220 results on '"Jan Kajstura"'
Search Results
2. Expression of Concern
- Author
-
David A. D'Alessandro, Jan Kajstura, Domenico D'Amario, Piero Anversa, Claudia Fiorini, Hanqiao Zheng, Toru Hosoda, Sergio Ottolenghi, F. Del Monte, Polina Goichberg, João Ferreira-Martins, Robert E. Michler, Yingnan Bai, Sergio Signore, Marcello Rota, and Annarosa Leri
- Subjects
Physiology ,Cardiac Stem Cell ,business.industry ,Cancer research ,Medicine ,Infarction ,Cardiology and Cardiovascular Medicine ,business ,medicine.disease ,EPH receptor A2 ,Ephrin a1 - Published
- 2019
3. Cardiac stem cells in patients with ischaemic cardiomyopathy - Authors' reply
- Author
-
Atul R. Chugh, Jan Kajstura, John H Loughran, Roberto Bolli, and Piero Anversa
- Subjects
medicine.medical_specialty ,business.industry ,General Medicine ,medicine.disease ,Clinical trial ,Cell therapy ,medicine.anatomical_structure ,Heart failure ,Concomitant ,Internal medicine ,medicine ,In patient ,Stage (cooking) ,Stem cell ,business ,Artery - Abstract
The paper by Roberto Bolli and colleagues (Nov 26, p 1847) on cardiac progenitor cell treatment for ischaemic heart failure is a milestone in the history of cardiac cell therapy. However, we noticed some points that possibly interfere with interpretation of the results. First, the trial is still ongoing, so why did Bolli and colleagues publish partial results at this time? Why not wait until the trial was completed? Second, 13 analysed patients were from the non-randomised stage of the trial—only ten had undergone random allocation. Moreover, the ratio of treated to control patients (24:32) was reversed (7:3) by the analysis stage, mainly because of patients’ withdrawal. Why did so many control patients lose interest in the study? We are cautious because of our own experience. When one of us (CS) started a cell therapy programme using CD133+ cells delivered during coronary artery bypass graft (CABG) surgery, a striking improvement in heart function was noted in the initial pilot trial. A partly randomised, open-label trial later showed a clear benefi t of the concomitant cell therapy over CABG surgery alone. Finally, we did a strictly double-blind, fully randomised, placebo-controlled trial (CARDIO133, NCT00462774) and recently analysed the results. Unfortunately, none of the previous positive eff ects were confi rmed. Essentially, we followed the wrong track for nearly 10 years, because we did not strictly adhere to the guidelines on how to do a randomised, controlled clinical trial. We hope this will not be the fate of the SCIPIO concept.
- Published
- 2018
4. Priming with Ligands Secreted by Human Stromal Progenitor Cells Promotes Grafts of Cardiac Stem/Progenitor Cells After Myocardial Infarction
- Author
-
Yoshitaka Iso, A.K.M. Tarikuz Zaman, Ingrid M. Curril, Jeffrey L. Spees, Burton E. Sobel, Charla N. Poole, Krithika S. Rao, Piero Anversa, and Jan Kajstura
- Subjects
Adult ,STAT3 Transcription Factor ,Stromal cell ,Cellular differentiation ,Myocardial Infarction ,Biology ,Ligands ,Protective Agents ,Article ,Cell Line ,Animals ,Humans ,Infusions, Intra-Arterial ,Insulin ,Progenitor cell ,Cell Proliferation ,Multipotent Stem Cells ,Myocardium ,Stem Cells ,Mesenchymal stem cell ,Connective Tissue Growth Factor ,Cell Differentiation ,Cell Biology ,Cell Hypoxia ,Rats ,Enzyme Activation ,CTGF ,Transplantation ,Multipotent Stem Cell ,Culture Media, Conditioned ,Immunology ,Cancer research ,Molecular Medicine ,Cattle ,Stromal Cells ,Stem cell ,Proto-Oncogene Proteins c-akt ,Stem Cell Transplantation ,Developmental Biology - Abstract
Transplantation of culture-expanded adult stem/progenitor cells often results in poor cellular engraftment, survival, and migration into sites of tissue injury. Mesenchymal cells including fibroblasts and stromal cells secrete factors that protect injured tissues, promote tissue repair, and support many types of stem/progenitor cells in culture. We hypothesized that secreted factors in conditioned medium (CdM) from adult bone marrow-derived multipotent stromal cells (MSCs) could be used to prime adult cardiac stem/progenitor cells (CSCs/CPCs) and improve graft success after myocardial infarction (MI). Incubation of adult rat CPCs in CdM from human MSCs isolated by plastic adherence or by magnetic sorting against CD271 (a.k.a., p75 low-affinity nerve growth factor receptor; p75MSCs) induced phosphorylation of STAT3 and Akt in CPCs, supporting their proliferation under normoxic conditions and survival under hypoxic conditions (1% oxygen). Priming CSCs with 30× p75MSC CdM for 30 minutes prior to transplantation into subepicardial tissue 1 day after MI markedly increased engraftment compared with vehicle priming. Screening CdM with neutralizing/blocking antibodies identified connective tissue growth factor (CTGF) and Insulin as key factors in p75MSC CdM that protected CPCs. Human CTGF peptide (CTGF-D4) and Insulin synergistically promoted CPC survival during hypoxia in culture. Similar to CdM priming, priming of CSCs with CTGF-D4 and Insulin for 30 minutes prior to transplantation promoted robust engraftment, survival, and migration of CSC derivatives at 1 week and 1 month after MI. Our results indicate that short-term priming of human CSCs with CTGF-D4 and Insulin may improve graft success and cardiac regeneration in patients with MI. Stem Cells 2014;32:674–683
- Published
- 2014
5. Retraction for Bearzi et al., Identification of a coronary vascular progenitor cell in the human heart
- Author
-
Jan Kajstura, Khaled Qanud, Andreas M. Zeiher, Thomas H. Hintze, Konrad Urbanek, Robert E. Michler, Caroline Ojaimi, Francesco Lo Monaco, David A. D'Alessandro, Domenico D'Amario, Piero Anversa, Martino Pepe, Silvana Bardelli, Marcello Rota, Annarosa Leri, Toru Hosoda, Stefanie Dimmeler, Claudia Bearzi, and Arantxa González
- Subjects
Multidisciplinary ,business.industry ,Medicine ,Human heart ,Identification (biology) ,Progenitor cell ,business ,Bioinformatics - Published
- 2019
6. Inositol 1, 4, 5-Trisphosphate Receptors and Human Left Ventricular Myocytes
- Author
-
Oriyanhan Wunimenghe, Marcello Rota, David A. D'Alessandro, Ramaswamy Kannappan, Xiaoxia Liu, Polina Goichberg, João Ferreira-Martins, Jan Kajstura, Fabio del Ben, Toru Hosoda, Sergio Signore, Piero Anversa, Robert E. Michler, Federica del Monte, Christian Arranto, Hanqiao Zheng, Andrea Sorrentino, Kazuya Isobe, Annarosa Leri, Barbara Ogorek, Ewa Wybieralska, Andrew Webster, Mehrdad Shafaie, and Fumihiro Sanada
- Subjects
medicine.medical_specialty ,Endoplasmic reticulum ,chemistry.chemical_element ,Calcium ,Biology ,medicine.disease ,chemistry.chemical_compound ,Endocrinology ,chemistry ,Physiology (medical) ,Internal medicine ,Heart failure ,medicine ,Inositol ,Ventricular myocytes ,Signal transduction ,Cardiology and Cardiovascular Medicine ,Receptor ,Calcium signaling - Abstract
Background— Little is known about the function of inositol 1,4,5-trisphosphate receptors (IP3Rs) in the adult heart experimentally. Moreover, whether these Ca 2+ release channels are present and play a critical role in human cardiomyocytes remains to be defined. IP3Rs may be activated after Gαq-protein–coupled receptor stimulation, affecting Ca 2+ cycling, enhancing myocyte performance, and potentially favoring an increase in the incidence of arrhythmias. Methods and Results— IP3R function was determined in human left ventricular myocytes, and this analysis was integrated with assays in mouse myocytes to identify the mechanisms by which IP3Rs influence the electric and mechanical properties of the myocardium. We report that IP3Rs are expressed and operative in human left ventricular myocytes. After Gαq-protein–coupled receptor activation, Ca 2+ mobilized from the sarcoplasmic reticulum via IP3Rs contributes to the decrease in resting membrane potential, prolongation of the action potential, and occurrence of early afterdepolarizations. Ca 2+ transient amplitude and cell shortening are enhanced, and extrasystolic and dysregulated Ca 2+ elevations and contractions become apparent. These alterations in the electromechanical behavior of human cardiomyocytes are coupled with increased isometric twitch of the myocardium and arrhythmic events, suggesting that Gαq-protein–coupled receptor activation provides inotropic reserve, which is hampered by electric instability and contractile abnormalities. Additionally, our findings support the notion that increases in Ca 2+ load by IP3Rs promote Ca 2+ extrusion by forward-mode Na + /Ca 2+ exchange, an important mechanism of arrhythmic events. Conclusions— The Gαq-protein/coupled receptor/IP3R axis modulates the electromechanical properties of the human myocardium and its propensity to develop arrhythmias.
- Published
- 2013
7. The Existence of Myocardial Repair
- Author
-
Piero Anversa, Annarosa Leri, Matthew Schoenfeld, William H. Frishman, and Jan Kajstura
- Subjects
Male ,Senescence ,medicine.medical_treatment ,Myocardial Infarction ,Bioinformatics ,Article ,medicine ,Humans ,Regeneration ,Myocardial infarction ,Bone Marrow Transplantation ,Heart Failure ,Clinical Trials as Topic ,business.industry ,Multipotent Stem Cells ,Regeneration (biology) ,Heart ,General Medicine ,Stem-cell therapy ,medicine.disease ,Cardiovascular physiology ,Multipotent Stem Cell ,Heart failure ,Female ,Stem cell ,Cardiology and Cardiovascular Medicine ,business ,Stem Cell Transplantation - Abstract
The lack of myocardial repair after myocardial infarction and the heart failure that eventually ensues was thought of as proof that myocardial cell regeneration and myocardial repair mechanisms do not exist. Recently, growing experimental and clinical evidence has proven this concept wrong. Cardiac stem cells and endogenous myocardial repair mechanisms do exist; however, they do not produce significant myocardial repair. Similarly, the preliminary results of stem cell therapy for myocardial repair have shown early promise but modest results. Preclinical studies are the key to understanding stem cell senescence and lack of cellular contact and vasculature in the infarcted region. Additional laboratory studies are sure to unlock the therapeutic mechanisms that will be required for significant myocardial repair.
- Published
- 2013
8. Mitotically Inactivated Embryonic Stem Cells Can Be Used as an In Vivo Feeder Layer to Nurse Damaged Myocardium after Acute Myocardial Infarction
- Author
-
Shankararao Navale, Larissa Verda, Ka Leung Ngai, You Hong Chen, James C. Carr, Xiaoqiang Han, Jon W. Lomasney, Jessa M. Baker, Richard K. Burt, Carolina Lucena, Jan Kajstura, Aya Kino, Piero Anversa, and Jesse Johnson
- Subjects
Cardiac function curve ,Pathology ,medicine.medical_specialty ,Physiology ,Penumbra ,In situ hybridization ,Biology ,medicine.disease ,Embryonic stem cell ,Transplantation ,cardiovascular system ,medicine ,Myocyte ,Myocardial infarction ,Stem cell ,Cardiology and Cardiovascular Medicine - Abstract
Rationale:Various types of viable stem cells have been reported to result in modest improvement in cardiac function after acute myocardial infarction. The mechanisms for improvement from different stem cell populations remain unknown. Objective:To determine whether irradiated (nonviable) embryonic stem cells (iESCs) improve postischemic cardiac function without adverse consequences. Methods and Results:After coronary artery ligation-induced cardiac infarction, either conditioned media or male murine or male human iESCs were injected into the penumbra of ischemic myocardial tissue of female mice or female rhesus macaque monkeys, respectively. Murine and human iESCs, despite irradiation doses that prevented proliferation and induced cell death, significantly improved cardiac function and decreased infarct size compared with untreated or media-treated controls. Fluorescent in situ hybridization of the Y chromosome revealed disappearance of iESCs within the myocardium, whereas 5-bromo-2′-deoxyuridine assays r...
