251
|
Noguchi K, Miwa Y, Sunohara M, Sato I. Analysis of vascular distribution and growth factors in human gingival tissue associated with periodontal probing depth. Okajimas Folia Anat Jpn 2011; 88:75-83. [PMID: 22184869 DOI: 10.2535/ofaj.88.75] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Vascular endothelial growth factor (VEGF) is a key regulator of blood vessel endothelium. Tissue levels of this angiogenesis marker are unknown in human gingival tissue, as is the correlation between vascular growth factors and hypoxia-inducible factor. We examined the expression of VEGF, type III tyrosine kinase receptors (VEGF-R2), platelet-endothelial cell adhesion molecule (CD31) and hypoxia-inducible factor (HIF) mRNA from human gingival tissue of the oral cavity. Tissue samples were from a small quantity of gingival sample biopsy with gingival sulcular depth (GSD) < 2 mm (Group 1), 2 to 4 mm (Group 2), and > 4 mm (Group 3). We found that the levels of VEGF-R2, CD31 and HIF mRNA were higher in the gingival tissue of Group 2 than that of Group 1, and VEGF in the Group 3 was also higher than that of Group 1. The different mRNA levels of these markers may reflect the mRNA levels reflect the vasculature state of gingival tissue based on GSD. VEGF-R2 and HIF also indicate the presence of an elongated blood vessel in the gingival tissue. In the early stage of angiogenesis, VEGF-R2 leads to expression of VEGF, and HIF-1 mediates increased VEGF expression in response to hypoxia in swollen tissues or during the expansion of periodontal tissues, which is useful in the early diagnosis of periodontal diseases.
Collapse
Affiliation(s)
- Kenzo Noguchi
- Department of Anatomy, School of Life Dentistry at Tokyo, Nippon Dental University, 1-9-20 Fujimi, Chiyoda-ku, Tokyo 102-8159, Japan
| | | | | | | |
Collapse
|
252
|
Forini F, Lionetti V, Ardehali H, Pucci A, Cecchetti F, Ghanefar M, Nicolini G, Ichikawa Y, Nannipieri M, Recchia FA, Iervasi G. Early long-term L-T3 replacement rescues mitochondria and prevents ischemic cardiac remodelling in rats. J Cell Mol Med 2011; 15:514-24. [PMID: 20100314 PMCID: PMC3922373 DOI: 10.1111/j.1582-4934.2010.01014.x] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
3,5,3′-Levo-triiodothyronine (L-T3) is essential for DNA transcription, mitochondrial biogenesis and respiration, but its circulating levels rapidly decrease after myocardial infarction (MI). The main aim of our study was to test whether an early and sustained normalization of L-T3 serum levels after MI exerts myocardial protective effects through a mitochondrial preservation. Seventy-two hours after MI induced by anterior interventricular artery ligation, rats were infused with synthetic L-T3 (1.2 μg/kg/day) or saline over 4 weeks. Compared to saline, L-T3 infusion restored FT3 serum levels at euthyroid state (3.0 ± 0.2 versus 4.2 ± 0.3 pg/ml), improved left ventricular (LV) ejection fraction (39.5 ± 2.5 versus 65.5 ± 6.9%), preserved LV end-systolic wall thickening in the peri-infarct zone (6.34 ± 3.1 versus 33.7 ± 6.21%) and reduced LV infarct-scar size by approximately 50% (all P < 0.05). Moreover, L-T3 significantly increased angiogenesis and cell survival and enhanced the expression of nuclear-encoded transcription factors involved in these processes. Finally, L-T3 significantly increased the expression of factors involved in mitochondrial DNA transcription and biogenesis, such as hypoxic inducible factor-1α, mitochondrial transcription factor A and peroxisome proliferator activated receptor γ coactivator-1α, in the LV peri-infarct zone. To further explore mechanisms of L-T3 protective effects, we exposed isolated neonatal cardiomyocytes to H2O2 and found that L-T3 rescued mitochondrial biogenesis and function and protected against cell death via a mitoKATP dependent pathway. Early and sustained physiological restoration of circulating L-T3 levels after MI halves infarct scar size and prevents the progression towards heart failure. This beneficial effect is likely due to enhanced capillary formation and mitochondrial protection.
Collapse
|
253
|
CXCL8 enhances the angiogenic activity of umbilical cord blood-derived outgrowth endothelial cells in vitro. Cell Biol Int 2011; 35:201-8. [PMID: 20958269 DOI: 10.1042/cbi20090225] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
OECs (outgrowth endothelial cells), also known as late-EPCs (late-endothelial progenitor cells), have a high proliferation potential in addition to in vitro tube formation capability. In ischaemic animal models, injected OECs were integrated into regenerating blood vessels and improved neovascularization. Previous reports have demonstrated the expression of CXCL8 to be up-regulated in ischaemic tissues. It has also been documented that CXCL8 stimulates the angiogenic activity of mature ECs (endothelial cells). Therefore, it has been suggested that CXCL8 plays an important role in neovascularization in ischaemic tissues. However, it is still uncertain whether CXCL8 also stimulates the angiogenic activity of OECs. This study evaluated the effects of CXCL8 on the angiogenic activity of OECs in vitro. OECs were isolated from human UCB (umbilical cord blood)-derived mononuclear cells. Phenotypes of the OECs were assessed by flow cytometry, immunostaining, and real-time RT (reverse transcription)-PCR. The effects of CXCL8 on OECs were investigated by transwell migration assay and capillary tube formation assay on Matrigel. The OEC clones isolated from UCB expressed OEC phenotypes. In addition, CXCL8 receptors (CXCR1 and CXCR2) were expressed on these OEC clones. CXCL8 significantly stimulated the transwell migration and capillary tube formation of OECs. Neutralizing antibody against CXCR2, but not CXCR1, abolished a transwell migration of OECs induced by CXCL8, suggesting the involvement of CXCL8/CXCR2 axis in transwell migration. These results demonstrate that CXCL8 stimulates the angiogenic activity of UCB-derived OECs in vitro.
Collapse
|
254
|
Bonios M, Chang CY, Terrovitis J, Pinheiro A, Barth A, Dong P, Santaularia M, Foster DB, Raman V, Abraham TP, Abraham MR. Constitutive HIF-1α expression blunts the beneficial effects of cardiosphere-derived cell therapy in the heart by altering paracrine factor balance. J Cardiovasc Transl Res 2011; 4:363-72. [PMID: 21538185 PMCID: PMC3104856 DOI: 10.1007/s12265-011-9265-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2010] [Accepted: 02/11/2011] [Indexed: 01/23/2023]
Abstract
Hypoxia-inducible factor-1alpha (HIF-1α) expression promotes angiogenesis and can influence stem cell engraftment. We investigated the effect of stable over-expression of constitutively active HIF-1α on cardiosphere-derived cell (CDC) engraftment and left ventricular function. CDCs were transduced with a lentivirus expressing a constitutively active mutant of human HIF-1α (LVHIF-1α). Two million male rat CDCs were injected into the infarct following ligation of the mid-LAD in female syngeneic rats. Left ventricular ejection fraction (EF) and circumferential strain were measured by echocardiography at 1 and 4 weeks post-MI in the following groups: PBS group (n = 7), CELL group (n = 7), and CELL-HIF group (n = 7). HIF-1α, VEGF, endothelin-1 expression, and CDC engraftment were measured by quantitative PCR. At 30 days, EF was unchanged in the CELL-HIF group (p = NS), increased in the CELL group (p = 0.025), and decreased in the PBS group (p = 0.021), but engraftment was similar (2.4% ± 3.3% vs 1.7% ± 0.8%, p = NS). Mean circumferential strain of the infarcted region was unchanged in the CELL-HIF group, but improved in the CELL group (p = 0.02). Endothelin-1 and VEGF expression were higher in HIF-CDCs exposed to hypoxia, compared with non-transduced CDCs. HIF-1α expression in CDCs blunted the beneficial functional effects of CDC transplantation, suggesting that paracrine factor balance may play an important role in cardiac regeneration.
Collapse
|
255
|
Saygili E, Pekassa M, Saygili E, Rackauskas G, Hommes D, Noor-Ebad F, Gemein C, Zink MDH, Schwinger RHG, Weis J, Marx N, Schauerte P, Rana OR. Mechanical stretch of sympathetic neurons induces VEGF expression via a NGF and CNTF signaling pathway. Biochem Biophys Res Commun 2011; 410:62-7. [PMID: 21640078 DOI: 10.1016/j.bbrc.2011.05.105] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Accepted: 05/17/2011] [Indexed: 10/18/2022]
Abstract
Mechanical stretch has been shown to increase vascular endothelial growth factor (VEGF) expression in cultured myocytes. Sympathetic neurons (SN) also possess the ability to express and secrete VEGF, which is mediated by the NGF/TrkA signaling pathway. Recently, we demonstrated that SN respond to stretch with an upregulation of nerve growth factor (NGF) and ciliary neurotrophic factor (CNTF). Whether stretch increases neuronal VEGF expression still remains to be clarified. Therefore, SN from the superior cervical ganglia of neonatal Sprangue Dawley rats were exposed to a gradual increase of stretch from 3% up to 13% within 3days (3%, 7% and 13%). Under these conditions, the expression and secretion of VEGF was analyzed. Mechanical stretch significantly increased VEGF mRNA and protein expression (mRNA: control=1 vs. stretch=3.1; n=3/protein: control=1 vs. stretch=2.7; n=3). ELISA experiments to asses VEGF content in the cell culture supernatant showed a time and dose dependency in VEGF increment due to stretch. NGF and CNTF neutralization decreased stretch-induced VEGF augmentation in a significant manner. This response was mediated in part by TrkA receptor activation. The stretch-induced VEGF upregulation was accompanied by an increase in HIF-1α expression. KDR levels remained unchanged under conditions of stretch, but showed a significant increase due to NGF neutralization. In summary, SN respond to stretch with an upregulation of VEGF, which is mediated by the NGF/CNTF and TrkA signaling pathway paralleled by HIF-1α expression. NGF signaling seems to play an important role in regulating neuronal KDR expression.
