151
|
Takimoto E, Koitabashi N, Hsu S, Ketner EA, Zhang M, Nagayama T, Bedja D, Gabrielson KL, Blanton R, Siderovski DP, Mendelsohn ME, Kass DA. Regulator of G protein signaling 2 mediates cardiac compensation to pressure overload and antihypertrophic effects of PDE5 inhibition in mice. J Clin Invest 2009; 119:408-20. [PMID: 19127022 DOI: 10.1172/jci35620] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2008] [Accepted: 11/12/2008] [Indexed: 01/19/2023] Open
Abstract
The heart initially compensates for hypertension-mediated pressure overload by enhancing its contractile force and developing hypertrophy without dilation. Gq protein-coupled receptor pathways become activated and can depress function, leading to cardiac failure. Initial adaptation mechanisms to reduce cardiac damage during such stimulation remain largely unknown. Here we have shown that this initial adaptation requires regulator of G protein signaling 2 (RGS2). Mice lacking RGS2 had a normal basal cardiac phenotype, yet responded rapidly to pressure overload, with increased myocardial Gq signaling, marked cardiac hypertrophy and failure, and early mortality. Swimming exercise, which is not accompanied by Gq activation, induced a normal cardiac response, while Rgs2 deletion in Galphaq-overexpressing hearts exacerbated hypertrophy and dilation. In vascular smooth muscle, RGS2 is activated by cGMP-dependent protein kinase (PKG), suppressing Gq-stimulated vascular contraction. In normal mice, but not Rgs2-/- mice, PKG activation by the chronic inhibition of cGMP-selective phosphodiesterase 5 (PDE5) suppressed maladaptive cardiac hypertrophy, inhibiting Gq-coupled stimuli. Importantly, PKG was similarly activated by PDE5 inhibition in myocardium from both genotypes, but PKG plasma membrane translocation was more transient in Rgs2-/- myocytes than in controls and was unaffected by PDE5 inhibition. Thus, RGS2 is required for early myocardial compensation to pressure overload and mediates the initial antihypertrophic and cardioprotective effects of PDE5 inhibitors.
Collapse
Affiliation(s)
- Eiki Takimoto
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
152
|
Sheikhi A, Jaberi Y, Esmaeilzadeh A, Khani M, Moosaeefard M, Shafaqatian M. The effect of cardiovascular drugs on pro-inflammatory cytokine secretion and natural killer activity of peripheral blood mononuclear cells of patients with chronic heart failure in vitro. Pak J Biol Sci 2009; 10:1580-7. [PMID: 19086502 DOI: 10.3923/pjbs.2007.1580.1587] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Recent studies have shown that patients with heart failure over-express pro-inflammatory cytokines which enhance natural killer (NK) activity and negatively influence contractility and contribute to the remodeling of myocardium. The question is that how cardiovascular drugs influence on the cytokines of Peripheral Blood Mononuclear Cells (PBMCs) in Chronic Heart Failure (CHF). To study the effect of cardiovascular drugs on PBMCs-cytokines and NK activity of CHF patients. PBMCs of CHF patients/normal controls collected by Ficoll-paque density centrifugation. NK activity against K562 target cell was measured with MTT colorimetric assay. PBMCs were cultivated in RPMI/FCS, stimulated with phytohaemaglutinin (PHA). Tumor necrosis factor (TNF)-alpha interleukin (IL)-6, IL-2 and IL-1beta of culture supernatants after 24 h incubation with/without furosemide, captopril and digoxin were measured with sandwitch ELISA. Patients had higher NK activity than controls (56.9% +/- 1.6 vs 50.9% +/- 1.2, p < 0.05). NK activity of patients who already consumed Captopril/Furosemide didn't show difference with controls. Captopril (3, 1, 0.3 microg mL(-1)) and Furosemide (5, 2.5, 1.25 microg mL(-1)) caused a dose dependent inhibition in TNF-alpha compared with control (329 +/- 23, 427 +/- 15, 519 +/- 19 and 343 +/- 19, 430 +/- 14, respectively vs. 562 +/- 24 pg mL(-1) p < 0.05). Furosemide caused a dose dependent decrease in IL-6 (421 +/- 31, 534 +/- 33 vs. 662 +/- 41 pg mL(-1) p < 0.05). Captopril and Furosemide didn't show any significant effect on IL-1beta/IL-2. Digoxin had no significant effect on PBMCs-cytokines. These data suggest that the immunomodulatory effects of Captopril and Furosemide may contribute to their beneficial and no long-term adverse effects on PBMCs.
Collapse
Affiliation(s)
- Abdolkarim Sheikhi
- Department of Immunology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | | | | | | | | | | |
Collapse
|
153
|
Hohensinner PJ, Kaun C, Rychli K, Niessner A, Pfaffenberger S, Rega G, Furnkranz A, Uhrin P, Zaujec J, Afonyushkin T, Bochkov VN, Maurer G, Huber K, Wojta J. The inflammatory mediator oncostatin M induces stromal derived factor‐1 in human adult cardiac cells. FASEB J 2008; 23:774-82. [DOI: 10.1096/fj.08-108035] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- P. J. Hohensinner
- Department of Internal Medicine II and and Thrombosis ResearchMedical University of ViennaViennaAustria
- Ludwig Boltzmann Cluster for Cardiovascular ResearchViennaAustria
| | - C. Kaun
- Department of Internal Medicine II and and Thrombosis ResearchMedical University of ViennaViennaAustria
| | - K. Rychli
- Department of Internal Medicine II and and Thrombosis ResearchMedical University of ViennaViennaAustria
| | - A. Niessner
- Department of Internal Medicine II and and Thrombosis ResearchMedical University of ViennaViennaAustria
| | - S. Pfaffenberger
- Department of Internal Medicine II and and Thrombosis ResearchMedical University of ViennaViennaAustria
| | - G. Rega
- Department of Internal Medicine II and and Thrombosis ResearchMedical University of ViennaViennaAustria
| | - A. Furnkranz
- Third Department of MedicineWilhelminenhospitalViennaAustria
| | - P. Uhrin
- Department of Vascular Biology and Thrombosis ResearchMedical University of ViennaViennaAustria
| | - J. Zaujec
- Department of Vascular Biology and Thrombosis ResearchMedical University of ViennaViennaAustria
| | - T. Afonyushkin
- Department of Vascular Biology and Thrombosis ResearchMedical University of ViennaViennaAustria
| | - V. N. Bochkov
- Department of Vascular Biology and Thrombosis ResearchMedical University of ViennaViennaAustria
| | - G. Maurer
- Department of Internal Medicine II and and Thrombosis ResearchMedical University of ViennaViennaAustria
| | - K. Huber
- Third Department of MedicineWilhelminenhospitalViennaAustria
| | - J. Wojta
- Department of Internal Medicine II and and Thrombosis ResearchMedical University of ViennaViennaAustria
- Ludwig Boltzmann Cluster for Cardiovascular ResearchViennaAustria
| |
Collapse
|
154
|
Chao W. Toll-like receptor signaling: a critical modulator of cell survival and ischemic injury in the heart. Am J Physiol Heart Circ Physiol 2008; 296:H1-12. [PMID: 19011041 DOI: 10.1152/ajpheart.00995.2008] [Citation(s) in RCA: 186] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Toll-like receptors (TLRs) represent the first line of host defense against microbial infection and play a pivotal role in both innate and adaptive immunity. TLRs recognize invading pathogens through molecular pattern recognition, transduce signals via distinct intracellular pathways involving a unique set of adaptor proteins and kinases, and ultimately lead to the activation of transcription factors and inflammatory responses. Among 10 TLRs identified in humans, at least two exist in the heart, i.e., TLR2 and TLR4. In addition to the critical role of these in mediating cardiac dysfunction in septic conditions, emerging evidence suggests that the TLRs can also recognize endogenous ligands and may play an important role in modulating cardiomyocyte survival and in ischemic myocardial injury. In animal models of ischemia-reperfusion injury or in hypoxic cardiomyocytes in vitro, the administration of a sublethal dose of lipopolysaccharide, which signals through TLR4, reduces subsequent myocardial infarction, improves cardiac functions, and attenuates cardiomyocyte apoptosis. By contrast, a systemic deficiency of TLR2, TLR4, or myeloid differentiation primary-response gene 88, an adaptor critical for all TLR signaling, except TLR3, leads to an attenuated myocardial inflammation, a smaller infarction size, a better preserved ventricular function, and a reduced ventricular remodeling after ischemic injury. These loss-of-function studies suggest that both TLRs contribute to myocardial inflammation and ischemic injury in the heart although the exact contribution of cardiac (vs. circulatory cell) TLRs remains to be defined. These recent studies demonstrate an emerging role for TLRs as a critical modulator in both cell survival and tissue injury in the heart.
Collapse
Affiliation(s)
- Wei Chao
- Dept. of Anesthesia & Critical Care, Massachusetts General Hospital, 55 Fruit St., Boston, MA 02114, USA.
| |
Collapse
|
155
|
Deletion of Shp2 tyrosine phosphatase in muscle leads to dilated cardiomyopathy, insulin resistance, and premature death. Mol Cell Biol 2008; 29:378-88. [PMID: 19001090 DOI: 10.1128/mcb.01661-08] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The intracellular signaling mechanisms underlying the pathogenesis of cardiac diseases are not fully understood. We report here that selective deletion of Shp2, an SH2-containing cytoplasmic tyrosine phosphatase, in striated muscle results in severe dilated cardiomyopathy in mice, leading to heart failure and premature mortality. Development of cardiomyopathy in this mouse model is coupled with insulin resistance, glucose intolerance, and impaired glucose uptake in striated muscle cells. Shp2 deficiency leads to upregulation of leukemia inhibitory factor-stimulated phosphatidylinositol 3-kinase/Akt, Erk5, and Stat3 pathways in cardiomyocytes. Insulin resistance and impaired glucose uptake in Shp2-deficient mice are at least in part due to impaired protein kinase C-zeta/lambda and AMP-kinase activities in striated muscle. Thus, we have generated a mouse line modeling human patients suffering from cardiomyopathy and insulin resistance. This study reinforces a concept that a compound disease with multiple cardiovascular and metabolic disturbances can be caused by a defect in a single molecule such as Shp2, which modulates multiple signaling pathways initiated by cytokines and hormones.
