Institute Affiliations


All Publications


  • Distribution of cardiomyocyte-selective adeno-associated virus serotype 9 vectors in swine following intracoronary and intravenous infusion. Physiological genomics Li, J., Kelly, S. C., Ivey, J. R., Thorne, P. K., Yamada, K. P., Aikawa, T., Mazurek, R., Turk, J. R., Silva, K. A., Amin, A. R., Tharp, D. L., Mueller, C. M., Thakur, H., Leary, E. V., Domeier, T. L., Rector, R. S., Fish, K., Cividini, F., Ishikawa, K., Emter, C. A., Kapiloff, M. S. 2022

    Abstract

    Limited reports exist regarding adeno-associated virus (AAV) biodistribution in swine. This study assessed biodistribution following antegrade intracoronary and intravenous delivery of two self-complementary serotype 9 AAV (AAV9sc) biologics designed to target signaling in the cardiomyocyte considered important for the development of heart failure. Under the control of a cardiomyocyte-specific promoter, AAV9sc.shmAKAP and AAV9sc.RBD express a small hairpin RNA for the perinuclear scaffold protein muscle A-kinase anchoring protein beta (mAKAPbeta) and an anchoring disruptor peptide for p90 ribosomal S6 kinase type 3 (RSK3), respectively. Quantitative PCR was used to assess viral genome (vg) delivery and transcript expression in Ossabaw and Yorkshire swine tissues. Myocardial viral delivery was 2-5 x 105 vg/g gDNA for both infusion techniques at a dose ~1013 vg/kg body weight, demonstrating a delivery of ~1-3 viral particles per cardiac diploid genome. Myocardial RNA levels for each expressed transgene were generally proportional to dose and genomic delivery, and comparable to levels for moderately expressed endogenous genes. Despite significant AAV9sc delivery to other tissues, including the liver, neither biologic induced toxic effects as assessed using functional, structural, and circulating cardiac and systemic markers. These results indicate successful targeted delivery of cardiomyocyte-selective viral vectors in swine without negative side effects, an important step in establishing efficacy in a preclinical experimental setting.

    View details for DOI 10.1152/physiolgenomics.00032.2022

    View details for PubMedID 35648460

  • Nuclear localized HDAC4 increases retinal ganglion cell survival after optic nerve crush injury Zhu, Y., Li, J., Bian, M., Xia, X., Nahmou, M., Sun, C., Goldberg, J. L., Kapiloff, M. ASSOC RESEARCH VISION OPHTHALMOLOGY INC. 2022
  • FGF21-FGFR4 signaling in cardiac myocytes promotes concentric cardiac hypertrophy in mouse models of diabetes. Scientific reports Yanucil, C., Kentrup, D., Li, X., Grabner, A., Schramm, K., Martinez, E. C., Li, J., Campos, I., Czaya, B., Heitman, K., Westbrook, D., Wende, A. R., Sloan, A., Roche, J. M., Fornoni, A., Kapiloff, M. S., Faul, C. 2022; 12 (1): 7326

    Abstract

    Fibroblast growth factor (FGF) 21, a hormone that increases insulin sensitivity, has shown promise as a therapeutic agent to improve metabolic dysregulation. Here we report that FGF21 directly targets cardiac myocytes by binding beta-klotho and FGF receptor (FGFR) 4. In combination with high glucose, FGF21 induces cardiac myocyte growth in width mediated by extracellular signal-regulated kinase 1/2 (ERK1/2) signaling. While short-term FGF21 elevation can be cardio-protective, we find that in type 2 diabetes (T2D) in mice, where serum FGF21 levels are elevated, FGFR4 activation induces concentric cardiac hypertrophy. As T2D patients are at risk for heart failure with preserved ejection fraction (HFpEF), we propose that induction of concentric hypertrophy by elevated FGF21-FGFR4 signaling may constitute a novel mechanism promoting T2D-associated HFpEF such that FGFR4 blockade might serve as a cardio-protective therapy in T2D. In addition, potential adverse cardiac effects of FGF21 mimetics currently in clinical trials should be investigated.

    View details for DOI 10.1038/s41598-022-11033-x

    View details for PubMedID 35513431

  • Targeting mAKAPbeta expression as a therapeutic approach for ischemic cardiomyopathy. Gene therapy Martinez, E. C., Li, J., Ataam, J. A., Tokarski, K., Thakur, H., Karakikes, I., Dodge-Kafka, K., Kapiloff, M. S. 1800

    Abstract

    Ischemic cardiomyopathy is a leading cause of death and an unmet clinical need. Adeno-associated virus (AAV) gene-based therapies hold great promise for treating and preventing heart failure. Previously we showed that muscle A-kinase Anchoring Protein beta (mAKAPbeta, AKAP6beta), a scaffold protein that organizes perinuclear signalosomes in the cardiomyocyte, is a critical regulator of pathological cardiac hypertrophy. Here, we show that inhibition of mAKAPbeta expression in stressed adult cardiomyocytes in vitro was cardioprotective, while conditional cardiomyocyte-specific mAKAP gene deletion in mice prevented pathological cardiac remodeling due to myocardial infarction. We developed a new self-complementary serotype 9 AAV gene therapy vector expressing a short hairpin RNA for mAKAPbeta under the control of a cardiomyocyte-specific promoter (AAV9sc.shmAKAP). This vector efficiently downregulated mAKAPbeta expression in the mouse heart in vivo. Expression of the shRNA also inhibited mAKAPbeta expression in human induced cardiomyocytes in vitro. Following myocardial infarction, systemic administration of AAV9sc.shmAKAP prevented the development of pathological cardiac remodeling and heart failure, providing long-term restoration of left ventricular ejection fraction. Our findings provide proof-of-concept for mAKAPbeta as a therapeutic target for ischemic cardiomyopathy and support the development of a translational pipeline for AAV9sc.shmAKAP for the treatment of heart failure.

    View details for DOI 10.1038/s41434-022-00321-w

    View details for PubMedID 35102273

  • Calcineurin Abeta-Specific Anchoring Confers Isoform-Specific Compartmentation and Function in Pathological Cardiac Myocyte Hypertrophy. Circulation Li, X., Li, J., Martinez, E. C., Froese, A., Passariello, C. L., Henshaw, K., Rusconi, F., Li, Y., Yu, Q., Thakur, H., Nikolaev, V. O., Kapiloff, M. S. 2020

    Abstract

    Background: The Ca2+/calmodulin-dependent phosphatase calcineurin is a key regulator of cardiac myocyte hypertrophy in disease. An unexplained paradox is how the Abeta isoform of calcineurin (CaNAbeta) is required for induction of pathological myocyte hypertrophy, despite calcineurin Aalpha expression in the same cells. In addition, it is unclear how the pleiotropic second messenger Ca2+ drives excitation-contraction coupling, while not stimulating hypertrophy via calcineurin in the normal heart. Elucidation of the mechanisms conferring this selectively in calcineurin signaling should reveal new strategies for targeting the phosphatase in disease. Methods: Primary adult rat ventricular myocytes were studied for morphology and intracellular signaling. New Forster Resonance Energy Transfer (FRET) reporters were used to assay Ca2+ and calcineurin activity in living cells. Conditional gene deletion and adeno-associated virus (AAV)-mediated gene delivery in the mouse were used to study calcineurin signaling following transverse aortic constriction in vivo. Results: Cdc42-interacting protein (CIP4/TRIP10) was identified as a new polyproline domain-dependent scaffold for CaNAbeta2 by yeast-2-hybrid screen. Cardiac myocyte-specific CIP4 gene deletion in mice attenuated pressure overload-induced pathological cardiac remodeling and heart failure. Accordingly, blockade of CaNAbeta polyproline-dependent anchoring using a competing peptide inhibited concentric hypertrophy in cultured myocytes, while disruption of anchoring in vivo using an AAV gene therapy vector inhibited cardiac hypertrophy and improved systolic function after pressure overload. Live cell FRET biosensor imaging of cultured myocytes revealed that Ca2+ levels and calcineurin activity associated with the CIP4 compartment were increased by neurohormonal stimulation, but minimally by pacing. Conversely, Ca2+ levels and calcineurin activity detected by non-localized FRET sensors were induced by pacing and minimally by neurohormonal stimulation, providing functional evidence for differential intracellular compartmentation of Ca2+ and calcineurin signal transduction. Conclusions: These results support a structural model for Ca2+ and CaNAbeta compartmentation in cells based upon an isoform-specific mechanism for calcineurin protein-protein interaction and localization. This mechanism provides an explanation for the specific role of CaNAbeta in hypertrophy and its selective activation under conditions of pathologic stress. Disruption of CaNAbeta polyproline-dependent anchoring constitutes a rational strategy for therapeutic targeting of CaNAbeta-specific signaling responsible for pathological cardiac remodeling in cardiovascular disease deserving of further pre-clinical investigation.