Collapse
Affiliation(s)
- Erol Saygili
- Department of Cardiology, University RWTH Aachen, Aachen, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
256
|
Abstract
This review shall familiarize the readers with various fundamental aspects of angiogenesis. Angiogenesis is a feature of a limited number of physiological processes like wound healing, ovulation, development of the corpus luteum, embryogenesis, lactating breast, during immune response, and during Inflammation. It is driven by a cocktail of growth factors and pro-angiogenic cytokines and is tempered by an equally diverse group of inhibitors of neovascularization. The properties and biological functions of angiogenic growth factors such as VEGF, FGF-2, nitric oxide, MMP, angiopoietin, TGF-β as well as various inhibitors such as angiostatin, endostatin, thrombospondin, canstatin, DII4, PEDF are discussed in this review with respect to their impact on angiogenic process. In recent years, it has become increasingly evident that excessive, insufficient, or abnormal angiogenesis contributes to the pathogenesis of many more disorders. A long list of disorders is characterized or caused by excessive or insufficient angiogenesis whereas several congenital or inherited diseases are also caused by abnormal vascular remodeling. It may be possible in the future to develop specific anti-angiogenic agents that offer a potential therapy for cancer and angiogenic diseases.
Collapse
Affiliation(s)
- Shraddha V Bhadada
- Institute of Pharmacy, Nirma University, Sarkhej-Gandhinagar Highway, Chharodi, Ahmedabad-382 481, Gujarat, India.
| | | | | |
Collapse
|
257
|
Korybalska K, Pyda M, Kawka E, Grajek S, Bręborowicz A, Witowski J. Interpretation of elevated serum VEGF concentrations in patients with myocardial infarction. Cytokine 2011; 54:74-8. [DOI: 10.1016/j.cyto.2011.01.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2010] [Revised: 10/26/2010] [Accepted: 01/04/2011] [Indexed: 11/16/2022]
|
258
|
Santos CX, Anilkumar N, Zhang M, Brewer AC, Shah AM. Redox signaling in cardiac myocytes. Free Radic Biol Med 2011; 50:777-93. [PMID: 21236334 PMCID: PMC3049876 DOI: 10.1016/j.freeradbiomed.2011.01.003] [Citation(s) in RCA: 239] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2010] [Revised: 01/05/2011] [Accepted: 01/05/2011] [Indexed: 02/07/2023]
Abstract
The heart has complex mechanisms that facilitate the maintenance of an oxygen supply-demand balance necessary for its contractile function in response to physiological fluctuations in workload as well as in response to chronic stresses such as hypoxia, ischemia, and overload. Redox-sensitive signaling pathways are centrally involved in many of these homeostatic and stress-response mechanisms. Here, we review the main redox-regulated pathways that are involved in cardiac myocyte excitation-contraction coupling, differentiation, hypertrophy, and stress responses. We discuss specific sources of endogenously generated reactive oxygen species (e.g., mitochondria and NADPH oxidases of the Nox family), the particular pathways and processes that they affect, the role of modulators such as thioredoxin, and the specific molecular mechanisms that are involved-where this knowledge is available. A better understanding of this complex regulatory system may allow the development of more specific therapeutic strategies for heart diseases.
Collapse
Key Words
- aif, apoptosis-inducing factor
- arc, apoptosis repressor with caspase recruitment domain
- camkii, calmodulin kinase ii
- ctgf, connective tissue growth factor
- eb, embryoid body
- ecc, excitation–contraction coupling
- er, endoplasmic reticulum
- es, embryonic stem
- etc, electron transport chain
- g6pdh, glucose-6-phosphate dehydrogenase
- gpcr, g-protein-coupled receptor
- hdac, histone deacetylase
- hif, hypoxia-inducible factor
- mao-a, monoamine oxidase-a
- mi, myocardial infarction
- mmp, matrix metalloproteinase
- mptp, mitochondrial permeability transition pore
- mtdna, mitochondrial dna
- ncx, na/ca exchanger
- nos, nitric oxide synthase
- phd, prolyl hydroxylase dioxygenase
- pka, protein kinase a
- pkc, protein kinase c
- pkg, protein kinase g
- ros, reactive oxygen species
- ryr, ryanodine receptor
- serca, sarcoplasmic reticulum calcium atpase
- sr, sarcoplasmic reticulum
- trx1, thioredoxin1
- tnfα, tumor necrosis factor-α
- vegf, vascular endothelial growth factor
- cardiac myocyte
- reactive oxygen species
- redox signaling
- hypertrophy
- heart failure
- nadph oxidase
- mitochondria
- free radicals
Collapse
|
259
|
Motwani MS, Rafiei Y, Tzifa A, Seifalian AM. In situ endothelialization of intravascular stents from progenitor stem cells coated with nanocomposite and functionalized biomolecules. Biotechnol Appl Biochem 2011; 58:2-13. [DOI: 10.1002/bab.10] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
260
|
Shah AP, Youngquist ST, McClung CD, Tzvetkova E, Hanif MA, Rosborough JP, Niemann JT. Markers of progenitor cell recruitment and differentiation rise early during ischemia and continue during resuscitation in a porcine acute ischemia model. J Interferon Cytokine Res 2011; 31:509-13. [PMID: 21332365 DOI: 10.1089/jir.2010.0133] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Clinical administration of bone marrow-derived stem cells in the setting of acute myocardial infarction (AMI) leads to improved left ventricular ejection fraction. Thymosin beta-4 (TB4) and vascular endothelial growth factor (VEGF) are linked to adult epicardial progenitor cell mobilization and neovascularization and is cardioprotective after myocardial ischemia. This study investigated the time course of TB4 and VEGF during AMI, cardiac arrest, and resuscitation. Fifteen anesthetized and instrumented domestic swine underwent balloon occlusion of the proximal left anterior descending coronary artery. During occlusion, venous blood samples were collected from the right atrium at 5-min intervals until 15 min after the onset of cardiopulmonary resuscitation (CPR). Plasma levels of TB4, VEGF, and matrix metalloproteinase-9 (MMP-9, selected as a marker for remodeling and repair) were measured by ELISA. Generalized linear mixed models were employed to model the time-dependent change in plasma concentration. All variables were natural log transformed, except TB4 values, to normalize distributions. Fifteen animals successfully underwent balloon occlusion of the left anterior descending coronary artery and samples were collected from these subjects. The average onset of spontaneous ventricular fibrillation was 28 min. TB4, VEGF, and MMP-9 demonstrated a statistically significant, time-dependent increase in concentration during ischemia. Following arrest and throughout the first 15 min of resuscitation, MMP-9 had an unchanged rate of rise when compared with the prearrest, ischemic period, with VEGF showing a deceleration in its time-dependent concentration trajectory and TB4 demonstrating an acceleration. Endogenous TB4 and VEGF increase shortly after the onset of AMI and increase through cardiac arrest and resuscitation in parallel to remodeling proteases. These markers continue to rise during successful resuscitation and may represent an endogenous mechanism to recruit undifferentiated stem cells to areas of myocardial injury.
Collapse
Affiliation(s)
- Atman P Shah
- Division of Cardiology, Department of Medicine, University of Chicago, Chicago, IL 60637, USA.
| | | | | | | | | | | | | |
Collapse
|
261
|
|
262
|
Dziankowska-Bartkowiak B, Gerlicz-Kowalczuk Z, Waszczykowska E. Angiogenin and SDF-1α serum concentration in patients with systemic sclerosis in relation to clinical status. Arch Med Sci 2011; 7:92-6. [PMID: 22291739 PMCID: PMC3258685 DOI: 10.5114/aoms.2011.20610] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2009] [Revised: 10/07/2009] [Accepted: 12/12/2009] [Indexed: 01/07/2023] Open
Abstract
INTRODUCTION Systemic sclerosis (SSc) is a connective tissue disorder characterized by tissue hypoxia due to vascular changes and excessive fibrosis of the skin and internal organs. Damage to blood vessels and endothelium, as well as imbalance of vascular homeostasis, impairment of angiogenesis and vasculogenesis are observed in the course of the disease. The aim of the study was to investigate the pro-angiogenic factors angiogenin and SDF-1α in patients with SSc. MATERIAL AND METHODS Serum samples were collected from 50 patients with dSSc (diffuse SSc) and lSSc (limited SSc) and from 38 patients used as a healthy control group. We explored: 1) how the serum concentrations of SDF-1α and angiogenin differ in the investigated groups; 2) the correlation among chemokines in SSc and the duration of the disease, Raynaud's phenomenon, sclerosis of the skin and TSS (total skin score). RESULTS Patients with SSc showed statistically significantly higher serum angiogenin concentration and there was no correlation between duration of the disease and Raynaud's phenomenon, skin sclerosis or TSS. There was also no difference or no correlation between serum level of SDF-1α and the investigated groups. CONCLUSIONS The increase in angiogenin concentration in the serum in patients with SSc may confirm endothelial damage caused by hypoxia and reduced vascular perfusion due to the course of SSc without contributing to compensatory revascularization.