Collapse
|
156
|
Hakim ZS, DiMichele LA, Rojas M, Meredith D, Mack CP, Taylor JM. FAK regulates cardiomyocyte survival following ischemia/reperfusion. J Mol Cell Cardiol 2008; 46:241-8. [PMID: 19028502 DOI: 10.1016/j.yjmcc.2008.10.017] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2008] [Revised: 10/10/2008] [Accepted: 10/11/2008] [Indexed: 10/21/2022]
Abstract
Myocyte apoptosis is central to myocardial dysfunction following ischemia/reperfusion (I/R) and during the transition from hypertrophy to heart failure. Focal adhesion kinase (FAK), a non-receptor tyrosine kinase regulates adhesion-dependent survival signals and unopposed FAK activation has been linked to tumor development. We previously showed that conditional myocyte-specific deletion of FAK (MFKO) in the adult heart did not affect basal cardiomyocyte survival or cardiac function but led to dilated cardiomyopathy and heart failure following pressure overload. In the present study, we sought to determine if FAK functions to limit stress-induced cardiomyocyte apoptosis. We reasoned that (I/R), which stimulates robust apoptotic cell death, might uncover an important cardioprotective function for FAK. We found that depletion of FAK markedly exacerbates hypoxia/re-oxygenation-induced cardiomyocyte cell death in vitro. Moreover, deletion of FAK in the adult myocardium resulted in significant increases in I/R-induced infarct size and cardiomyocyte apoptosis with a concomitant reduction in left ventricular function. Finally, our results suggest that NF-kappaB signaling may play a key role in modulating FAK-dependent cardioprotection, since FAK inactivation blunted activation of the NF-kappaB survival signaling pathway and reduced levels of the NF-kappaB target genes, Bcl2 and Bcl-xl. Since the toggling between pro-survival and pro-apoptotic signals remains central to preventing irreversible damage to the heart, we conclude that targeted FAK activation may be beneficial for protecting stress-dependent cardiac remodeling.
Collapse
Affiliation(s)
- Zeenat S Hakim
- Department of Pathology, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | | | | | | | | |
Collapse
|
157
|
Chun HJ, Narula J, Hofstra L, Wu JC. Intracellular and extracellular targets of molecular imaging in the myocardium. ACTA ACUST UNITED AC 2008; 5 Suppl 2:S33-41. [PMID: 18641605 DOI: 10.1038/ncpcardio1161] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2007] [Accepted: 01/08/2008] [Indexed: 11/09/2022]
Abstract
Utilization of molecular imaging has significantly advanced the field of cardiovascular medicine. In addition to the targets currently in use, novel targets are being developed, including those involved in the processes of myocardial metabolism, myocardial injury, cardiac neurotransmission, and interstitial dysregulation. Further development of these imaging targets may lead to improved characterization of disease processes and guide provision of individualized therapies.
Collapse
Affiliation(s)
- Hyung J Chun
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305-5344, USA
| | | | | | | |
Collapse
|
158
|
Abstract
Apoptosis is an evolutionarily conserved mode of cell death that is tightly regulated and critical for multicellular organism development and cellular homeostasis. Specific biochemical and morphological changes characterise cells undergoing apoptosis, and reflect the specificity in which activated apoptotic pathways follow. The two best-characterized apoptotic pathways are the extrinsic pathway and the intrinsic pathway, which involve cell surface death receptors and the mitochondria and endoplasmic reticulum respectively. Apoptotic stimuli lead to activation of either or both of these pathways, and involve sequential activation of different cysteine proteases (caspases), and in the case of the intrinsic pathway, activation of a family of Bcl-2 proteins that critically regulate cell death. Conversely, dis-inhibition of endogenous inhibitors is often required for effective apoptotic cell death. Furthermore, an interesting recurring protein-protein interaction within this framework of apoptotic cascades involves interactions between death domain motifs that are present on many of the regulatory proteins in both apoptotic pathways. Cardiomyocyte apoptosis has been demonstrated in human heart failure and in rodents, apoptosis itself directly causes dilated cardiomyopathy. Understanding the intricacies of apoptotic death pathways and determining the relevance of these to cardiomyopathy is therefore essential if cardiomyocyte apoptosis is to be a pharmacological target for heart failure therapy.
Collapse
|
159
|
Fasnacht N, Müller W. Conditional gp130 deficient mouse mutants. Semin Cell Dev Biol 2008; 19:379-84. [DOI: 10.1016/j.semcdb.2008.07.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2008] [Revised: 06/27/2008] [Accepted: 07/10/2008] [Indexed: 01/06/2023]
|
160
|
González V, Salgueiro E, Jimeno FJ, Hidalgo A, Rubio T, Manso G. Post‐marketing safety of antineoplasic monoclonal antibodies: rituximab and trastuzumab. Pharmacoepidemiol Drug Saf 2008; 17:714-21. [DOI: 10.1002/pds.1587] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
161
|
Willey CD, Palanisamy AP, Johnston RK, Mani SK, Shiraishi H, Tuxworth WJ, Zile MR, Balasubramanian S, Kuppuswamy D. STAT3 activation in pressure-overloaded feline myocardium: role for integrins and the tyrosine kinase BMX. Int J Biol Sci 2008; 4:184-99. [PMID: 18612371 PMCID: PMC2443357 DOI: 10.7150/ijbs.4.184] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2008] [Accepted: 06/24/2008] [Indexed: 11/23/2022] Open
Abstract
Growth, survival and cytoskeletal rearrangement of cardiomyocytes are critical for cardiac hypertrophy. Signal transducer and activator of transcription-3 (STAT3) activation is an important cardioprotective factor associated with cardiac hypertrophy. Although STAT3 activation has been reported via signaling through Janus Kinase 2 (JAK2) in several cardiac models of hypertrophy, the importance of other nonreceptor tyrosine kinases (NTKs) has not been explored. Utilizing an in vivo feline right ventricular pressure-overload (RVPO) model of hypertrophy, we demonstrate that in 48 h pressure-overload (PO) myocardium, STAT3 becomes phosphorylated and redistributed to detergent-insoluble fractions with no accompanying JAK2 activation. PO also caused increased levels of phosphorylated STAT3 in both cytoplasmic and nuclear fractions. To investigate the role of other NTKs, we used our established in vitro cell culture model of hypertrophy where adult feline cardiomyocytes are embedded three-dimensionally (3D) in type-I collagen and stimulated with an integrin binding peptide containing an Arg-Gly-Asp (RGD) motif that we have previously shown to recapitulate the focal adhesion complex (FAC) formation of 48 h RVPO. RGD stimulation of adult cardiomyocytes in vitro caused both STAT3 redistribution and activation that were accompanied by the activation and redistribution of c-Src and the TEC family kinase, BMX, but not JAK2. However, infection with dominant negative c-Src adenovirus was unable to block RGD-stimulated changes on either STAT3 or BMX. Further analysis in vivo in 48 h PO myocardium showed the presence of both STAT3 and BMX in the detergent-insoluble fraction with their complex formation and phosphorylation. Therefore, these studies indicate a novel mechanism of BMX-mediated STAT3 activation within a PO model of cardiac hypertrophy that might contribute to cardiomyocyte growth and survival.
Collapse
Affiliation(s)
- Christopher D Willey
- Cardiology Division of the Department of Medicine, Gazes Cardiac Research Institute, Medical University of South Carolina, Charleston, SC 29425-2221, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
162
|
Loyer X, Gómez AM, Milliez P, Fernandez-Velasco M, Vangheluwe P, Vinet L, Charue D, Vaudin E, Zhang W, Sainte-Marie Y, Robidel E, Marty I, Mayer B, Jaisser F, Mercadier JJ, Richard S, Shah AM, Bénitah JP, Samuel JL, Heymes C. Cardiomyocyte overexpression of neuronal nitric oxide synthase delays transition toward heart failure in response to pressure overload by preserving calcium cycling. Circulation 2008; 117:3187-98. [PMID: 18541744 DOI: 10.1161/circulationaha.107.741702] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Defects in cardiomyocyte Ca(2+) cycling are a signature feature of heart failure (HF) that occurs in response to sustained hemodynamic overload, and they largely account for contractile dysfunction. Neuronal nitric oxide synthase (NOS1) influences myocyte excitation-contraction coupling through modulation of Ca(2+) cycling, but the potential relevance of this in HF is unknown. METHODS AND RESULTS We generated a transgenic mouse with conditional, cardiomyocyte-specific NOS1 overexpression (double-transgenic [DT]) and studied cardiac remodeling, myocardial Ca(2+) handling, and contractility in DT and control mice subjected to transverse aortic constriction (TAC). After TAC, control mice developed eccentric hypertrophy with evolution toward HF as revealed by a significantly reduced fractional shortening. In contrast, DT mice developed a greater increase in wall thickness (P<0.0001 versus control+TAC) and less left ventricular dilatation than control+TAC mice (P<0.0001 for both end-systolic and end-diastolic dimensions). Thus, DT mice displayed concentric hypertrophy with fully preserved fractional shortening (43.7+/-0.6% versus 30.3+/-2.6% in control+TAC mice, P<0.05). Isolated cardiomyocytes from DT+TAC mice had greater shortening, intracellular Ca(2+) transients, and sarcoplasmic reticulum Ca(2+) load (P<0.05 versus control+TAC for all parameters). These effects could be explained, at least in part, through modulation of phospholamban phosphorylation status. CONCLUSIONS Cardiomyocyte NOS1 may be a useful target against cardiac deterioration during chronic pressure-overload-induced HF through modulation of calcium cycling.