- Published
- 2012
9. Biased DNA Segregation During Stem Cell Division
- Author
-
Annarosa Leri, Piero Anversa, and Jan Kajstura
- Subjects
Genetics ,Stem cell division ,Cell division ,DNA synthesis ,Physiology ,DNA replication ,Chromatid ,Biology ,Stem cell ,Cardiology and Cardiovascular Medicine ,Immortal DNA strand hypothesis ,Cell biology ,Telomere - Abstract
A Subpopulation of Adult Skeletal Muscle Stem Cells Retains All Template DNA Strands After Cell Division Rocheteau et al Cell . 2012;148:112–125. Adult skeletal muscle stem cells are a heterogeneous cell population characterized by a small subset of undifferentiated cells that express at high levels the paired/homeodomain gene Pax7 . This category of satellite cells divides predominantly by asymmetric chromatid segregation, generating a daughter cell that carries the mother DNA and retains stem cell property, and a daughter cell that inherits the newly synthesized DNA and acquires the myocyte lineage.1 Recently, the established modalities of stem cell self-renewal, that is, generation of a daughter stem cell and a daughter-committed cell, have been questioned, and an old theory that may have important implications in stem cell function and senescence has been reconsidered. The possibility that nonrandom chromatid segregation regulates stem cell division has gained interest, promoting intense discussion in the field.2–6 The immortal DNA strand hypothesis advanced by John Cairns in 19752 suggests that stem cell division is characterized by asymmetric segregation of chromatids so that one daughter cell contains only the old intact DNA and the other carries chromatids composed exclusively of the newly synthesized DNA (Figure 1). This process would attenuate the accumulation of replication errors in the parental DNA strand.2,7 In the event that deleterious mutations are acquired during DNA replication, they would be transmitted to daughter cells, which may undergo senescence and apoptosis,8–11 having a reduced capacity to repair DNA damage.2,12,13 Moreover, telomeric shortening dictated by DNA synthesis would affect only partly the actual stem cells retaining the old DNA, and telomere attrition would be largely restricted to the newly synthesized strands and become templates in subsequent descendants.6,14 Figure 1. Nonrandom and random chromatid segregation. With asymmetric chromatid segregation, a dividing mother …
- Published
- 2012
10. Role of stem cells in cardiovascular biology
- Author
-
Piero Anversa, Marcello Rota, Jan Kajstura, Annarosa Leri, and Toru Hosoda
- Subjects
Cell division ,Stem Cells ,Hematology ,Anatomy ,Cell cycle ,Biology ,Cardiovascular System ,Article ,Review article ,medicine ,Humans ,Myocyte ,Stem cell ,Progenitor cell ,medicine.symptom ,Myopathy ,Neuroscience ,Organism - Abstract
This review article addresses the controversy as to whether the adult heart possesses an intrinsic growth reserve. If myocyte renewal takes place in healthy and diseased organs, the reconstitution of the damaged tissue lost upon pathological insults might be achieved by enhancing a natural occurring process. Evidence in support of the old and new view of cardiac biology is critically discussed in an attempt to understand whether the heart is a static or dynamic organ. According to the traditional concept, the heart exerts its function until death of the organism with the same or lesser number of cells that are present at birth. This paradigm was challenged by documentation of the cell cycle activation and nuclear and cellular division in a subset of myocytes. These observations raised the important question of the origin of replicating myocytes. Several theories have been proposed and are presented in this review article. Newly formed myocytes may derive from a pre-existing pool of cells that has maintained the ability to divide. Alternatively, myocytes may be generated by activation and commitment of resident cardiac stem cells or by migration of progenitor cells from distant organs. In all cases, parenchymal cell turnover throughout lifespan results in a heterogeneous population consisting of young, adult, and senescent myocytes. With time, accumulation of old myocytes has detrimental effects on cardiac performance and may cause the development of an aging myopathy.
- Published
- 2011
11. Evidence for Human Lung Stem Cells
- Author
-
Federico Quaini, Christian Arranto, Sean Hall, Silvana Bardelli, Marcello Rota, Joseph Loscalzo, Barbara Ogorek, Xiaoli Liu, Jan Kajstura, Piero Anversa, Mark A. Perrella, Giovanna Giordano, Carlos Rondon-Clavo, Fumihiro Sanada, Hussein Rayatzadeh, Hanqiao Zheng, Ronglih Liao, Annarosa Leri, Alex Matsuda, Kathleen J. Haley, Toru Hosoda, João Ferreira-Martins, Polina Goichberg, and Domenico D'Amario
- Subjects
Adult ,Pluripotent Stem Cells ,Pathology ,medicine.medical_specialty ,Biology ,Article ,Mice ,Cancer stem cell ,medicine ,Animals ,Humans ,Regeneration ,Progenitor cell ,Induced pluripotent stem cell ,Lung ,Stem cell transplantation for articular cartilage repair ,Induced stem cells ,Fetal Stem Cells ,business.industry ,Stem Cells ,Amniotic stem cells ,General Medicine ,respiratory system ,Clone Cells ,respiratory tract diseases ,Mice, Inbred C57BL ,Endothelial stem cell ,Proto-Oncogene Proteins c-kit ,Amniotic epithelial cells ,Cancer research ,Female ,Stem cell ,business ,Biomarkers ,Stem Cell Transplantation ,Adult stem cell - Abstract
BACKGROUND Although progenitor cells have been described in distinct anatomical regions of the lung, description of resident stem cells has remained elusive. METHODS Surgical lung-tissue specimens were studied in situ to identify and characterize human lung stem cells. We defined their phenotype and functional properties in vitro and in vivo. RESULTS Human lungs contain undifferentiated human lung stem cells nested in niches in the distal airways. These cells are self-renewing, clonogenic, and multipotent in vitro. After injection into damaged mouse lung in vivo, human lung stem cells form human bronchioles, alveoli, and pulmonary vessels integrated structurally and functionally with the damaged organ. The formation of a chimeric lung was confirmed by detection of human transcripts for epithelial and vascular genes. In addition, the self-renewal and long-term proliferation of human lung stem cells was shown in serial-transplantation assays. CONCLUSIONS Human lungs contain identifiable stem cells. In animal models, these cells participate in tissue homeostasis and regeneration. They have the undemonstrated potential to promote tissue restoration in patients with lung disease. (Funded by the National Institutes of Health.).
- Published
- 2011
12. Cardiomyogenesis in the Adult Human Heart
- Author
-
Barbara Ogorek, Fumihiro Sanada, Piero Anversa, Jan Kajstura, Hanqiao Zheng, John F. Tisdale, Annarosa Leri, Federica del Monte, Shira Perl, João Ferreira-Martins, Donald Orlic, Toru Hosoda, Carlos Rondon-Clavo, Domenico D'Amario, Polina Goichberg, Konrad Urbanek, and Marcello Rota
- Subjects
Programmed cell death ,Pathology ,medicine.medical_specialty ,Cell fusion ,medicine.diagnostic_test ,Physiology ,Cell growth ,Regeneration (biology) ,Biology ,Article ,Flow cytometry ,Andrology ,chemistry.chemical_compound ,chemistry ,Circulatory system ,medicine ,Myocyte ,Cardiology and Cardiovascular Medicine ,Thymidine - Abstract
Rationale: The ability of the human heart to regenerate large quantities of myocytes remains controversial, and the extent of myocyte renewal claimed by different laboratories varies from none to nearly 20% per year. Objective: To address this issue, we examined the percentage of myocytes, endothelial cells, and fibroblasts labeled by iododeoxyuridine in postmortem samples obtained from cancer patients who received the thymidine analog for therapeutic purposes. Additionally, the potential contribution of DNA repair, polyploidy, and cell fusion to the measurement of myocyte regeneration was determined. Methods and Results: The fraction of myocytes labeled by iododeoxyuridine ranged from 2.5% to 46%, and similar values were found in fibroblasts and endothelial cells. An average 22%, 20%, and 13% new myocytes, fibroblasts, and endothelial cells were generated per year, suggesting that the lifespan of these cells was approximately 4.5, 5, and 8 years, respectively. The newly formed cardiac cells showed a full...
- Published
- 2010
13. Progenitor Cells From the Explanted Heart Generate Immunocompatible Myocardium Within the Transplanted Donor Heart
- Author
-
Marcello Rota, Hanqiao Zheng, Piero Anversa, Konrad Urbanek, David A. D'Alessandro, Domenico D'Amario, Federico Mosna, David Stern, Michael O. Zembala, Jan Kajstura, Silvana Bardelli, Toru Hosoda, Ornella Rimoldi, Robert E. Michler, Ricardo Bello, Adriana Bastos Carvalho, Annarosa Leri, Alessandro Gatti, and M. Elena Padin-Iruegas
- Subjects
Heart transplantation ,medicine.medical_specialty ,Physiology ,medicine.medical_treatment ,cardiac progenitor cells ,Biology ,heart transplantation ,medicine.disease ,Article ,Transplantation ,Internal medicine ,Circulatory system ,Myocardial scarring ,cardiovascular system ,medicine ,Cardiology ,Myocardial infarction ,cell therapy ,medicine.symptom ,Stem cell ,Progenitor cell ,Cardiology and Cardiovascular Medicine ,Coronary atherosclerosis - Abstract
Rationale: Chronic rejection, accelerated coronary atherosclerosis, myocardial infarction, and ischemic heart failure determine the unfavorable evolution of the transplanted heart in humans. Objective: Here we tested whether the pathological manifestations of the transplanted heart can be corrected partly by a strategy that implements the use of cardiac progenitor cells from the recipient to repopulate the donor heart with immunocompatible cardiomyocytes and coronary vessels. Methods and Results: A large number of cardiomyocytes and coronary vessels were created in a rather short period of time from the delivery, engraftment, and differentiation of cardiac progenitor cells from the recipient. A proportion of newly formed cardiomyocytes acquired adult characteristics and was integrated structurally and functionally within the transplant. Similarly, the regenerated arteries, arterioles, and capillaries were operative and contributed to the oxygenation of the chimeric myocardium. Attenuation in the extent of acute damage by repopulating cardiomyocytes and vessels decreased significantly the magnitude of myocardial scarring preserving partly the integrity of the donor heart. Conclusions: Our data suggest that tissue regeneration by differentiation of recipient cardiac progenitor cells restored a significant portion of the rejected donor myocardium. Ultimately, immunosuppressive therapy may be only partially required improving quality of life and lifespan of patients with cardiac transplantation.
- Published
- 2009
14. Disruption of Striated Preferentially Expressed Gene Locus Leads to Dilated Cardiomyopathy in Mice
- Author
-
Mark A. Perrella, Piero Anversa, Tripurasundari Ramjiganesh, Kate G. Ackerman, Robert F. Padera, Yen-Hsu Chen, Sean Hall, Xiaoli Liu, Scott L. Schissel, Su Wol Chung, Jan Kajstura, Ronglih Liao, Shaw-Fang Yet, Matthew D. Layne, and Annarosa Leri
- Subjects
Cardiomyopathy, Dilated ,Mice, Knockout ,Gene isoform ,Cell type ,Vascular smooth muscle ,Days post coitum ,Muscle Proteins ,Gene targeting ,Mice, Transgenic ,Biology ,Molecular biology ,Article ,Mice, Inbred C57BL ,Mice ,Exon ,Animals, Newborn ,Physiology (medical) ,Gene Targeting ,Gene expression ,Mutagenesis, Site-Directed ,Animals ,Myocyte ,Cardiology and Cardiovascular Medicine ,Myosin-Light-Chain Kinase - Abstract
Background— The striated preferentially expressed gene (Speg) generates 4 different isoforms through alternative promoter use and tissue-specific splicing. Depending on the cell type, Speg isoforms may serve as markers of striated or smooth muscle differentiation. Methods and Results— To elucidate function of Speg gene isoforms, we disrupted the Speg gene locus in mice by replacing common exons 8, 9, and 10 with a lacZ gene. β-Galactosidase activity was detected in cardiomyocytes of the developing heart starting at day 11.5 days post coitum (dpc). β-Galactosidase activity in other cell types, including vascular smooth muscle cells, did not begin until 18.5 dpc. In the developing heart, protein expression of only Spegα and Spegβ isoforms was present in cardiomyocytes. Homozygous Speg mutant hearts began to enlarge by 16.5 dpc, and by 18.5 dpc, they demonstrated dilation of right and left atria and ventricles. These cardiac abnormalities in the absence of Speg were associated with a cellular hypertrophic response, myofibril degeneration, and a marked decrease in cardiac function. Moreover, Speg mutant mice exhibited significant neonatal mortality, with increased death occurring by 2 days after birth. Conclusions— These findings demonstrate that mutation of the Speg locus leads to cardiac dysfunction and a phenotype consistent with a dilated cardiomyopathy.
- Published
- 2009
15. Cardiac stem cells and myocardial disease
- Author
-
Claudia Bearzi, Jan Kajstura, Roberto Bolli, Marcello Rota, Konrad Urbanek, Toru Hosoda, Piero Anversa, Annarosa Leri, Kajstura, J, Urbanek, K, Rota, M, Bearzi, C, Hosoda, T, Bolli, R, Anversa, P, and Leri, A
- Subjects
education.field_of_study ,medicine.medical_specialty ,Heart Diseases ,business.industry ,Multipotent Stem Cells ,Myocardium ,Regeneration (biology) ,Population ,Endothelial stem cell ,Internal medicine ,Myocardial scarring ,Cardiology ,Animals ,Humans ,Medicine ,Myocyte ,Progenitor cell ,Stem cell ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,education ,business ,Molecular Biology ,Stem Cell Transplantation ,Adult stem cell - Abstract
Recent data indicate that the heart is a self-renewing organ and contains a pool of progenitor cells (PCs). According to the new paradigm, this resident population of multipotent undifferentiated cells gives rise to myocytes, endothelial cells, smooth muscle cells and fibroblasts. Understanding the function of cardiac PCs is critical for the implementation of these cells in the treatment of the diseased human heart. However, cardiac repair is an extremely complex phenomenon. Efficient myocardial regeneration requires restoration of segmental and focal areas of myocardial scarring, replacement of damaged coronary arteries, arterioles and capillaries, and substitution of hypertrophied poorly contracting myocytes with smaller better functioning parenchymal cells. To achieve these goals, the acquisition of a more profound knowledge of the biology of cardiac PCs cells and their fate following pathologic insults represents an essential need.