Collapse
|
263
|
Ito T, Tanabe K, Nakamura A, Funamoto K, Aoyagi A, Sato K, Hoshiai T, Suenaga K, Sugawara J, Nagase S, Okamura K, Yaegashi N, Kimura Y. Aberrant Expression of Hypoxia-Inducible Factor 1.ALPHA. in the Fetal Heart Is Associated with Maternal Undernutrition. TOHOKU J EXP MED 2011; 224:163-71. [DOI: 10.1620/tjem.224.163] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Takuya Ito
- Innovation of New Biomedical Engineering Center, Tohoku University
| | - Kaori Tanabe
- International Advanced research and Education Organization, Tohoku University
| | - Ai Nakamura
- International Advanced research and Education Organization, Tohoku University
| | - Kiyoe Funamoto
- International Advanced research and Education Organization, Tohoku University
| | - Ayako Aoyagi
- International Advanced research and Education Organization, Tohoku University
| | - Kazuyo Sato
- Division of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine
| | - Tetsuro Hoshiai
- Division of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine
| | - Kaori Suenaga
- Division of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine
| | - Junichi Sugawara
- Division of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine
| | - Satoru Nagase
- Division of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine
| | | | - Nobuo Yaegashi
- Division of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine
| | - Yoshitaka Kimura
- International Advanced research and Education Organization, Tohoku University
| |
Collapse
|
264
|
Kishore R, Tkebuchava T, Sasi SP, Silver M, Gilbert HY, Yoon YS, Park HY, Thorne T, Losordo DW, Goukassian DA. Tumor necrosis factor-α signaling via TNFR1/p55 is deleterious whereas TNFR2/p75 signaling is protective in adult infarct myocardium. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 691:433-48. [PMID: 21153348 PMCID: PMC3076133 DOI: 10.1007/978-1-4419-6612-4_45] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Raj Kishore
- Cardiovascular Division, Caritas St. Elizabeth’s Medical Center, Tufts University School of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Tengiz Tkebuchava
- Cardiovascular Division, Caritas St. Elizabeth’s Medical Center, Tufts University School of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Sharath P. Sasi
- Cardiovascular Division, Caritas St. Elizabeth’s Medical Center, Tufts University School of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Marcy Silver
- Cardiovascular Division, Caritas St. Elizabeth’s Medical Center, Tufts University School of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Hu-Ya Gilbert
- Cardiovascular Division, Caritas St. Elizabeth’s Medical Center, Tufts University School of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Young-Sup Yoon
- Cardiovascular Division, Caritas St. Elizabeth’s Medical Center, Tufts University School of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Hee-Young Park
- Cardiovascular Division, Caritas St. Elizabeth’s Medical Center, Tufts University School of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Tina Thorne
- Cardiovascular Division, Caritas St. Elizabeth’s Medical Center, Tufts University School of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Douglas W. Losordo
- Cardiovascular Division, Caritas St. Elizabeth’s Medical Center, Tufts University School of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - David A. Goukassian
- Cardiovascular Division, Caritas St. Elizabeth’s Medical Center, Tufts University School of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
265
|
Abstract
The glycemic index (GI) indicates how fast blood glucose is raised after consuming a carbohydrate-containing food. Human metabolic studies indicate that GI is related to patho-physiological responses after meals. Compared with a low-GI meal, a high-GI meal is characterized with hyperglycemia during the early postprandial stage (0-2h) and a compensatory hyperlipidemia associated with counter-regulatory hormone responses during late postprandial stage (4-6h). Over the past three decades, several human health disorders have been related to GI. The strongest relationship suggests that consuming low-GI foods prevents diabetic complications. Diabetic retinopathy (DR) is a complication of diabetes. In this aspect, GI appears to be useful as a practical guideline to help diabetic people choose foods. Abundant epidemiological evidence also indicates positive associations between GI and risk for type 2 diabetes, cardiovascular disease, and more recently, age-related macular degeneration (AMD) in people without diabetes. Although data from randomized controlled intervention trials are scanty, these observations are strongly supported by evolving molecular mechanisms which explain the pathogenesis of hyperglycemia. This wide range of evidence implies that dietary hyperglycemia is etiologically related to human aging and diseases, including DR and AMD. In this context, these diseases can be considered as metabolic retinal diseases. Molecular theories that explain hyperglycemic pathogenesis involve a mitochondria-associated pathway and four glycolysis-associated pathways, including advanced glycation end products formation, protein kinase C activation, polyol pathway, and hexosamine pathway. While the four glycolysis-associated pathways appear to be universal for both normoxic and hypoxic conditions, the mitochondria-associated mechanism appears to be most relevant to the hyperglycemic, normoxic pathogenesis. For diseases that affect tissues with highly active metabolism and that frequently face challenge from low oxygen tension, such as retina in which metabolism is determined by both glucose and oxygen homeostases, these theories appear to be insufficient. Several lines of evidence indicate that the retina is particularly vulnerable when hypoxia coincides with hyperglycemia. We propose a novel hyperglycemic, hypoxia-inducible factor (HIF) pathway, to complement the current theories regarding hyperglycemic pathogenesis. HIF is a transcription complex that responds to decrease oxygen in the cellular environment. In addition to playing a significant role in the regulation of glucose metabolism, under hyperglycemia HIF has been shown to increase the expression of HIF-inducible genes, such as vascular endothelial growth factor (VEGF) leading to angiogenesis. To this extent, we suggest that HIF can also be described as a hyperglycemia-inducible factor. In summary, while management of dietary GI appears to be an effective intervention for the prevention of metabolic diseases, specifically AMD and DR, more interventional data is needed to evaluate the efficacy of GI management. There is an urgent need to develop reliable biomarkers of exposure, surrogate endpoints, as well as susceptibility for GI. These insights would also be helpful in deciphering the detailed hyperglycemia-related biochemical mechanisms for the development of new therapeutic agents.
Collapse
Affiliation(s)
- Chung-Jung Chiu
- Jean Mayer United States Department of Agriculture Human Nutrition Research Center on Aging, Department of Ophthalmology, School of Medicine, Tufts University, 711 Washington Street, Boston, MA 02111, United States.
| | | |
Collapse
|
266
|
Saito Y, Kondo H, Hojo Y. Granzyme B as a novel factor involved in cardiovascular diseases. J Cardiol 2010; 57:141-7. [PMID: 21168312 DOI: 10.1016/j.jjcc.2010.10.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2010] [Revised: 10/05/2010] [Accepted: 10/13/2010] [Indexed: 01/25/2023]
Abstract
Apoptosis plays an important role in cardiovascular diseases such as atherosclerosis, ischemic heart disease, and congestive heart failure. Previous studies have demonstrated that oxidative stress, physiological stress, and inflammatory cytokines such as tumor necrosis factor and Fas ligand are involved in apoptosis of cardiovascular system. We demonstrate that another apoptosis-related pathway, i.e. granzyme B/perforin system is involved in cardiovascular diseases. Expression of granzyme B, a member of serine protease family is increased in acute coronary syndrome, coronary artery disease with end-stage renal disease, and subacute stage of acute myocardial infarction. Although granzyme B is extensively researched in immunological disorders, the role of granzyme B/perforin system was not clear in the cardiovascular field. In addition, little is known regarding the inhibition of granzyme B system in the clinical situation. In this review we demonstrate recent findings of granzyme B in cardiovascular diseases and possible therapeutic applications of inhibiting the granzyme B/perforin system.
Collapse
Affiliation(s)
- Yuji Saito
- Department of Cardiology, Catholic Health System, Buffalo, NY, USA
| | | | | |
Collapse
|
267
|
Wu Y, Zhao RCH, Tredget EE. Concise review: bone marrow-derived stem/progenitor cells in cutaneous repair and regeneration. Stem Cells 2010; 28:905-15. [PMID: 20474078 PMCID: PMC2964514 DOI: 10.1002/stem.420] [Citation(s) in RCA: 194] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Our understanding of the role of bone marrow (BM)-derived cells in cutaneous homeostasis and wound healing had long been limited to the contribution of inflammatory cells. Recent studies, however, suggest that the BM contributes a significant proportion of noninflammatory cells to the skin, which are present primarily in the dermis in fibroblast-like morphology and in the epidermis in a keratinocyte phenotype; and the number of these BM-derived cells increases markedly after wounding. More recently, several studies indicate that mesenchymal stem cells derived from the BM could significantly impact wound healing in diabetic and nondiabetic animals, through cell differentiation and the release of paracrine factors, implying a profound therapeutic potential. This review discusses the most recent understanding of the contribution of BM-derived noninflammatory cells to cutaneous homeostasis and wound healing.
Collapse
Affiliation(s)
- Yaojiong Wu
- Life Science Division, Graduate School at Shenzhen, Tsinghua University, Beijing, People's Republic of China
| | | | | |
Collapse
|
268
|
Heidrich F, Sossalla S, Schotola H, Vorkamp T, Ortmann P, Popov AF, Coskun KO, Rajab TK, Friedrich M, Sohns C, Hinz J, Bauer M, Quintel M, Schöndube FA, Schmitto JD. The Role of Phospho-Adenosine Monophosphate-Activated Protein Kinase and Vascular Endothelial Growth Factor in a Model of Chronic Heart Failure. Artif Organs 2010; 34:969-79. [DOI: 10.1111/j.1525-1594.2010.01121.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
269
|
Miyata T, Takizawa S, van Ypersele de Strihou C. Hypoxia. 1. Intracellular sensors for oxygen and oxidative stress: novel therapeutic targets. Am J Physiol Cell Physiol 2010; 300:C226-31. [PMID: 20980551 DOI: 10.1152/ajpcell.00430.2010] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
A variety of human disorders, e.g., ischemic heart disease, stroke, kidney disease, eventually share the deleterious consequences of a common, hypoxic and oxidative stress pathway. In this review, we utilize recent information on the cellular defense mechanisms against hypoxia and oxidative stress with the hope to propose new therapeutic tools. The hypoxia-inducible factor (HIF) is a key player as it activates a broad range of genes protecting cells against hypoxia. Its level is determined by its degradation rate by intracellular oxygen sensors prolyl hydroxylases (PHDs). There are three different PHD isoforms (PHD1-3). Small molecule PHD inhibitors improve hypoxic injury in experimental animals but, unfortunately, may induce adverse effects associated with PHD2 inhibition, e.g., angiogenesis. As yet, no inhibitor specific for a distinct PHD isoform is currently available. Still, the specific disruption of the PHD1 gene is known to induce hypoxic tolerance, without angiogenesis and erythrocytosis, by reprogramming basal oxygen metabolism with an attendant decreased oxidative stress in hypoxic mitochondria. A specific PHD1 inhibitor might therefore offer a novel therapy against hypoxia. The nuclear factor-erythroid 2 p45-related factor 2 (Nrf2) regulates the basal and inducible expression of numerous antioxidant stress genes. Disruption of its gene exacerbates oxidative tissue injury. Nrf2 activity is modulated by Kelch-like ECH-associated protein 1 (Keap1), an intracellular sensor for oxidative stress. Inhibitors of Keap 1 may prove therapeutic against oxidative tissue injury.
Collapse
Affiliation(s)
- Toshio Miyata
- United Centers for Advanced Research and Translational Medicine, Tohoku University School of Medicine, 2-1 Seiryo-Machi, Aoba-ku, Sendai, 980-8575, Japan.
| | | | | |
Collapse
|
270
|
Moslehi J, Minamishima YA, Shi J, Neuberg D, Charytan D, Padera RF, Signoretti S, Liao R, Kaelin WG. Loss of hypoxia-inducible factor prolyl hydroxylase activity in cardiomyocytes phenocopies ischemic cardiomyopathy. Circulation 2010; 122:1004-16. [PMID: 20733101 PMCID: PMC2971656 DOI: 10.1161/circulationaha.109.922427] [Citation(s) in RCA: 136] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
BACKGROUND Ischemic cardiomyopathy is the major cause of heart failure and a significant cause of morbidity and mortality. The degree of left ventricular dysfunction in this setting is often out of proportion to the amount of overtly infarcted tissue, and how decreased delivery of oxygen and nutrients leads to impaired contractility remains incompletely understood. The Prolyl Hydroxylase Domain-Containing Protein (PHD) prolyl hydroxylases are oxygen-sensitive enzymes that transduce changes in oxygen availability into changes in the stability of the hypoxia-inducible factor transcription factor, a master regulator of genes that promote survival in a low-oxygen environment. METHODS AND RESULTS We found that cardiac-specific PHD inactivation causes ultrastructural, histological, and functional changes reminiscent of ischemic cardiomyopathy over time. Moreover, long-term expression of a stabilized hypoxia-inducible factor alpha variant in cardiomyocytes also led to dilated cardiomyopathy. CONCLUSIONS Sustained loss of PHD activity and subsequent hypoxia-inducible factor activation, as would occur in the setting of chronic ischemia, are sufficient to account for many of the changes in the hearts of individuals with chronic coronary artery disease.