Collapse
Affiliation(s)
- Xavier Loyer
- Institut National de Santé et de Recherché Médicale (INSERM), Unit 689, Centre de Recherché Cardiovasculaire Lariboisière, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
163
|
Petretto E, Sarwar R, Grieve I, Lu H, Kumaran MK, Muckett PJ, Mangion J, Schroen B, Benson M, Punjabi PP, Prasad SK, Pennell DJ, Kiesewetter C, Tasheva ES, Corpuz LM, Webb MD, Conrad GW, Kurtz TW, Kren V, Fischer J, Hubner N, Pinto YM, Pravenec M, Aitman TJ, Cook SA. Integrated genomic approaches implicate osteoglycin (Ogn) in the regulation of left ventricular mass. Nat Genet 2008; 40:546-52. [PMID: 18443592 PMCID: PMC2742198 DOI: 10.1038/ng.134] [Citation(s) in RCA: 132] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2008] [Accepted: 02/20/2008] [Indexed: 01/19/2023]
Abstract
Left ventricular mass (LVM) and cardiac gene expression are complex traits regulated by factors both intrinsic and extrinsic to the heart. To dissect the major determinants of LVM, we combined expression quantitative trait locus1 and quantitative trait transcript (QTT) analyses of the cardiac transcriptome in the rat. Using these methods and in vitro functional assays, we identified osteoglycin (Ogn) as a major candidate regulator of rat LVM, with increased Ogn protein expression associated with elevated LVM. We also applied genome-wide QTT analysis to the human heart and observed that, out of 22,000 transcripts, OGN transcript abundance had the highest correlation with LVM. We further confirmed a role for Ogn in the in vivo regulation of LVM in Ogn knockout mice. Taken together, these data implicate Ogn as a key regulator of LVM in rats, mice and humans, and suggest that Ogn modifies the hypertrophic response to extrinsic factors such as hypertension and aortic stenosis.
Collapse
Affiliation(s)
- Enrico Petretto
- Medical Research Council Clinical Sciences Centre, Faculty of Medicine, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
164
|
Lazaridis G, Pentheroudakis G, Pavlidis N. Integrating trastuzumab in the neoadjuvant treatment of primary breast cancer: Accumulating evidence of efficacy, synergy and safety. Crit Rev Oncol Hematol 2008; 66:31-41. [PMID: 17766143 DOI: 10.1016/j.critrevonc.2007.07.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2007] [Revised: 07/07/2007] [Accepted: 07/24/2007] [Indexed: 11/23/2022] Open
Abstract
Neoadjuvant chemotherapy is used in non-metastatic breast cancer in order to eradicate micrometastatic deposits early during disease course, as well as in order to decrease tumour bulk and render surgery feasible or breast-conserving. Moreover, it offers promise to serve as an in vivo chemosensitivity assay and as a powerful predictive factor for outcome. Trastuzumab, a monoclonal antibody targeting an epitope in the extracellular domain of the Human Epidermal Growth Factor Receptor-2 (HER2/erbB-2), was found to be active in HER2-overexpressing metastatic as well as in resected breast cancer when given post-operatively. In this review, we summarise the evidence on the activity and safety of trastuzumab-containing neoadjuvant chemotherapy for the management of women with localised, irresectable or resectable breast cancer. Twenty-three published studies enrolling a total of 585 patients reported pathologic complete responses (pCR) ranging from 7 to 78% with a favourable adverse event profile, data that are presented and discussed in this review. The impact of trastuzumab on long-term outcome, the identification of surrogate biomarkers for sensitivity or resistance to antineoplastic therapy, the optimal schedule of trastuzumab administration and the more active chemotherapeutic regimen for synergism are only a few of the key points needing elucidation so as to rationalise trastuzumab-based approaches.
Collapse
Affiliation(s)
- George Lazaridis
- Department of Medical Oncology, Ioannina University Hospital, Niarxou Avenue, 45500 Ioannina, Greece
| | | | | |
Collapse
|
165
|
Hauck L, Harms C, An J, Rohne J, Gertz K, Dietz R, Endres M, von Harsdorf R. Protein kinase CK2 links extracellular growth factor signaling with the control of p27(Kip1) stability in the heart. Nat Med 2008; 14:315-24. [PMID: 18311148 DOI: 10.1038/nm1729] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2007] [Accepted: 01/22/2008] [Indexed: 12/26/2022]
Abstract
p27(Kip1) (p27) blocks cell proliferation through the inhibition of cyclin-dependent kinase-2 (Cdk2). Despite its robust expression in the heart, little is known about both the function and regulation of p27 in this and other nonproliferative tissues, in which the expression of its main target, cyclin E-Cdk2, is known to be very low. Here we show that angiotensin II, a major cardiac growth factor, induces the proteasomal degradation of p27 through protein kinase CK2-alpha'-dependent phosphorylation. Conversely, unphosphorylated p27 potently inhibits CK2-alpha'. Thus, the p27-CK2-alpha' interaction is regulated by hypertrophic signaling events and represents a regulatory feedback loop in differentiated cardiomyocytes analogous to, but distinct from, the feedback loop arising from the interaction of p27 with Cdk2 that controls cell proliferation. Our data show that extracellular growth factor signaling regulates p27 stability in postmitotic cells, and that inactivation of p27 by CK2-alpha' is crucial for agonist- and stress-induced cardiac hypertrophic growth.
Collapse
Affiliation(s)
- Ludger Hauck
- Division of Cardiology, University Network Hospitals and Toronto General Research Institute, 200 Elizabeth Street, Toronto M5G 2C4, Canada
| | | | | | | | | | | | | | | |
Collapse
|
166
|
Anselmi A, Gaudino M, Baldi A, Vetrovec GW, Bussani R, Possati G, Abbate A. Role of apoptosis in pressure-overload cardiomyopathy. J Cardiovasc Med (Hagerstown) 2008; 9:227-32. [DOI: 10.2459/jcm.0b013e328277f1d7] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
167
|
Barry SP, Davidson SM, Townsend PA. Molecular regulation of cardiac hypertrophy. Int J Biochem Cell Biol 2008; 40:2023-39. [PMID: 18407781 DOI: 10.1016/j.biocel.2008.02.020] [Citation(s) in RCA: 218] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2007] [Revised: 02/13/2008] [Accepted: 02/15/2008] [Indexed: 01/05/2023]
Abstract
Heart failure is one of the leading causes of mortality in the western world and encompasses a wide spectrum of cardiac pathologies. When the heart experiences extended periods of elevated workload, it undergoes hypertrophic enlargement in response to the increased demand. Cardiovascular disease, such as that caused by myocardial infarction, obesity or drug abuse promotes cardiac myocyte hypertrophy and subsequent heart failure. A number of signalling modulators in the vasculature milieu are known to regulate heart mass including those that influence gene expression, apoptosis, cytokine release and growth factor signalling. Recent evidence using genetic and cellular models of cardiac hypertrophy suggests that pathological hypertrophy can be prevented or reversed and has promoted an enormous drive in drug discovery research aiming to identify novel and specific regulators of hypertrophy. In this review we describe the molecular characteristics of cardiac hypertrophy such as the aberrant re-expression of the fetal gene program. We discuss the various molecular pathways responsible for the co-ordinated control of the hypertrophic program including: natriuretic peptides, the adrenergic system, adhesion and cytoskeletal proteins, IL-6 cytokine family, MEK-ERK1/2 signalling, histone acetylation, calcium-mediated modulation and the exciting recent discovery of the role of microRNAs in controlling cardiac hypertrophy. Characterisation of the signalling pathways leading to cardiac hypertrophy has led to a wealth of knowledge about this condition both physiological and pathological. The challenge will be translating this knowledge into potential pharmacological therapies for the treatment of cardiac pathologies.
Collapse
Affiliation(s)
- Sean P Barry
- Medical Molecular Biology Unit, Institute of Child Health, University College London, 30 Guilford Street, London WC1N IEH, United Kingdom.
| | | | | |
Collapse
|
168
|
Fischer P, Hilfiker-Kleiner D. Role of gp130-mediated signalling pathways in the heart and its impact on potential therapeutic aspects. Br J Pharmacol 2008; 153 Suppl 1:S414-27. [PMID: 18246092 DOI: 10.1038/bjp.2008.1] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
IL-6-type cytokines bind to plasma membrane receptor complexes containing the common signal transducing receptor chain gp130 that is ubiquitously expressed in most tissues including the heart. The two major signalling cascades activated by the gp130 receptor, SHP2/ERK and STAT pathways, have been demonstrated to play important roles in cardiac development, hypertrophy, protection and remodelling in response to physiological and pathophysiological stimuli. Experimental data, both in vivo and in vitro, imply beneficial effects of gp130 signalling on cardiomyocytes in terms of growth and survival. In contrast, it has been reported that elevated serum levels of IL-6 cytokines and gp130 proteins are strong prognostic markers for morbidity and mortality in patients with heart failure or after myocardial infarction. Moreover, it has been shown that the local gp130 receptor system is altered in failing human hearts. In the present review, we summarize the basic principles of gp130 signalling, which requires simultaneous activation of STAT and ERK pathways under the tight control of positive and negative intracellular signalling modulators to provide a balanced biological outcome. Furthermore, we highlight the key role of the gp130 receptor and its major downstream effectors in the heart in terms of development and regeneration and in response to various physiological and pathophysiological stress situations. Finally, we comment on tissue-specific diversity and challenges in targeted pharmacological interference with components of the gp130 receptor system.