- Published
- 2008
16. IGF-I alleviates diabetes-induced RhoA activation, eNOS uncoupling, and myocardial dysfunction
- Author
-
Feng Gao, Jinhong Duan, Jan Kajstura, James R. Sowers, Jun Ren, Nair Sreejayan, Xiaoping Yang, Piero Anversa, D. Paul Thomas, and Annarosa Leri
- Subjects
rho GTP-Binding Proteins ,medicine.medical_specialty ,RHOA ,Nitric Oxide Synthase Type III ,Physiology ,Mice, Transgenic ,Cell Separation ,medicine.disease_cause ,p38 Mitogen-Activated Protein Kinases ,Diabetes Mellitus, Experimental ,Nitric oxide ,Mice ,chemistry.chemical_compound ,Physiology (medical) ,Diabetes mellitus ,Internal medicine ,Kv1.2 Potassium Channel ,medicine ,Animals ,Myocyte ,Myocytes, Cardiac ,Insulin-Like Growth Factor I ,Phosphorylation ,Protein kinase B ,Rho-associated protein kinase ,Ultrasonography ,rho-Associated Kinases ,Myosin Heavy Chains ,biology ,medicine.disease ,Biopterin ,Myocardial Contraction ,Oncogene Protein v-akt ,Tetrahydrofolate Dehydrogenase ,Endocrinology ,chemistry ,Mechanism of action ,biology.protein ,Folic Acid Antagonists ,Mitogen-Activated Protein Kinases ,medicine.symptom ,Cardiomyopathies ,rhoA GTP-Binding Protein ,Oxidation-Reduction ,Oxidative stress - Abstract
IGF-I rescues diabetic heart defects and oxidative stress, although the underlying mechanism of action remains poorly understood. This study was designed to delineate the beneficial effects of IGF-I with a focus on RhoA, Akt, and eNOS coupling. Echocardiography was performed in normal or diabetic Friend Virus-B type (FVB) and IGF-I transgenic mice. Cardiomyocyte contractile properties were evaluated using peak shortening (PS), time-to-90% relengthening (TR90), and intracellular Ca2+ rise and decay. Diabetes reduced fraction shortening, PS, and intracellular Ca2+; it increased chamber size, prolonged TR90, and intracellular Ca2+ decay. Levels of RhoA mRNA, active RhoA, and O2− were elevated, whereas nitric oxide (NO) levels were reduced in diabetes. Diabetes-induced O2− accumulation was ablated by the NO synthase (NOS) inhibitor nitro-l-arginine methyl ester (l-NAME), indicating endothelial NOS (eNOS) uncoupling, all of which except heart size were negated by IGF-I. The IGF-I-elicited beneficial effects were mimicked by the Rho kinase inhibitor Y27632 and BH4. Diabetes depressed expression of Kv1.2 and dihydrofolate reductase (DHFR), increased β-myosin heavy-chain expression, stimulated p38 MAPK, and reduced levels of total Akt and phosphorylated Akt/eNOS, all of which with the exception of myosin heavy chain were attenuated by IGF-I. In addition, Y27632 and the eNOS coupler folate abrogated glucose toxicity-induced PS decline, TR90 prolongation, while it increased O2− and decreased NO and Kv1.2 levels. The DHFR inhibitor methotrexate impaired myocyte function, NO/O2− balance, and rescued Y27632-induced cardiac protection. These results revealed that IGF-I benefits diabetic hearts via Rho inhibition and antagonism of diabetes-induced decrease in pAkt, eNOS uncoupling, and K+ channel expression.
- Published
- 2008
17. Myocardial Regeneration and Stem Cell Repair
- Author
-
Jan Kajstura, Annarosa Leri, William H. Frishman, and Piero Anversa
- Subjects
Heart Failure ,Pathology ,medicine.medical_specialty ,business.industry ,Myocardium ,Regeneration (biology) ,Clinical uses of mesenchymal stem cells ,General Medicine ,Embryonic stem cell ,Endothelial stem cell ,Precursor cell ,Cancer research ,Humans ,Medicine ,Stem cell ,Cardiomyopathies ,Cardiology and Cardiovascular Medicine ,business ,Embryonic Stem Cells ,Bone Marrow Transplantation ,Adult stem cell ,Stem cell transplantation for articular cartilage repair - Abstract
Recent evidence would suggest that the heart is not a terminally differentiated organ and has the ability to regenerate itself under normal and pathophysiologic conditions. A major effort has been made to identify precursor cells that are capable of differentiating into cell lineages different from their organ of origin. Embryonic stem cells and bone marrow-derived cells (BMCs) have been studied and characterized, and BM precursor cells are currently being utilized as therapy in clinical trials of patients with heart failure of ischemic and nonischemic etiologies. Controversy remains, however, whether BMCs are the best cells to be used for replacement therapy. The existence of a cardiac stem cell (CSC) has also been described, which has the ability to generate new cardiac myocytes and blood vessels, raising the possibility of rebuilding a damaged heart with the organ's own precursor stem cell population. Animal studies have suggested such a possibility, and a clinical trial using CSCs is in progress. This monograph discusses our current understanding of myocardial regeneration and the roles that endogenous and exogenous stem cells may have in the future therapy of cardiovascular disease.
- Published
- 2008
18. Pim-1 regulates cardiomyocyte survival downstream of Akt
- Author
-
Weitao Wu, Steven R. Houser, Sailay Siddiqi, Erik Schaefer, Gregory Emmanuel, Francesca Delucchi, Christopher C. Glembotski, Jenna Fransioli, Piero Anversa, Yu Misao, Mark A. Sussman, Joshua J. Martindale, Annarosa Leri, Kimberlee M. Fischer, Roberto Alvarez, Jan Kajstura, Remus M Beretta, Michael L. Arcarese, John A. Muraski, Grazia Esposito, Natalie Gude, Nancy S. Magnuson, Christopher T. Cottage, Anton Berns, and Marcello Rota
- Subjects
medicine.medical_specialty ,Transgene ,bcl-X Protein ,Apoptosis ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Mice ,Proto-Oncogene Proteins c-pim-1 ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Animals ,Humans ,Myocyte ,Myocytes, Cardiac ,Protein kinase B ,Cell Nucleus ,Mice, Knockout ,Cardioprotection ,Pressure overload ,Kinase ,Myocardium ,General Medicine ,Rats ,Endocrinology ,Proto-Oncogene Proteins c-bcl-2 ,Phosphorylation ,Proto-Oncogene Proteins c-akt - Abstract
The serine-threonine kinases Pim-1 and Akt regulate cellular proliferation and survival. Although Akt is known to be a crucial signaling protein in the myocardium, the role of Pim-1 has been overlooked. Pim-1 expression in the myocardium of mice decreased during postnatal development, re-emerged after acute pathological injury in mice and was increased in failing hearts of both mice and humans. Cardioprotective stimuli associated with Akt activation induced Pim-1 expression, but compensatory increases in Akt abundance and phosphorylation after pathological injury by infarction or pressure overload did not protect the myocardium in Pim-1-deficient mice. Transgenic expression of Pim-1 in the myocardium protected mice from infarction injury, and Pim-1 expression inhibited cardiomyocyte apoptosis with concomitant increases in Bcl-2 and Bcl-X(L) protein levels, as well as in Bad phosphorylation levels. Relative to nontransgenic controls, calcium dynamics were significantly enhanced in Pim-1-overexpressing transgenic hearts, associated with increased expression of SERCA2a, and were depressed in Pim-1-deficient hearts. Collectively, these data suggest that Pim-1 is a crucial facet of cardioprotection downstream of Akt.
- Published
- 2007
19. Myocardial regeneration by exogenous and endogenous progenitor cells
- Author
-
Marcello Rota, Toru Hosoda, Jan Kajstura, Piero Anversa, and Annarosa Leri
- Subjects
Cardiac progenitors ,business.industry ,Regeneration (biology) ,Endogeny ,Mutually exclusive events ,Article ,Cell biology ,medicine.anatomical_structure ,Drug Discovery ,cardiovascular system ,Molecular Medicine ,Medicine ,Compartment (development) ,Bone marrow ,Progenitor cell ,business ,Homeostasis - Abstract
A problem in need of resolution concerns the origin of cardiac progenitor cells and the mechanisms by which these cells are preserved within the cardiac niches. This might be accomplished by migration of progenitor cells from the bone marrow to the myocardium. Alternatively, the progenitor cell compartment in the heart may be maintained by asymmetric division of resident cells. These two possibilities are not mutually exclusive and both exogenous and endogenous progenitor cells may contribute to cardiac homeostasis.
- Published
- 2007
20. The Young Mouse Heart Is Composed of Myocytes Heterogeneous in Age and Function
- Author
-
Antonella De Angelis, Piero Anversa, Marcello Rota, Roberto Rizzi, Ezio Musso, Jochen Tillmanns, Annarosa Leri, Michael L. Arcarese, Grazia Esposito, Toru Hosoda, Ornella Rimoldi, Claudia Bearzi, Jan Kajstura, Derin Tugal, Konrad Urbanek, Rota, M., Hosoda, T., DE ANGELIS, Antonella, Arcarese, M. L., Esposito, G., Rizzi, R., Tillmanns, J., Tugal, D., Musso, E., Rimoldi, O., Bearzi, C., Urbanek, K., Anversa, P., Leri, A., Kajstura, J., Rota, M, Hosoda, T, De Angelis, A, Arcarese, Ml, Esposito, G, Rizzi, R, Tillmanns, J, Tugal, D, Musso, E, Rimoldi, O, Bearzi, C, Urbanek, K, Anversa, P, Leri, A, and Kajstura, J
- Subjects
Cardiac function curve ,Senescence ,Aging ,medicine.medical_specialty ,Physiology ,Action Potentials ,Biology ,Mice ,Internal medicine ,Myocyte volume ,medicine ,Animals ,Homeostasis ,Myocyte ,Cell Lineage ,Myocytes, Cardiac ,Cellular Senescence ,Cyclin-Dependent Kinase Inhibitor p16 ,Cell Size ,Calcium metabolism ,Senescence-associated proteins ,Telomere length ,Stem Cells ,Heart ,Telomere ,Myocardial Contraction ,Action potential profile ,Excitation-contraction coupling ,Cardiovascular physiology ,Mice, Inbred C57BL ,Electrophysiology ,Endocrinology ,Circulatory system ,Potassium ,Calcium ,Cardiology and Cardiovascular Medicine - Abstract
The recognition that the adult heart continuously renews its myocyte compartment raises the possibility that the age and lifespan of myocytes does not coincide with the age and lifespan of the organ and organism. If this were the case, myocyte turnover would result at any age in a myocardium composed by a heterogeneous population of parenchymal cells which are structurally integrated but may contribute differently to myocardial performance. To test this hypothesis, left ventricular myocytes were isolated from mice at 3 months of age and the contractile, electrical, and calcium cycling characteristics of these cells were determined together with the expression of the senescence-associated protein p16 INK4a and telomere length. The heart was characterized by the coexistence of young, aged, and senescent myocytes. Old nonreplicating, p16 INK4a -positive, hypertrophied myocytes with severe telomeric shortening were present together with young, dividing, p16 INK4a -negative, small myocytes with long telomeres. A class of myocytes with intermediate properties was also found. Physiologically, evidence was obtained in favor of the critical role that action potential (AP) duration and I CaL play in potentiating Ca 2+ cycling and the mechanical behavior of young myocytes or in decreasing Ca 2+ transients and the performance of senescent hypertrophied cells. The characteristics of the AP appeared to be modulated by the transient outward K + current I to which was influenced by the different expression of the K + channels subunits. Collectively, these observations at the physiological and structural cellular level document that by necessity the heart has to constantly repopulate its myocyte compartment to replace senescent poorly contracting myocytes with younger more efficient cells. Thus, cardiac homeostasis and myocyte turnover regulate cardiac function.
