrophoresis, Heart Ventricles metabolism, Heart Ventricles ultrastructure, Humans, Microscopy, Fluorescence, Myocardial Ischemia pathology, Myocardial Ischemia therapy, Myocardium metabolism, Nitric Oxide Synthase metabolism, Severity of Illness Index, Tumor Necrosis Factor-alpha metabolism, Atrial Natriuretic Factor ultrastructure, Cardiomyopathy, Dilated metabolism, Heart-Assist Devices, Myocardial Ischemia metabolism, Myocardium ultrastructure, Nitric Oxide Synthase ultrastructure, Tumor Necrosis Factor-alpha ultrastructure
- Abstract
Background: The heart undergoes repair and initiates protective mechanisms via ventricular unloading. We examined the presence of 2 markers in pre-unloaded and post-unloaded human cardiac tissue that are important indicators of cardiac failure, tumor necrosis factor-alpha and inducible nitric oxide synthase. We also measured 2 nuclear transcription factors, NFkappaB50 and NFkappaB65, comparing quantities and localizations to determine if mechanical unloading reduced their presence, as these markers are also thought to be indicators of impending heart failure. Amounts and localizations in patients that had been diagnosed with either ischemic or non-ischemic cardiomyopathy were compared after mechanical unloading with a left ventricular assist device. To establish that unloading had been achieved, levels of atrial natriuretic protein were determined., Methods: Core biopsies were harvested at assist device implantation and removal. Fluorescence deconvolution microscopy image reconstructions of fluorescence probes were correlated with data obtained by western Blot and electrobility shift assays., Results: Statistically significant differences in localization and amounts of tumor necrosis factor and nitric oxide synthase were seen between pre- and post-assist device samples. Amounts of tumor necrosis factor and nitric oxide synthase in ischemic tissue were increased at the time of assist device removal, but decreased in dilated or idiomyopathic samples. Ventricular unloading resulted in reduced levels of natriuretic protein, with the greatest reduction being seen in ischemic tissue. Both NFkappaB50 and NFkappaB65 increased in ischemic tissue, but only NFkappaB50 in non-ischemic samples., Conclusions: Changes in localization of the factors and altered levels of cytokine and nitric oxide synthase indicate that the heart switches to a "protective and repair" mode, and mechanical unloading allows this transition to occur. Observed changes were dependent on the etiology of the disease.
- Published
- 2005
- Full Text
- View/download PDF
43. Placement of a left ventricular assist device in a patient with dextrocardia.
- Author
-
Southard RE, Nelson JC, Joyce DL, Thompson LO, Skrabal CA, Torre-Amione G, Youker KA, Noon GP, and Loebe M
- Subjects
- Comorbidity, Humans, Male, Middle Aged, Dextrocardia epidemiology, Heart Failure therapy, Heart-Assist Devices
- Abstract
Dextrocardia most commonly presents in the setting of situs inversus, but it may occur as an isolated anomaly with normal position of the abdominal organs. Herein we present a 54-year-old man with ischemic cardiomyopathy and dextrocardia with normal position of the abdominal organs who presented with an exacerbation of congestive heart failure requiring inotropic support as well as mechanical ventilation. An implantable, wearable left ventricular assist device was placed in this patient to allow for ambulation and eventual discharge home. The patient survived 4 months before he developed pneumonia and expired.