    View details for DOI 10.1161/CIRCULATIONAHA.119.044893

    View details for PubMedID 32611257

  • Signalosome-Regulated SRF Phosphorylation Determining Myocyte Growth in Width versus Length as a Therapeutic Target for Heart Failure. Circulation Li, J. n., Tan, Y. n., Passariello, C. L., Martinez, E. C., Kritzer, M. D., Li, X. n., Li, X. n., Li, Y. n., Yu, Q. n., Ohgi, K. n., Thakur, H. n., MacArthur, J. W., Ivey, J. R., Woo, Y. J., Emter, C. A., Dodge-Kafka, K. n., Rosenfeld, M. G., Kapiloff, M. S. 2020

    Abstract

    Background: Concentric and eccentric cardiac hypertrophy are associated with pressure and volume overload, respectively, in cardiovascular disease both conferring an increased risk of heart failure. These contrasting forms of hypertrophy are characterized by asymmetric growth of the cardiac myocyte in mainly width or length, respectively. The molecular mechanisms determining myocyte preferential growth in width versus length remain poorly understood. Identification of the mechanisms governing asymmetric myocyte growth could provide new therapeutic targets for the prevention or treatment of heart failure. Methods: Primary adult rat ventricular myocytes, adeno-associated virus (AAV)-mediated gene delivery in mice, and human tissue samples are used to define a regulatory pathway controlling pathological myocyte hypertrophy. Chromatin Immunoprecipitation Assays with Sequencing (ChIP-seq) and Precision Nuclear Run-On Sequencing (PRO-seq) are used to define a transcriptional mechanism. Results: Here we report that asymmetric cardiac myocyte hypertrophy is modulated by serum response factor (SRF) phosphorylation, constituting an epigenomic switch balancing the growth in width versus length of adult ventricular myocytes in vitro and in vivo. SRF Ser103 phosphorylation is bidirectionally regulated by p90 ribosomal S6 kinase type 3 (RSK3) and protein phosphatase 2A (PP2A) at signalosomes organized by the scaffold protein muscle A-kinase anchoring protein β (mAKAPβ), such that increased SRF phosphorylation activates Activator Protein 1 (AP1)-dependent enhancers that direct myocyte growth in width. AAV are used to express in vivo mAKAPβ-derived RSK3 and PP2A anchoring disruptor peptides that block the association of the enzymes with the mAKAPβ scaffold. Inhibition of RSK3 signaling prevents concentric cardiac remodeling due to pressure overload, while inhibition of PP2A signaling prevents eccentric cardiac remodeling induced by myocardial infarction, in each case improving cardiac function. SRF Ser103 phosphorylation is significantly decreased in dilated human hearts, supporting the notion that modulation of the mAKAPβ-SRF signalosome could be a new therapeutic approach for human heart failure. Conclusions: We have identified a new molecular switch, namely mAKAPβ signalosome-regulated SRF phosphorylation, that controls a transcriptional program responsible for modulating changes in cardiac myocyte morphology that occur secondary to pathological stressors. Complementary AAV-based gene therapies constitute rationally-designed strategies for a new translational modality for heart failure.

    View details for DOI 10.1161/CIRCULATIONAHA.119.044805

    View details for PubMedID 32933333

  • Regulation of Neuronal Survival and Axon Growth by a Perinuclear cAMP Compartment JOURNAL OF NEUROSCIENCE Boczek, T., Cameron, E. G., Yu, W., Xia, X., Shah, S. H., Chabeco, B., Galvao, J., Nahmou, M., Li, J., Thakur, H., Goldberg, J. L., Kapiloff, M. S. 2019; 39 (28): 5466–80
  • Muscle A-kinase-anchoring protein-beta-bound calcineurin toggles active and repressive transcriptional complexes of myocyte enhancer factor 2D JOURNAL OF BIOLOGICAL CHEMISTRY Li, J., Paris, S., Thakur, H., Kapiloff, M. S., Dodge-Kafka, K. L. 2019; 294 (7): 2543–54
  • Regulation of Neuronal Survival and Axon Growth by a Perinuclear cAMP Compartment. The Journal of neuroscience : the official journal of the Society for Neuroscience Boczek, T. n., Cameron, E. G., Yu, W. n., Xia, X. n., Shah, S. H., Castillo Chabeco, B. n., Galvao, J. n., Nahmou, M. n., Li, J. n., Thakur, H. n., Goldberg, J. L., Kapiloff, M. S. 2019

    Abstract

    Cyclic-AMP (cAMP) signaling is known to be critical in neuronal survival and axon growth. Increasingly the subcellular compartmentation of cAMP signaling has been appreciated, but outside of dendritic synaptic regulation, few cAMP compartments have been defined in terms of molecular composition or function in neurons. Specificity in cAMP signaling is conferred in large part by A-kinase anchoring proteins (AKAPs) that localize protein kinase A (PKA) and other signaling enzymes to discrete intracellular compartments. We now reveal that cAMP signaling within a perinuclear neuronal compartment organized by the large multivalent scaffold protein mAKAPα promotes neuronal survival and axon growth. mAKAPα signalosome function is explored using new molecular tools designed to specifically alter local cAMP levels as studied by live cell FRET imaging. In addition, enhancement of mAKAPα-associated cAMP signaling by isoform-specific displacement of bound phosphodiesterase is demonstrated to increase retinal ganglion cell survival in vivo in mice of both sexes following optic nerve crush injury. These findings define a novel neuronal compartment that confers cAMP regulation of neuroprotection and axon growth and that may be therapeutically targeted in disease.SIGNIFICANCE STATEMENTcAMP is a second messenger responsible for the regulation of diverse cellular processes including neuronal neurite extension and survival following injury. Signal transduction by cAMP is highly compartmentalized in large part due to the formation of discrete, localized multimolecular signaling complexes by A-kinase anchoring proteins. Although the concept of cAMP compartmentation is well-established, the function and identity of these compartments remain poorly understood in neurons. In this study, we provide evidence for a neuronal perinuclear cAMP compartment organized by the scaffold protein mAKAPα that is necessary and sufficient for the induction of neurite outgrowth in vitro and for the survival of retinal ganglion cells in vivo following optic nerve injury.