- Published
- 2007
21. If I Can Stop One Heart From Breaking
- Author
-
Piero Anversa, Jan Kajstura, and Annarosa Leri
- Subjects
medicine.medical_specialty ,business.industry ,Human life ,medicine.disease ,humanities ,Surgery ,Tissue therapy ,Cardiac regeneration ,Physiology (medical) ,Internal medicine ,Cardiac repair ,medicine ,Long labor ,Cardiology ,Myocardial infarction ,Cardiology and Cardiovascular Medicine ,Ischemic heart ,business ,Beneficial effects - Abstract
If I can stop one heart from breaking — -Emily Dickinson 1 In the Ode to Broken Things , Pablo Neruda tenders a powerful metaphor of existence.2 It is not difficult to recognize the unwinding of human life in the broken clock that was once “the secret thread of our weeks” and now, with its “blue guts” exposed and its “wide heart unsprung,” is the symbol of agony and death. The beginning and ending of life are embraced in a poem about simple and ordinary things that accompany human beings throughout life and then are lost, together with the feelings and significance that were assigned to them. All things break, even the heart. What to do for broken things and broken hearts? The poet suggests collecting our treasures and sinking them in the ocean with the hope that the “long labor of its tides” may give back wholeness to the fragments. While we wait for the sea to reveal its strength and miraculous effects, the discovery that stem cells repair broken organs projects a more hopeful view of medicine and the way it is practiced. Article p 896 Soon after the first experimental evidence that bone marrow cells (BMCs) induce cardiac repair in the postinfarcted heart,3 unfractionated mononuclear BMCs and CD34-positive cells were given to patients affected by acute myocardial infarction or chronic ischemic heart failure.4,5 Results accumulated so far have documented the feasibility of this therapeutic approach with indications of potential beneficial effects on cardiac function and critical clinical end points. Although the mechanisms by which BMCs improve ventricular performance in humans are currently unknown, experimental evidence favors cardiac regeneration and the production of paracrine factors by these cells.6 New myocytes and coronary vessels are formed by transdifferentiation of the delivered cells, a process that …
- Published
- 2007
22. Concise Review: Stem Cells, Myocardial Regeneration, and Methodological Artifacts
- Author
-
Marcello Rota, Piero Anversa, Konrad Urbanek, Roberto Bolli, Claudia Bearzi, Toru Hosoda, Annarosa Leri, Jan Kajstura, Anversa, P, Leri, A, Rota, M, Hosoda, T, Bearzi, C, Urbanek, K, Kajstura, J, and Bolli, R
- Subjects
Cardiomyopathy, Dilated ,Heart Diseases ,Stem Cells ,Regeneration (biology) ,Myocardial Ischemia ,Heart ,Cell Biology ,Biology ,Mammalian heart ,Cell biology ,Haematopoiesis ,Humans ,Regeneration ,Molecular Medicine ,Progenitor cell ,Stem cell ,Artifacts ,Developmental Biology - Abstract
This review discusses the current controversy about the role that endogenous and exogenous progenitor cells have in cardiac homeostasis and myocardial regeneration following injury. Although great enthusiasm was created by the possibility of reconstituting the damaged heart, the opponents of this new concept of cardiac biology have interpreted most of the findings supporting this possibility as the product of technical artifacts. This article challenges this established, static view of cardiac growth and favors the notion that the mammalian heart has the inherent ability to replace its cardiomyocytes through the activation of a pool of resident primitive cells or the administration of hematopoietic stem cells.
- Published
- 2006
23. The Telomere–Telomerase Axis and the Heart
- Author
-
Piero Anversa, Toru Hosoda, Konrad Urbanek, Claudia Bearzi, Annarosa Leri, Jan Kajstura, Roberto Bolli, Marcello Rota, Kajstura, J, Rota, M, Urbanek, K, Hosoda, T, Bearzi, C, Anversa, P, Bolli, R, and Leri, A
- Subjects
Programmed cell death ,Telomerase ,Cell division ,Physiology ,Clinical Biochemistry ,Cell ,Biology ,Biochemistry ,Regenerative medicine ,medicine ,Animals ,Homeostasis ,Humans ,Myocyte ,Molecular Biology ,Cellular Senescence ,General Environmental Science ,Myocardium ,Cell Biology ,Telomere ,Cell cycle ,Cell biology ,Oxidative Stress ,medicine.anatomical_structure ,Cardiovascular Diseases ,General Earth and Planetary Sciences ,Reactive Oxygen Species - Abstract
The preservation of myocyte number and cardiac mass throughout life is dependent on the balance between cell death and cell division. Rapidly emerging evidence indicates that new myocytes can be formed through the activation and differentiation of resident cardiac progenitor cells. The critical issue is the identification of mechanisms that define the aging of cardiac progenitor cells and, ultimately, their inability to replace dying myocytes. The most reliable marker of cellular senescence is the modification of the telomere-telomerase axis, together with the expression of the cell cycle inhibitors p16INK4a and p53. Cellular senescence is characterized by biochemical events that occur within the cell. In this regard, one of the most relevant processes is represented by repeated oxidative stress that may evolve into the activation of the cell death program or result in the development of a senescent phenotype. Thus, the modulation of telomerase activity and the control of telomeric length, together with the attenuation of the formation of reactive oxygen species, may represent important therapeutic tools in regenerative medicine and in prevention of aging and diabetic cardiomyopathies.
- Published
- 2006
24. Akt Promotes Increased Cardiomyocyte Cycling and Expansion of the Cardiac Progenitor Cell Population
- Author
-
Natalie Gude, Marta Rubio, Jan Kajstura, Mark A. Sussman, John A. Muraski, Erik Schaefer, and Piero Anversa
- Subjects
medicine.medical_specialty ,Physiology ,Population ,Mice, Transgenic ,Biology ,Polymerase Chain Reaction ,Mice ,Paracrine signalling ,Internal medicine ,medicine ,Animals ,Progenitor cell ,education ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Oligonucleotide Array Sequence Analysis ,Muscle Cells ,education.field_of_study ,Microscopy, Confocal ,Cell growth ,Myocardium ,Stem Cells ,Cell Cycle ,Cell biology ,Proto-Oncogene Proteins c-kit ,Endocrinology ,Animals, Newborn ,Hepatocyte growth factor ,Stem cell ,Cardiology and Cardiovascular Medicine ,Proto-Oncogene Proteins c-akt ,medicine.drug - Abstract
Activation of Akt is associated with enhanced cell cycling and cellular proliferation in nonmyocytes, but this effect of nuclear Akt accumulation has not been explored in the context of the myocardium. Cardiac-specific expression of nuclear-targeted Akt (Akt/nuc) in transgenics prolongs postnatal cell cycling as evidenced by increased numbers of Ki67 + cardiomyocytes at 2 to 3 weeks after birth. Similarly, nuclear-targeting of Akt promotes expansion of the presumptive cardiac progenitor cell population as assessed by immunolabeling for c-kit in combination with myocyte-specific markers Nkx 2.5 or MEF 2C. Increases in pro-proliferative cytokines, including tumor-necrosis superfamily 8, interleukin-17e, and hepatocyte growth factor, were found in nuclear-targeted Akt myocardial samples. Concurrent signaling mediated by paracrine factors downstream of Akt/nuc expression may be responsible for phenotypic effects of nuclear-targeted Akt in the myocardium, including enhanced cell proliferation and expansion of the stem cell population.
- Published
- 2006
25. Nuclear targeting of Akt antagonizes aspects of cardiomyocyte hypertrophy
- Author
-
Piero Anversa, Yasuyuki Tsujita, Takahiro Kato, John A. Muraski, Jan Kajstura, Isao Shiraishi, and Mark A. Sussman
- Subjects
medicine.medical_specialty ,Systole ,Active Transport, Cell Nucleus ,Mice, Transgenic ,Biology ,Muscle hypertrophy ,Mice ,Phosphatidylinositol 3-Kinases ,Paracrine signalling ,fluids and secretions ,Atrial natriuretic peptide ,Internal medicine ,medicine ,Animals ,Myocytes, Cardiac ,Kinase activity ,Protein kinase A ,Autocrine signalling ,Protein kinase B ,Cell Nucleus ,Microscopy, Confocal ,Multidisciplinary ,Kinase ,Myocardium ,Hypertrophy ,Biological Sciences ,Cell biology ,Endocrinology ,Gene Expression Regulation ,Proto-Oncogene Proteins c-akt - Abstract
The serine/threonine kinase Akt regulates cellular survival, proliferation, gene transcription, protein translation, metabolism, and differentiation. Although Akt substrates are found throughout the cell, activated Akt normally accumulates in the nucleus, suggesting that biologically relevant targets are located there. Consequences of nuclear Akt signaling in cardiomyocytes were explored by using nuclear-targeted Akt (Akt-nuc). Accumulation of Akt-nuc did not provoke hypertrophy, unlike constitutively activated Akt. Instead, Akt-nuc inhibited hypertrophy concurrent with increased atrial natriuretic peptide (ANP) expression that depended upon phosphatidylinositol-3 kinase activity. Akt-nuc antihypertrophic effects were blocked by inhibition of either guanylyl cyclase A receptor or cyclic guanosine monophosphate-dependent protein kinase in cultured cardiomyocytes. Corroborating evidence showed blunted acute hypertrophic remodeling in Akt-nuc transgenic mice after transverse aortic constriction coincident with higher ANP expression and smaller myocyte volume. In addition, Akt-nuc expression improved systolic function and survival in the chronic phase of transverse aortic constriction-induced hypertrophy. Thus, Akt-nuc antagonizes certain aspects of hypertrophy through autocrine/paracrine stimulation of a phosphatidylinositol-3 kinase-dependent signaling cascade that promotes ANP expression, resulting in a unique combination of prosurvival coupled with antihypertrophic signaling.
- Published
- 2006
26. Cause of death: suicide
- Author
-
Jan Kajstura, Piero Anversa, Edmund H. Sonnenblick, Annarosa Leri, and Roberto Bolli
- Subjects
Programmed cell death ,Ventricular Remodeling ,Myocardium ,Apoptosis ,Biology ,medicine.disease ,medicine.disease_cause ,Structure and function ,Renin-Angiotensin System ,Oxidative Stress ,medicine ,Animals ,Humans ,Myocyte ,Myocytes, Cardiac ,Cardiology and Cardiovascular Medicine ,Ventricular remodeling ,Molecular Biology ,Neuroscience ,Oxidative stress ,Cause of death - Abstract
In the last decade, apoptosis has gained recognition as an important event determining the structure and function of the myocardium. The purpose of this review is to provide insights into the mechanisms controlling programmed cell death of myocytes. In this regard, the role of the local renin-angiotensin system and oxidative stress is emphasized. Moreover, the effect of cell death on ventricular remodeling and performance is analyzed. The impact of the identification of apoptosis in the heart on the current view of myocardium as a dynamic organ is discussed.
- Published
- 2006
27. Heart failure and regenerative cardiology
- Author
-
Piero Anversa, Annarosa Leri, Toru Hosoda, and Jan Kajstura
- Subjects
Heart Failure ,Embryology ,medicine.medical_specialty ,Myocardium ,Stem Cells ,media_common.quotation_subject ,Biomedical Engineering ,Bone Marrow Cells ,Heart ,Pavilion ,Art ,Mice ,Internal medicine ,Cardiology ,medicine ,Animals ,Humans ,Regeneration ,Bone Marrow Transplantation ,Stem Cell Transplantation ,media_common - Abstract
Annarosa Leri1, Toru Hosoda1, Jan Kajstura1 & Piero Anversa2† †Author for correspondence 1Cardiovascular Research Institute, Department of Medicine, New York Medical College, Valhalla, New York, NY 10595, USA 2Cardiovascular Research Institute, Department of Medicine, Vosburgh Pavilion, Room 302, New York Medical College, Valhalla, New York, NY 10595, USA Tel.: +1 914 594 4168; Fax: +1 914 594 4406; E-mail: piero_anversa@ nymc.edu ‘The bizarre behavior of hematopoietic stem cells has surprised and distressed many of us; they disobey the dogma of embryonic specification and with elegant and plastic arabesques undergo unexpected metamorphoses.’
- Published
- 2006
28. Endothelial progenitor cells: Neovascularization or more?
- Author
-
Annarosa Leri, Jan Kajstura, Roberto Bolli, and Piero Anversa
- Subjects
Cell fusion ,Regeneration (biology) ,Transdifferentiation ,Biology ,Cardiovascular physiology ,Endothelial stem cell ,Neovascularization ,Vasculogenesis ,Cancer research ,medicine ,Progenitor cell ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,Molecular Biology - Published
- 2006
29. Myocardial aging
- Author
-
Toru Hosoda, Roberto Bolli, Annarosa Leri, Marcello Rota, Jan Kajstura, Edmund H. Sonnenblick, Piero Anversa, Konrad Urbanek, Anversa, P, Rota, M, Urbanek, K, Hosoda, T, Sonnenblick, Eh, Leri, A, Kajstura, J, and Bolli, R
- Subjects
Senescence ,Aging ,education.field_of_study ,Physiology ,business.industry ,Myocardium ,Regeneration (biology) ,Population ,Stimulation ,medicine.disease ,Cell biology ,Physiology (medical) ,Heart failure ,medicine ,Animals ,Humans ,Myocyte ,Stem cell ,Cardiomyopathies ,Cardiology and Cardiovascular Medicine ,education ,business ,Homeostasis ,Stem Cell Transplantation - Abstract
This review questions the old paradigm that describes the heart as a post-mitotic organ and introduces the notion of the heart as a self-renewing organ regulated by a compartment of multipotent cardiac stem cells (CSCs) capable of regenerating myocytes and coronary vessels throughout life. Because of this dramatic change in cardiac biology, the objective is to provide an alternative perspective of the aging process of the heart and stimulate research in an area that pertains to all of us without exception. The recent explosion of the field of stem cell biology, with the recognition that the possibility exists for extrinsic and intrinsic regeneration of myocytes and coronary vessels, necessitates reevaluation of cardiac homeostasis and myocardial aging. From birth to senescence, the mammalian heart is composed of non-dividing and dividing cells. Loss of telomeric DNA is minimal in fetal and neonatal myocardium but rather significant in the senescent heart. Aging affects the growth and differentiation potential of CSCs interfering not only with their ability to sustain physiological cell turnover but also with their capacity to adapt to increases in pressure and volume loads. The recognition of factors enhancing the activation of the CSC pool, their mobilization, and translocation, however, suggests that the detrimental effects of aging on the heart might be prevented or reversed by local stimulation of CSCs or the intramyocardial delivery of CSCs following their expansion and rejuvenation in vitro. CSC therapy may become, perhaps, a novel strategy for the devastating problem of heart failure in the old population.