Collapse
Affiliation(s)
- Javid Moslehi
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115
| | - Yoji Andrew Minamishima
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115
| | - Jianru Shi
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115
| | - Donna Neuberg
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute and Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115
| | - David Charytan
- Division of Nephrology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115
| | - Robert F. Padera
- Department of Medicine Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA,02115
| | - Sabina Signoretti
- Department of Medicine Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA,02115
| | - Ronglih Liao
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115
| | - William G. Kaelin
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115
- Howard Hughes Medical Institute, Chevy Chase, MD, 20815
| |
Collapse
|
271
|
Abstract
Since its discovery in early 1990s, hypoxia inducible factor 1 (HIF-1) has been increasingly recognized for its key role in transcriptional control of more than a hundred genes that regulate a wide-spectrum of cellular functional events, including angiogenesis, vasomotor control, glucose and energy metabolism, erythropoiesis, iron homeostasis, pH regulation, cell proliferation and viability. Evidence accumulated during the past 7 years suggests a critical role for HIF-1alpha in mediating cardioprotection. The purpose of our present article is to provide an updated overview on this important regulator of gene expression in the cellular stress-responsive and adaptive process. We have particularly emphasized the involvement of HIF-1 in the induction of cardioprotective molecules, such as inducible nitric oxide synthase (iNOS), hemeoxygenase 1 (HO-1), and erythropoietin (EPO), which in turn alleviate myocardial damages caused by harmful events such as ischemia-reperfusion injury. Despite these advances, further in-depth studies are needed to elucidate the possible coordination or interaction between HIF-1alpha and other key transcription factors in regulating protein expression that leads to cardioprotection.
Collapse
|
272
|
Fam NP, Arab S, Billia F, Han R, Proteau G, Latter D, Errett L, Bonneau D, Dunne R, Liu PP, Stewart DJ. Increased myocardial expression of angiopoietin-2 in patients undergoing urgent surgical revascularization for acute coronary syndromes. Can J Cardiol 2010; 26:365-70. [PMID: 20847963 PMCID: PMC2950740 DOI: 10.1016/s0828-282x(10)70417-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2008] [Accepted: 01/12/2009] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Myocardial ischemia triggers the expression of multiple angiogenic factors including vascular endothelial growth factor and its receptors. However, vascular endothelial growth factor does not act in isolation. OBJECTIVE To identify other genes important in the angiogenic response to clinically relevant myocardial ischemia. METHODS AND RESULTS Paired intraoperative biopsies of ischemic and nonischemic myocardium were obtained from 12 patients with acute coronary syndromes (ACS) undergoing urgent coronary artery bypass graft surgery. Real-time polymerase chain reaction demonstrated significant upregulation of angiopoietin-2 (Ang-2) in ischemic myocardium, to a greater extent than other classical angiogenic factors. Microarray gene profiling identified Ang-2 to be among the top 10 differentially upregulated genes, in addition to genes involved in inflammation, cell signalling, remodelling and apoptosis. CONCLUSIONS The present document is the first report of microarray analysis of patients with ACS, and supports an important role for Ang-2 in the angiogenic response to severe ischemia in the human heart. Common gene expression patterns in ACS may provide opportunities for targeted pharmacological and cellular intervention.
Collapse
Affiliation(s)
- Neil P Fam
- Terrence Donnelly Heart Centre, St Michael’s Hospital
| | - Sara Arab
- University Health Network
- Heart and Stroke/Richard Lewar Centre, University of Toronto, Toronto
| | - Filio Billia
- Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario
| | - Robin Han
- Terrence Donnelly Heart Centre, St Michael’s Hospital
| | | | - David Latter
- Terrence Donnelly Heart Centre, St Michael’s Hospital
| | - Lee Errett
- Terrence Donnelly Heart Centre, St Michael’s Hospital
| | | | | | - Peter P Liu
- University Health Network
- Heart and Stroke/Richard Lewar Centre, University of Toronto, Toronto
| | - Duncan J Stewart
- Terrence Donnelly Heart Centre, St Michael’s Hospital
- McLaughlin Centre for Molecular Medicine
- Heart and Stroke/Richard Lewar Centre, University of Toronto, Toronto
- Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario
| |
Collapse
|
273
|
Ogi H, Nakano Y, Niida S, Dote K, Hirai Y, Suenari K, Tonouchi Y, Oda N, Makita Y, Ueda S, Kajihara K, Imai K, Sueda T, Chayama K, Kihara Y. Is structural remodeling of fibrillated atria the consequence of tissue hypoxia? Circ J 2010; 74:1815-21. [PMID: 20631454 DOI: 10.1253/circj.cj-09-0969] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Matrix metalloproteinases (MMPs) play an important role in degradation of the extracellular matrix of injured tissue. MMP-9 expression increases in fibrillating atrial tissue; however, the mechanism for this increase has not been clarified. METHODS AND RESULTS Changes in the expression of vascular endothelial growth factor (VEGF), VEGF receptors, and hypoxia-induced transcription factor-1alpha (HIF-1alpha) in fibrillating atrial tissue were investigated. Atrial tissue samples were obtained from 13 patients with atrial fibrillation (AF) and 25 patients without a history of AF (regular sinus rhythm, RSR) undergoing cardiac operations. Western blot, real-time polymerase chain reaction, and immunofluorescence analyses of the expression of VEGF, VEGF receptors, and HIF-1alpha were performed. The VEGF mRNA and protein levels increased significantly in the AF group compared with the RSR group (P<0.05), and the expression of HIF-1alpha protein was also significantly higher in the AF group. VEGF receptor-1 mRNA, a high-affinity receptor for VEGF, but not VEGF receptor-2 mRNA, was upregulated in the atria of the AF group (P<0.05). Immunofluorescence staining revealed excess production and co-localization of HIF-1alpha, VEGF and MMP-9 in the endothelium of the atrial arteries in the AF group. CONCLUSIONS It is possible that upregulation of HIF-1/VEGF is involved in the enhancement of MMP-9 expression under hypoxic conditions.
Collapse
Affiliation(s)
- Hiroshi Ogi
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical Science, Hiroshima, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
274
|
Complex role of the HIF system in cardiovascular biology. J Mol Med (Berl) 2010; 88:1101-11. [DOI: 10.1007/s00109-010-0646-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2010] [Revised: 05/17/2010] [Accepted: 06/11/2010] [Indexed: 12/18/2022]
|
275
|
Cardiomyocyte-specific inactivation of thyroid hormone in pathologic ventricular hypertrophy: an adaptative response or part of the problem? Heart Fail Rev 2010; 15:133-42. [PMID: 19107595 PMCID: PMC2820687 DOI: 10.1007/s10741-008-9133-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Recent studies in various rodent models of pathologic ventricular hypertrophy report the re-expression of deiodinase type 3 (D3) in cardiomyocytes. D3 inactivates thyroid hormone (T3) and is mainly expressed in tissues during development. The stimulation of D3 activity in ventricular hypertrophy and subsequent heart failure is associated with severe impairment of cardiac T3 signaling. Hypoxia-induced signaling appears to drive D3 expression in the hypertrophic cardiomyocyte, but other signaling cascades implicated in hypertrophy are also capable of stimulating transcription of the DIO3 gene. Many cardiac genes are transcriptionally regulated by T3 and impairment of T3 signaling will not only reduce energy turnover, but also lead to changes in gene expression that contribute to contractile dysfunction in pathologic remodeling. Whether stimulation of D3 activity and the ensuing local T3-deficiency is an adaptive response of the stressed heart or part of the pathologic signaling network leading to heart failure, remains to be established.
Collapse
|
276
|
Majumder S, Ilayaraja M, Seerapu HR, Sinha S, Siamwala JH, Chatterjee S. Chick embryo partial ischemia model: a new approach to study ischemia ex vivo. PLoS One 2010; 5:e10524. [PMID: 20479865 PMCID: PMC2866318 DOI: 10.1371/journal.pone.0010524] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2009] [Accepted: 04/16/2010] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Ischemia is a pathophysiological condition due to blockade in blood supply to a specific tissue thus damaging the physiological activity of the tissue. Different in vivo models are presently available to study ischemia in heart and other tissues. However, no ex vivo ischemia model has been available to date for routine ischemia research and for faster screening of anti-ischemia drugs. In the present study, we took the opportunity to develop an ex vivo model of partial ischemia using the vascular bed of 4(th) day incubated chick embryo. METHODOLOGY/PRINCIPAL FINDINGS Ischemia was created in chick embryo by ligating the right vitelline artery using sterile surgical suture. Hypoxia inducible factor- 1 alpha (HIF-1alpha), creatine phospho kinase-MB and reactive oxygen species in animal tissues and cells were measured to confirm ischemia in chick embryo. Additionally, ranolazine, N-acetyl cysteine and trimetazidine were administered as an anti-ischemic drug to validate the present model. Results from the present study depicted that blocking blood flow elevates HIF-1alpha, lipid peroxidation, peroxynitrite level in ischemic vessels while ranolazine administration partially attenuates ischemia driven HIF-1alpha expression. Endothelial cell incubated on ischemic blood vessels elucidated a higher level of HIF-1alpha expression with time while ranolazine treatment reduced HIF-1alpha in ischemic cells. Incubation of caprine heart strip on chick embryo ischemia model depicted an elevated creatine phospho kinase-MB activity under ischemic condition while histology of the treated heart sections evoked edema and disruption of myofibril structures. CONCLUSIONS/SIGNIFICANCE The present study concluded that chick embryo partial ischemia model can be used as a novel ex vivo model of ischemia. Therefore, the present model can be used parallel with the known in vivo ischemia models in understanding the mechanistic insight of ischemia development and in evaluating the activity of anti-ischemic drug.