- Published
- 2005
- Full Text
- View/download PDF
44. Organ-specific regulation of pro-inflammatory molecules in heart, lung, and kidney following brain death.
- Author
-
Skrabal CA, Thompson LO, Potapov EV, Southard RE, Joyce DL, Youker KA, Noon GP, and Loebe M
- Subjects
- Animals, Cytokines analysis, Female, Gene Expression Profiling, Intercellular Adhesion Molecule-1 genetics, Interleukin-1 genetics, Interleukin-6 genetics, Male, Organ Specificity, RNA, Messenger analysis, Receptors, Interleukin-6 genetics, Swine, Tumor Necrosis Factor-alpha genetics, Brain Death metabolism, Cytokines genetics, Kidney metabolism, Lung metabolism, Myocardium metabolism
- Abstract
Background: Nonspecific inflammatory events following brain death may increase the intensity of the immunological host response. The present study investigated the course of pro-inflammatory molecules in heart, lung, kidney, and plasma after brain death induction., Materials and Methods: Brain death was induced in five pigs by inflation of an intracranial Foley catheter and five pigs were sham-operated as controls. Each experiment was terminated 6 h after brain death/sham operation and the organs were harvested. We measured the mRNA and protein levels for TNF-alpha, IL-1beta, and IL-6 in heart, lung, kidney, and plasma. Additionally, the mRNA expression for IL-6R, ICAM-1, MCP-1, and TGF-beta was determined in each organ., Results: After 6 h, the plasma cytokine levels were higher in the brain-dead animals than in the sham-operated. In heart, lung, and kidney there was an increase in IL-6 and IL-1beta following brain death, while TNF-alpha was up-regulated in lung only (P < 0.05). MCP-1 and TGF-beta were significantly higher in heart and lung and IL-6R increased in heart after brain death (P < 0.05)., Conclusions: Brain death was associated with non-uniform cytokine expression patterns in the investigated organs. These expression patterns may cause variable pro-inflammatory priming resulting in different degrees of damage and explain the organ-specific variation in outcomes after transplantations.
- Published
- 2005
- Full Text
- View/download PDF
45. Plasma neurohormone levels correlate with left ventricular functional and morphological improvement in LVAD patients.
- Author
-
Thompson LO, Skrabal CA, Loebe M, Lafuente JA, Roberts RR, Akgul A, Jones V, Bruckner BA, Thohan V, Noon GP, and Youker KA
- Subjects
- Adult, Aged, Female, Fibrosis, Humans, Male, Middle Aged, Myocardium pathology, Brain-Derived Neurotrophic Factor blood, Endothelin-1 blood, Heart-Assist Devices, Ventricular Function, Left
- Abstract
Objective: Brain natriuretic peptide (BNP) and endothelin-1 (ET-1) have been shown to be markers of left ventricular (LV) function. To determine the feasibility of using serial assays of these neurohormones in the assessment of cardiac status in the left ventricular assist device (LVAD) setting, we examined the relationship between LV function, myocardial morphology, and plasma levels of these hormones in LVAD recipients., Methods: Plasma BNP and ET-1 levels were serially assayed in 19 end-stage congestive heart failure (CHF) patients before and after LVAD implantation with various devices (i.e., MicroMed DeBakeyVAD/DVAD, Novacor/NVAD, TCI Heartmate/TCI, Thoratec/TVAD). Echocardiography performed correspondingly at the time points of the hormonal assays and immunohistochemical collagen staining of left ventricular tissue samples, derived from six non-failing hearts as well as from LVAD patients at the time of device insertion and removal, were then contrasted. Patients were grouped according to device used and etiology of heart disease (ischemic or dilated cardiomyopathy, ICM/DCM)., Results: LVAD therapy significantly improved LV ejection fraction (EF%: 21 +/- 3.8% to 28.11 +/- 3.57%), cardiac output (CO: 3.49 +/- 1.3 to 7.3 +/- 0.2 l/m), and left ventricular end-diastolic diameter (LVEDD: 6.68 +/- 0.92 versus 4.79 +/- 1.54 cm, P < 0.0001) in all patients. Absolute BNP and ET-1 plasma levels remained significantly lower in all patients after LVAD implantation (both P < 0.001). The NVAD group exhibited the most BNP reduction and EF% increase (P < 0.0004 and P < 0.038, respectively). Average collagen levels were reduced in all patients (P < 0.0005). Among the devices, the NVAD group demonstrated the most evident change (P < 0.0036), while there was comparable reduction in the DCM and ICM groups (both P < 0.03). In general, postoperative BNP and ET-1 trends exhibited a notable parallelism with both manifesting bi-phasic tendencies and an inverse proportionality to corresponding EF% measurements., Conclusions: Device selection appears to influence the cardiac morphological and neurohormonal expressive tendencies exhibited by recipients. Plasma BNP and ET-1 levels correlate with both LV function and myocardial morphological improvement. Alterations in the levels of these hormones during LVAD support may be real-time indicators of prevailing myocardial autocrine/paracrine activity and as such may be of potential use in future algorithms of cardiac assessment and therapeutic decision-making with regard to transplant urgency and/or possible device explantation.