    View details for PubMedID 31097623

  • Muscle A-kinase-anchoring protein-beta-bound calcineurin toggles active and repressive transcriptional complexes of myocyte enhancer factor 2D. The Journal of biological chemistry Li, J., Aponte Paris, S., Thakur, H., Kapiloff, M. S., Dodge-Kafka, K. L. 2018

    Abstract

    Myocyte enhancer factor 2 (MEF2) transcription factors are key regulators of the development and adult phenotype of diverse tissues including skeletal and cardiac muscles. Controlled by multiple post-translational modifications, MEF2D is an effector for the Ca2+/calmodulin-dependent protein phosphatase calcineurin (CaN, PP2B, and PPP3). CaN-catalyzed dephosphorylation promotes the desumoylation and acetylation of MEF2D increasing its transcriptional activity. Both MEF2D and CaN bind the scaffold protein muscle A-kinase anchoring protein beta (mAKAPbeta), which is localized to the nuclear envelope, such that C2C12 skeletal myoblast differentiation and neonatal rat ventricular myocyte hypertrophy are inhibited by mAKAPbeta signalosome targeting. Using immunoprecipitation and DNA-binding assays, we now show that the formation of mAKAPbeta signalosomes is required for MEF2D dephosphorylation, desumoylation, and acetylation in C2C12 cells. Reduced MEF2D phosphorylation was coupled to a switch from type IIa histone deacetylase to p300 histone acetylase binding that correlated with increased MEF2D-dependent gene expression and ventricular myocyte hypertrophy. Together these results highlight the importance of mAKAPbeta signalosomes for regulating MEF2D activity in striated muscle, affirming mAKAPbeta as a nodal regulator in the myocyte intracellular signaling network.

    View details for PubMedID 30523159

  • Bidirectional regulation of HDAC5 by mAKAP beta signalosomes in cardiac myocytes JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY Dodge-Kafka, K. L., Gildart, M., Li, J., Thakur, H., Kapiloff, M. S. 2018; 118: 13–25

    Abstract

    Class IIa histone deacetylases (HDACs) are transcriptional repressors whose nuclear export in the cardiac myocyte is associated with the induction of pathological gene expression and cardiac remodeling. Class IIa HDACs are regulated by multiple, functionally opposing post-translational modifications, including phosphorylation by protein kinase D (PKD) that promotes nuclear export and phosphorylation by protein kinase A (PKA) that promotes nuclear import. We have previously shown that the scaffold protein muscle A-kinase anchoring protein β (mAKAPβ) orchestrates signaling in the cardiac myocyte required for pathological cardiac remodeling, including serving as a scaffold for both PKD and PKA. We now show that mAKAPβ is a scaffold for HDAC5 in cardiac myocytes, forming signalosomes containing HDAC5, PKD, and PKA. Inhibition of mAKAPβ expression attenuated the phosphorylation of HDAC5 by PKD and PKA in response to α- and β-adrenergic receptor stimulation, respectively. Importantly, disruption of mAKAPβ-HDAC5 anchoring prevented the induction of HDAC5 nuclear export by α-adrenergic receptor signaling and PKD phosphorylation. In addition, disruption of mAKAPβ-PKA anchoring prevented the inhibition by β-adrenergic receptor stimulation of α-adrenergic-induced HDAC5 nuclear export. Together, these data establish that mAKAPβ signalosomes serve to bidirectionally regulate the nuclear-cytoplasmic localization of class IIa HDACs. Thus, the mAKAPβ scaffold serves as a node in the myocyte regulatory network controlling both the repression and activation of pathological gene expression in health and disease, respectively.

    View details for PubMedID 29522762

    View details for PubMedCentralID PMC5940533

  • An AKAP-Lbc-RhoA interaction inhibitor promotes the translocation of aquaporin-2 to the plasma membrane of renal collecting duct principal cells. PloS one Schrade, K., Tröger, J., Eldahshan, A., Zühlke, K., Abdul Azeez, K. R., Elkins, J. M., Neuenschwander, M., Oder, A., Elkewedi, M., Jaksch, S., Andrae, K., Li, J., Fernandes, J., Müller, P. M., Grunwald, S., Marino, S. F., Vukićević, T., Eichhorst, J., Wiesner, B., Weber, M., Kapiloff, M., Rocks, O., Daumke, O., Wieland, T., Knapp, S., von Kries, J. P., Klussmann, E. 2018; 13 (1): e0191423

    Abstract

    Stimulation of renal collecting duct principal cells with antidiuretic hormone (arginine-vasopressin, AVP) results in inhibition of the small GTPase RhoA and the enrichment of the water channel aquaporin-2 (AQP2) in the plasma membrane. The membrane insertion facilitates water reabsorption from primary urine and fine-tuning of body water homeostasis. Rho guanine nucleotide exchange factors (GEFs) interact with RhoA, catalyze the exchange of GDP for GTP and thereby activate the GTPase. However, GEFs involved in the control of AQP2 in renal principal cells are unknown. The A-kinase anchoring protein, AKAP-Lbc, possesses GEF activity, specifically activates RhoA, and is expressed in primary renal inner medullary collecting duct principal (IMCD) cells. Through screening of 18,431 small molecules and synthesis of a focused library around one of the hits, we identified an inhibitor of the interaction of AKAP-Lbc and RhoA. This molecule, Scaff10-8, bound to RhoA, inhibited the AKAP-Lbc-mediated RhoA activation but did not interfere with RhoA activation through other GEFs or activities of other members of the Rho family of small GTPases, Rac1 and Cdc42. Scaff10-8 promoted the redistribution of AQP2 from intracellular vesicles to the periphery of IMCD cells. Thus, our data demonstrate an involvement of AKAP-Lbc-mediated RhoA activation in the control of AQP2 trafficking.

    View details for DOI 10.1371/journal.pone.0191423

    View details for PubMedID 29373579

    View details for PubMedCentralID PMC5786306

  • RSK3 is required for concentric myocyte hypertrophy in an activated Raf1 model for Noonan syndrome. Journal of molecular and cellular cardiology Passariello, C. L., Martinez, E. C., Thakur, H., Cesareo, M., Li, J., Kapiloff, M. S. 2016; 93: 98-105

    Abstract

    Noonan syndrome (NS) is a congenital disorder resulting from mutations of the Ras-Raf signaling pathway. Hypertrophic cardiomyopathy associated with RAF1 "RASopathy" mutations is a major risk factor for heart failure and death in NS and has been attributed to activation of MEK1/2-ERK1/2 mitogen-activated protein kinases. We recently discovered that type 3 p90 ribosomal S6 kinase (RSK3) is an ERK effector that is required, like ERK1/2, for concentric myocyte hypertrophy in response to pathological stress such as pressure overload. In order to test whether RSK3 also contributes to NS-associated hypertrophic cardiomyopathy, RSK3 knock-out mice were crossed with mice bearing the Raf1(L613V) human NS mutation. We confirmed that Raf1(L613V) knock-in confers a NS-like phenotype, including cardiac hypertrophy. Active RSK3 was increased in Raf1(L613V) mice. Constitutive RSK3 gene deletion prevented the Raf1(L613V)-dependent concentric growth in width of the cardiac myocyte and attenuated cardiac hypertrophy in female mice. These results are consistent with RSK3 being an important mediator of ERK1/2-dependent growth in RASopathy. In conjunction with previously published data showing that RSK3 is important for pathological remodeling of the heart, these data suggest that targeting of this downstream MAP-kinase pathway effector should be considered in the treatment of RASopathy-associated hypertrophic cardiomyopathy.

    View details for DOI 10.1016/j.yjmcc.2016.02.020

    View details for PubMedID 26940993

    View details for PubMedCentralID PMC4846495

  • Muscle A-Kinase Anchoring Protein-a is an Injury-Specific Signaling Scaffold Required for Neurotrophic- and Cyclic Adenosine Monophosphate-Mediated Survival. EBioMedicine Wang, Y., Cameron, E. G., Li, J., Stiles, T. L., Kritzer, M. D., Lodhavia, R., Hertz, J., Nguyen, T., Kapiloff, M. S., Goldberg, J. L. 2015; 2 (12): 1880-1887

    Abstract

    Neurotrophic factor and cAMP-dependent signaling promote the survival and neurite outgrowth of retinal ganglion cells (RGCs) after injury. However, the mechanisms conferring neuroprotection and neuroregeneration downstream to these signals are unclear. We now reveal that the scaffold protein muscle A-kinase anchoring protein-α (mAKAPα) is required for the survival and axon growth of cultured primary RGCs. Although genetic deletion of mAKAPα early in prenatal RGC development did not affect RGC survival into adulthood, nor promoted the death of RGCs in the uninjured adult retina, loss of mAKAPα in the adult increased RGC death after optic nerve crush. Importantly, mAKAPα was required for the neuroprotective effects of brain-derived neurotrophic factor and cyclic adenosine-monophosphate (cAMP) after injury. These results identify mAKAPα as a scaffold for signaling in the stressed neuron that is required for RGC neuroprotection after optic nerve injury.