Collapse
Affiliation(s)
- Syamantak Majumder
- Vascular Biology Lab, AU-KBC Research Centre, Anna University, Chennai, India
| | - M. Ilayaraja
- Vascular Biology Lab, AU-KBC Research Centre, Anna University, Chennai, India
| | | | - Swaraj Sinha
- Vascular Biology Lab, AU-KBC Research Centre, Anna University, Chennai, India
| | - Jamila H. Siamwala
- Vascular Biology Lab, AU-KBC Research Centre, Anna University, Chennai, India
| | - Suvro Chatterjee
- Vascular Biology Lab, AU-KBC Research Centre, Anna University, Chennai, India
- * E-mail:
| |
Collapse
|
277
|
Devaux Y, Azuaje F, Vausort M, Yvorra C, Wagner DR. Integrated protein network and microarray analysis to identify potential biomarkers after myocardial infarction. Funct Integr Genomics 2010; 10:329-37. [PMID: 20414696 DOI: 10.1007/s10142-010-0169-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2010] [Revised: 03/22/2010] [Accepted: 03/28/2010] [Indexed: 01/19/2023]
Abstract
A significant proportion of patients develop left ventricular (LV) dysfunction and heart failure (HF) after acute myocardial infarction (MI). Existing biomarkers of HF provide limited information after MI. To identify new prognostic biomarkers in MI patients, we designed an approach combining protein interaction networks and microarray analysis of blood cells. Blood samples for RNA and protein analysis were taken from 127 acute MI patients. Echocardiography was performed at one month. Assuming that angiogenesis is related to cardiac repair after MI, a protein-protein interaction network of angiogenesis was constructed and analyzed. Among the 556 proteins and 686 interactions of this network, a cluster of 53 proteins highly specialized in regulation of cell growth was identified. Of these 53 proteins, 38 were found differentially expressed by microarrays between low (< or = 40%) and high (>40%) LV ejection fraction (EF) patients (n = 32). Among these 38 genes, prediction analysis identified a set of three genes able to predict significant LV dysfunction (EF < or = 40%) with an area under the receiver operating characteristic curve (AUC) of 0.82. These three genes-vascular endothelial growth factor B, thrombospondin-1 and placental growth factor-had a stronger predictive value than brain natriuretic peptide and troponin T (AUC of 0.63). Independent validations on protein expression and quantitative PCR datasets confirmed the results. In conclusion, a new strategy is described that allows identifying new potential biomarkers. The three specific biomarkers described here remain to be validated in a larger patient population.
Collapse
Affiliation(s)
- Yvan Devaux
- Laboratory of Cardiovascular Research, Centre de Recherche Public-Santé, 120 route d'Arlon, 1150, Luxembourg, Luxembourg.
| | | | | | | | | |
Collapse
|
278
|
Abstract
Blood vessels function as conduits for the delivery of O(2) and nutrients. Hypoxia-inducible factor 1 (HIF-1) mediates adaptive transcriptional responses to hypoxia/ischemia that include expression of angiogenic cytokines/growth factors by hypoxic cells and expression of cognate receptors for these ligands by vascular cells and their progenitors. Impairment of HIF-1-dependent responses to hypoxia is a major factor contributing to the impaired vascular responses to ischemia that are associated with aging and diabetes.
Collapse
Affiliation(s)
- Gregg L Semenza
- Institute for Cell Engineering, Departments of Pediatrics, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
279
|
Affiliation(s)
- John D Scott
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA 98195, USA.
| | | |
Collapse
|
280
|
Probst-Cousin S, Neundörfer B, Heuss D. Microvasculopathic neuromuscular diseases: Lessons from hypoxia-inducible factors. Neuromuscul Disord 2010; 20:192-7. [DOI: 10.1016/j.nmd.2010.01.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2009] [Revised: 12/21/2009] [Accepted: 01/07/2010] [Indexed: 10/19/2022]
|
281
|
Differential DNA methylation correlates with differential expression of angiogenic factors in human heart failure. PLoS One 2010; 5:e8564. [PMID: 20084101 PMCID: PMC2797324 DOI: 10.1371/journal.pone.0008564] [Citation(s) in RCA: 154] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2009] [Accepted: 11/30/2009] [Indexed: 11/19/2022] Open
Abstract
Epigenetic mechanisms such as microRNA and histone modification are crucially responsible for dysregulated gene expression in heart failure. In contrast, the role of DNA methylation, another well-characterized epigenetic mark, is unknown. In order to examine whether human cardiomyopathy of different etiologies are connected by a unifying pattern of DNA methylation pattern, we undertook profiling with ischaemic and idiopathic end-stage cardiomyopathic left ventricular (LV) explants from patients who had undergone cardiac transplantation compared to normal control. We performed a preliminary analysis using methylated-DNA immunoprecipitation-chip (MeDIP-chip), validated differential methylation loci by bisulfite-(BS) PCR and high throughput sequencing, and identified 3 angiogenesis-related genetic loci that were differentially methylated. Using quantitative RT-PCR, we found that the expression of these genes differed significantly between CM hearts and normal control (p<0.01). Moreover, for each individual LV tissue, differential methylation showed a predicted correlation to differential expression of the corresponding gene. Thus, differential DNA methylation exists in human cardiomyopathy. In this series of heterogenous cardiomyopathic LV explants, differential DNA methylation was found in at least 3 angiogenesis-related genes. While in other systems, changes in DNA methylation at specific genomic loci usually precede changes in the expression of corresponding genes, our current findings in cardiomyopathy merit further investigation to determine whether DNA methylation changes play a causative role in the progression of heart failure.
Collapse
|
282
|
Abstract
Molecular imaging is a new and evolving field that employs a targeted approach to noninvasively assess biologic processes in vivo. By assessing key elements in specific cellular processes prior to irreversible end-organ damage, molecular tools will allow for earlier detection and intervention, improving management and outcomes associated with cardiovascular diseases. The goal of those working to expand this field is not just to provide diagnostic and prognostic information, but rather to guide an individual's pharmacological, cell-based, or genetic therapeutic regimen. This article will review molecular imaging tools in the context of our current understanding of biological processes of the myocardium, including angiogenesis, ventricular remodeling, inflammation, and apoptosis. The focus will be on radiotracer-based molecular imaging modalities with an emphasis on clinical application. Though this field is still in its infancy and may not be fully ready for widespread use, molecular imaging of myocardial biology has begun to show promise of clinical utility in acute and chronic ischemia, acute myocardial infarction, congestive heart failure, as well as in more global inflammatory and immune-mediated responses in the heart-like myocarditis and allogeneic cardiac transplant rejection. With continued research and development, molecular imaging promises to be an important tool for the optimization of cardiovascular care.
Collapse
Affiliation(s)
- Alan R. Morrison
- Section of Cardiovascular Medicine, Department of Medicine, Yale University School of Medicine, New Haven, CT
| | - Albert J. Sinusas
- Section of Cardiovascular Medicine, Department of Medicine, Yale University School of Medicine, New Haven, CT
- Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
283
|
Samuel SM, Akita Y, Paul D, Thirunavukkarasu M, Zhan L, Sudhakaran PR, Li C, Maulik N. Coadministration of adenoviral vascular endothelial growth factor and angiopoietin-1 enhances vascularization and reduces ventricular remodeling in the infarcted myocardium of type 1 diabetic rats. Diabetes 2010; 59:51-60. [PMID: 19794062 PMCID: PMC2797944 DOI: 10.2337/db09-0336] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Hyperglycemia impairs angiogenesis in response to ischemia, leading to ventricular remodeling. Although the effects of overexpressing angiogenic growth factors have been studied in inducing angiogenesis, the formation of functional vessels remains a challenge. The present study evaluates the reversal of diabetes-mediated impairment of angiogenesis in the infarcted diabetic rat myocardium by proangiogenic gene therapy. RESEARCH DESIGN AND METHODS Ad*VEGF and Ad*Ang1 were intramyocardially administered in combination immediately after myocardial infarction to nondiabetic and diabetic rats. Ad*LacZ was similarly administered to the respective control groups. The hearts were excised for molecular and immunohistochemical analysis at predetermined time points. The myocardial function was measured by echocardiography 30 days after the intervention. RESULTS We observed reduced fibrosis and increased capillary/arteriolar density along with reduced ventricular remodeling, as assessed by echocardiography in the treated diabetic animals compared with the nontreated diabetic controls. We also observed increased phosphorylated mitogen-activated protein kinase-activated protein kinase-2, 2 days after the treatment and increased expression of vascular endothelial growth factor (VEGF), Flk-1, angiopoietin-1 (Ang-1), Tie-2, and survivin, 4 days after treatment in the diabetic animals. Gel shift analysis revealed that the combination gene therapy stimulated the DNA binding activity of nuclear factor-kappaB in the diabetic animals. CONCLUSIONS Our preclinical data demonstrate the efficacy of coadministration of adenoviral VEGF and Ang-1 in increasing angiogenesis and reducing ventricular remodeling in the infarcted diabetic myocardium. These unique results call for the initiation of a clinical trial to assess the efficacy of this therapeutic strategy in the treatment of diabetes-related human heart failure.