- Published
- 2005
30. Stem cells in the dog heart are self-renewing, clonogenic, and multipotent and regenerate infarcted myocardium, improving cardiac function
- Author
-
Axel Linke, Annarosa Leri, Piero Anversa, Angelo Nascimbene, Daria Nurzynska, Michael Böhm, Konrad Urbanek, Patrick Müller, Jan Kajstura, Daniele Torella, Claudia Casarsa, Stefano Cascapera, Federico Quaini, Thomas H. Hintze, Clotilde Castaldo, Linke, A, Muller, P, Nurzynska, DARIA ANNA, Casarsa, C, Torella, D, Nascimbene, A, Castaldo, Clotilde, Cascapera, S, Bohm, M, Quaini, F, Urbanek, K, Leri, A, Hintze, Th, Kajstura, J, and Anversa, P.
- Subjects
Cardiac function curve ,Cell Survival ,medicine.medical_treatment ,Cellular differentiation ,Myocardial Infarction ,Biology ,Muscle, Smooth, Vascular ,Receptor, IGF Type 1 ,Electrocardiography ,Dogs ,Cell Movement ,medicine ,Animals ,Regeneration ,Myocyte ,Myocardial infarction ,Insulin-Like Growth Factor I ,Clonogenic assay ,Multidisciplinary ,Hepatocyte Growth Factor ,MEF2 Transcription Factors ,Myocardium ,Stem Cells ,Growth factor ,Cell Differentiation ,Heart ,Anatomy ,Biological Sciences ,Proto-Oncogene Proteins c-met ,medicine.disease ,Myocardial Contraction ,Cell biology ,Myogenic Regulatory Factors ,cardiovascular system ,Hepatocyte growth factor ,Stem cell ,Cell Division ,medicine.drug - Abstract
The purpose of this study was to determine whether the heart in large mammals contains cardiac progenitor cells that regulate organ homeostasis and regenerate dead myocardium after infarction. We report that the dog heart possesses a cardiac stem cell pool characterized by undifferentiated cells that are self-renewing, clonogenic, and multipotent. These clonogenic cells and early committed progeny possess a hepatocyte growth factor (HGF)–c-Met and an insulin-like growth factor 1 (IGF-1)-IGF-1 receptor system that can be activated to induce their migration, proliferation, and survival. Therefore, myocardial infarction was induced in chronically instrumented dogs implanted with sonomicrometric crystals in the region of the left ventricular wall supplied by the occluded left anterior descending coronary artery. After infarction, HGF and IGF-1 were injected intramyocardially to stimulate resident cardiac progenitor cells. This intervention led to the formation of myocytes and coronary vessels within the infarct. Newly generated myocytes expressed nuclear and cytoplasmic proteins specific of cardiomyocytes: MEF2C was detected in the nucleus, whereas α-sarcomeric actin, cardiac myosin heavy chain, troponin I, and α-actinin were identified in the cytoplasm. Connexin 43 and N-cadherin were also present. Myocardial reconstitution resulted in a marked recovery of contractile performance of the infarcted heart. In conclusion, the activation of resident primitive cells in the damaged dog heart can promote a significant restoration of dead tissue, which is paralleled by a progressive improvement in cardiac function. These results suggest that strategies capable of activating the growth reserve of the myocardium may be important in cardiac repair after ischemic injury.
- Published
- 2005
31. Identity Deception: Not a Crime for a Stem Cell
- Author
-
Jan Kajstura, Annarosa Leri, and Piero Anversa
- Subjects
Induced stem cells ,Physiology ,Cellular differentiation ,Stem cell theory of aging ,Clinical uses of mesenchymal stem cells ,Cell Differentiation ,Biology ,Hematopoietic Stem Cells ,Immunology ,Animals ,Humans ,Progenitor cell ,Stem cell ,Neuroscience ,Adult stem cell ,Stem cell transplantation for articular cartilage repair - Abstract
Stem cell transdifferentiation in the adult organism is the most common and questioned mechanism of growth and repair. Recent data suggest that adult stem cells are capable of generating mature cells beyond their own tissue boundaries, a process called developmental plasticity. To date, the most versatile cell discovered is the bone marrow progenitor cell.
- Published
- 2005
32. Bone Marrow Cells Differentiate in Cardiac Cell Lineages After Infarction Independently of Cell Fusion
- Author
-
Toru Hosoda, Annarosa Leri, Federico Quaini, Daniele Torella, Massimiliano Bonafè, Elias Zias, Hideko Kasahara, Marcello Rota, Claudia Bearzi, Piero Anversa, Daria Nurzynska, Bernardo Nadal-Ginard, Jan Kajstura, Brian Whang, Stefano Cascapera, Angelo Nascimbene, Konrad Urbanek, Kajstura, J., Rota, M., Whang, B., Cascapera, S., Hosoda, T., Bearzi, C., Nurzynska, DARIA ANNA, Kasahara, H., Zias, E., Bonafe, M., Nadal Ginard, B., Torella, D., Nascimbene, A., Quaini, F., Urbanek, K., Leri, A., Anversa, P., J. Kajstura, M. Rota, B. Whang, S. Cascapera, T. Hosoda, C. Bearzi, D. Nurzynska, H. Kasahara, E. Zia, M. Bonafe, B. Nadal-Ginard B, D. Torella, A. Nascimbene, F. Quaini, K. Urbanek A. Leri, and P. Anversa
- Subjects
Male ,Pathology ,medicine.medical_specialty ,Physiology ,Green Fluorescent Proteins ,Myocytes, Smooth Muscle ,Cell ,Myocardial Infarction ,Bone Marrow Cells ,Mice, Transgenic ,Injections, Intralesional ,Biology ,Ventricular Function, Left ,Cell Fusion ,Mice ,Paracrine signalling ,Genes, Reporter ,Y Chromosome ,Paracrine Communication ,medicine ,Animals ,Humans ,Regeneration ,Myocyte ,Cell Lineage ,Myocytes, Cardiac ,Cell fusion ,Regeneration (biology) ,Graft Survival ,Transdifferentiation ,Hematopoietic Stem Cell Transplantation ,Endothelial Cells ,Cell Differentiation ,Heart ,Myocardial Contraction ,Capillaries ,Arterioles ,Proto-Oncogene Proteins c-kit ,Haematopoiesis ,medicine.anatomical_structure ,Organ Specificity ,Female ,Bone marrow ,Artifacts ,Cardiology and Cardiovascular Medicine ,Stem Cell Transplantation - Abstract
Recent studies in mice have challenged the ability of bone marrow cells (BMCs) to differentiate into myocytes and coronary vessels. The claim has also been made that BMCs acquire a cell phenotype different from the blood lineages only by fusing with resident cells. Technical problems exist in the induction of myocardial infarction and the successful injection of BMCs in the mouse heart. Similarly, the accurate analysis of the cell populations implicated in the regeneration of the dead tissue is complex and these factors together may account for the negative findings. In this study, we have implemented a simple protocol that can easily be reproduced and have reevaluated whether injection of BMCs restores the infarcted myocardium in mice and whether cell fusion is involved in tissue reconstitution. For this purpose, c-kit–positive BMCs were obtained from male transgenic mice expressing enhanced green fluorescence protein (EGFP). EGFP and the Y-chromosome were used as markers of the progeny of the transplanted cells in the recipient heart. By this approach, we have demonstrated that BMCs, when properly administrated in the infarcted heart, efficiently differentiate into myocytes and coronary vessels with no detectable differentiation into hemopoietic lineages. However, BMCs have no apparent paracrine effect on the growth behavior of the surviving myocardium. Within the infarct, in 10 days, nearly 4.5 million biochemically and morphologically differentiated myocytes together with coronary arterioles and capillary structures were generated independently of cell fusion. In conclusion, BMCs adopt the cardiac cell lineages and have an important therapeutic impact on ischemic heart failure.
- Published
- 2005
33. Nuclear Targeting of Akt Enhances Kinase Activity and Survival of Cardiomyocytes
- Author
-
Youngkeun Ahn, Jan Kajstura, Elizabeth Murphy, Sara Welch, Isao Shiraishi, Annarosa Leri, Jaime Melendez, Kenneth Walsh, Mark A. Sussman, Erik Schaefer, Daniele Torella, Daria Nurzynska, Maryanne Skavdahl, Piero Anversa, Anthony Rosenzweig, Shiraishi, I., Melendez, J., Ahn, Y., Skavdahl, M., Murphy, E., Welch, S., Schaefer, E., Walsh, K., Rosenzweig, A., Torella, D., Nurzynska, DARIA ANNA, Kajstura, J., Leri, A., Anversa, P., and Sussman, M. A.
- Subjects
Programmed cell death ,Physiology ,Heart Ventricles ,Recombinant Fusion Proteins ,Genetic Vectors ,Nuclear Localization Signals ,Active Transport, Cell Nucleus ,Genes, myc ,Myocardial Infarction ,Apoptosis ,Mice, Transgenic ,Myocardial Reperfusion Injury ,Protein Serine-Threonine Kinases ,Biology ,Transfection ,Polymerase Chain Reaction ,Adenoviridae ,Rats, Sprague-Dawley ,Mice ,Proto-Oncogene Proteins ,Genes, Synthetic ,Animals ,Myocyte ,Myocytes, Cardiac ,Kinase activity ,Protein kinase B ,Cells, Cultured ,PI3K/AKT/mTOR pathway ,Cell Nucleus ,Kinase ,Gene Expression Profiling ,Cell Hypoxia ,Rats ,Cell biology ,Enzyme Activation ,Phosphorylation ,Cardiology and Cardiovascular Medicine ,Proto-Oncogene Proteins c-akt - Abstract
Heart failure is associated with death of cardiomyocytes leading to loss of contractility. Previous studies using membrane-targeted Akt (myristolated-Akt), an enzyme involved in antiapoptotic signaling, showed inhibition of cell death and prevention of pathogenesis induced by cardiomyopathic stimuli. However, recent studies by our group have found accumulation of activated Akt in the nucleus, suggesting that biologically relevant target(s) of Akt activity may be located there. To test this hypothesis, a targeted Akt construct was created to determine the antiapoptotic action of nuclear Akt accumulation. Nuclear localization of the adenovirally encoded Akt construct was confirmed by confocal microscopy. Cardiomyocytes expressing nuclear-targeted Akt showed no evidence of morphological remodeling such as altered myofibril density or hypertrophy. Nuclear-targeted Akt significantly elevated levels of phospho-Akt and kinase activity and inhibited apoptosis as effectively as myristolated-Akt in hypoxia-induced cell death. Transgenic overexpression of nuclear-targeted Akt did not result in hypertrophic remodeling, altered cardiomyocyte DNA content or nucleation, or enhanced phosphorylation of typical cytoplasmic Akt substrates, yet transgenic hearts were protected from ischemia-reperfusion injury. Gene array analyses demonstrated changes in the transcriptional profile of Akt/nuc hearts compared with nontransgenic controls distinct from prior characterizations of Akt expression in transgenic hearts. Collectively, these experiments show that targeting of Akt to the nucleus mediates inhibition of apoptosis without hypertrophic remodeling, opening new possibilities for therapeutic applications of nuclear-targeted Akt to inhibit cell death associated with heart disease.
- Published
- 2004
34. Primitive Cells and Tissue Regeneration
- Author
-
Jan Kajstura, Annarosa Leri, Piero Anversa, and Bernardo Nadal-Ginard
- Subjects
Pathology ,medicine.medical_specialty ,Physiology ,Cellular differentiation ,Mesenchymal stem cell ,Clinical uses of mesenchymal stem cells ,Bone Marrow Stem Cell ,Biology ,Cell biology ,Cell therapy ,medicine ,Stem cell ,Cardiology and Cardiovascular Medicine ,Adult stem cell ,Stem cell transplantation for articular cartilage repair - Abstract
In the last few years, several experimental studies have used stem cells of different sources to reconstitute damaged tissues.1 The brain and the heart have been the most investigated organs because of the long-standing view of the lack of regenerating potential of neurons and myocytes.2,3 Bone marrow stem cells (BMSCs) have been reported capable of transdifferentiating in various cell lineages distinct from the site of origin4 and, because of this property, they may constitute a new form of cellular therapy. Neuronal and myocardial growth mediated by bone marrow cells (BMCs) has been demonstrated, but these results have been challenged5,6 and the issue of BMSC transdifferentiation has become highly controversial. Heated debates at scientific meetings, letters in high-profile journals, and reports with contradicting observations have raised questions on the plasticity of BMSCs.5–7 If negative results would be more cautiously interpreted instead of being blown out of context, it is likely that the actual role that adult stem cells play in the repair of tissues and organs would be better understood and appreciated. This is particularly relevant when negative data are dropped as “valid” statements from the podium and are quoted before they undergo peer review and publication.6 A good example of the opposite approach is found in the study of Chen and colleagues8 in this issue of Circulation Research . The authors have utilized, among other sophisticated techniques, confocal microscopy to identify and characterize an important new function of human pluripotent adult mesenchymal BMSCs. In this report, an unequivocal demonstration was obtained on the ability of these cells to deliver vascular endothelial growth factor (VEGF) to an ischemic region of the brain. VEGF accumulation coupled with endogenous activation of endothelial cells and VEGF synthesis promoted vessel formation after stroke. BMSCs via VEGF secretion acted …
- Published
- 2003
35. Myocyte Death, Growth, and Regeneration in Cardiac Hypertrophy and Failure
- Author
-
Bernardo Nadal-Ginard, Annarosa Leri, Piero Anversa, and Jan Kajstura
- Subjects
Senescence ,Programmed cell death ,medicine.medical_specialty ,Physiology ,Cardiomegaly ,Biology ,Muscle hypertrophy ,Internal medicine ,medicine ,Animals ,Humans ,Regeneration ,Myocyte ,Myocytes, Cardiac ,Cellular Senescence ,Bone Marrow Transplantation ,Heart Failure ,Cell Death ,Myocardium ,Stem Cells ,Regeneration (biology) ,medicine.disease ,Cell biology ,Endocrinology ,Heart failure ,Stem cell ,Cardiology and Cardiovascular Medicine ,Cell Division ,Homeostasis - Abstract
The accepted paradigm considers the adult mammalian heart as a postmitotic organ, which possesses a relatively constant number of myocytes from shortly after birth to adulthood and senescence. This notion is questioned by the demonstration that although most adult myocytes are terminally differentiated, there is a small and continuously renewed subpopulation of cycling myocytes produced by the differentiation of cardiac stem-like cells. Myocyte death and myocyte regeneration are introduced as major determinants of cardiac homeostasis and alterations of ventricular anatomy and function in physiological and pathological states. The possibility of reconstituting dead myocardium by stem-like cells is advanced and proposed as a major area of future research.