    View details for DOI 10.1016/j.ebiom.2015.10.025

    View details for PubMedID 26844267

  • RSK3: A regulator of pathological cardiac remodeling. IUBMB life Martinez, E. C., Passariello, C. L., Li, J., Matheson, C. J., Dodge-Kafka, K., Reigan, P., Kapiloff, M. S. 2015; 67 (5): 331-7

    Abstract

    The family of p90 ribosomal S6 kinases (RSKs) are pleiotropic effectors for extracellular signal-regulated kinase signaling pathways. Recently, RSK3 was shown to be important for pathological remodeling of the heart. Although cardiac myocyte hypertrophy can be compensatory for increased wall stress, in chronic heart diseases, this nonmitotic cell growth is usually associated with interstitial fibrosis, increased cell death, and decreased cardiac function. Although RSK3 is less abundant in the cardiac myocyte than other RSK family members, RSK3 appears to serve a unique role in cardiac myocyte stress responses. A potential mechanism conferring the unique function of RSK3 in the heart is anchoring by the scaffold protein muscle A-kinase anchoring protein β (mAKAPβ). Recent findings suggest that RSK3 should be considered as a therapeutic target for the prevention of heart failure, a clinical syndrome of major public health significance.

    View details for DOI 10.1002/iub.1383

    View details for PubMedID 25988524

    View details for PubMedCentralID PMC4449288

  • S-nitrosoglutathione reductase-dependent PPARγ denitrosylation participates in MSC-derived adipogenesis and osteogenesis. The Journal of clinical investigation Cao, Y., Gomes, S. A., Rangel, E. B., Paulino, E. C., Fonseca, T. L., Li, J., Teixeira, M. B., Gouveia, C. H., Bianco, A. C., Kapiloff, M. S., Balkan, W., Hare, J. M. 2015; 125 (4): 1679-91

    Abstract

    Bone marrow-derived mesenchymal stem cells (MSCs) are a common precursor of both adipocytes and osteoblasts. While it is appreciated that PPARγ regulates the balance between adipogenesis and osteogenesis, the roles of additional regulators of this process remain controversial. Here, we show that MSCs isolated from mice lacking S-nitrosoglutathione reductase, a denitrosylase that regulates protein S-nitrosylation, exhibited decreased adipogenesis and increased osteoblastogenesis compared with WT MSCs. Consistent with this cellular phenotype, S-nitrosoglutathione reductase-deficient mice were smaller, with reduced fat mass and increased bone formation that was accompanied by elevated bone resorption. WT and S-nitrosoglutathione reductase-deficient MSCs exhibited equivalent PPARγ expression; however, S-nitrosylation of PPARγ was elevated in S-nitrosoglutathione reductase-deficient MSCs, diminishing binding to its downstream target fatty acid-binding protein 4 (FABP4). We further identified Cys 139 of PPARγ as an S-nitrosylation site and demonstrated that S-nitrosylation of PPARγ inhibits its transcriptional activity, suggesting a feedback regulation of PPARγ transcriptional activity by NO-mediated S-nitrosylation. Together, these results reveal that S-nitrosoglutathione reductase-dependent modification of PPARγ alters the balance between adipocyte and osteoblast differentiation and provides checkpoint regulation of the lineage bifurcation of these 2 lineages. Moreover, these findings provide pathophysiological and therapeutic insights regarding MSC participation in adipogenesis and osteogenesis.

    View details for DOI 10.1172/JCI73780

    View details for PubMedID 25798618

    View details for PubMedCentralID PMC4396480

  • mAKAP-a master scaffold for cardiac remodeling. Journal of cardiovascular pharmacology Passariello, C. L., Li, J., Dodge-Kafka, K., Kapiloff, M. S. 2015; 65 (3): 218-25

    Abstract

    Cardiac remodeling is regulated by an extensive intracellular signal transduction network. Each of the many signaling pathways in this network contributes uniquely to the control of cellular adaptation. In the last few years, it has become apparent that multimolecular signaling complexes or "signalosomes" are important for fidelity in intracellular signaling and for mediating crosstalk between the different signaling pathways. These complexes integrate upstream signals and control downstream effectors. In the cardiac myocyte, the protein mAKAPβ serves as a scaffold for a large signalosome that is responsive to cAMP, calcium, hypoxia, and mitogen-activated protein kinase signaling. The main function of mAKAPβ signalosomes is to modulate stress-related gene expression regulated by the transcription factors NFATc, MEF2, and HIF-1α and type II histone deacetylases that control pathological cardiac hypertrophy.

    View details for DOI 10.1097/FJC.0000000000000206

    View details for PubMedID 25551320

    View details for PubMedCentralID PMC4355281

  • The scaffold protein muscle A-kinase anchoring protein β orchestrates cardiac myocyte hypertrophic signaling required for the development of heart failure. Circulation. Heart failure Kritzer, M. D., Li, J., Passariello, C. L., Gayanilo, M., Thakur, H., Dayan, J., Dodge-Kafka, K., Kapiloff, M. S. 2014; 7 (4): 663-72

    Abstract

    Cardiac myocyte hypertrophy is regulated by an extensive intracellular signal transduction network. In vitro evidence suggests that the scaffold protein muscle A-kinase anchoring protein β (mAKAPβ) serves as a nodal organizer of hypertrophic signaling. However, the relevance of mAKAPβ signalosomes to pathological remodeling and heart failure in vivo remains unknown.Using conditional, cardiac myocyte-specific gene deletion, we now demonstrate that mAKAPβ expression in mice is important for the cardiac hypertrophy induced by pressure overload and catecholamine toxicity. mAKAPβ targeting prevented the development of heart failure associated with long-term transverse aortic constriction, conferring a survival benefit. In contrast to 29% of control mice (n=24), only 6% of mAKAPβ knockout mice (n=31) died in the 16 weeks of pressure overload (P=0.02). Accordingly, mAKAPβ knockout inhibited myocardial apoptosis and the development of interstitial fibrosis, left atrial hypertrophy, and pulmonary edema. This improvement in cardiac status correlated with the attenuated activation of signaling pathways coordinated by the mAKAPβ scaffold, including the decreased phosphorylation of protein kinase D1 and histone deacetylase 4 that we reveal to participate in a new mAKAP signaling module. Furthermore, mAKAPβ knockout inhibited pathological gene expression directed by myocyte-enhancer factor-2 and nuclear factor of activated T-cell transcription factors that associate with the scaffold.mAKAPβ orchestrates signaling that regulates pathological cardiac remodeling in mice. Targeting of the underlying physical architecture of signaling networks, including mAKAPβ signalosome formation, may constitute an effective therapeutic strategy for the prevention and treatment of pathological remodeling and heart failure.