Collapse
Affiliation(s)
- P Fischer
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | | |
Collapse
|
169
|
Kong L, Andrassy M, Chang JS, Huang C, Asai T, Szabolcs MJ, Homma S, Liu R, Zou YS, Leitges M, Yan SD, Ramasamy R, Schmidt AM, Yan SF. PKCbeta modulates ischemia-reperfusion injury in the heart. Am J Physiol Heart Circ Physiol 2008; 294:H1862-70. [PMID: 18245560 DOI: 10.1152/ajpheart.01346.2007] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Protein kinase C-betaII (PKCbetaII) is an important modulator of cellular stress responses. To test the hypothesis that PKCbetaII modulates the response to myocardial ischemia-reperfusion (I/R) injury, we subjected mice to occlusion and reperfusion of the left anterior descending coronary artery. Homozygous PKCbeta-null (PKCbeta(-/-)) and wild-type mice fed the PKCbeta inhibitor ruboxistaurin displayed significantly decreased infarct size and enhanced recovery of left ventricular (LV) function and reduced markers of cellular necrosis and serum creatine phosphokinase and lactate dehydrogenase levels compared with wild-type or vehicle-treated animals after 30 min of ischemia followed by 48 h of reperfusion. Our studies revealed that membrane translocation of PKCbetaII in LV tissue was sustained after I/R and that gene deletion or pharmacological blockade of PKCbeta protected ischemic myocardium. Homozygous deletion of PKCbeta significantly diminished phosphorylation of c-Jun NH(2)-terminal mitogen-activated protein kinase and expression of activated caspase-3 in LV tissue of mice subjected to I/R. These data implicate PKCbeta in I/R-mediated myocardial injury, at least in part via phosphorylation of JNK, and suggest that blockade of PKCbeta may represent a potent strategy to protect the vulnerable myocardium.
Collapse
Affiliation(s)
- Linghua Kong
- Dept. of Surgery, College of Physicians and Surgeons of Columbia University, New York, NY 10032, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
170
|
Rohrbach S, Engelhardt S, Lohse MJ, Werdan K, Holtz J, Muller-Werdan U. Activation of AP-1 contributes to the beta-adrenoceptor-mediated myocardial induction of interleukin-6. Mol Med 2008; 13:605-14. [PMID: 17948064 DOI: 10.2119/2007-00071.rohrbach] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2007] [Accepted: 10/05/2007] [Indexed: 01/08/2023] Open
Abstract
The induction of proinflammatory cytokines in stressed myocardium is considered an innate immune response, but the role of beta-adrenergic signaling in this proinflammatory response and the mechanisms of cardioprotection by beta-blockers are not fully understood. In the present study, we analyzed interleukin-6 (IL-6) formation and promoter activation in beta-adrenoceptor-stimulated neonatal rat cardiomyocytes, in transgenic mice with cardiac overexpression of beta1-adrenoceptors, and in failing human myocardium. IL-6 formation and release in cultured cardiomyocytes under beta-adrenoceptor stimulation requires the activation of activating protein-1 (AP-1) binding sites and of cAMP response elements (CRE) in the IL-6 promoter, but this release (140 +/- 6 pg/mL medium under 10(-6) M isoproterenol vs. 81 +/- 3 pg/mL unstimulated, P < 0.05) is moderate compared with that under inflammatory stimulation (855 +/- 44 pg/mL, endotoxin 0.1microg/mL). Similarly, IL-6 is induced together with CRE- and AP-1 activation in the left ventricle (LV) of beta1-transgenic mice before the onset of failure. However, we observed IL-6 induction with activation of NF-kappaB in addition to CRE and AP-1 in beta1-transgenic mice at the age of 22 weeks and in explanted human LV after full development of failure. Treatment with beta-blockers lowered myocardial IL-6 as well as AP-1, NF-kappaB, and CRE activation. Therefore, the activation of AP-1 and CRE is part of beta-adrenergic signal transduction for IL-6 induction in nonfailing and failing cardiomyocytes, whereas NF-kappaB activation contributes only in overloaded failing myocardium.
Collapse
Affiliation(s)
- Susanne Rohrbach
- Institute of Pathophysiology, Martin Luther University Halle-Wittenberg, Germany.
| | | | | | | | | | | |
Collapse
|
171
|
A non-conservative polymorphism in the IL-6 signal transducer (IL6ST)/gp130 is associated with myocardial infarction in a hypertensive population. ACTA ACUST UNITED AC 2008; 146:189-96. [DOI: 10.1016/j.regpep.2007.09.031] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2007] [Revised: 09/03/2007] [Accepted: 09/05/2007] [Indexed: 12/19/2022]
|
172
|
Qian JY, Harding P, Liu Y, Shesely E, Yang XP, LaPointe MC. Reduced cardiac remodeling and function in cardiac-specific EP4 receptor knockout mice with myocardial infarction. Hypertension 2008; 51:560-6. [PMID: 18180401 PMCID: PMC3115709 DOI: 10.1161/hypertensionaha.107.102590] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2007] [Accepted: 12/06/2007] [Indexed: 01/18/2023]
Abstract
We have shown previously that cyclooxygenase-2 inhibition reduces cardiac hypertrophy and fibrosis postmyocardial infarction (MI) in a mouse model and that prostaglandin E(2) stimulates cardiomyocyte hypertrophy in vitro through its EP(4) receptor. Because the role of cardiac myocyte EP(4) in cardiac function and hypertrophy in vivo is unknown, we generated mice lacking EP(4) only in cardiomyocytes (CM- EP(4) knockout [KO]). Twelve- to 14-week-old mice were evaluated using echocardiography and histology. There were no differences in ejection fraction, myocyte cross-sectional area, and interstitial collagen fraction between KO mice and littermate controls. To test the hypothesis that EP(4) is involved in cardiac remodeling after MI, we induced MI by ligating the left anterior descending coronary artery. Two weeks later, the mice were subjected to echocardiography, and hearts were removed for histology and Western blot. There was no difference in infarct size between KO mice and controls; however, KO mice showed less myocyte cross-sectional area and interstitial collagen fraction than controls. Also, CM-EP4 KO mice had reduced ejection fraction. Because the transcription factor Stat-3 is involved in hypertrophy and protection from ischemic injury, we tested whether it was activated in control and KO mouse hearts after MI. Western blot indicated that Stat-3 was activated in control hearts after MI but not in KO hearts. Thus, CM-EP4 deletion decreased hypertrophy, fibrosis, and activation of Stat-3. However, cardiac function was unexpectedly worsened in these mice. We conclude that cardiac myocyte EP(4) plays a role in hypertrophy via activation of Stat-3, a process that seems to be cardioprotective.
Collapse
Affiliation(s)
- Jian-Yong Qian
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, 2799 W Grand Blvd, Detroit, MI 48202-2689, USA
| | | | | | | | | | | |
Collapse
|
173
|
Diwan A, Wansapura J, Syed FM, Matkovich SJ, Lorenz JN, Dorn GW. Nix-mediated apoptosis links myocardial fibrosis, cardiac remodeling, and hypertrophy decompensation. Circulation 2008; 117:396-404. [PMID: 18178777 PMCID: PMC2538800 DOI: 10.1161/circulationaha.107.727073] [Citation(s) in RCA: 142] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND Pathological cardiac hypertrophy inevitably remodels, leading to functional decompensation. Although modulation of apoptosis-regulating genes occurs in cardiac hypertrophy, a causal role for programmed cardiomyocyte death in left ventricular (LV) remodeling has not been established. METHODS AND RESULTS We targeted the gene for proapoptotic Nix, which is transcriptionally upregulated in pressure overload and Gq-dependent hypertrophies, in the mouse germ line or specifically in cardiomyocytes (knockout [KO]) and conditionally overexpressed it in the heart (transgenic [TG]). Conditional forced Nix expression acted synergistically with the prohypertrophic Gq transgene to increase cardiomyocyte apoptosis (0.8+/-0.1% in GqTG versus 7.8+/-0.6% in GqTG+NixTG; P<0.001), causing lethal cardiomyopathy with LV dilation and depressed systolic function (percent fractional shortening, 39+/-4 versus 23+/-4; P=0.042). In the reciprocal experiment, germ-line Nix ablation significantly reduced cardiomyocyte apoptosis (4.8+/-0.2% in GqTG+NixKO versus 8.4+/-0.5% in GqTG; P=0.001), which improved percent fractional shortening (43+/-3% versus 27+/-3%; P=0.017), attenuated LV remodeling, and largely prevented lethality in the Gq peripartum model of apoptotic cardiomyopathy. Cardiac-specific (Nkx2.5-Cre) Nix KO mice subjected to transverse aortic constriction developed significantly less LV dilation by echocardiography and magnetic resonance imaging, maintained concentric remodeling, and exhibited preserved LV ejection fraction (61+/-2% in transverse aortic constriction cardiac Nix KO versus 36+/-6% in transverse aortic constriction wild-type mice; P=0.003) at 9 weeks, with reduced cardiomyocyte apoptosis at day 4 (1.70+/-0.21% versus 2.73+/-0.35%; P=0.032). CONCLUSIONS Nix-induced cardiomyocyte apoptosis is a major determinant of adverse remodeling in pathological hypertrophies, a finding that suggests therapeutic value for apoptosis inhibition to prevent cardiomyopathic decompensation.