- Published
- 2003
36. Ablation of telomerase and telomere loss leads to cardiac dilatation and heart failure associated with p53 upregulation
- Author
-
Laura Barlucchi, Sonia Franco, Piero Anversa, Stefano Chimenti, Federica Limana, Jan Kajstura, Bernardo Nadal-Ginard, Antonella Zacheo, Maria A. Blasco, and Annarosa Leri
- Subjects
medicine.medical_specialty ,Telomerase ,Population ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Muscle hypertrophy ,Mice ,Internal medicine ,medicine ,Animals ,Humans ,Myocyte ,education ,Molecular Biology ,Cells, Cultured ,Heart Failure ,Mice, Knockout ,Muscle Cells ,education.field_of_study ,General Immunology and Microbiology ,General Neuroscience ,Cardiac myocyte ,Heart ,Articles ,Anatomy ,Telomere ,Genes, p53 ,medicine.disease ,Vasodilation ,Disease Models, Animal ,Gene Expression Regulation ,Heart failure ,Knockout mouse ,cardiovascular system ,Cardiology ,Tumor Suppressor Protein p53 - Abstract
Cardiac failure is a frequent cause of death in the aging human population. Telomere attrition occurs with age, and is proposed to be causal for the aging process. To determine whether telomere shortening leads to a cardiac phenotype, we studied heart function in the telomerase knockout mouse, Terc–/–. We studied Terc–/– mice at the second, G2, and fifth, G5, generation. Telomere shortening in G2 and G5 Terc–/– mice was coupled with attenuation in cardiac myocyte proliferation, increased apoptosis and cardiac myocyte hypertrophy. On a single-cell basis, telomere shortening was coincidental with increased expression of p53, indicating the presence of dysfunctional telomeres in cardiac myocytes from G5 Terc–/– mice. The impairment in cell division, the enhanced cardiac myocyte death and cellular hypertrophy, are concomitant with ventricular dilation, thinning of the wall and cardiac dysfunction. Thus, inhibition of cardiac myocyte replication provoked by telomere shortening, results in de-compensated eccentric hypertrophy and heart failure in mice. Telomere shortening with age could also contribute to cardiac failure in humans, opening the possibility for new therapies.
- Published
- 2003
37. Myocyte proliferation and ventricular remodeling
- Author
-
Jan Kajstura, Annarosa Leri, and Piero Anversa
- Subjects
Heart Failure ,Cardiac function curve ,medicine.medical_specialty ,Ventricular Remodeling ,business.industry ,Myocardium ,Myocyte proliferation ,Cardiomegaly ,medicine.disease ,Muscle hypertrophy ,Internal medicine ,Heart failure ,medicine ,Cardiology ,Humans ,Myocyte ,Cardiac myocyte proliferation ,Myocytes, Cardiac ,Myocyte hypertrophy ,Cardiology and Cardiovascular Medicine ,business ,Ventricular remodeling ,Cell Division - Abstract
Improvement in the methodological approach to the analysis of the myocardium has provided clear evidence of cardiac myocyte proliferation, questioning the general belief that the growth of the adult heart under physiological and pathological conditions can occur only by cellular hypertrophy. Myocyte regeneration contributes via myocyte death to the physiological turnover of myocytes and via myocyte hypertrophy to cardiac remodeling. Several questions, however, remain to be answered. Among them, it is still unknown whether myocyte multiplication exerts a positive and/or negative effect on ventricular anatomy and cardiac function. The addition of newly generated myocytes leads to cavitary dilation with relative thinning of the wall. Conversely, myocyte proliferation, characterized by the parallel addition of cells, can be expected to increase wall thickness, decrease chamber size, and ameliorate cardiac performance.
- Published
- 2002
38. Cardiac-Specific IGF-1 Expression Attenuates Dilated Cardiomyopathy in Tropomodulin-Overexpressing Transgenic Mice
- Author
-
Erik Schaefer, Stefano Chimenti, David M. Plank, Betty J. Glascock, Sandra A. Witt, Anna Maria Andreoli, Federica Limana, Piero Anversa, Sara Welch, Annarosa Leri, Jan Kajstura, and Mark A. Sussman
- Subjects
Cardiomyopathy, Dilated ,Genetically modified mouse ,medicine.medical_specialty ,Heart disease ,Physiology ,Cardiomyopathy ,Apoptosis ,Cell Count ,Mice, Transgenic ,Mice ,Internal medicine ,medicine ,Animals ,Myocyte ,Insulin-Like Growth Factor I ,Cell Size ,Ventricular Remodeling ,biology ,Microfilament Proteins ,Dilated cardiomyopathy ,medicine.disease ,Ki-67 Antigen ,Endocrinology ,Gene Expression Regulation ,Heart failure ,Circulatory system ,biology.protein ,Calcium ,Carrier Proteins ,Cardiology and Cardiovascular Medicine ,Tropomodulin - Abstract
To test the hypothesis that early interventional treatment with insulin-like growth factor-1 (IGF-1) alleviates subsequent development of dilated cardiomyopathy, cardiac-specific IGF-1 expression was introduced by selective cross-breeding into a transgenic mouse model of heart failure that displays phenotypic characteristics of severe dilation. Hemodynamic, structural, and cellular parameters of the heart were compared between nontransgenic, tropomodulin-overexpressing cardiomyopathic, and the hybrid tropomodulin/IGF-1-overexpressing mice. Beneficial effects of IGF-1 were apparent by multiple indices of cardiac structure and function, including normalization of heart mass, anatomy, hemodynamics, and apoptosis. IGF-1 expression also acted as a proliferative stimulus as evidenced by calculated increases in myocyte number as well as expression of Ki67, a nuclear marker of cellular replication. Cellular analyses revealed that IGF-1 inhibited characteristic cardiomyocyte elongation in dilated hearts and restored calcium dynamics comparable to that observed in normal cells. Collectively, these results provide novel information regarding the ability of IGF-1 to inhibit progression of cardiomyopathic disease in a defined model system and suggest that heart failure may benefit from early interventional IGF-1 treatment.
- Published
- 2002
39. Response to Letter Regarding Article 'Inositol 1,4,5-Trisphosphate Receptors and Human Left Ventricular Myocytes'
- Author
-
Mehrdad Shafaie, Fumihiro Sanada, David A. D'Alessandro, Andrew Webster, Barbara Ogorek, Robert E. Michler, Piero Anversa, Annarosa Leri, Polina Goichberg, Oriyanhan Wunimenghe, Ewa Wybieralska, Xiaoxia Liu, Christian Arranto, João Ferreira-Martins, Toru Hosoda, Federica del Monte, Hanqiao Zheng, Ramaswamy Kannappan, Sergio Signore, Andrea Sorrentino, Kazuya Isobe, Jan Kajstura, Fabio del Ben, and Marcello Rota
- Subjects
Inotrope ,Adult ,Male ,medicine.medical_specialty ,Heart Ventricles ,Action Potentials ,Endogeny ,Stimulation ,Article ,chemistry.chemical_compound ,Mice ,Physiology (medical) ,Internal medicine ,medicine ,Myocyte ,Animals ,Humans ,Inositol 1,4,5-Trisphosphate Receptors ,Inositol ,Myocytes, Cardiac ,Calcium Signaling ,Receptor ,Cells, Cultured ,Calcium signaling ,Heart Failure ,business.industry ,Endoplasmic reticulum ,Arrhythmias, Cardiac ,Ryanodine Receptor Calcium Release Channel ,Middle Aged ,Myocardial Contraction ,Mice, Inbred C57BL ,Sarcoplasmic Reticulum ,Endocrinology ,chemistry ,GTP-Binding Protein alpha Subunits, Gq-G11 ,Female ,Cardiology and Cardiovascular Medicine ,business ,Signal Transduction - Abstract
Little is known about the function of inositol 1,4,5-trisphosphate receptors (IP3Rs) in the adult heart experimentally. Moreover, whether these Ca(2+) release channels are present and play a critical role in human cardiomyocytes remains to be defined. IP3Rs may be activated after Gαq-protein-coupled receptor stimulation, affecting Ca(2+) cycling, enhancing myocyte performance, and potentially favoring an increase in the incidence of arrhythmias.IP3R function was determined in human left ventricular myocytes, and this analysis was integrated with assays in mouse myocytes to identify the mechanisms by which IP3Rs influence the electric and mechanical properties of the myocardium. We report that IP3Rs are expressed and operative in human left ventricular myocytes. After Gαq-protein-coupled receptor activation, Ca(2+) mobilized from the sarcoplasmic reticulum via IP3Rs contributes to the decrease in resting membrane potential, prolongation of the action potential, and occurrence of early afterdepolarizations. Ca(2+) transient amplitude and cell shortening are enhanced, and extrasystolic and dysregulated Ca(2+) elevations and contractions become apparent. These alterations in the electromechanical behavior of human cardiomyocytes are coupled with increased isometric twitch of the myocardium and arrhythmic events, suggesting that Gαq-protein-coupled receptor activation provides inotropic reserve, which is hampered by electric instability and contractile abnormalities. Additionally, our findings support the notion that increases in Ca(2+) load by IP3Rs promote Ca(2+) extrusion by forward-mode Na(+)/Ca(2+) exchange, an important mechanism of arrhythmic events.The Gαq-protein/coupled receptor/IP3R axis modulates the electromechanical properties of the human myocardium and its propensity to develop arrhythmias.
- Published
- 2014
40. Progenitor Cells and Cardiac Homeostasis and Regeneration
- Author
-
Annarosa Leri, Piero Anversa, and Jan Kajstura
- Subjects
Endothelial stem cell ,Programmed cell death ,Regeneration (biology) ,Immunology ,Stem cell theory of aging ,Myocyte ,Biology ,Progenitor cell ,Stem cell ,Homeostasis ,Cell biology - Abstract
This chapter discusses the fundamental observations that have profoundly changed our view of the adult human heart, resulting in the experimental use of stem/progenitor cells as a potential form of therapy for the failing heart. The progressive decline in myocyte number as a function of age and the formation of scarred tissue following myocardial infarction have been interpreted as irrefutable proofs of the post-mitotic characteristic of the heart. Emerging evidence supports a more dynamic view of the heart, in which cell death and renewal are vital components of the remodeling process that governs cardiac homeostasis, aging and disease. The identification of dividing myocytes raises the important question concerning the origin of the newly formed cells. Myocyte renewal can be accomplished by commitment of stem cells, and replication or dedifferentiation of pre-existing myocytes. In vitro and in vivo findings strongly suggest that replicating myocytes correspond to transit amplifying cells derived from the lineage determination of primitive cells, supporting the notion that cardiomyogenesis is controlled by activation and differentiation of stem cells.
- Published
- 2014
41. REMOVED: Stem Cells and Heart Disease
- Author
-
Piero Anversa, Jan Kajstura, Annarosa Leri, and Bernardo Nadal-Ginard
- Subjects
medicine.medical_specialty ,Heart disease ,Stem cell theory of aging ,Clinical uses of mesenchymal stem cells ,Amniotic stem cells ,Biology ,medicine.disease ,Muscle hypertrophy ,Internal medicine ,Amniotic epithelial cells ,Immunology ,medicine ,Cardiology ,Stem cell ,Adult stem cell - Abstract
A fundamental issue concerning the ability of the heart to sustain cardiac diseases of ischemic and nonischemic origin is whether myocardial regeneration can occur in the adult organ, or whether this growth adaptation is restricted to prenatal life, severely limiting the response of the heart to pathologic states. The concept of the heart as a terminally differentiated organ unable to replace working myocytes has been at the center of cardiovascular research and therapeutic developments for the last 50 years. The accepted view has been and remains that the heart reacts to an increase in workload only by hypertrophy of the existing myocytes during postnatal maturation, adulthood, and senility. In contrast to the widespread acceptance of this dogma, numerous findings indicate that the mammalian heart has replication potential and its response to abnormal elevations in load can be accompanied by myocyte proliferation.