    View details for DOI 10.1161/CIRCHEARTFAILURE.114.001266

    View details for PubMedID 24812305

    View details for PubMedCentralID PMC4277867

  • p90 ribosomal S6 kinase 3 contributes to cardiac insufficiency in α-tropomyosin Glu180Gly transgenic mice. American journal of physiology. Heart and circulatory physiology Passariello, C. L., Gayanilo, M., Kritzer, M. D., Thakur, H., Cozacov, Z., Rusconi, F., Wieczorek, D., Sanders, M., Li, J., Kapiloff, M. S. 2013; 305 (7): H1010-9

    Abstract

    Myocardial interstitial fibrosis is an important contributor to the development of heart failure. Type 3 p90 ribosomal S6 kinase (RSK3) was recently shown to be required for concentric myocyte hypertrophy under in vivo pathological conditions. However, the role of RSK family members in myocardial fibrosis remains uninvestigated. Transgenic expression of α-tropomyosin containing a Glu180Gly mutation (TM180) in mice of a mixed C57BL/6:FVB/N background induces a cardiomyopathy characterized by a small left ventricle, interstitial fibrosis, and diminished systolic and diastolic function. Using this mouse model, we now show that RSK3 is required for the induction of interstitial fibrosis in vivo. TM180 transgenic mice were crossed to RSK3 constitutive knockout (RSK3(-/-)) mice. Although RSK3 knockout did not affect myocyte growth, the decreased cardiac function and mild pulmonary edema associated with the TM180 transgene were attenuated by RSK3 knockout. The improved cardiac function was consistent with reduced interstitial fibrosis in the TM180;RSK3(-/-) mice as shown by histology and gene expression analysis, including the decreased expression of collagens. The specific inhibition of RSK3 should be considered as a potential novel therapeutic strategy for improving cardiac function and the prevention of sudden cardiac death in diseases in which interstitial fibrosis contributes to the development of heart failure.

    View details for DOI 10.1152/ajpheart.00237.2013

    View details for PubMedID 23913705

    View details for PubMedCentralID PMC3798750

  • CIP4 is required for the hypertrophic growth of neonatal cardiac myocytes. Journal of biomedical science Rusconi, F., Thakur, H., Li, J., Kapiloff, M. S. 2013; 20: 56

    Abstract

    CIP4 is a scaffold protein that regulates membrane deformation and tubulation, organization of the actin cytoskeleton, endocytosis of growth factor receptors, and vesicle trafficking. Although expressed in the heart, CIP4 has not been studied with regards to its potential function in cardiac myocytes.We now show using RNA interference that CIP4 expression in neonatal rat ventricular myocytes is required for the induction of non-mitotic, hypertrophic growth by the α-adrenergic agonist phenylephrine, the IL-6 cytokine leukemia inhibitor factor, and fetal bovine serum, as assayed using morphometry, immunocytochemistry for the hypertrophic marker atrial natriuretic factor and [3H]leucine incorporation for de novo protein synthesis. This requirement was consistent with the induction of CIP4 expression by hypertrophic stimulation. The inhibition of myocyte hypertrophy by CIP4 small interfering oligonucleotides (siRNA) was rescued by expression of a recombinant CIP4 protein, but not by a mutant lacking the N-terminal FCH domain responsible for CIP4 intracellular localization.These results imply that CIP4 plays a significant role in the intracellular hypertrophic signal transduction network that controls the growth of cardiac myocytes in heart disease.

    View details for DOI 10.1186/1423-0127-20-56

    View details for PubMedID 23915320

    View details for PubMedCentralID PMC3750294

  • Regulation of MEF2 transcriptional activity by calcineurin/mAKAP complexes. Experimental cell research Li, J., Vargas, M. A., Kapiloff, M. S., Dodge-Kafka, K. L. 2013; 319 (4): 447-54

    Abstract

    The calcium/calmodulin-dependent protein phosphatase calcineurin is required for the induction of transcriptional events that initiate and promote myogenic differentiation. An important effector for calcineurin in striated muscle is the transcription factor myocyte enhancer factor 2 (MEF2). The targeting of the enzyme and substrate to specific intracellular compartments by scaffold proteins often confers specificity in phosphatase activity. We now show that the scaffolding protein mAKAP organizes a calcineurin/MEF2 signaling complex in myocytes, regulating gene transcription. A calcineurin/mAKAP/MEF2 complex can be isolated from C2C12 cells and cardiac myocytes, and the calcineurin/MEF2 association is dependent on mAKAP expression. We have identified a peptide comprising the calcineurin binding domain in mAKAP that can disrupt the binding of the phosphatase to the scaffold in vivo. Dominant interference of calcineurin/mAKAP binding blunts the increase in MEF2 transcriptional activity seen during myoblast differentiation, as well as the expression of endogenous MEF2-target genes. Furthermore, disruption of calcineurin binding to mAKAP in cardiac myocytes inhibits adrenergic-induced cellular hypertrophy. Together these data illustrate the importance of calcineurin anchoring by the mAKAP scaffold for MEF2 regulation.

    View details for DOI 10.1016/j.yexcr.2012.12.016

    View details for PubMedID 23261540

    View details for PubMedCentralID PMC3563847

  • Anchored p90 ribosomal S6 kinase 3 is required for cardiac myocyte hypertrophy. Circulation research Li, J., Kritzer, M. D., Michel, J. J., Le, A., Thakur, H., Gayanilo, M., Passariello, C. L., Negro, A., Danial, J. B., Oskouei, B., Sanders, M., Hare, J. M., Hanauer, A., Dodge-Kafka, K., Kapiloff, M. S. 2013; 112 (1): 128-39

    Abstract

    Cardiac myocyte hypertrophy is the main compensatory response to chronic stress on the heart. p90 ribosomal S6 kinase (RSK) family members are effectors for extracellular signal-regulated kinases that induce myocyte growth. Although increased RSK activity has been observed in stressed myocytes, the functions of individual RSK family members have remained poorly defined, despite being potential therapeutic targets for cardiac disease.To demonstrate that type 3 RSK (RSK3) is required for cardiac myocyte hypertrophy.RSK3 contains a unique N-terminal domain that is not conserved in other RSK family members. We show that this domain mediates the regulated binding of RSK3 to the muscle A-kinase anchoring protein scaffold, defining a novel kinase anchoring event. Disruption of both RSK3 expression using RNA interference and RSK3 anchoring using a competing muscle A-kinase anchoring protein peptide inhibited the hypertrophy of cultured myocytes. In vivo, RSK3 gene deletion in the mouse attenuated the concentric myocyte hypertrophy induced by pressure overload and catecholamine infusion.Taken together, these data demonstrate that anchored RSK3 transduces signals that modulate pathologic myocyte growth. Targeting of signaling complexes that contain select kinase isoforms should provide an approach for the specific inhibition of cardiac myocyte hypertrophy and for the development of novel strategies for the prevention and treatment of heart failure.

    View details for DOI 10.1161/CIRCRESAHA.112.276162

    View details for PubMedID 22997248

    View details for PubMedCentralID PMC3537852

  • AKAPs: the architectural underpinnings of local cAMP signaling. Journal of molecular and cellular cardiology Kritzer, M. D., Li, J., Dodge-Kafka, K., Kapiloff, M. S. 2012; 52 (2): 351-8

    Abstract

    The cAMP-dependent protein kinase A (PKA) is targeted to specific compartments in the cardiac myocyte by A-kinase anchoring proteins (AKAPs), a diverse set of scaffold proteins that have been implicated in the regulation of excitation-contraction coupling and cardiac remodeling. AKAPs bind not only PKA, but also a large variety of structural and signaling molecules. In this review, we discuss the basic concepts underlying compartmentation of cAMP and PKA signaling, as well as a few of the individual AKAPs that have been shown to be functionally relevant in the heart. This article is part of a Special Issue entitled "Local Signaling in Myocytes".