Collapse
Affiliation(s)
- Abhinav Diwan
- Center for Molecular Cardiovascular Research, University of Cincinnati, Cincinnati, OH
| | - Janaka Wansapura
- Imaging Research Center, Department of Radiology, Children's Hospital Medical Center, Cincinnati, OH
| | - Faisal M. Syed
- Center for Molecular Cardiovascular Research, University of Cincinnati, Cincinnati, OH
| | - Scot J. Matkovich
- Center for Molecular Cardiovascular Research, University of Cincinnati, Cincinnati, OH
| | - John N. Lorenz
- Department of Molecular and Cellular Physiology, University of Cincinnati, Cincinnati, OH
| | - Gerald W. Dorn
- Center for Molecular Cardiovascular Research, University of Cincinnati, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH
| |
Collapse
|
174
|
Cutler MJ, Rosenbaum DS, Dunlap ME. Structural and electrical remodeling as therapeutic targets in heart failure. J Electrocardiol 2008; 40:S1-7. [PMID: 17993305 DOI: 10.1016/j.jelectrocard.2007.05.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2007] [Accepted: 05/30/2007] [Indexed: 10/22/2022]
Abstract
Heart failure is a progressive clinical syndrome that is characterized by remodeling of the myocardium in response to various stress signals. The past several years has seen remarkable progress in unraveling the molecular and cellular mechanisms of structural and electrical remodeling in HF. Improved understanding of the molecular mechanism of myocardial remodeling has resulted in improved HF therapies and revealed potentially novel therapeutic targets. This review discusses the mechanisms of myocardial remodeling in HF and their clinical manifestations. Current and investigational HF therapies targeting these mechanisms also will be discussed.
Collapse
Affiliation(s)
- Michael J Cutler
- Heart and Vascular Research Center, MetroHealth Campus, Case Western Reserve University, Cleveland, OH 44109-1998, USA
| | | | | |
Collapse
|
175
|
Beeri R, Yosefy C, Guerrero JL, Nesta F, Abedat S, Chaput M, del Monte F, Handschumacher MD, Stroud R, Sullivan S, Pugatsch T, Gilon D, Vlahakes GJ, Spinale FG, Hajjar RJ, Levine RA. Mitral Regurgitation Augments Post-Myocardial Infarction Remodeling. J Am Coll Cardiol 2008; 51:476-86. [PMID: 18222360 DOI: 10.1016/j.jacc.2007.07.093] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2007] [Revised: 07/09/2007] [Accepted: 07/16/2007] [Indexed: 12/01/2022]
Affiliation(s)
- Ronen Beeri
- Cardiac Ultrasound Laboratory, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
176
|
Kurdi M, Booz GW. Can the protective actions of JAK-STAT in the heart be exploited therapeutically? Parsing the regulation of interleukin-6-type cytokine signaling. J Cardiovasc Pharmacol 2007; 50:126-41. [PMID: 17703129 DOI: 10.1097/fjc.0b013e318068dd49] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Activation of the transcription factor signal transducers and activators of transcription (STAT) 3 is a defining feature of the interleukin (IL)-6 family of cytokines, which include IL-6, leukemia inhibitory factor, and cardiotrophin-1. These cytokines, as well as STAT3 activation, have been shown to be protective for cardiac myocytes and necessary for ischemia preconditioning. However, the mechanisms that regulate IL-6-type cytokine signaling in cardiac myocytes are largely unexplored. We propose that the protective character of IL-6-type cytokine signaling in cardiac myocytes is determined principally by three mechanisms: redox status of the nonreceptor tyrosine kinase Janus kinase 1 (JAK) 1 that activates STAT3, phosphorylation of STAT3 within the transcriptional activation domain on serine 727, and STAT3-mediated induction of suppressor of cytokine signaling (SOCS) 3 that terminates IL-6-type cytokine signaling. Moreover, we hypothesize that hyperactivation of the JAK kinases, particularly JAK2, mismatched STAT3 serine-tyrosine phosphorylation or heightened STAT3 transcriptional activity, and SOCS3 induction may ultimately prove detrimental. Here we summarize recent evidence that supports this hypothesis, as well as additional possible mechanisms of JAK-STAT regulation. Understanding how IL-6-type cytokine signaling is regulated in cardiac myocytes has great significance for exploiting the therapeutic potential of these cytokines and the phenomenon of preconditioning.
Collapse
Affiliation(s)
- Mazen Kurdi
- Division of Molecular Cardiology, Cardiovascular Research Institute, College of Medicine, The Texas A&M University System Health Science Center, College Station, TX 76504, USA
| | | |
Collapse
|
177
|
|
178
|
González A, Ravassa S, Loperena I, López B, Beaumont J, Querejeta R, Larman M, Díez J. Association of depressed cardiac gp130-mediated antiapoptotic pathways with stimulated cardiomyocyte apoptosis in hypertensive patients with heart failure. J Hypertens 2007; 25:2148-57. [PMID: 17885560 DOI: 10.1097/hjh.0b013e32828626e2] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To investigate whether the glycoprotein (gp130)-mediated survival pathway, which protects cardiomyocytes from apoptosis, is depressed in left ventricular hypertrophy hypertensive patients with chronic heart failure. METHODS Transvenous endomyocardial biopsies were obtained in 52 hypertensive patients with left ventricular hypertrophy: 28 without heart failure and 24 with heart failure. gp130 and gp130-dependent antiapoptotic pathways p42/44 mitogen-activated protein kinase (MAPK) and phosphatidylinositol-3 kinase (PI3K)/protein kinase B (Akt) as well as gp130 agonist cardiotrophin-1 were analyzed by reverse transcriptase-polymerase chain reaction and western blot. Apoptosis was assessed by DNA end-labeling (TUNEL), caspase-3 immunostaining and caspase substrate poly(ADP-ribose) polymerase cleavage. RESULTS gp130 protein expression (P < 0.05) and p42/44 MAPK and PI3K/Akt activation (P < 0.01) were decreased in heart-failure hypertensive patients compared with nonheart-failure hypertensive individuals. No changes in gp130 mRNA expression were found between the two groups. Cardiotrophin-1 was increased (P < 0.05) at both the mRNA and protein levels in heart-failure hypertensive individuals compared with nonheart-failure hypertensive individuals. Cardiomyocyte apoptosis was increased (P < 0.01) in heart-failure hypertensive individuals compared with nonheart-failure hypertensive individuals. Inverse correlations (P < 0.01) occurred between cardiomyocyte apoptosis and p42/44 MAPK and PI3K/Akt activation in all hypertensive patients. Cardiotrophin-1 correlated inversely (r = -0.554, P < 0.05) with gp130 in all hypertensive individuals. In cultured HL-1 cardiomyocytes, cardiotrophin-1 decreased (P < 0.05) the gp130:phosphorylated gp130 (at Ser782) ratio and increased (P < 0.05) gp130ubiquitination. CONCLUSIONS An association exists between depression of the gp130 cytoprotective pathway and stimulation of cardiomyocyte apoptosis in hypertensive patients that develop heart failure. Whether the excess of cardiotrophin-1 induces ligand-induced receptor down-regulation in these patients requires further study.
Collapse
Affiliation(s)
- Arantxa González
- Division of Cardiovascular Sciences, Centre for Applied Medical Research, University of Navarra, Pamplona, Spain
| | | | | | | | | | | | | | | |
Collapse
|
179
|
Beeri R, Yosefy C, Guerrero JL, Abedat S, Handschumacher MD, Stroud RE, Sullivan S, Chaput M, Gilon D, Vlahakes GJ, Spinale FG, Hajjar RJ, Levine RA. Early Repair of Moderate Ischemic Mitral Regurgitation Reverses Left Ventricular Remodeling: A Functional and Molecular Study. Circulation 2007; 116:I288-93. [PMID: 17846319 DOI: 10.1161/circulationaha.106.681114] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Mitral regurgitation (MR) doubles postmyocardial infarction (MI) mortality. We have shown that moderate MR augments remodeling in an apical MI model (no intrinsic MR) with independent left ventricle-to-left atrial MR-type flow. We hypothesized that repairing moderate MR 1 month after MI reverses this remodeling. METHODS AND RESULTS Anteroapical MIs were created in 18 sheep, and a left ventricle-to-left atrial shunt implanted in 12 (regurgitant fraction, 30%). Six sheep had the shunt closed at 1 month (repair group). Sheep were compared at baseline, and at 1 and 3 months. Sheep in the MI+MR (unrepaired) and repaired groups remodeled during the first month (120% increased left ventricular end-systolic volume [ESV; P<0.01]), but shunt closure reversed remodeling at 3 months, with end-diastolic volume (EDV) and ESV 135% and 128% of baseline versus 220% and 280% without repair (P<0.001). At 3 months, dP/dt and preload-recruitable stroke work were relatively maintained in the repaired and MI-only groups versus nearly 50% decreases without repair. Prohypertrophic gp130 and antiapoptotic pAkt increased followed by exhaustion below baseline without repair, but remained elevated at 3 months with repair or MI only. With repair, matrix metalloproteinase-2 decreased to < or = 50% that without repair in remote and border zones at 3 months, and the matrix metalloproteinase inhibitor TIMP-4 increased dramatically. CONCLUSIONS Early repair of moderate MR in the setting of apical MI substantially reverses the otherwise progressive remodeling process, with reduced left ventricular volumes, relatively maintained contractility, persistently activated intracellular signals promoting hypertrophy and opposing apoptosis, and reduced matrix proteolytic activity. These findings are of interest for the current controversy regarding potential benefits of repair of MR after MI.