Collapse
Affiliation(s)
- Samson Mathews Samuel
- Molecular Cardiology and Angiogenesis Laboratory, Department of Surgery, University of Connecticut Health Center, Farmington, Connecticut
- Department of Biochemistry, University of Kerala, Trivandrum, Kerala, India
| | - Yuzo Akita
- Molecular Cardiology and Angiogenesis Laboratory, Department of Surgery, University of Connecticut Health Center, Farmington, Connecticut
| | - Debayon Paul
- Molecular Cardiology and Angiogenesis Laboratory, Department of Surgery, University of Connecticut Health Center, Farmington, Connecticut
| | - Mahesh Thirunavukkarasu
- Molecular Cardiology and Angiogenesis Laboratory, Department of Surgery, University of Connecticut Health Center, Farmington, Connecticut
| | - Lijun Zhan
- Molecular Cardiology and Angiogenesis Laboratory, Department of Surgery, University of Connecticut Health Center, Farmington, Connecticut
| | | | - Chuanfu Li
- Department of Surgery, East Tennessee State University, Johnson City, Tennessee
| | - Nilanjana Maulik
- Molecular Cardiology and Angiogenesis Laboratory, Department of Surgery, University of Connecticut Health Center, Farmington, Connecticut
- Corresponding author: Nilanjana Maulik,
| |
Collapse
|
284
|
Fuster MM, Wang L. Endothelial heparan sulfate in angiogenesis. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2010; 93:179-212. [PMID: 20807646 DOI: 10.1016/s1877-1173(10)93009-3] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Heparan sulfate (HS) is a linear polysaccharide composed of 50-200 glucosamine and uronic acid (glucuronic acid or iduronic acid) disaccharide repeats with epimerization and various sulfation modifications. HS is covalently attached to core proteins to form HS-proteoglycans. Most of the functions of HS-proteoglycans are mediated by their HS moieties. The biosynthesis of HS is initiated by chain polymerization and is followed by stepwise modification reactions, including sulfation and epimerization. These modifications generate ligand-binding sites that modulate cell functions and activities of proteinases and/or proteinase inhibitors. HS is abundantly expressed in developing and mature vasculature, and understanding its roles in vascular biology and related human diseases is an area of intense investigation. In this chapter, we summarize the significant recent advances in our understanding of the roles of HS in developmental and pathological angiogenesis with a major focus on studies using transgenic as well as gene knockout/knockdown models in mice and zebrafish. These studies have revealed that HS critically regulates angiogenesis by playing a proangiogenic role, and this regulatory function critically depends on HS fine structure. The latter is responsible for facilitating cell-surface binding of various proangiogenic growth factors that in turn mediate endothelial growth signaling. In cancer, mouse studies have revealed important roles for endothelial cell-surface HS as well as matrix-associated HS, wherein signaling by multiple growth factors as well as matrix storage of growth factors may be regulated by HS. We also discuss important mediators that may fine-tune such regulation, such as heparanase and sulfatases; and models wherein targeting HS (or core protein) biosynthesis may affect tumor growth and vascularization. Finally, the importance of targeting HS in other human diseases wherein angiogenesis may play pathophysiologic (or even therapeutic) roles is considered.
Collapse
Affiliation(s)
- Mark M Fuster
- Department of Medicine, Division of Pulmonary and Critical Care, University of California San Diego, La Jolla, California, USA
| | | |
Collapse
|
285
|
Nesselmann C, Li W, Ma N, Steinhoff G. Stem cell-mediated neovascularization in heart repair. Ther Adv Cardiovasc Dis 2009; 4:27-42. [PMID: 20042449 DOI: 10.1177/1753944709353338] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Accumulating clinical and experimental evidence indicates that stem cells from various sources are promising in the treatment of cardiac dysfunction. They may be incorporated into neovascular foci and thus contribute to postnatal physiological and pathological vasculogenesis and/or produce a variety of growth factors for angiogenesis and cytokines that home other stem cells from other organs for cardiac regeneration. This review focuses on the neovascularization of stem cells from different sources in cardiac repair, with emphasis on adult stem cells.
Collapse
|
286
|
Gyöngyösi M, Hemetsberger R, Posa A, Charwat S, Pavo N, Petnehazy O, Petrasi Z, Pavo IJ, Hemetsberger H, Benedek I, Benedek T, Benedek I, Kovacs I, Kaun C, Maurer G. Hypoxia-inducible factor 1-alpha release after intracoronary versus intramyocardial stem cell therapy in myocardial infarction. J Cardiovasc Transl Res 2009; 3:114-21. [PMID: 20560024 DOI: 10.1007/s12265-009-9154-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2009] [Accepted: 11/06/2009] [Indexed: 01/06/2023]
Abstract
We have investigated the effect of stem cell delivery on the release of hypoxia-inducible factor 1 alpha (HIF-1alpha) in peripheral circulation and myocardium in experimental myocardial ischemia. Closed-chest, reperfused myocardial infarction (MI) was created in domestic pigs. Porcine mesenchymal stem cells (MSCs) were cultured and delivered (9.8 +/- 1.2 x 10(6)) either percutaneously NOGA-guided transendocardially (Group IM) or intracoronary (Group IC) 22 +/- 4 days post-MI. Pigs without MSC delivery served as sham control (Group S). Plasma HIF-1alpha was measured at baseline, immediately post- and at follow-up (FUP; 2 h or 24 h) post-MSC delivery by ELISA kit. Myocardial HIF-1alpha expression of infarcted, normal myocardium, or border zone was determined by Western blot. Plasma level of HIF-1alpha increased immediately post-MI (from 278 +/- 127 to 631 +/- 375 pg/ml, p < 0.05). Cardiac delivery of MSCs elevated the plasma levels of HIF-1alpha significantly (p < 0.05) in groups IC and IM immediately post-MSC delivery, and returned to baseline level at FUP, without difference between the groups IC and IM. The myocardial tissue HIF-1alpha expression in the infarcted area was higher in Group IM than in Group IC or S (1,963 +/- 586 vs. 1,307 +/- 392 vs. 271 +/- 110 activity per square millimeter, respectively, p < 0.05), while the border zone contained similarly lower level of HIF-1alpha, but still significantly higher as compared with Group S. Trend towards increase in myocardial expression of HIF-1alpha was measured in Group IM at 24 h, in contrast to Group IC. In conclusion, both stem cell delivery modes increase the systemic and myocardial level of HIF-1alpha. Intramyocardial delivery of MSC seems to trigger the release of angiogenic HIF-1alpha more effectively than does intracoronary delivery.
Collapse
Affiliation(s)
- Mariann Gyöngyösi
- Department of Cardiology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
287
|
Vajanto I, Korpisalo P, Karjalainen J, Hakala T, Mäkinen K, Ylä-Herttuala S. Antegrade flow and peripheral resistance determine the level of endogenous arteriogenesis in patients with superficial femoral artery occlusion. Eur J Clin Invest 2009; 39:1048-54. [PMID: 19807785 DOI: 10.1111/j.1365-2362.2009.02208.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OBJECTIVES Occlusion in a limb artery leads to impaired blood supply and ischaemia. Collateral artery growth (arteriogenesis) is one of the most effective natural response mechanisms to compensate this pathologic situation. However, it is unknown if clinically important features, like poor run-off, have an impact on compensatory vessel growth. METHODS Study population of this retrospective study consisted of 70 patients who suffered from lower limb ischaemia and underwent bypass surgery because of an occlusion of the superficial femoral artery. Clinical data were collected and pre- and postoperative angiograms were reviewed. Number of collateral vessels bypassing the occluded segment was counted. Features of inflow and outflow vessels were recorded. RESULTS The mean number of collaterals was 13 + or - 0.5 per patient. In univariate analysis, short daily walking distance, chronic critical leg ischaemia, low ankle brachial index, low number of patent calf arteries and stenosed inflow arteries predicted low number of collateral arteries. In the multivariate analysis, only the quality of inflow and the number of patent calf vessels demonstrated an independent association (P < 0.05) with the number of collaterals. CONCLUSIONS Ankle-brachial index, grade of symptoms and daily walking capacity could be used to predict collateral density. Importantly, a good antegrade flow and peripheral runoff seem to have a significant effect on collateral density, implying an impact on the activation of arteriogenesis.
Collapse
Affiliation(s)
- I Vajanto
- Department of Cardiothoracic Surgery, Kuopio University Hospital, Kuopio, Finland
| | | | | | | | | | | |
Collapse
|
288
|
|
289
|
van Rooij E, Fielitz J, Sutherland LB, Thijssen VL, Crijns HJ, Dimaio MJ, Shelton J, De Windt LJ, Hill JA, Olson EN. Myocyte enhancer factor 2 and class II histone deacetylases control a gender-specific pathway of cardioprotection mediated by the estrogen receptor. Circ Res 2009; 106:155-65. [PMID: 19893013 DOI: 10.1161/circresaha.109.207084] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
RATIONALE Gender differences in cardiovascular disease have long been recognized and attributed to beneficial cardiovascular actions of estrogen. Class II histone deacetylases (HDACs) act as key modulators of heart disease by repressing the activity of the myocyte enhancer factor (MEF)2 transcription factor, which promotes pathological cardiac remodeling in response to stress. Although it is proposed that HDACs additionally influence nuclear receptor signaling, the effect of class II HDACs on gender differences in cardiovascular disease remains unstudied. OBJECTIVE We aimed to examine the effect of class II HDACs on post-myocardial infarction remodeling in male and female mice. METHODS AND RESULTS Here we show that the absence of HDAC5 or -9 in female mice protects against maladaptive remodeling following myocardial infarction, during which there is an upregulation of estrogen-responsive genes in the heart. This genetic reprogramming coincides with a pronounced increase in expression of the estrogen receptor (ER)alpha gene, which we show to be a direct MEF2 target gene. ERalpha also directly interacts with class II HDACs. Cardioprotection resulting from the absence of HDAC5 or -9 in female mice can be attributed, at least in part, to enhanced neoangiogenesis in the infarcted region via upregulation of the ER target gene vascular endothelial growth factor-a. CONCLUSIONS Our results reveal a novel gender-specific pathway of cardioprotection mediated by ERalpha and its regulation by MEF2 and class II HDACs.
Collapse
Affiliation(s)
- Eva van Rooij
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9148, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
290
|
Brehm M, Picard F, Ebner P, Turan G, Bölke E, Köstering M, Schüller P, Fleissner T, Ilousis D, Augusta K, Peiper M, Schannwell C, Strauer BE. Effects of exercise training on mobilization and functional activity of blood-derived progenitor cells in patients with acute myocardial infarction. Eur J Med Res 2009; 14:393-405. [PMID: 19748858 PMCID: PMC3351971 DOI: 10.1186/2047-783x-14-9-393] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The aim of the present study was to determine whether regular exercise training (ET) is effective at promoting the mobilization of CPCs and improving their functional activity in patients with recently acquired myocardial infarction (STEMI). Regular physical training has been shown to improve myocardial perfusion and cardiovascular function. This may be related in part to a mobilization of bone marrow-derived circulating progenitor cells (CPCs) as well as an enhanced vascularisation. METHODS 37 patients with STEMI were randomly assigned to an ET group or a non-ET group (controls). Two weeks after STEMI, three weeks after regular ET and three months after ET, BNP levels, exercise echocardiography and exercise spiroergometry were evaluated. The number of CD34+/CD45+ and CD133+/CD45+ CPCs was measured by flow cytometry analysis. The migration capacity of the CPCs was determined with a boyden chamber and the clonogenic capacity by CFU-assay. RESULTS In the ET-group the number and migration capacity of CPCs increased significantly after regular exercise training. The BNP level decreased significantly from 121 +/- 94 to 75 +/- 47 pg/ml (p<0.001) after the ET period, the left ventricular ejection fraction raised in parallel at peak exercise, and the cardiorespiratory condition improved as demonstrated by an increase of VO2max (from 1641 +/- 522 to 1842 +/- 724 ml/min, p<0.02). These three effects persist till three months after the ET period. CONCLUSIONS Regular physical activity appears to predispose the mobilization and enhanced functional activity of CPCs, a phenomenon which might lead to an improved cardiac function in patients with recently acquired acute myocardial infarction.