    View details for DOI 10.1016/j.yjmcc.2011.05.002

    View details for PubMedID 21600214

    View details for PubMedCentralID PMC3168680

  • A-kinase anchoring proteins: scaffolding proteins in the heart. American journal of physiology. Heart and circulatory physiology Diviani, D., Dodge-Kafka, K. L., Li, J., Kapiloff, M. S. 2011; 301 (5): H1742-53

    Abstract

    The pleiotropic cyclic nucleotide cAMP is the primary second messenger responsible for autonomic regulation of cardiac inotropy, chronotropy, and lusitropy. Under conditions of prolonged catecholaminergic stimulation, cAMP also contributes to the induction of both cardiac myocyte hypertrophy and apoptosis. The formation of localized, multiprotein complexes that contain different combinations of cAMP effectors and regulatory enzymes provides the architectural infrastructure for the specialization of the cAMP signaling network. Scaffolds that bind protein kinase A are called "A-kinase anchoring proteins" (AKAPs). In this review, we discuss recent advances in our understanding of how PKA is compartmentalized within the cardiac myocyte by AKAPs and how AKAP complexes modulate cardiac function in both health and disease.

    View details for DOI 10.1152/ajpheart.00569.2011

    View details for PubMedID 21856912

    View details for PubMedCentralID PMC3213966

  • The mAKAPbeta scaffold regulates cardiac myocyte hypertrophy via recruitment of activated calcineurin. Journal of molecular and cellular cardiology Li, J., Negro, A., Lopez, J., Bauman, A. L., Henson, E., Dodge-Kafka, K., Kapiloff, M. S. 2010; 48 (2): 387-94

    Abstract

    mAKAPbeta is the scaffold for a multimolecular signaling complex in cardiac myocytes that is required for the induction of neonatal myocyte hypertrophy. We now show that the pro-hypertrophic phosphatase calcineurin binds directly to a single site on mAKAPbeta that does not conform to any of the previously reported consensus binding sites. Calcineurin-mAKAPbeta complex formation is increased in the presence of Ca(2+)/calmodulin and in norepinephrine-stimulated primary cardiac myocytes. This binding is of functional significance because myocytes exhibit diminished norepinephrine-stimulated hypertrophy when expressing a mAKAPbeta mutant incapable of binding calcineurin. In addition to calcineurin, the transcription factor NFATc3 also associates with the mAKAPbeta scaffold in myocytes. Calcineurin bound to mAKAPbeta can dephosphorylate NFATc3 in myocytes, and expression of mAKAPbeta is required for NFAT transcriptional activity. Taken together, our results reveal the importance of regulated calcineurin binding to mAKAPbeta for the induction of cardiac myocyte hypertrophy. Furthermore, these data illustrate how scaffold proteins organizing localized signaling complexes provide the molecular architecture for signal transduction networks regulating key cellular processes.

    View details for DOI 10.1016/j.yjmcc.2009.10.023

    View details for PubMedID 19883655

    View details for PubMedCentralID PMC2813376

  • An adenylyl cyclase-mAKAPbeta signaling complex regulates cAMP levels in cardiac myocytes. The Journal of biological chemistry Kapiloff, M. S., Piggott, L. A., Sadana, R., Li, J., Heredia, L. A., Henson, E., Efendiev, R., Dessauer, C. W. 2009; 284 (35): 23540-6

    Abstract

    Protein kinase A-anchoring proteins (AKAPs) play important roles in the compartmentation of cAMP signaling, anchoring protein kinase A (PKA) to specific cellular organelles and serving as scaffolds that assemble localized signaling cascades. Although AKAPs have been recently shown to bind adenylyl cyclase (AC), the functional significance of this association has not been studied. In cardiac myocytes, the muscle protein kinase A-anchoring protein beta (mAKAPbeta) coordinates cAMP-dependent, calcium, and MAP kinase pathways and is important for cellular hypertrophy. We now show that mAKAPbeta selectively binds type 5 AC in the heart and that mAKAPbeta-associated AC activity is absent in AC5 knock-out hearts. Consistent with its known inhibition by PKA phosphorylation, AC5 is inhibited by association with mAKAPbeta-PKA complexes. AC5 binds to a unique N-terminal site on mAKAP-(245-340), and expression of this peptide disrupts endogenous mAKAPbeta-AC association. Accordingly, disruption of mAKAPbeta-AC5 complexes in neonatal cardiac myocytes results in increased cAMP and hypertrophy in the absence of agonist stimulation. Taken together, these results show that the association of AC5 with the mAKAPbeta complex is required for the regulation of cAMP second messenger controlling cardiac myocyte hypertrophy.

    View details for DOI 10.1074/jbc.M109.030072

    View details for PubMedID 19574217

    View details for PubMedCentralID PMC2749128

  • Cardiomyogenic stem and progenitor cell plasticity and the dissection of cardiopoiesis. Journal of molecular and cellular cardiology Perino, M. G., Yamanaka, S., Li, J., Wobus, A. M., Boheler, K. R. 2008; 45 (4): 475-94

    Abstract

    Cell-based therapies hold promise of repairing an injured heart, and the description of stem and progenitor cells with cardiomyogenic potential is critical to its realization. At the vanguard of these efforts are analyses of embryonic stem cells, which clearly have the capacity to generate large numbers of cardiomyocytes in vitro. Through the use of this model system, a number of signaling mechanisms have been worked out that describes at least partially the process of cardiopoiesis. Studies on adult stem and on progenitor cells with cardiomyogenic potential are still in their infancy, and much less is known about the molecular signals that are required to induce the differentiation to cardiomyocytes. It is also unclear whether the pathways are similar or different between embryonic and adult cell-induced cardiomyogenesis, partly because of the continued controversies that surround the stem cell theory of cardiac self-renewal. Irrespective of any perceived or actual limitations, the study of stem and progenitor cells has provided important insights into the process of cardiomyogenesis, and it is likely that future research in this area will turn the promise of repairing an injured heart into a reality.

    View details for DOI 10.1016/j.yjmcc.2008.05.002

    View details for PubMedID 18565538

    View details for PubMedCentralID PMC2597345

  • B-MYB is essential for normal cell cycle progression and chromosomal stability of embryonic stem cells. PloS one Tarasov, K. V., Tarasova, Y. S., Tam, W. L., Riordon, D. R., Elliott, S. T., Kania, G., Li, J., Yamanaka, S., Crider, D. G., Testa, G., Li, R. A., Lim, B., Stewart, C. L., Liu, Y., Van Eyk, J. E., Wersto, R. P., Wobus, A. M., Boheler, K. R. 2008; 3 (6): e2478

    Abstract

    The transcription factor B-Myb is present in all proliferating cells, and in mice engineered to remove this gene, embryos die in utero just after implantation due to inner cell mass defects. This lethal phenotype has generally been attributed to a proliferation defect in the cell cycle phase of G1.In the present study, we show that the major cell cycle defect in murine embryonic stem (mES) cells occurs in G2/M. Specifically, knockdown of B-Myb by short-hairpin RNAs results in delayed transit through G2/M, severe mitotic spindle and centrosome defects, and in polyploidy. Moreover, many euploid mES cells that are transiently deficient in B-Myb become aneuploid and can no longer be considered viable. Knockdown of B-Myb in mES cells also decreases Oct4 RNA and protein abundance, while over-expression of B-MYB modestly up-regulates pou5f1 gene expression. The coordinated changes in B-Myb and Oct4 expression are due, at least partly, to the ability of B-Myb to directly modulate pou5f1 gene promoter activity in vitro. Ultimately, the loss of B-Myb and associated loss of Oct4 lead to an increase in early markers of differentiation prior to the activation of caspase-mediated programmed cell death.Appropriate B-Myb expression is critical to the maintenance of chromosomally stable and pluripotent ES cells, but its absence promotes chromosomal instability that results in either aneuploidy or differentiation-associated cell death.