Collapse
Affiliation(s)
- Ronen Beeri
- Cardiac Ultrasound Laboratory, Cardiovascular Research Center, and Cardiothoracic Surgery Department, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
180
|
|
181
|
Qu J, Zhou J, Yi XP, Dong B, Zheng H, Miller LM, Wang X, Schneider MD, Li F. Cardiac-specific haploinsufficiency of beta-catenin attenuates cardiac hypertrophy but enhances fetal gene expression in response to aortic constriction. J Mol Cell Cardiol 2007; 43:319-26. [PMID: 17673255 PMCID: PMC2084259 DOI: 10.1016/j.yjmcc.2007.06.006] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2007] [Revised: 04/26/2007] [Accepted: 06/08/2007] [Indexed: 11/21/2022]
Abstract
In addition to its role in cell adhesion, beta-catenin is an important signaling molecule in the Wnt/Wingless signaling pathway. Recent studies have indicated that beta-catenin is stabilized by hypertrophic stimuli and may regulate cardiac hypertrophic responses. To explore the role and requirement of beta-catenin in cardiac development and hypertrophy, we deleted the beta-catenin gene specifically in cardiac myocytes by crossing loxP-floxed beta-catenin mice with transgenic mice expressing a Cre recombinase under the control of the alpha-myosin heavy chain promoter. No homozygous beta-catenin-deleted mice were born alive and died before embryonic day 14.5, indicating significant and irreplaceable roles of beta-catenin in embryonic heart development. Heterozygous beta-catenin-deleted mice, however, demonstrated no structural and functional abnormality. The response of heterozygous beta-catenin-deleted mice to transverse aortic constriction, however, was significantly attenuated with decreased heart weight and heart weight/body weight ratio compared to controls with intact beta-catenin genes. Hemodynamic analysis revealed that there was no difference in cardiac function between wild-type and heterozygous beta-catenin-deleted mice. On the other hand, the expression of fetal genes, beta-myosin heavy chain, atrial and brain natriuretic peptides was significantly higher in heterozygous beta-catenin-deleted mice when compared to wild-type beta-catenin mice. These results suggest that the cytoplasmic level of beta-catenin modulates hypertrophic response and fetal gene reprogramming after pressure overload.
Collapse
Affiliation(s)
- Jiaxiang Qu
- Sanford Research/USD, Cardiovascular Research Institute, Sanford School of Medicine of The University of South Dakota and Sanford Health, Sioux Falls, SD 57105
| | - Jibin Zhou
- Sanford Research/USD, Cardiovascular Research Institute, Sanford School of Medicine of The University of South Dakota and Sanford Health, Sioux Falls, SD 57105
| | - Xian Ping Yi
- Sanford Research/USD, Cardiovascular Research Institute, Sanford School of Medicine of The University of South Dakota and Sanford Health, Sioux Falls, SD 57105
- Department of Pathology, Zhongshan University the Fifth Affiliated Hospital, 52 Meihua East Road, Zhuhai, Guangdong Province 519000, P. R. China
| | - Baojun Dong
- Sanford Research/USD, Cardiovascular Research Institute, Sanford School of Medicine of The University of South Dakota and Sanford Health, Sioux Falls, SD 57105
| | - Hanqiao Zheng
- Sanford Research/USD, Cardiovascular Research Institute, Sanford School of Medicine of The University of South Dakota and Sanford Health, Sioux Falls, SD 57105
- Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, 414 East Clark Street, Lee Medical Building, Vermillion, SD 57069
| | - Lisa M. Miller
- Sanford Research/USD, Cardiovascular Research Institute, Sanford School of Medicine of The University of South Dakota and Sanford Health, Sioux Falls, SD 57105
| | - Xuejun Wang
- Sanford Research/USD, Cardiovascular Research Institute, Sanford School of Medicine of The University of South Dakota and Sanford Health, Sioux Falls, SD 57105
- Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, 414 East Clark Street, Lee Medical Building, Vermillion, SD 57069
| | - Michael D. Schneider
- Department of Medicine, Department of Cell Biology, and Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas 77030
| | - Faqian Li
- Sanford Research/USD, Cardiovascular Research Institute, Sanford School of Medicine of The University of South Dakota and Sanford Health, Sioux Falls, SD 57105
| |
Collapse
|
182
|
Smart N, Risebro CA, Melville AAD, Moses K, Schwartz RJ, Chien KR, Riley PR. Thymosin beta-4 is essential for coronary vessel development and promotes neovascularization via adult epicardium. Ann N Y Acad Sci 2007; 1112:171-88. [PMID: 17495252 DOI: 10.1196/annals.1415.000] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Ischemic heart disease leading to myocardial infarction causes irreversible cell loss and scarring and is a major cause of morbidity and mortality in humans. Significant effort in the field of cardiovascular medicine has been invested in the search for adult cardiac progenitor cells that may replace damaged muscle cells and/or contribute to new vessel formation (neovascularization) and in the identification of key factors, which may induce such progenitor cells to contribute to myocardial repair and collateral vessel growth. We recently demonstrated that the actin monomer-binding protein, thymosin beta-4 (Tbeta-4), when secreted from the myocardium provides a paracrine stimulus to the cells of the epicardium-derived cells (EPDCs) to promote their inward migration and differentiation into endothelial and smooth muscle cells to form the coronary vasculature. Translating this essential role for Tbeta-4 in coronary vessel development to the adult, we found that treatment of cultured adult explants with Tbeta-4 stimulated extensive outgrowth of epicardin-positive epicardial cells, which, as they migrated away from the explant, differentiated into procollagen type I, SMalphaA, and Flk1-positive cells indicative of fibroblasts, smooth muscle, and endothelial cells; thus releasing the adult epicardium from a quiescent state and restoring pluripotency. The ability of Tbeta-4 to promote coronary vessel development and potentially induce new vasculature in the adult is essential for cardiomyocyte survival and could contribute significantly toward the reported Tbeta4-induced cardioprotection and repair in the adult heart. Tbeta-4 is currently subject to multicenter phase 1 clinical trials for treatment of cardiovascular disease (http://www.regenerx.com), therefore, insight into the repair mechanism(s) induced by Tbeta-4 is an essential step toward harnessing therapeutic survival, migration, and repair properties of the peptide in the context of acute myocardial damage.
Collapse
Affiliation(s)
- Nicola Smart
- Molecular Medicine Unit, UCL Institute of Child Health, London, WC1N 1EH, UK
| | | | | | | | | | | | | |
Collapse
|
183
|
Yndestad A, Damås JK, Øie E, Ueland T, Gullestad L, Aukrust P. Role of inflammation in the progression of heart failure. Curr Cardiol Rep 2007; 9:236-41. [PMID: 17470337 DOI: 10.1007/bf02938356] [Citation(s) in RCA: 132] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Chronic heart failure (HF) is a disorder characterized in part by immune activation and inflammation. Thus, patients with HF have elevated levels of a number of inflammatory cytokines, both in the circulation and in the failing heart itself. Several mechanisms for this immune activation, which are not mutually exclusive, have been suggested, including neurohormonal activation, hemodynamic overload, and activation of the innate immune system secondary to cardiac stress. Importantly, experimental studies have shown that inflammatory cytokines such as tumor necrosis factor-alpha, interleukin-1b, and monocyte chemoattractant peptide-1 may contribute to the development and progression of HF by promoting myocardial hypertrophy, activating matrix metalloproteinases, provoking contractile dysfunction, and inducing apoptosis. However, inflammatory cytokines may also have adaptive and cardioprotective effects. This important aspect of cytokine biology must be kept in mind when designing new immunomodulatory treatment modalities in HF.
Collapse
Affiliation(s)
- Arne Yndestad
- Section of Clinical Immunology and Infectious Diseases, Medical Department, Rikshospitalet-Radiumhospitalet Medical Center, Sognsvannsveien 20, N-0027 Oslo, Norway
| | | | | | | | | | | |
Collapse
|
184
|
Mercadier JJ. Approches physiopathologiques actuelles de l'insuffisance cardiaque. Presse Med 2007; 36:979-84. [PMID: 17433611 DOI: 10.1016/j.lpm.2007.02.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2007] [Accepted: 02/12/2007] [Indexed: 11/20/2022] Open
Abstract
Heart failure is the consequence of cardiac remodeling that affects all the structural and functional aspects of the heart, from its ventricular geometry to the molecular components of myocytes and other myocardial cells. This remodeling is activated by biomechanical stress from the onset of the causal disease (sudden in cases of myocardial infarction and more progressively in cases of hypertension, for example). This biomechanical stress combines, depending on the cause, diverse degrees of diastolic stretching and systolic overload of the ventricles with systemic and tissular neurohormonal modifications. These mechanical and neurohormonal factors activate numerous intracellular signaling pathways, interconnected in a complex web, and lead to reprogramming the genome of the myocytes and other myocardial cells. Activation of some of these pathways leads to a beneficial adaptive remodeling (growth, cardiac hypertrophy of pregnancy and of athletes) or on the contrary to harmful remodeling (heart disease). The predominance of the stimulation of the harmful pathways over that of the beneficial pathways in heart disease is responsible for progression towards heart failure. Current research aims at identifying new pathways and participants in the beneficial and harmful remodeling of the myocardium in order to develop new drugs that will block ever more specifically the harmful pathways but also stimulate the beneficial ones, to prevent progression towards heart failure.