- Published
- 2005
- Full Text
- View/download PDF
46. Activation of cardiac Cdk9 represses PGC-1 and confers a predisposition to heart failure.
- Author
-
Sano M, Wang SC, Shirai M, Scaglia F, Xie M, Sakai S, Tanaka T, Kulkarni PA, Barger PM, Youker KA, Taffet GE, Hamamori Y, Michael LH, Craigen WJ, and Schneider MD
- Subjects
- Animals, Apoptosis, Cells, Cultured, Cyclin T, Cyclins metabolism, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Enzyme Activation, Gene Expression, Heart Failure genetics, Heart Failure metabolism, Humans, Mice, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology, NF-E2-Related Factor 1, Nuclear Respiratory Factor 1, Nuclear Respiratory Factors, Promoter Regions, Genetic, RNA Polymerase II metabolism, Rats, Trans-Activators genetics, Trans-Activators metabolism, Transcription Factors genetics, Transcription, Genetic, Cyclin-Dependent Kinase 9 metabolism, Heart Failure etiology, Myocardium metabolism, Transcription Factors metabolism
- Abstract
Hypertrophy allows the heart to adapt to workload but culminates in later pump failure; how it is achieved remains uncertain. Previously, we showed that hypertrophy is accompanied by activation of cyclin T/Cdk9, which phosphorylates the C-terminal domain of the large subunit of RNA polymerase II, stimulating transcription elongation and pre-mRNA processing; Cdk9 activity was required for hypertrophy in culture, whereas heart-specific activation of Cdk9 by cyclin T1 provoked hypertrophy in mice. Here, we report that alphaMHC-cyclin T1 mice appear normal at baseline yet suffer fulminant apoptotic cardiomyopathy when challenged by mechanical stress or signaling by the G-protein Gq. At pathophysiological levels, Cdk9 activity suppresses many genes for mitochondrial proteins including master regulators of mitochondrial function (peroxisome proliferator-activated receptor gamma coactivator 1 (PGC-1), nuclear respiratory factor-1). In culture, cyclin T1/Cdk9 suppresses PGC-1, decreases mitochondrial membrane potential, and sensitizes cardiomyocytes to apoptosis, effects rescued by exogenous PGC-1. Cyclin T1/Cdk9 inhibits PGC-1 promoter activity and preinitiation complex assembly. Thus, chronic activation of Cdk9 causes not only cardiomyocyte enlargement but also defective mitochondrial function, via diminished PGC-1 transcription, and a resulting susceptibility to apoptotic cardiomyopathy.