    View details for DOI 10.1371/journal.pone.0002478

    View details for PubMedID 18575582

    View details for PubMedCentralID PMC2423619

  • Pluripotency of embryonic stem cells. Cell and tissue research Yamanaka, S., Li, J., Kania, G., Elliott, S., Wersto, R. P., Van Eyk, J., Wobus, A. M., Boheler, K. R. 2008; 331 (1): 5-22

    Abstract

    Embryonic stem (ES) cells derived from pre-implantation embryos have the potential to differentiate into any cell type derived from the three germ layers of ectoderm (epidermal tissues and nerves), mesoderm (muscle, bone, blood), and endoderm (liver, pancreas, gastrointestinal tract, lungs), including fetal and adult cells. Alone, these cells do not develop into a viable fetus or adult animal because they do not retain the potential to contribute to extraembryonic tissue, and in vitro, they lack spatial and temporal signaling cues essential to normal in vivo development. The basis of pluripotentiality resides in conserved regulatory networks composed of numerous transcription factors and multiple signaling cascades. Together, these regulatory networks maintain ES cells in a pluripotent and undifferentiated form; however, alterations in the stoichiometry of these signals promote differentiation. By taking advantage of this differentiation capacity in vitro, ES cells have clearly been shown to possess the potential to generate multipotent stem and progenitor cells capable of differentiating into a limited number of cell fates. These latter types of cells may prove to be therapeutically viable, but perhaps more importantly, the studies of these cells have led to a greater understanding of mammalian development.

    View details for DOI 10.1007/s00441-007-0520-5

    View details for PubMedID 18026755

  • The pro-angiogenic cytokine pleiotrophin potentiates cardiomyocyte apoptosis through inhibition of endogenous AKT/PKB activity. The Journal of biological chemistry Li, J., Wei, H., Chesley, A., Moon, C., Krawczyk, M., Volkova, M., Ziman, B., Margulies, K. B., Talan, M., Crow, M. T., Boheler, K. R. 2007; 282 (48): 34984-93

    Abstract

    Pleiotrophin is a development-regulated cytokine and growth factor that can promote angiogenesis, cell proliferation, or differentiation, and it has been reported to have neovasculogenic effects in damaged heart. Developmentally, it is prominently expressed in fetal and neonatal hearts, but it is minimally expressed in normal adult heart. Conversely, we show in a rat model of myocardial infarction and in human dilated cardiomyopathy that pleiotrophin is markedly up-regulated. To elucidate the effects of pleiotrophin on cardiac contractile cells, we employed primary cultures of rat neonatal and adult cardiomyocytes. We show that pleiotrophin is released from cardiomyocytes in vitro in response to hypoxia and that the addition of recombinant pleiotrophin promotes caspase-mediated genomic DNA fragmentation in a dose- and time-dependent manner. Functionally, it potentiates the apoptotic response of neonatal cardiomyocytes to hypoxic stress and to ultraviolet irradiation and of adult cardiomyocytes to hypoxia-reoxygenation. Moreover, UV-induced apoptosis in neonatal cardiomyocytes can be partially inhibited by small interfering RNA-mediated knockdown of endogenous pleiotrophin. Mechanistically, pleiotrophin antagonizes IGF-1 associated Ser-473 phosphorylation of AKT/PKB, and it concomitantly decreases both BAD and GSK3beta phosphorylation. Adenoviral expression of constitutively active AKT and lithium chloride-mediated inhibition of GSK3beta reduce the potentiated programmed cell death elicited by pleiotrophin. These latter data indicate that pleiotrophin potentiates cardiomyocyte cell death, at least partially, through inhibition of AKT signaling. In conclusion, we have uncovered a novel function for pleiotrophin on heart cells following injury. It fosters cardiomyocyte programmed cell death in response to pro-apoptotic stress, which may be critical to myocardial injury repair.

    View details for DOI 10.1074/jbc.M703513200

    View details for PubMedID 17925408

  • Embryonic stem cells and cardiomyocyte differentiation: phenotypic and molecular analyses. Journal of cellular and molecular medicine Wei, H., Juhasz, O., Li, J., Tarasova, Y. S., Boheler, K. R. 2005; 9 (4): 804-17

    Abstract

    Embryonic stem (ES) cell lines, derived from the inner cell mass (ICM) of blastocyst-stage embryos, are pluripotent and have a virtually unlimited capacity for self-renewal and differentiation into all cell types of an embryoproper. Both human and mouse ES cell lines are the subject of intensive investigation for potential applications in developmental biology and medicine. ES cells from both sources differentiate in vitro into cells of ecto-, endoand meso-dermal lineages, and robust cardiomyogenic differentiation is readily observed in spontaneously differentiating ES cells when cultured under appropriate conditions. Molecular, cellular and physiologic analyses demonstrate that ES cell-derived cardiomyocytes are functionally viable and that these cell derivatives exhibit characteristics typical of heart cells in early stages of cardiac development. Because terminal heart failure is characterized by a significant loss of cardiomyocytes, the use of human ES cell-derived progeny represents one possible source for cell transplantation therapies. With these issues in mind, this review will focus on the differentiation of pluripotent embryonic stem cells into cardiomyocytes as a developmental model, and the possible use of ES cell-derived cardiomyocytes as source of donor cells.

    View details for DOI 10.1111/j.1582-4934.2005.tb00381.x

    View details for PubMedID 16364192

    View details for PubMedCentralID PMC6740270

  • Gene expression profiles in response to the activation of adrenoceptors in A7r5 aortic smooth muscle cells. Clinical and experimental pharmacology & physiology Wang, Y., Hou, R., Li, P., Li, J., Yan, J., Yin, F., Han, C., Zhang, Y. 2004; 31 (9): 602-7

    Abstract

    1. Vascular adrenoceptors play an important role in vascular physiology and pathophysiology, such as hypertension, atherosclerosis and restenosis after angioplasty. To define the changes in the ene expression in vascular smooth muscle cells in response to the activation of alpha1- or beta-adrenoceptors, a DNA microarray was used. 2. First, the existence of alpha1- and beta-adrenoceptors in A7r5 aortic smooth muscle cells was confirmed by radioligand binding. Then, the inhibitory effects of phenylephrine (an alpha1-adrenoceptor agonist) and isoproterenol (a beta-adrenoceptor agonist) on the proliferation of A7r5 cells were determined by [3H]-thymidine incorporation. 3. The A7r5 cells were treated with 10 micromol/L phenylephrine or 1 micromol/L isoproterenol for 24 h and changes in gene expression were detected with the DNA microarray. Only 14 and 20 genes were identified after treatment of cells with phenylephrine and isoproterenol, respectively, and most genes displayed decreased expression. The changed genes could be grouped into five major functional categories: cell signalling/communication, cell structure/motility, cell/organism defence, gene/protein expression and metabolism. The gene expression profile in response to the activation of alpha1-adrenoceptors was very different from that following activation of beta-adrenoceptors. Interestingly, many phenylephrine-responsive genes were associated with metabolism, whereas many isoproterenol-responsive genes encoded cell signalling and structure proteins. This means that adrenoceptors may modulate multiple aspects of biological function in vascular smooth muscle cells. 4. Collectively, the activation of both alpha1-adrenoceptors (with phenylephrine) and beta-adrenoceptors (with isoproterenol) inhibited the proliferation of A7r5 cells, but microarray data revealed that the mechanisms may be different: the activation of alpha1-adrenoceptors could induce the expression of metabolic genes, resulting in the inhibition of proliferation, whereas activation of beta-adrenoceptors altered the expression of genes that encoded cell signalling and structure proteins to inhibit cell proliferation.