Collapse
Affiliation(s)
- Jean-Jacques Mercadier
- Services d'explorations fonctionnelles et de cardiologie, AP-HP, Inserm U698, Groupe hospitalier Bichat - Claude Bernard, Paris (75), France.
| |
Collapse
|
185
|
Fischer P, Hilfiker-Kleiner D. Survival pathways in hypertrophy and heart failure: the gp130-STAT3 axis. Basic Res Cardiol 2007; 102:279-97. [PMID: 17530315 DOI: 10.1007/s00395-007-0658-z] [Citation(s) in RCA: 125] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2007] [Revised: 04/23/2007] [Accepted: 04/24/2007] [Indexed: 12/26/2022]
Abstract
Circulating levels of interleukin (IL)-6 and related cytokines are elevated in patients with congestive heart failure and after myocardial infarction. Serum IL-6 concentrations are related to decreasing functional status of these patients and provide important prognostic information.Moreover, in the failing human heart, multiple components of the IL-6- glycoprotein (gp)130 receptor system are impaired, implicating an important role of this system in cardiac pathophysiology.Experimental studies have shown that the common receptor subunit of IL-6 cytokines is phosphorylated in response to pressure overload and myocardial infarction and that it subsequently activates at least three different downstream signaling pathways, the signal transducers and activators of transcription 1 and 3 (STAT1/3), the Src-homology tyrosine phosphatase 2 (SHP2)-Ras-ERK, and the PI3K-Akt system. Gp130 receptor mediated signaling promotes cardiomyocyte survival, induces hypertrophy, modulates cardiac extracellular matrix and cardiac function. In this regard, the gp130 receptor system and its main downstream mediator STAT3 play a key role in cardioprotection. This review summarizes the current knowledge of IL-6 cytokines, gp130 receptor and STAT3 signaling in the heart exposed to physiological (aging, pregnancy) and pathophysiological stress (ischemia, pressure overload, inflammation and cardiotoxic agents) with a special focus on the potential role of individual IL-6 cytokines.
Collapse
Affiliation(s)
- P Fischer
- Dept. of Cardiology & Angiology, Medical School Hannover, Hannover, Germany
| | | |
Collapse
|
186
|
|
187
|
Hoshijima M, Knöll R, Pashmforoush M, Chien KR. Reversal of calcium cycling defects in advanced heart failure toward molecular therapy. J Am Coll Cardiol 2007; 48:A15-23. [PMID: 17084280 DOI: 10.1016/j.jacc.2006.06.070] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2006] [Revised: 05/22/2006] [Accepted: 06/22/2006] [Indexed: 02/04/2023]
Abstract
Heart failure is a growing major cause of human morbidity and mortality worldwide. A wave of new insights from diverse laboratories has begun to uncover new therapeutic strategies that affect the molecular pathways within cardiomyocytes that drive heart failure progression. Using an integrative approach that employs insights from genetic-based studies in mouse and humans and in vivo somatic gene transfer studies, we have uncovered a new link between stress signals mediated by mechanical stretch and defects in sarcoplasmic reticulum (SR) calcium cycling. An intrinsic mechanical stress sensing system is embedded in the Z disc of cardiomyocytes, and defects in stretch responses can lead to heart failure progression and associated increases in wall stress. Reversal of the chronic increases in wall stress by promoting SR calcium cycling can prevent and partially reverse heart failure progression in multiple genetic and acquired model systems of heart failure in both small and large animals. We propose that reversal of advanced heart failure is possible by targeting the defects in SR calcium cycling, which may be a final common pathway for the progression of many forms of heart failure.
Collapse
Affiliation(s)
- Masahiko Hoshijima
- Institute of Molecular Medicine, University of California San Diego, La Jolla, California, USA
| | | | | | | |
Collapse
|
188
|
Kimura R, Maeda M, Arita A, Oshima Y, Obana M, Ito T, Yamamoto Y, Mohri T, Kishimoto T, Kawase I, Fujio Y, Azuma J. Identification of cardiac myocytes as the target of interleukin 11, a cardioprotective cytokine. Cytokine 2007; 38:107-15. [PMID: 17629706 DOI: 10.1016/j.cyto.2007.05.011] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2007] [Revised: 04/30/2007] [Accepted: 05/25/2007] [Indexed: 11/23/2022]
Abstract
Interleukin (IL)-6 family cytokines, which share glycoprotein 130 (gp130) as a signal-transducing receptor component, play important roles in the maintenance of cardiac homeostasis. IL-11, a member of IL-6 family cytokines, is expressed in cardiac myocytes, though it remains to be elucidated how IL-11 functions in the hearts. In the present study, first, we showed that IL-11 administration reduced the ischemia/reperfusion injury in the hearts. IL-11 receptor alpha was expressed in cardiomyocytes. IL-11 treatment rapidly activated signal transducer and activator of transcription 3 (STAT3) and extracellular signal-regulated kinase (ERK) 1/2 in cardiac myocytes. IL-11 stimulation resulted in the translocation of phosphorylated STAT3 into nuclei. Immunofluorescence microscopic analyses revealed that IL-11 treatment led to the cell elongation, as is the case with other cardiotrophic members of IL-6 family, such as leukemia inhibitory factor. Finally we showed that IL-11 treatment conferred the resistance to cell death induced by hydrogen peroxide, which was abrogated by adenoviral transfer of dominant negative STAT3, but not by the inhibition of ERK1/2 with U0126. These findings indicate that IL-11 mediates cytoprotective signals in cardiomyocytes, proposing that IL-11 has the potential to exhibit cardioprotection as a novel biological function.
Collapse
Affiliation(s)
- Ryusuke Kimura
- Department of Clinical Pharmacology and Pharmacogenomics, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita City, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
189
|
|
190
|
|
191
|
Abstract
Understanding the cytokine network is a very complex task. One way is the dissection of the network by the generation and analysis of mutant mice. As the technology advances more sophisticated approaches toward this goal become available and proof to disclose an even more complex picture of the cytokine network as we initially anticipated. This increase in complexity leads to fascinating challenges in the future.
Collapse
Affiliation(s)
- Werner Müller
- University of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK.
| |
Collapse
|
192
|
Abstract
Cardiac myocytes rapidly proliferate during fetal life but exit the cell cycle soon after birth in mammals. Although the extent to which adult cardiac myocytes are capable of cell cycle reentry is controversial and species-specific differences may exist, it appears that for the vast majority of adult cardiac myocytes the predominant form of growth postnatally is an increase in cell size (hypertrophy) not number. Unfortunately, this limits the ability of the heart to restore function after any significant injury. Interest in novel regenerative therapies has led to the accumulation of much information on the mechanisms that regulate the rapid proliferation of cardiac myocytes in utero, their cell cycle exit in the perinatal period, and the permanent arrest (terminal differentiation) in adult myocytes. The recent identification of cardiac progenitor cells capable of giving rise to cardiac myocyte-like cells has challenged the dogma that the heart is a terminally differentiated organ and opened new prospects for cardiac regeneration. In this review, we summarize the current understanding of cardiomyocyte cell cycle control in normal development and disease. In addition, we also discuss the potential usefulness of cardiomyocyte self-renewal as well as feasibility of therapeutic manipulation of the cardiac myocyte cell cycle for cardiac regeneration.
Collapse
Affiliation(s)
- Preeti Ahuja
- Division of Cardiology, University of California at Los Angeles
- The Cardiovascular Research Laboratories, Departments of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Patima Sdek
- Division of Cardiology, University of California at Los Angeles
- The Cardiovascular Research Laboratories, Departments of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - W. Robb Maclellan
- Corresponding author: W. Robb MacLellan, Cardiovascular Research Laboratories, David Geffen school of Medicine at UCLA, 675 C.E. Young Dr., MRL 3-645, Los Angeles, California, 90095-1760; Phone: (310) 825-2556; Fax: (310) 206-5777; e-mail:
| |
Collapse
|
193
|
Iso Y, Spees JL, Serrano C, Bakondi B, Pochampally R, Song YH, Sobel BE, Delafontaine P, Prockop DJ. Multipotent human stromal cells improve cardiac function after myocardial infarction in mice without long-term engraftment. Biochem Biophys Res Commun 2007; 354:700-6. [PMID: 17257581 PMCID: PMC1851899 DOI: 10.1016/j.bbrc.2007.01.045] [Citation(s) in RCA: 295] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2007] [Accepted: 01/07/2007] [Indexed: 12/20/2022]
Abstract
The aim of this study was to determine whether intravenously administered multipotent stromal cells from human bone marrow (hMSCs) can improve cardiac function after myocardial infarction (MI) without long-term engraftment and therefore whether transitory paracrine effects or secreted factors are responsible for the benefit conferred. hMSCs were injected systemically into immunodeficient mice with acute MI. Cardiac function and fibrosis after MI in the hMSC-treated group were significantly improved compared with controls. However, despite the cardiac improvement, there was no evident hMSC engraftment in the heart 3 weeks after MI. Microarray assays and ELISAs demonstrated that multiple protective factors were expressed and secreted from the hMSCs in culture. Factors secreted by hMSCs prevented cell death of cultured cardiomyocytes and endothelial cells under conditions that mimicked tissue ischemia. The favorable effects of hMSCs appear to reflect the impact of secreted factors rather than engraftment, differentiation, or cell fusion.