- Published
- 2004
- Full Text
- View/download PDF
47. Localizing and quantifying ablation lesions in the left ventricle by myocardial contrast echocardiography.
- Author
-
Khoury DS, Rao L, Ding C, Sun H, Youker KA, Panescu D, and Nagueh SF
- Subjects
- Animals, Contrast Media, Dogs, Feasibility Studies, Female, Heart Conduction System, Male, Prospective Studies, Tachycardia, Ventricular diagnostic imaging, Tachycardia, Ventricular surgery, Catheter Ablation, Echocardiography, Heart Ventricles diagnostic imaging, Heart Ventricles surgery, Myocardium
- Abstract
Introduction: The inability to determine the extent and intramural depth of ablation lesions can hamper the success of catheter ablation. The study tested the feasibility of differentiating radiofrequency ablation lesions from normal myocardium and quantifying their dimensions by myocardial contrast echocardiography (MCE)., Methods and Results: In 11 normal dogs, we created 14 focal and 4 linear lesions at different left ventricular sites. MCE was performed both before and after ablation by using an intracardiac echocardiography catheter (9 MHz) and infusing contrast microbubbles through the left coronary artery. We initially used two-dimensional MCE to image focal lesions and subsequently three-dimensional MCE to image linear lesions. An independent observer examined the lesion pathology. We found that intracardiac echocardiography alone could not delineate lesion dimensions. However, after ablation, MCE localized the lesions as well-defined, low-contrast areas within the normally opacified myocardium. Lesion dimensions by MCE immediately after ablation and 30 minutes later were similar. In 12 focal lesions, the average maximum depth (5.55 +/- 1.38 mm) and average maximum diameter (10.38 +/- 2.09 mm) by MCE were in excellent agreement with the pathologic depth (5.20 +/- 1.45 mm) and diameter (10.61 +/- 1.67 mm). Two focal lesions could not be detected by MCE and later were found to be superficial. Three-dimensional MCE correctly reconstructed the extent and shape of linear lesions compared to pathology (length: 18.7 +/- 5.7 vs 18.5 +/- 5.6 mm; maximum longitudinal cross-sectional area: 81.2 +/- 9.6 vs 76.0 +/- 10.3 mm2)., Conclusion: MCE accurately localized and quantified radiofrequency ablation lesions in the normal left ventricle. This new application of MCE may advance ablation for managing ventricular arrhythmias that involve intramural or epicardial regions by providing instantaneous anatomic feedback on the effects of ablation during catheterization.
- Published
- 2004
- Full Text
- View/download PDF
48. Role of mast cells and their mediators in failing myocardium under mechanical ventricular support.
- Author
-
Akgul A, Skrabal CA, Thompson LO, Loebe M, Lafuente JA, Noon GP, and Youker KA
- Subjects
- Adult, Case-Control Studies, Cell Count, Extracellular Space, Female, Fibrosis, Heart Failure metabolism, Heart Failure pathology, Humans, Male, Middle Aged, Time Factors, Fibroblast Growth Factor 2 metabolism, Heart Failure therapy, Heart-Assist Devices, Mast Cells physiology, Myocardium metabolism, Myocardium pathology
- Abstract
Background: Mast cells have been implicated in tissue remodeling and fibroblast stimulation. We explored the effect of mechanical support by left ventricular assist device (LVAD) in failing myocardium and looked into grade and distribution of interstitial fibrosis, mast cell density, mast cell phenotypes and basic fibroblast growth factor (bFGF) expression pre- and post-LVAD., Methods: Myocardial tissue was obtained from 20 patients with end-stage cardiomyopathy at the time of LVAD implantation and LVAD removal and from 7 donor hearts not used for transplantation. Tissue sections were stained for mast cells using tryptase as a marker and the myocardial fibrosis was measured. Double staining for tryptase and chymase was performed for detection of chymase-positive mast cells. Fluorescent microscopy showed the relationship of mast cells to bFGF, and bFGF expression was quantified by Western blot., Results: There was a significant increase in mast cells in heart failure vs normal myocardium. A secondary increase in mast cells occurred after long-term (>40 days) support compared with matched pre-LVAD samples (mean +/- SEM; 57.4 +/- 8.6 cells/10 fields vs 45.1 +/- 7.6 SEM cells/10 fields, p < 0.01). The secondary increase in mast cells was associated specifically with an increase in chymase-negative mast cells (p < 0.01). These findings are statistically significant with concurrent decreased expression of bFGF and decreased fibrosis in the same patient tissues (p < 0.01)., Conclusions: We suggest that, under long-term support, there is a change in phenotypic expression in mast cells, which can alter fibroblast functions. The decreased myocardial bFGF levels might be the result of these phenotypically altered mast cells.