Collapse
Affiliation(s)
- Michael Brehm
- Department of Internal Medicine, Division of Cardiology, Pneumology and Angiology, Heinrich-Heine-University, Moorenstr. 5, 40 225 Düsseldorf, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
291
|
Abstract
The success of liver transplantation has led to an ever-increasing demand for liver grafts. Since the first successful living donor liver transplantation, this surgical innovation has been well established in children and has significantly relieved the crisis of donor organ shortage for children. However, the extension of living donor liver transplantation to adult recipients is limited by the graft volume. The major concern of adult-to-adult living donor liver transplantation is the adequate graft that can be harvested from a living donor. Small-for-size graft injury is frequently observed. To develop novel effective treatments attenuating small-for-size liver graft injury during living donor liver transplantation, it is important to explore the precise mechanism of acute phase small-for-size graft damage. Recently, a number of clinical studies and animal experiments have been conducted to investigate the possible key issues on acute phase small-for-size liver graft injury, such as mechanical injury from shear stress, subsequent inflammatory responses, and imbalance of vasoregulatory factors. This review focuses on the mechanism of small-for-size liver graft injury based on the number of clinical and experimental studies. The latest research findings of the significance of acute phase liver graft injury on late phase tumor recurrence and metastasis are also addressed.
Collapse
Affiliation(s)
- Kendrick Co Shih
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | | |
Collapse
|
292
|
Obata JE, Nakamura T, Kitta Y, Kodama Y, Sano K, Kawabata KI, Saitoh Y, Fujioka D, Kobayashi T, Yano T, Watanabe Y, Watanabe K, Kugiyama K. Treatment of Acute Myocardial Infarction With Sirolimus-Eluting Stents Results in Chronic Endothelial Dysfunction in the Infarct-Related Coronary Artery. Circ Cardiovasc Interv 2009; 2:384-91. [DOI: 10.1161/circinterventions.108.821371] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background—
Sirolimus-eluting stent (SES) implantation aggravated endothelial vasomotor dysfunction in infarct-related coronary arteries.
Methods and Results—
This study examined the effect of SES implantation on the duration of reperfusion-induced endothelial vasomotor dysfunction in infarct-related coronary arteries and on postinfarct left ventricular dysfunction in acute myocardial infarction (AMI). Patients with a first AMI due to occlusion of the left anterior descending coronary artery and successful reperfusion using SES (n=15) or bare metal stents (BMS; n=18) were examined. The vasomotor response of the left anterior descending coronary artery to acetylcholine and left ventriculography were examined 2 weeks and 6 months after AMI. At 6 months after AMI, the impairment of epicardial coronary artery dilation and coronary blood flow increase in response to acetylcholine was recovered from 2 weeks after AMI in BMS-treated patients, whereas the responses of SES-treated patients improved but remained impaired compared with BMS-treated patients (% increase in blood flow, 77�12% in SES versus 116�15% in BMS at 10 μg/min of acetylcholine,
P
<0.01). Left ventricular regional wall dysfunction in the left anterior descending coronary artery territory improved from 2 weeks to 6 months after AMI in BMS-treated patients but not in SES-treated patients (% improvement of average SD/chord, 6% in SES versus 19% in BMS,
P
<0.05), although left ventricular global ejection fraction was similar between the groups at any time points.
Conclusions—
SES implantation may delay recovery of reperfusion-induced endothelial vasomotor dysfunction in infarct-related coronary arteries and left ventricular regional dysfunction for at least 6 months after AMI.
Collapse
Affiliation(s)
- Jyun-ei Obata
- From the Department of Internal Medicine II, Faculty of Medicine, University of Yamanashi, Chuo City, Japan
| | - Takamitsu Nakamura
- From the Department of Internal Medicine II, Faculty of Medicine, University of Yamanashi, Chuo City, Japan
| | - Yoshinobu Kitta
- From the Department of Internal Medicine II, Faculty of Medicine, University of Yamanashi, Chuo City, Japan
| | - Yasushi Kodama
- From the Department of Internal Medicine II, Faculty of Medicine, University of Yamanashi, Chuo City, Japan
| | - Keita Sano
- From the Department of Internal Medicine II, Faculty of Medicine, University of Yamanashi, Chuo City, Japan
| | - Ken-Ichi Kawabata
- From the Department of Internal Medicine II, Faculty of Medicine, University of Yamanashi, Chuo City, Japan
| | - Yukio Saitoh
- From the Department of Internal Medicine II, Faculty of Medicine, University of Yamanashi, Chuo City, Japan
| | - Daisuke Fujioka
- From the Department of Internal Medicine II, Faculty of Medicine, University of Yamanashi, Chuo City, Japan
| | - Tsuyoshi Kobayashi
- From the Department of Internal Medicine II, Faculty of Medicine, University of Yamanashi, Chuo City, Japan
| | - Toshiaki Yano
- From the Department of Internal Medicine II, Faculty of Medicine, University of Yamanashi, Chuo City, Japan
| | - Yosuke Watanabe
- From the Department of Internal Medicine II, Faculty of Medicine, University of Yamanashi, Chuo City, Japan
| | - Kazuhiro Watanabe
- From the Department of Internal Medicine II, Faculty of Medicine, University of Yamanashi, Chuo City, Japan
| | - Kiyotaka Kugiyama
- From the Department of Internal Medicine II, Faculty of Medicine, University of Yamanashi, Chuo City, Japan
| |
Collapse
|
293
|
Zhao H, Wang Y, Wu Y, Li X, Yang G, Ma X, Zhao R, Liu H. Hyperlipidemia does not prevent the cardioprotection by postconditioning against myocardial ischemia/reperfusion injury and the involvement of hypoxia inducible factor-1alpha upregulation. Acta Biochim Biophys Sin (Shanghai) 2009; 41:745-53. [PMID: 19727523 DOI: 10.1093/abbs/gmp063] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Hyperlipidemia is regarded as an independent risk factor in the development of ischemic heart disease, and it can increase the myocardial susceptibility to ischemia/reperfusion (I/R) injury. Ischemic postconditioning (Postcon) has been demonstrated to attenuate the myocardial injury induced by I/R in normal conditions. But the effect of ischemic Postcon on hyperlipidemic animals is unknown. Hypoxia inducible factor-1 (HIF-1) has been demonstrated to play a central role in the cardioprotection by preconditioning, which is one of the protective strategies except for Postcon. The aim of this study was to determine whether Postcon could reduce myocardial injury in hyperlipidemic animals and to assess whether HIF-1 was involved in Postcon mechanisms. Male Wistar rats underwent the left anterior descending coronary occlusion for 30 min followed by 180 min of reperfusion with or without Postcon after fed with high fat diet or normal diet for 8 weeks. The detrimental indices induced by the I/R insult included infarct size, plasma creatine kinase activity and caspase-3 activity. Results showed that hyperlipidemia remarkably enhanced the myocardial injury induced by I/R, while Postcon significantly decreased the myocardial injury in both normolipidemic and hyperlipidemic rats. Moreover, both hyperlipidemia and I/R promoted the HIF-1alpha expression. Most importantly, we have for the first time demonstrated that Postcon further induced a significant increase in HIF-1alpha protein level not only in normolipidemic but also in hyperlipidemic conditions. Thus, Postcon reduces the myocardial injury induced by I/R in normal and hyperlipidemic animals, and HIF-1alpha upregulation may involve in the Postcon-mediated cardioprotective mechanisms.
Collapse
Affiliation(s)
- Huanxin Zhao
- Department of Physiology, Shanxi Medical University, Taiyuan 030001, China
| | | | | | | | | | | | | | | |
Collapse
|
294
|
Kusumanto YH, Tio RA, Loef BG, Sluiter WJ, Mulder NH, Hospers GAP. Systemic VEGF levels after coronary artery bypass graft surgery reflects the extent of inflammatory response. ACTA ACUST UNITED AC 2009; 8:41-5. [PMID: 16720427 DOI: 10.1080/14628840500480449] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
BACKGROUND Circulating vascular endothelial growth factor (VEGF) was studied as a substitute endpoint for treatment response after VEGF plasmid therapy. The effect of coronary artery bypass surgery (CABG) with cardiopulmonary bypass (CPB) on systemic VEGF levels are however largely unknown, therefore, we studied the effect of this procedure to measure VEGF levels after surgery alone. METHODS Fourteen patients requiring CABG were included. VEGF165 levels, ischemic markers, and hematology were measured before, directly after six days after surgery. RESULTS VEGF165 in serum and whole blood levels were increased up to six days after CABG, respectively 249.6+/-50.4 to 451.7+/-56.4 (day 6) and 581.9+/-105.1 to 783.4+/-97.7 (day six). There was a close correlation of circulating VEGF165 with leukocyte counts and platelets and not with ischemic markers. CONCLUSION Following surgery and in case of activated leukocyte and platelet counts care must be taken in the interpretation of systemic VEGF165 levels.
Collapse
Affiliation(s)
- Yoka H Kusumanto
- Department of Medical Oncology, University of Groningen and University Medical Center Groningen, The Netherlands
| | | | | | | | | | | |
Collapse
|
295
|
Hahn RG, Yin L, Ekengren J, Sandfeldt L. Vascular endothelial growth factor in serum indicates cardiovascular risk in urology patients. ACTA ACUST UNITED AC 2009; 40:144-8. [PMID: 16608813 DOI: 10.1080/00365590500368666] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
OBJECTIVE We hypothesized that a high serum concentration of vascular endothelial growth factor (VEGF), a cytokine involved in prostate growth which is also upregulated in chronic ischemia, indicates an increased risk of cardiovascular disease and death in urology patients. MATERIAL AND METHODS The serum VEGF concentration was measured in 219 males (mean age 72 years) who sought medical attention because of lower urinary tract symptoms. Data on cardiovascular events and survival were obtained from the hospital registry of Stockholm County and the Death Registry over a period of up to 10 years (mean 6.2 years). RESULTS After adjusting for the effects of age and smoking, patients with a serum VEGF level in the upper 20% of the cohort (>500 pg/ml) had an increased risk of developing cardiovascular disease (hazard ratio 2.18; 95% CI 1.04-4.60), including acute myocardial infarction (3.36; 1.35-8.41) and stroke (3.98; 1.61-9.86). They also had an increased risk of death (1.74; 1.01-3.00). These differences from patients with a lower serum VEGF concentration (<300 pg/ml) were manifested 2-5 years after the blood sample was taken. CONCLUSION An elevated VEGF level in peripheral blood was a risk factor for subsequent development of cardiovascular disease.