    View details for DOI 10.1111/j.1440-1681.2004.04058.x

    View details for PubMedID 15479167

  • [Comparison of changes in left ventricular gene expression profiles from different cardiac hypertrophy models in rats]. Sheng li xue bao : [Acta physiologica Sinica] Li, P., Li, J. L., Feng, X. H., Li, Z. P., Yin, F., Yan, J., Hou, R., Han, Q. D., Zhang, Y. Y. 2004; 56 (2): 210-8

    Abstract

    To get insights into the principles of gene expression changes during cardiac hypertrophy, three rat cardiac hypertrophy models were prepared, i.e., suprarenal abdominal aortic stenosis (SRS), arterial-vein fistula (AVF) and continuous jugular vein infusion of norepinephrine (NEi). The cardiac function and structure were analyzed by echocardiograph as well as histological examination. Total RNA of left ventricles was extracted and gene expression profiles were analyzed by cDNA microarray. SRS and NEi induced concentric cardiac hypertrophy and AVF induced eccentric hypertrophy in rats, among which NEi caused obvious cardiac fibrosis. The changes of gene expression profiles were compared comprehensively across different pathologic cardiac hypertrophy models. While gene expression profiles of different cardiac hypertrophy models compared with pairs, parts of the genes involved were found overlapped, and mostly the gene expression changed in the same direction between two models, but some of them changed in the opposite directions. Expression levels of 19 genes were found changed across all cardiac hypertrophy models, and genes relatively regulated in a specific model was also found when comparison of all the three models was carried out. Novel clues for further study might derive from the results mentioned above, and some genes might be the marker genes of cardiac hypertrophy or the targets of therapy.

    View details for PubMedID 15127132

  • Effects of losartan on pressure overload-induced cardiac gene expression profiling in rats. Clinical and experimental pharmacology & physiology Li, J., Li, P., Feng, X., Li, Z., Hou, R., Han, C., Zhang, Y. 2003; 30 (11): 827-32

    Abstract

    1. In the present study, the effects of losartan on myocardial gene expression changes following cardiac hypertrophy were investigated. 2. Male Wistar rats were randomized to receive 5 or 30 mg/kg per day losartan (i.p.) 1 day after suprarenal abdominal aortic constriction. Two weeks later, cardiac morphology and function were recorded with echocardiography and mean arterial central pressure was measured using carotid catheters. Myocardial gene expression was assessed with cDNA microarrays. 3. The ratios of left ventricular weights to bodyweights, the posterior thickness of the left ventricle and mean arterial central pressure were significantly increased by aortic constriction and attenuated by losartan in a dose-related manner. Genes in different functional categories were regulated in pressure overload-induced cardiac hypertrophy and the majority of changes in gene expression were inhibited by losartan in a dose-dependent manner. 4. However, there were still some genes that were unaffected by losartan, even at a higher dose. In contrast, losartan, especially at a lower dose, was able to induce changes in the expression of several additional genes that were unregulated in simple aortic constriction. 5. In conclusion, losartan is able to inhibit pressure overload-induced cardiac hypertrophy, as well as the majority of pressure overload-related changes in gene expression. The genes that remained unaffected or those that were additionally induced by losartan are likely to be new targets for investigation or therapy.

    View details for DOI 10.1046/j.1440-1681.2003.03917.x

    View details for PubMedID 14678244

  • Gene expression profile of cardiomyocytes in hypertrophic heart induced by continuous norepinephrine infusion in the rats. Cellular and molecular life sciences : CMLS Li, P., Li, J., Feng, X., Li, Z., Hou, R., Han, C., Zhang, Y. 2003; 60 (10): 2200-9

    Abstract

    Catecholamines play an important role in the development of cardiac hypertrophy. To observe cardiomyocyte-specific gene expression changes induced by catecholamines in vivo, left ventricular cardiomyocytes were isolated from male Sprague-Dawley rats after continuous infusion of norepinephrine (NE; 0.2 mg/kg per hour intravenously) for 0.5, 1, 2, 3 and 7 days. The gene expression profiles of these cells during different NE infusion stages were assessed by using a cDNA microarray, and the microarray data were further analyzed by a clustering method. Cardiac hypertrophy was induced upon continuous NE infusion, with the peak at 3 days. Meanwhile, manifest changes in gene expression profile within cardiomyocytes over the time course were revealed, most of the genes never having been reported to be involved in cardiac hypertrophy. The number of genes displaying differential expression also peaked at the middle stage of infusion (2-3 days), and the majority of the signaling molecules were found differentially expressed mainly at this stage, including phosphatidylinositol 3-kinase, calcium/calmodulin-dependent protein kinase II and non-receptor tyrosine kinases, etc. The tumor suppressor p53 was found up-regulated at very early (0.5 days) and late stages (7 days) of NE infusion. Self-organization clustering analysis revealed subsets of coordinate regulated genes. One set consisted of several enzymes involved in energy metabolism, including carnitine octanoyltransferase, ATP synthase subunit c, pancreatic lipase and glycogen phosphorylase, possessing a similar expression pattern with a rapidly elevated expression level at the early stage of NE infusion. This is the first study to provide transcriptional information for cardiomyocytes, a single cell type, in the heart during the development of cardiac hypertrophy in vivo, and may provide accurate clues to elaborate hypotheses about the evolution of this pathology.

    View details for DOI 10.1007/s00018-003-3178-5

    View details for PubMedID 14618266

  • [Alterations of Axin protein expression during cardiac remodeling in rats]. Sheng li xue bao : [Acta physiologica Sinica] Li, P., Li, J. L., Yin, F., Yan, J., Feng, X. H., Li, Z. P., Han, Q. D., Zhang, Y. Y. 2003; 55 (3): 331-5

    Abstract

    The purpose of the present study was to observe the expression of Axin protein during cardiac remodeling in rats. Cardiac remodeling animal models were prepared with the methods of jugular venous norepinephrine (NE)-infusion or arterial-vein fistula (AVF). The ultrasonic parameters of rat hearts were recorded before sacrifice. The expressions of Axin protein were determined by Western blot in rat hearts from different remodeling models as well as cultured cardiac fibroblasts from adult rats. Cardiac concentric hypertrophy and fibrosis was induced by 3-day jugular vein infusion of NE in rats. The expression of Axin in the left ventricles increased significantly compared with that of the control group. Cardiac eccentric hypertrophy without fibrosis was induced by A-V fistula for one week in rats, and no change in Axin protein expression in the left ventricles was observed. In cultured adult rat cardiac fibroblasts, NE treatment for 24 h increased significantly the Axin protein level. It is therefore concluded that Axin protein was expressed in rat heart and increased significantly in left ventricles during NE-induced rat cardiac remodeling, which may be relevant to cardiac fibrosis.

    View details for PubMedID 12817302

  • [Changes in the gene expression profile of the left heart ventricle during growth in the rat]. Sheng li xue bao : [Acta physiologica Sinica] Li, P., Li, J. L., Hou, R., Han, Q. D., Zhang, Y. Y. 2003; 55 (2): 191-6

    Abstract

    Wistar rats of 8, 10 and 12-week-old were chosen for study of the relationship between cardiac growth and its gene expression profile changes during maturation. The ultrasonic parameters of rat hearts were recorded before sacrifice, then total RNA of left ventricle were extracted and gene expression profiles were analyzed by cDNA microarray. During growth from 8 weeks to 12 weeks, the body weight increased by 45.5% (287+/-13 g vs 197+/-10 g), and the increment in the first two-week period was equal to that of the second two-week period. The mass of left ventricle and the posterior wall thickness increased by 27.7% (0.60+/-0.03 g vs 0.47+/-0.02 g) and 23.6% (2.04+/-0.04 mm vs 1.65+/-0.13 mm), respectively, and their increment in the first two-week period was much more than that in the second one. Meanwhile, the gene expression profile of the left ventricle changed significantly, which involved cellular structure, metabolism, oxidative stress, signal transduction, etc. Compared with the 8-week-old rats, these genes were mostly up-regulated in 10-week-old rats, while for 12-week-old rats, the gene expression profile of the left ventricle recovered to the pattern of 8-week-old rats again on the whole. These results suggest that the relationship between the changes in cardiac function and gene expression profile can be analyzed comprehensively with the technique of microarray, and that the changes in gene expression profile of the left ventricle during rat maturation adapt to the physiological growth of heart, which is of benefit for keeping the metabolism balance between materials and energy.

    View details for PubMedID 12715110