- Published
- 2004
- Full Text
- View/download PDF
49. Interaction between isolated human myocardial mast cells and cultured fibroblasts.
- Author
-
Skrabal CA, Thompson LO, Southard RE, Joyce DL, Noon GP, Loebe M, and Youker KA
- Subjects
- Cardiac Output, Low pathology, Cardiac Output, Low therapy, Cell Separation, Cells, Cultured, Coculture Techniques, Fibroblasts chemistry, HSP47 Heat-Shock Proteins, Heart-Assist Devices, Heat-Shock Proteins analysis, Humans, Ventricular Function, Left, Cardiac Output, Low physiopathology, Cell Communication, Fibroblasts physiology, Mast Cells physiology, Myocardium pathology
- Abstract
Introduction: Previously, we reported an increase in interstitial collagen and total mast-cell numbers in heart failure versus normal myocardium. A secondary increase, primarily in chymase-negative mast cells, occurred following LVAD support compared to matched pre-LVAD tissue samples and was associated with a decrease in interstitial collagen and bFGF. To further elucidate the changes in interstitial collagen, we investigated the direct interaction between mast cells, isolated from failing myocardium with or without previous LVAD support, and human fibroblasts in a coculture model. Additionally, the expression of HSP-47, the pro-collagen-specific chaperone protein, was determined in the particular myocardium., Materials and Methods: Myocardial tissue was obtained from 10 patients with end-stage dilated cardiomyopathy (DCM) at the time of transplantation. Five patients were transplanted following LVAD support, five patients without previous LVAD support. Mast cells were isolated according to a standard protocol, including collagenase digestion and cell separation. The isolated mast cells were co-cultured with human fibroblasts for 12 h, with or without stimulation of degranulation, and protein synthesis was measured by [(3)H]-proline incorporation. HSP-47 immunostaining was performed in the different myocardial samples and the positive cells were quantified., Results: Stimulated mast cells isolated from DCM tissue (without previous LVAD support) caused a 92% increase in [(3)H]-proline incorporation and consequently in protein production in fibroblasts compared to mast-cell free culture (P < 0.01), while conversely stimulated mast cells isolated from LVAD supported myocardium decreased the [(3)H]-proline incorporation by 63% (P < 0.01) below baseline. Nonstimulated mast cells did not significantly alter the protein production over baseline. There was also a significant increase in the number of HSP-47-positive cells in DCM myocardium compared to normal (P < 0.01) and there was a shift toward normal after LVAD support (P < 0.01)., Conclusion: We demonstrate that fibroblast protein production in vitro is significantly altered by mast cells and that the direction of change is dependent on whether myocardium was supported by LVAD. We suggest that under long-term LVAD support there is a phenotypic alteration in myocardial mast cells, which leads to a change in concentration and/or composition of mediators, capable of re-remodeling the myocardial matrix.
- Published
- 2004
- Full Text
- View/download PDF
50. Apicoaortic conduit in a patient with severe hemolysis after three aortic valve replacements.
- Author
-
Skrabal CA, Loebe M, Thompson LO, Buergler JM, Youker KA, Lafuente JA, and Noon GP
- Subjects
- Adult, Aortic Valve Stenosis diagnostic imaging, Echocardiography, Transesophageal, Follow-Up Studies, Heart Valve Prosthesis Implantation methods, Hemolysis, Humans, Male, Reoperation statistics & numerical data, Risk Assessment, Severity of Illness Index, Treatment Outcome, Ventricular Outflow Obstruction diagnosis, Aortic Valve Stenosis surgery, Bioprosthesis adverse effects, Heart Valve Prosthesis adverse effects, Heart Valve Prosthesis Implantation adverse effects, Ventricular Outflow Obstruction surgery
- Published
- 2004
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.