Collapse
|
296
|
Yamaki T, Iwai-Takano M, Yaoita H, Ogawa K, Tajima H, Takeishi Y, Maruyama Y. Participation of mast cells in angiogenesis in the border zone of myocardial infarction in rats. J Med Ultrason (2001) 2009; 36:119-27. [PMID: 27277224 DOI: 10.1007/s10396-009-0229-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2008] [Accepted: 06/02/2009] [Indexed: 11/28/2022]
Abstract
BACKGROUND We hypothesized that mast cells may participate in coronary angiogenesis in acute myocardial infarction, contributing to myocardial salvage. METHODS The left coronary artery was occluded in control (n = 30) and Ws rats (n = 30), which genetically lacked c-kit, resulting in a mast cell deficiency. Four weeks later, the infarct area, i.e., infarct core and surrounding infarct areas, and the non-infarct area were assessed histopathologically. The mast cell and small vessel densities were assessed using toluidine blue and alkaline phosphatase staining. Myocardial perfusion was assessed by myocardial contrast echocardiography (MCE). RESULTS In Ws rats, the percentage infarct core area increased (p < 0.05) compared with the controls, whereas the percentage surrounding infarct area decreased (p < 0.01). Mast cell density increased most in the surrounding infarct area (p < 0.01) in control rats, whereas mast cells were absent in Ws rats. Compared with the controls, coronary microvessel density decreased in the surrounding infarct area in Ws rats (p < 0.01). MCE showed that the percentage infarct core area, i.e., perfusion defect, increased (p < 0.05) and the percentage surrounding infarct area, i.e., reduced perfusion area, decreased (p < 0.01) in Ws rats. CONCLUSION Mast cells may participate in promoting coronary angiogenesis in the infarct area surrounding the infarct core, contributing to attenuation of left ventricular dysfunction.
Collapse
Affiliation(s)
- Takayoshi Yamaki
- Department of Cardiology and Hematology, Fukushima Medical University, Hikarigaoka 1, Fukushima, 960-1295, Japan
| | - Masumi Iwai-Takano
- Department of Cardiology and Hematology, Fukushima Medical University, Hikarigaoka 1, Fukushima, 960-1295, Japan.
| | - Hiroyuki Yaoita
- Department of Cardiology and Hematology, Fukushima Medical University, Hikarigaoka 1, Fukushima, 960-1295, Japan
| | - Kazuei Ogawa
- Department of Cardiology and Hematology, Fukushima Medical University, Hikarigaoka 1, Fukushima, 960-1295, Japan
| | - Hiroko Tajima
- Department of Cardiology and Hematology, Fukushima Medical University, Hikarigaoka 1, Fukushima, 960-1295, Japan
| | - Yasuchika Takeishi
- Department of Cardiology and Hematology, Fukushima Medical University, Hikarigaoka 1, Fukushima, 960-1295, Japan
| | - Yukio Maruyama
- Department of Cardiology and Hematology, Fukushima Medical University, Hikarigaoka 1, Fukushima, 960-1295, Japan
| |
Collapse
|
297
|
Tokudome T, Kishimoto I, Yamahara K, Osaki T, Minamino N, Horio T, Sawai K, Kawano Y, Miyazato M, Sata M, Kohno M, Nakao K, Kangawa K. Impaired recovery of blood flow after hind-limb ischemia in mice lacking guanylyl cyclase-A, a receptor for atrial and brain natriuretic peptides. Arterioscler Thromb Vasc Biol 2009; 29:1516-21. [PMID: 19628785 DOI: 10.1161/atvbaha.109.187526] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Atrial and brain natriuretic peptides (ANP and BNP, respectively) function via guanylyl cyclase (GC)-A, resulting in diuresis, natriuresis, and blood vessel dilation. Here, we investigated the role of endogenous ANP/BNP-GC-A signaling on reparative vascular remodeling using a hind-limb ischemia model. METHODS AND RESULTS In GC-A-deficient mice (GC-A-KO), hind-limb ischemia resulted in autoamputation or severe ulcers in 60% of mice (6/10) during the 28-day observation period. In wild-type (WT) mice, partial amputation or mild ulcers were detected in only 20% of mice (2/10). Laser Doppler perfusion imaging revealed that the recovery of blood flow in the ischemic limb was significantly inhibited in GC-A-KO mice compared with WT mice. Immunostainings with anti-PECAM-1 antibody demonstrated that, in GC-A-KO, the capillary density of the ischemic tissue was significantly diminished compared to WT. Furthermore, bone marrow transplantation showed the predominant role of GC-A on local ischemic tissue rather than on vascular progenitor cells mobilized from bone marrow during vascular remodeling. In cultured human endothelial cells, ANP treatment significantly stimulated mRNA expressions of vascular endothelial growth factor and endothelial nitric oxide synthase via Erk1/2-dependent mechanism. CONCLUSIONS These results suggest that endogenous ANP and BNP play important roles in reparative vascular remodeling in ischemic tissue.
Collapse
Affiliation(s)
- Takeshi Tokudome
- Research Institute, National Cardiovascular Center, Suita, Osaka 565-8565, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
298
|
Zhao W, Han Q, Lin H, Sun W, Gao Y, Zhao Y, Wang B, Wang X, Chen B, Xiao Z, Dai J. Human basic fibroblast growth factor fused with Kringle4 peptide binds to a fibrin scaffold and enhances angiogenesis. Tissue Eng Part A 2009; 15:991-8. [PMID: 18771415 DOI: 10.1089/ten.tea.2008.0240] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Appropriate three-dimensional (3D) scaffolds and signal molecules could accelerate tissue regeneration and wound repair. In this work, we targeted human basic fibroblast growth factor (bFGF), a potent angiogenic factor, to a fibrin scaffold to improve therapeutic angiogenesis. We fused bFGF to the Kringle4 domain (K4), a fibrin-binding peptide from human plasminogen, to endow bFGF with specific fibrin-binding ability. The recombinant K4bFGF bound specifically to the fibrin scaffold so that K4bFGF was delivered in a site-specific manner, and the fibrin scaffold provided 3D support for cell migration and proliferation. Subcutaneous implantation of the fibrin scaffolds bound with K4bFGF but not with bFGF induced neovascularization. Immunohistochemical analysis showed significantly more proliferation cells in the fibrin scaffolds incorporated with K4bFGF than in those with bFGF. Moreover, the regenerative tissues were integrated well with the fibrin scaffolds, suggesting its good biocompatibility. In summary, targeted delivery of K4bFGF could potentially improve therapeutic angiogenesis.
Collapse
Affiliation(s)
- Wenxue Zhao
- Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
299
|
Attenuation of myocardial injury by postconditioning: role of hypoxia inducible factor-1alpha. Basic Res Cardiol 2009; 105:109-18. [PMID: 19597757 DOI: 10.1007/s00395-009-0044-0] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2009] [Revised: 06/19/2009] [Accepted: 06/26/2009] [Indexed: 01/12/2023]
Abstract
Postconditioning (PostC) has regenerated interest as a mechanical intervention against myocardial ischemia/reperfusion injury, but its molecular mechanisms remain elusive. This study tested the hypothesis that hypoxia inducible factor-1alpha (HIF-1alpha) plays a role in PostC-induced cardioprotection. Male Wistar rats were subjected to 30 min ischemia followed by 3 h of reperfusion (Control). PostC with 3 cycles of 10 s reperfusion and 10 s re-occlusion was applied at the onset of reperfusion. Relative to the Sham group, HIF-1alpha protein level was increased by 2.9-fold in the Control group, but its level was enhanced by 5.8-fold with PostC (P < 0.01 vs. Control). However, HIF-1alpha protein level was further augmented by 2.0-fold and 3.3-fold, respectively, when the prolyl hydroxylase inhibitor, dimethyloxalylglycine (DMOG, 40 mg/kg, i.p.) was given at 24 h before ischemia in both Control and PostC groups. PostC reduced infarct size by 24% compared with the Control (27 +/- 4.2% vs. 36 +/- 5.2%, P < 0.01), consistent with significant lower levels of plasma creatine kinase activity, index of cardiomyocyte apoptosis and caspase-3 activity. Although pretreatment with DMOG significantly reduced infarct size relative to the Control, the infarct-sparing effect of PostC was remarkably enhanced when DMOG was given before PostC (18 +/- 2.0% vs. 27 +/- 4.2% in PostC alone, P < 0.05). There was a significant linear inverse relationship between HIF-1alpha protein level and infarct size (r = -0.799, P < 0.01) among all groups. Furthermore, along with up-regulated HIF-1alpha expression, the levels of iNOS mRNA and protein were significantly increased in the PostC alone and DMOG plus PostC groups. In conclusion, these data suggest that HIF-1alpha is involved in cardioprotection by PostC and pharmacological augmentation of HIF-1alpha expression that enhances the infarct-sparing effect of PostC; iNOS, the downstream gene of HIF-1alpha, may participate in signaling pathways in mediating PostC's protection.
Collapse
|
300
|
Sundset R, Cooper M, Mikalsen SO, Ytrehus K. Ischemic Preconditioning Protects Against Gap Junctional Uncoupling in Cardiac Myofibroblasts. ACTA ACUST UNITED AC 2009; 11:51-66. [PMID: 16247851 DOI: 10.1080/15419060490951772] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Ischemic preconditioning increases the heart's tolerance to a subsequent longer ischemic period. The purpose of this study was to investigate the role of gap junction communication in simulated preconditioning in cultured neonatal rat cardiac myofibroblasts. Gap junctional intercellular communication was assessed by Lucifer yellow dye transfer. Preconditioning preserved intercellular coupling after prolonged ischemia. An initial reduction in coupling in response to the preconditioning stimulus was also observed. This may protect neighboring cells from damaging substances produced during subsequent regional ischemia in vivo, and may preserve gap junctional communication required for enhanced functional recovery during subsequent reperfusion.
Collapse
Affiliation(s)
- Rune Sundset
- Department of Medical Physiology, Faculty of Medicine, University of Tromsø, Tromsø, Norway.
| | | | | | | |
Collapse
|