Collapse
Affiliation(s)
- Yoshitaka Iso
- Center for Gene Therapy, Tulane University Health Sciences Center, 1430 Tulane Avenue, New Orleans, LA 70112
- Cardiovascular Research Institute, Department of Medicine, University of Vermont, 208 South Park Drive, Colchester, VT 05446
| | - Jeffrey L. Spees
- Center for Gene Therapy, Tulane University Health Sciences Center, 1430 Tulane Avenue, New Orleans, LA 70112
- Cardiovascular Research Institute, Department of Medicine, University of Vermont, 208 South Park Drive, Colchester, VT 05446
| | - Claudia Serrano
- Section of Cardiology, Department of Medicine, Tulane University Health Sciences Center, 1430 Tulane Avenue, New Orleans, LA 70112
| | - Benjamin Bakondi
- Cardiovascular Research Institute, Department of Medicine, University of Vermont, 208 South Park Drive, Colchester, VT 05446
| | - Radhika Pochampally
- Center for Gene Therapy, Tulane University Health Sciences Center, 1430 Tulane Avenue, New Orleans, LA 70112
| | - Yao-Hua Song
- Section of Cardiology, Department of Medicine, Tulane University Health Sciences Center, 1430 Tulane Avenue, New Orleans, LA 70112
| | - Burton E. Sobel
- Cardiovascular Research Institute, Department of Medicine, University of Vermont, 208 South Park Drive, Colchester, VT 05446
| | - Patrick Delafontaine
- Section of Cardiology, Department of Medicine, Tulane University Health Sciences Center, 1430 Tulane Avenue, New Orleans, LA 70112
| | - Darwin J. Prockop
- Center for Gene Therapy, Tulane University Health Sciences Center, 1430 Tulane Avenue, New Orleans, LA 70112
| |
Collapse
|
194
|
Dai RP, Xu JM, Tao LJ, Li L, Li ZP, Zhang JY. Induction of tumor necrosis-alpha, p38 and JNK in the spinal cord following acute heart injury in the rat model. Acta Anaesthesiol Scand 2007; 51:365-71. [PMID: 17257180 DOI: 10.1111/j.1399-6576.2006.01218.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND It is still not known whether the spinal cytokine signaling pathways are involved in the pathophysiologic mechanism of the acute phase of heart disease. This study examines the expression pattern of tumor necrosis factor-alpha (TNF-alpha) and its two related mitogenic-activated protein kinases, p38 and Jun-N-terminal kinase (JNK), in the spinal cord in response to acute cardiac injury (ACI). METHODS The ACI rat model was established by intra-myocardial injection of formalin. At the indicated times after the establishment of ACI, the thoracic segments of the spinal cord were harvested and Western blot was performed to determine the expression of TNF-alpha, p38 and JNK. The localization of the cytokine and the kinases was determined by immunohistochemistry and double immunofluorescence. RESULTS In response to ACI, TNF-alpha protein was up-regulated and reached a peak level at 6 h after ACI. The up-regulated TNF-alpha was distributed in all the laminae in the spinal cord and mainly localized in the neurons, as determined by immunohistochemistry and double immunofluorescence. In response to ACI, p38 and JNK were also up-regulated in the spinal cord. The expression profiles of p38 and JNK were similar to that of activated TNF-alpha following ACI. CONCLUSIONS This study shows that cardiac injury can induce the activation of spinal TNF-alpha, p38 and JNK. The activated spinal cytokine signaling may contribute to disease progression in the acute phase of cardiac injury in clinical practice.
Collapse
Affiliation(s)
- R-P Dai
- Department of Anesthesia, Second Xiang-Ya Hospital, Central South University, Changsha, China.
| | | | | | | | | | | |
Collapse
|
195
|
Abstract
Chronic heart failure (CHF) is often subdivided based on left ventricular ejection fraction (LVEF) in 2 distinct forms, usually specified as "diastolic heart failure" and "systolic heart failure." In this review, arguments are provided against an LVEF-based bimodal view, and CHF is presented as one pathophysiological identity encompassing a continuous spectrum of closely related phenotypes. Most importantly, there is currently no pathophysiological basis to support a bimodal view. As a result, conceptual presentations of CHF, such as the vicious circle paradigm of CHF, become obsolete. Furthermore, the binary view of CHF is the unfortunate result of selection biases that has confounded practically all clinical trials of CHF. Unfortunately, current investigations still introduce selection bias when studying heart failure at preserved or reduced LVEF. Future investigations should analyze CHF as one disease and focus on the mechanisms through which disease modifiers such as sex, diabetes, and hypertension induce phenotypic diversity.
Collapse
Affiliation(s)
- Gilles W De Keulenaer
- Department of Physiology, University of Antwerp, and Division of Cardiology, Middelheim Hospital, Antwerp, Belgium.
| | | |
Collapse
|
196
|
Gong KZ, Zhang H, Du JH, Zhang YY. Crosstalk between signaling pathways of adrenoreceptors and signal transducers and activators of transcription 3 (STAT3) in heart. Acta Pharmacol Sin 2007; 28:153-65. [PMID: 17241516 DOI: 10.1111/j.1745-7254.2007.00525.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Recently, there have been important advancements in our understanding of the signaling mechanisms of adrenoreceptors (AR) and signal transducers and activators of transcription 3 (STAT3). While their crucial roles in the pathological processes of the heart are well established, accumulating evidence suggests there is a complex pattern of crosstalk between these 2 signaling pathways. Moreover, the potential for crosstalk occurs at multiple levels in each signaling cascade and involves receptor transactivation, G proteins, small GTPases, cyclic adenosine 3',5'-monophosphate/protein kinase A, protein kinase C, scaffold/adaptor proteins, protein tyrosine kinases, and mitogen-activated protein kinases. In addition, post-translational modification (eg acetylation) of STAT3 may provide a link between STAT3 and AR signaling. In particular, crosstalk between these 2 systems in the heart would appear to be dependent upon the species/tissue studied, developmental stage, and eliciting stimulus. This at least partly accounts for the epigenetic effects on biological function that is mediated by the 2 signaling pathways. Elucidation of these mechanisms will provide new targets in the development of novel clinical strategies for heart disorders.
Collapse
Affiliation(s)
- Kai-zheng Gong
- Institute of Vascular Medicine, Peking University Third Hospital, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing 100083, China
| | | | | | | |
Collapse
|
197
|
Diwan A, Dorn GW. Decompensation of Cardiac Hypertrophy: Cellular Mechanisms and Novel Therapeutic Targets. Physiology (Bethesda) 2007; 22:56-64. [PMID: 17289931 DOI: 10.1152/physiol.00033.2006] [Citation(s) in RCA: 131] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Cardiac hypertrophy leads to heart failure, and both conditions can ultimately prove lethal. Here, traditional and novel mechanisms relating hypertrophy and heart failure are described at the physiological, cellular, and molecular levels. The rational application of these mechanistic considerations to therapeutics targeting hypertrophy and heart failure is discussed.
Collapse
Affiliation(s)
- Abhinav Diwan
- Center for Molecular Cardiovascular Research, University of Cincinnati, Cincinnati, OH, USA
| | | |
Collapse
|
198
|
Abboud J, Murad Y, Chen-Scarabelli C, Saravolatz L, Scarabelli TM. Peripartum cardiomyopathy: a comprehensive review. Int J Cardiol 2007; 118:295-303. [PMID: 17208320 DOI: 10.1016/j.ijcard.2006.08.005] [Citation(s) in RCA: 112] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2006] [Revised: 07/17/2006] [Accepted: 08/03/2006] [Indexed: 01/11/2023]
Abstract
Peripartum cardiomyopathy (PPCM) is a rare disorder in which left ventricular dysfunction and symptoms of heart failure occur in the peripartum period in previously healthy women. Incidence of PPCM ranges from 1 in 1300 to 1 in 15,000 pregnancies. The etiology of PPCM is unknown, but viral, autoimmune, and idiopathic causes may contribute. The diagnostic criteria are onset of heart failure in the last month of pregnancy or in first 5 months postpartum, absence of determinable cause for cardiac failure, and absence of a demonstrable heart disease before the last month of pregnancy. Risk factors for PPCM include advanced maternal age, multiparity, African race, twinning, gestational hypertension, and long-term tocolysis. The clinical presentation of patients with PPCM is similar to that of patients with dilated cardiomyopathy. Early diagnosis and initiation of treatment are essential to optimize pregnancy outcome. Treatment is similar to medical therapy for other forms of dilated cardiomyopathy. About half the patients of PPCM recover without complications. The prognosis is poor in patients with persistent cardiomyopathy. Persistence of disease after 6 months indicates irreversible cardiomyopathy and portends worse survival.
Collapse
Affiliation(s)
- John Abboud
- Center for Heart and Vessel Preclinical Studies, Division of Cardiology, St John Hospital and Medical Center, Wayne State University, Detroit, MI 48236, USA
| | | | | | | | | |
Collapse
|
199
|
Cardiac Development: Toward a Molecular Basis for Congenital Heart Disease. CARDIOVASCULAR MEDICINE 2007. [DOI: 10.1007/978-1-84628-715-2_52] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
200
|
Barry SP, Townsend PA, Latchman DS, Stephanou A. Role of the JAK-STAT pathway in myocardial injury. Trends Mol Med 2006; 13:82-9. [PMID: 17194625 DOI: 10.1016/j.molmed.2006.12.002] [Citation(s) in RCA: 132] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2006] [Revised: 11/24/2006] [Accepted: 12/14/2006] [Indexed: 01/03/2023]
Abstract
Cardiovascular pathologies are an enormous burden in human health and despite the vast amount of research; the molecular mechanisms and pathways that control the underlying pathologies are still not fully appreciated. The Janus kinase (JAK)-signal transducers and activators of transcription (STAT) pathway has recently been shown to be an integral part of the response of the myocardium to various cardiac insults, including myocardial infarction, oxidative damage, myocarditis, hypertrophy and remodeling, in addition to having a prominent role in cardioprotective therapies such as ischaemic preconditioning. Here, recent advances in the understanding of how the JAK-STAT pathway orchestrates the response to cellular damage in the myocardium are discussed, along with the potential benefits and challenges in manipulating this pathway in cardiovascular therapy.
Collapse
Affiliation(s)
- Seán P Barry
- Medical Molecular Biology Unit, The Institute of Child Health, University College London, 30 Guilford Street, London, UK
| | | | | | | |
Collapse
|