- Published
- 2014
42. List of Contributors
- Author
-
Russell C. Addis, Piero Anversa, Judith Arcidiacono, Anthony Atala, Joyce Axelman, Ashok Batra, Helen M. Blau, Susan Bonner-Weir, Mairi Brittan, Hal E. Broxmeyer, Mara Cananzi, Constance Cepko, Tao Cheng, Susana M. Chuva de Sousa Lopes, Gregory O. Clark, Maegen Colehour, Paolo de Coppi, Giulio Cossu, George Q. Daley, Jiyoung M. Dang, Natalie Direkze, Yuval Dor, Gregory R. Dressler, Charles N. Durfor, Ewa C.S. Ellis, Martin Evans, Donna M. Fekete, Donald Fink, Elaine Fuchs, Margaret T. Fuller, Richard L. Gardner, Zulma Gazit, Dan Gazit, John D. Gearhart, Victor M. Goldberg, Rodolfo Gonzalez, Deborah Lavoie Grayeski, Ronald M. Green, Markus Grompe, Stephen L. Hilbert, Marko E. Horb, Jerry I. Huang, Jaimie Imitola, D. Leanne Jones, Jan Kajstura, David S. Kaplan, Pritinder Kaur, Kathleen C. Kent, Candace L. Kerr, Ali Khademhosseini, Nadav Kimelman, Irina Klimanskaya, Jennifer N. Kraszewski, Mark A. LaBarge, Robert Langer, Robert Lanza, Ellen Lazarus, Jean Pyo Lee, Mark H. Lee, Annarosa Leri, Shulamit Levenberg, S. Robert Levine, John W. Littlefield, Richard McFarland, Jill McMahon, Douglas A. Melton, Mary Tyler Moore, Franz-Josef Mueller, Christine L. Mummery, Bernardo Nadal-Ginard, Hitoshi Niwa, Keisuke Okita, Jitka Ourednik, Vaclav Ourednik, Kook I. Park, Ethan S. Patterson, Gadi Pelled, Christopher S. Potten, Sean Preston, Philip R. Roelandt, Valerie D. Roobrouck, Nadia Rosenthal, Janet Rossant, Maurilio Sampaolesi, Maria Paola Santini, David T. Scadden, Holger Schlüter, Gunter Schuch, Michael J. Shamblott, Dima Sheyn, Richard L. Sidman, Evan Y. Snyder, Shay Soker, Stephen C. Strom, Lorenz Studer, M. Azim Surani, Francesco Saverio Tedesco, Yang D. Teng, David Tosh, Alan Trounson, Tudorita Tumbar, Edward Upjohn, George Varigos, Catherine M. Verfaillie, Zhan Wang, Gordon C. Weir, Kevin J. Whittlesey, J. Koudy Williams, James W. Wilson, Celia Witten, Nicholas A. Wright, Shinya Yamanaka, and Jung U. Yoo
- Published
- 2014
43. c-Kit-positive cardiac stem cells nested in hypoxic niches are activated by stem cell factor reversing the aging myopathy
- Author
-
Emily Mangano, Maria Cimini, Chiara Mangiaracina, Polina Goichberg, Ada Pesapane, Emeka Ifedigbo, Giulia Borghetti, Andrea Sorrentino, Kazuya Isobe, Mario Ricciardi, Sergio Signore, Anna Czarna, Toru Hosoda, Marcello Rota, Junghyun Kim, Ewa Wybieralska, Augustine M.K. Choi, Annarosa Leri, Donato Cappetta, Piero Anversa, Noel Yan-Ki Chan, Fumihiro Sanada, Barbara Ogorek, Jan Kajstura, Mark A. Perrella, Sanada F, Kim J, Czarna A, Chan NY, Signore S, Ogórek B, Isobe K, Wybieralska E, Borghetti G, Pesapane A, Sorrentino A, Mangano E, Cappetta D, Mangiaracina C, Ricciardi M, Cimini M, Ifedigbo E, Perrella MA, Goichberg P, Choi AM, Kajstura J, Hosoda T, Rota M, Anversa P, Leri A, Sanada, F, Kim, J, Czarna, A, Chan, Ny, Signore, S, Ogórek, B, Isobe, K, Wybieralska, E, Borghetti, G, Pesapane, A, Sorrentino, A, Mangano, E, Cappetta, D, Mangiaracina, C, Ricciardi, M, Cimini, M, Ifedigbo, E, Perrella, Ma, Goichberg, P, Choi, Am, Kajstura, J, Hosoda, T, Rota, M, Anversa, P, and Leri, A
- Subjects
Aging ,Physiology ,Stem cell theory of aging ,Population ,Stem cell factor ,Biology ,Article ,Mice ,stem cell factor ,Animals ,Myocyte ,Cell Lineage ,Myocytes, Cardiac ,Stem Cell Niche ,Hypoxia ,education ,Cellular Senescence ,Cell Proliferation ,education.field_of_study ,Cell growth ,Myocardium ,Cell Cycle ,Telomere Homeostasis ,Cell cycle ,Cell biology ,Mice, Inbred C57BL ,Proto-Oncogene Proteins c-kit ,Immunology ,Stem cell ,Cardiomyopathies ,Cardiology and Cardiovascular Medicine ,Cell aging ,Myoblasts, Cardiac - Abstract
Rationale: Hypoxia favors stem cell quiescence, whereas normoxia is required for stem cell activation, but whether cardiac stem cell (CSC) function is regulated by the hypoxic/normoxic state of the cell is currently unknown. Objective: A balance between hypoxic and normoxic CSCs may be present in the young heart, although this homeostatic control may be disrupted with aging. Defects in tissue oxygenation occur in the old myocardium, and this phenomenon may expand the pool of hypoxic CSCs, which are no longer involved in myocyte renewal. Methods and Results: Here, we show that the senescent heart is characterized by an increased number of quiescent CSCs with intact telomeres that cannot re-enter the cell cycle and form a differentiated progeny. Conversely, myocyte replacement is controlled only by frequently dividing CSCs with shortened telomeres; these CSCs generate a myocyte population that is chronologically young but phenotypically old. Telomere dysfunction dictates their actual age and mechanical behavior. However, the residual subset of quiescent young CSCs can be stimulated in situ by stem cell factor reversing the aging myopathy. Conclusions: Our findings support the notion that strategies targeting CSC activation and growth interfere with the manifestations of myocardial aging in an animal model. Although caution has to be exercised in the translation of animal studies to human beings, our data strongly suggest that a pool of functionally competent CSCs persists in the senescent heart and that this stem cell compartment can promote myocyte regeneration effectively, partly correcting the aging myopathy.
- Published
- 2014
44. List of Contributors
- Author
-
Robby D. Bowles, Anthony J. (Tony) Smith, Jon D. Ahlstrom, Julie Albon, Peter G. Alexander, Richard A. Altschuler, Pedro Alvarez, A. Amendola, Rachael Anatol, Nasim Annabi, Piero Anversa, Judith Arcidiacono, Anthony Atala, Kyriacos A. Athanasiou, François A. Auger, Debra T. Auguste, Hani A. Awad, Stephen F. Badylak, Alexander M. Bailey, Michael P. Barry, Daniel Becker, Visar Belegu, Jonathan Bernhard, Timothy Bertram, Valérie Besnard, Z.F. Bhat, Hina Bhat, Sangeeta N. Bhatia, Sarindr Bhumiratana, Paolo Bianco, Catherine Clare Blackburn, Thomas Bollenbach, Lawrence A. Bonassar, Mike Boulton, Amy D. Bradshaw, Christopher K. Breuer, Luke Brewster, Eric M. Brey, Mairi Brittan, Bryan N. Brown, T. Brown, J.A. Buckwalter, Deborah Buffington, Karen J.L. Burg, Timothy C. Burg, Stéphane Chabaud, Thomas Ming Swi Chang, Yunchao Chang, Robert G. Chapman, Fa-Ming Chen, Una Chen, Elisa Cimetta, Richard A.F. Clark, Karen L. Clark, Muriel A. Cleary, Réjean Cloutier, Clark K. Colton, George Cotsarelis, Ronald G. Crystal, Gislin Dagnelie, Lino da Silva Ferreira, Jeffrey M. Davidson, Thomas F. Deuel, Natalie Direkze, Gregory R. Dressler, Charles N. Durfor, Craig L. Duvall, George Eng, George Engelmayr, Thomas Eschenhagen, Mark Eu-Kien Wong, Vincent Falanga, Katie Faria, Denise L. Faustman, Dario O. Fauza, Qiang Feng, Lino Ferreira, Donald W. Fink, William Fissell, Lisa E. Freed, Mark E. Furth, Denise Gay, Sharon Gerecht-Nir, Lucie Germain, Charles A. Gersbach, Francine Goulet, Ritu Goyal, Maria B. Grant, Howard P. Greisler, Farshid Guilak, Brendan A.C. Harley, David A. Hart, Abdelkrim Hmadcha, Steve J. Hodges, Heidi R. Hofer, Jeffrey O. Hollinger, Patricia Holobaugh, Jeffrey A. Hubbell, H. David Humes, Donald E. Ingber, Beau Inskeep, Xingyu Jiang, Jan Kajstura, Ravi S. Kane, Jeffrey M. Karp, F. Kurtis Kasper, Ali Khademhosseini, Sven Kili, Erin A. Kimbrel, Irina Klimanskaya, Joachim Kohn, Shaun M. Kunisaki, Themis R. Kyriakides, Eric Lagasse, Jean Lamontagne, Robert Langer, Robert Lanza, Shimon Lecht, Benjamin W. Lee, Chang H. Lee, Mark H. Lee, Peter I. Lelkes, Annarosa Leri, David W. Levine, Feng Li, Michael T. Longaker, Javier López, Shi-Jiang Lu, Ying Luo, Ben D. MacArthur, Nancy Ruth Manley, Rohan Manohar, Jonathan Mansbridge, Athanasios Mantalaris, Jeremy J. Mao, J.L. Marsh, David C. Martin, J.A. Martin, M. Martins-Green, Koichi Masuda, Mark W. Maxfield, Kathryn L. McCabe, John W. McDonald, Richard McFarland, Antonios G. Mikos, José del R. Millán, Josef M. Miller, Shari Mills, Kristen L. Moffat, Mark J. Mondrinos, Daniel T. Montoro, Malcolm A.S. Moore, Rebekah A. Neal, Robert M. Nerem, Shengyong Ng, Craig Scott Nowell, Haruko Obokata, Bjorn Reino Olsen, Richard O.C. Oreffo, Regis J. O’Keefe, Kathy O’Neill, Ophir Ortiz, Carolyn K. Pan, Vikas Pathak, M. Petreaca, Daniela Pezzolla, Maksim V. Plikus, Julia M. Polak, Mark Post, Sean Preston, Aleš Prokop, Milica Radisic, Egon Ranghini, Yehoash Raphael, A.H. Reddi, Herrmann Reichenspurner, Ellen Richie, Pamela Gehron Robey, Becky Robinson, Anabel Rojas, Shuvo Roy, Alan J. Russell, Rajiv Saigal, W. Mark Saltzman, Ali Samadikuchaksaraei, Athanassios Sambanis, Jochen Schacht, Stacey C. Schutte, Lyndsey Schutte, Steven D. Schwartz, Robert E. Schwartz, Lori A. Setton, Su-Hua Sha, Jing Shan, Paul T. Sharpe, Songtao Shi, Arun R. Shrivats, Franck Simon, Dario Sirabella, J.M.W. Slack, Bernat Soria, Patrick Spicer, Kelly R. Stevens, Frank E. Stockdale, H. Christiaan Stronks, Lorenz Studer, Shuichi Takayama, James A. Thomson, Jordan E. Trachtenberg, Elsa Treffeisen, Rocky S. Tuan, Charles A. Vacanti, Joseph P. Vacanti, Cor van der Weele, Matthew Vincent, Gordana Vunjak-Novakovic, Lars U. Wahlberg, Derrick C. Wan, Anne Wang, Angela J. Westover, George M. Whitesides, Jeffrey A. Whitsett, Steve Winitsky, Celia Witten, Stefan Worgall, Nicholas A. Wright, Ioannis V. Yannas, Simon Young, Junying Yu, Zheng Zhang, Wenfu Zheng, Wolfram Hubertus Zimmermann, and Laurie Zoloth
- Published
- 2014
45. Telomerase expression and activity are coupled with myocyte proliferation and preservation of telomeric length in the failing heart
- Author
-
Annarosa Leri, Thomas H. Hintze, Piero Anversa, Federica Limana, Laura Barlucchi, Zbigniew Darzynkiewicz, A. Deptala, Bernardo Nadal-Ginard, and Jan Kajstura
- Subjects
medicine.medical_specialty ,Telomerase ,Population ,Biology ,Muscle hypertrophy ,Dogs ,Internal medicine ,medicine ,Animals ,Humans ,Myocyte ,education ,Heart Failure ,education.field_of_study ,Multidisciplinary ,Cell growth ,Myocardium ,Dilated cardiomyopathy ,Biological Sciences ,Telomere ,medicine.disease ,Cell biology ,DNA-Binding Proteins ,Ki-67 Antigen ,Endocrinology ,Heart failure ,Cell Division - Abstract
The role and even the existence of myocyte proliferation in the adult heart remain controversial. Documentation of cell cycle regulators, DNA synthesis, and mitotic images has not modified the view that myocardial growth can only occur from hypertrophy of an irreplaceable population of differentiated myocytes. To improve understanding the biology of the heart and obtain supportive evidence of myocyte replication, three indices of cell proliferation were analyzed in dogs affected by a progressive deterioration of cardiac performance and dilated cardiomyopathy. The magnitude of cycling myocytes was evaluated by the expression of Ki67 in nuclei. Ki67 labeling of left ventricular myocytes increased 5-fold, 12-fold, and 17-fold with the onset of moderate and severe ventricular dysfunction and overt failure, respectively. Telomerase activity in vivo is present only in multiplying cells; this enzyme increased 2.4-fold and 3.1-fold in the decompensated heart, preserving telomeric length in myocytes. The contribution of cycling myocytes to telomerase activity was determined by the colocalization of Ki67 and telomerase in myocyte nuclei. More than 50% of Ki67-positive cells expressed telomerase in the overloaded myocardium, suggesting that these myocytes were the morphological counterpart of the biochemical assay of enzyme activity. Moreover, we report that 20–30% of canine myocytes were telomerase competent, and this value was not changed by cardiac failure. In conclusion, the enhanced expression of Ki67 and telomerase activity, in combination with Ki67-telomerase labeling of myocyte nuclei, support the notion that myocyte proliferation contributes to cardiac hypertrophy of the diseased heart.
- Published
- 2001
46. Myocardial Akt Activation and Gender
- Author
-
Jan Kajstura, Piero Anversa, Erik Schaefer, Angela Walker, Kenneth D. R. Setchell, Isao Shiraishi, Mark A. Sussman, Sara Welch, and Dreama Camper-Kirby
- Subjects
Adult ,Male ,medicine.medical_specialty ,Cell Survival ,Physiology ,medicine.medical_treatment ,Active Transport, Cell Nucleus ,Genistein ,Mice, Inbred Strains ,Stimulation ,Protein Serine-Threonine Kinases ,Biology ,Mice ,chemistry.chemical_compound ,Cytosol ,Sex Factors ,Risk Factors ,Proto-Oncogene Proteins ,Internal medicine ,medicine ,Animals ,Humans ,Insulin-Like Growth Factor I ,Phosphorylation ,Protein kinase A ,Protein kinase B ,Cells, Cultured ,Aged ,Aged, 80 and over ,Cell Nucleus ,Estradiol ,Kinase ,Myocardium ,Growth factor ,Nuclear Proteins ,Forkhead Transcription Factors ,Cell nucleus ,medicine.anatomical_structure ,Endocrinology ,chemistry ,Cardiovascular Diseases ,Female ,Cardiology and Cardiovascular Medicine ,Proto-Oncogene Proteins c-akt ,Subcellular Fractions ,Transcription Factors - Abstract
Abstract —Cardiovascular disease risk is higher in men than women, but the basis for this discrepancy remains controversial. Estrogenic stimulation of the myocardium or isolated cardiomyocytes has been purported to exert multiple beneficial effects associated with inhibition of maladaptive responses to pathogenic insults. This report describes a significant difference between the sexes in myocardial activation of Akt, a protein kinase that regulates a broad range of physiological responses including metabolism, gene transcription, and cell survival. We find that young women possess higher levels of nuclear-localized phospho-Akt 473 relative to comparably aged men or postmenopausal women. Both localization of phospho-Akt 473 in myocardial nuclei of sexually mature female mice versus males and Akt kinase activity in nuclear extracts of hearts from female mice versus males are elevated. Cytosolic localization of phospho-forkhead, a downstream nuclear target of Akt, is also increased in female relative to male mice, suggesting a potential mechanism for cardioprotective nuclear signaling resulting from Akt activation. Phospho-Akt 473 levels and localization at cardiac nuclei are similarly increased in transgenic mice with myocardium-specific expression of insulin-like growth factor I, a proven stimulus for Akt activation. Phospho-Akt 473 is also localized to the nucleus of cultured cardiomyocytes after exposure to 17β-estradiol or genistein (a phytoestrogen in soy protein–based diets), and neonatal exposure of litters to genistein elevated nuclear phospho-Akt 473 localization. The activation of Akt in a gender-dependent manner may help explain differences observed in cardiovascular disease risk between the sexes and supports the potential beneficial effects of estrogenic stimulation.
- Published
- 2001
47. Canine Ventricular Myocytes Possess a Renin-Angiotensin System That Is Upregulated With Heart Failure
- Author
-
Hideo Tada, Annarosa Leri, David E. Dostal, Bernardo Nadal-Ginard, Jan Kajstura, Piero Anversa, Laura Barlucchi, Fabio Fiordaliso, and Thomas H. Hintze
- Subjects
medicine.medical_specialty ,Physiology ,Heart Ventricles ,Blotting, Western ,Cathepsin D ,Peptidyl-Dipeptidase A ,Biology ,Binding, Competitive ,Receptor, Angiotensin, Type 2 ,Receptor, Angiotensin, Type 1 ,Renin-Angiotensin System ,Chymases ,Dogs ,Internal medicine ,Renin ,Renin–angiotensin system ,medicine ,Animals ,Ventricular Function ,Myocyte ,RNA, Messenger ,Receptor ,Heart Failure ,Microscopy, Confocal ,Receptors, Angiotensin ,Reverse Transcriptase Polymerase Chain Reaction ,Angiotensin II ,Serine Endopeptidases ,Cardiac Pacing, Artificial ,Chymase ,medicine.disease ,Immunohistochemistry ,Actins ,Up-Regulation ,Blot ,Endocrinology ,Heart failure ,Tumor Suppressor Protein p53 ,Cardiology and Cardiovascular Medicine ,Protein Binding - Abstract
Abstract —Ventricular pacing leads to a dilated myopathy in which cell death and myocyte hypertrophy predominate. Because angiotensin II (Ang II) stimulates myocyte growth and triggers apoptosis, we tested whether canine myocytes express the components of the renin-angiotensin system (RAS) and whether the local RAS is upregulated with heart failure. p53 modulates transcription of angiotensinogen (Aogen) and AT 1 receptors in myocytes, raising the possibility that enhanced p53 function in the decompensated heart potentiates Ang II synthesis and Ang II–mediated responses. Therefore, the presence of mRNA transcripts for Aogen, renin, angiotensin-converting enzyme, chymase, and AT 1 and AT 2 receptors was evaluated by reverse transcriptase–polymerase chain reaction in myocytes. Changes in the protein expression of these genes were then determined by Western blot in myocytes from control dogs and dogs affected by congestive heart failure. p53 binding to the promoter of Aogen and AT 1 receptor was also determined. Ang II in myocytes was measured by ELISA and by immunocytochemistry and confocal microscopy. Myocytes expressed mRNAs for all the constituents of RAS, and heart failure was characterized by increased p53 DNA binding to Aogen and AT 1 . Additionally, protein levels of Aogen, renin, cathepsin D, angiotensin-converting enzyme, and AT 1 were markedly increased in paced myocytes. Conversely, chymase and AT 2 proteins were not altered. Ang II quantity and labeling of myocytes increased significantly with cardiac decompensation. In conclusion, dog myocytes synthesize Ang II, and activation of p53 function with ventricular pacing upregulates the myocyte RAS and the generation and secretion of Ang II. Ang II may promote myocyte growth and death, contributing to the development of heart failure.
- Published
- 2001
48. Immunohistochemical Analysis of Cardiac Tissue
- Author
-
Barbara Ogorek, Donato Cappetta, and Jan Kajstura
- Subjects
Von Willebrand factor ,biology ,α smooth muscle actin ,Antigen ,business.industry ,biology.protein ,Immunohistochemistry ,Medicine ,business ,Molecular biology - Published
- 2013
49. Inhibition of p53 Function Prevents Renin-Angiotensin System Activation and Stretch-Mediated Myocyte Apoptosis
- Author
-
T. Jan Kajstura, Keith A. Webster, Nanette H. Bishopric, Piero Anversa, Fabio Fiordaliso, Federica Limana, Annarosa Leri, and Manabu Setoguchi
- Subjects
medicine.medical_specialty ,Heart Ventricles ,Blotting, Western ,Angiotensinogen ,Apoptosis ,Endogeny ,Stimulation ,Receptor, Angiotensin, Type 2 ,Receptor, Angiotensin, Type 1 ,Pathology and Forensic Medicine ,Rats, Sprague-Dawley ,Renin-Angiotensin System ,Bcl-2-associated X protein ,Downregulation and upregulation ,Proto-Oncogene Proteins ,Internal medicine ,Renin–angiotensin system ,In Situ Nick-End Labeling ,medicine ,Animals ,Myocyte ,Cells, Cultured ,bcl-2-Associated X Protein ,Microscopy, Confocal ,Receptors, Angiotensin ,biology ,Angiotensin II ,Myocardium ,Genes, p53 ,Rats ,Up-Regulation ,Endocrinology ,Proto-Oncogene Proteins c-bcl-2 ,biology.protein ,Stress, Mechanical ,DNA Probes ,Regular Articles - Abstract
To determine whether stretch-induced activation of p53 is necessary for the up-regulation of the local renin-angiotensin system and angiotensin II (Ang II)-induced apoptosis, ventricular myocytes were infected with an adenoviral vector carrying mutated p53, Adp53m, before 12 hours of stretch. Noninfected myocytes and myocytes infected with AdLacZ served as controls. Stretching of Adp53m-infected myocytes prevented stimulation of p53 function that conditioned the expression of p53-dependent genes; quantity of angiotensinogen (Aogen), AT(1), and Bax decreased, whereas Bcl-2 increased. Ang II generation was not enhanced by stretch. Conversely, stretch produced opposite changes in noninfected and AdLacZ-infected myocytes: Aogen increased twofold, AT(1) increased 2. 1-fold, Bax increased 2.5-fold, and Ang II increased 2.4-fold. These responses were coupled with 4.5-fold up-regulation of wild-type p53. Stretch elicited comparable adaptations in p53-independent genes, in the presence or absence of mutated p53; renin increased threefold, angiotensin-converting enzyme increased ninefold, and AT(2) increased 1.7-fold. Infection with Adp53m inhibited myocyte apoptosis after stretch. Conversely, stretch increased apoptosis by 6.2-fold in myocytes with elevated endogenous wild-type p53. Thus, a competitor of p53 function interfered with both stretch-induced Ang II formation and apoptosis, indicating that p53 is a major modulator of myocyte renin-angiotensin system and cell survival after mechanical deformation.
- Published
- 2000
50. Myocyte Death in Streptozotocin-Induced Diabetes in Rats Is Angiotensin II- Dependent
- Author
-
Baosheng Li, Jan Kajstura, Fabio Fiordaliso, Roberto Latini, Annarosa Leri, Edmund H Sonnenblick, and Piero Anversa
- Subjects
medicine.medical_specialty ,Programmed cell death ,Necrosis ,Apoptosis ,Streptozocin ,Diabetes Mellitus, Experimental ,Pathology and Forensic Medicine ,Internal medicine ,Diabetic cardiomyopathy ,Renin–angiotensin system ,Animals ,Medicine ,Myocyte ,Molecular Biology ,Receptors, Angiotensin ,Angiotensin II receptor type 1 ,business.industry ,Angiotensin II ,Myocardium ,Cell Biology ,Streptozotocin ,medicine.disease ,Rats ,Endocrinology ,medicine.symptom ,business ,medicine.drug - Abstract
To determine whether myocyte death and angiotensin II (AT II) formation are implicated in the development of diabetic cardiomyopathy, rats were injected with streptozotocin, and apoptosis and necrosis were measured at 3, 10, and 28 days. Expression of the components of the renin-angiotensin system (RAS) and AT II levels were assessed at 3 days. The percentage of AT II-labeled myocytes and the number and distribution of AT II sites in myocytes were measured at 3 and 10 days. The effects of AT1 blockade on local RAS and cell death were examined at 3 days. Diabetes was characterized by myocyte apoptosis that peaked at 3 days and decreased at 10 and 28 days, in spite of high concentrations of blood glucose. Cell necrosis was absent throughout. Angiotensinogen, renin, and AT1 receptor increased in myocytes from diabetic rat hearts, while angiotensin-converting enzyme and AT2 remained constant. AT II quantity increased severalfold, as did the fraction of AT II positive cells and the number of AT II sites per myocyte. However, AT II labeling decreased at 10 days, which paralleled the reduction in myocyte death. AT1 antagonist inhibited upregulation of this receptor and angiotensinogen, which prevented AT II synthesis and myocyte death at their peaks with diabetes. An aggregate 30% myocyte loss and a 14% increase in the volume of viable cells were found in diabetic rats at 28 days. Thus diabetic cardiomyopathy may be viewed as an AT II-dependent process in which that peptide plays a critical role in myocyte death and hypertrophy.
- Published
- 2000
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.