Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Neth Heart J ; 30(7-8): 377-382, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35099775

ABSTRACT

BACKGROUND: Infections with potentially cardiotropic viruses are associated with the development of atrial fibrillation (AF). However, whether direct viral infection of the atria is involved in the pathogenesis of AF is unclear. We have therefore analysed the presence of cardiotropic viral genomes in AF patients. METHODS: Samples of left atrial tissue were obtained from 50 AF patients (paroxysmal, n = 20; long-standing persistent/permanent, n = 30) during cardiac surgery and from autopsied control patients (n = 14). Herein, the presence of PVB19, EBV, CMV, HHV­6, adenovirus and enterovirus genomes was determined by polymerase chain reaction. The densities of CD45+ and CD3+ cells and fibrosis in the atria were quantified by (immuno)histochemistry. RESULTS: Of the tested viruses only the PVB19 genome was detected in the atria of 10% of patients, paroxysmal AF (2 of 20) and long-standing persistent/permanent AF (3 of 30). Conversely, in 50% of controls (7 of 14) PVB19 genome was found. No significant association was found between PVB19 and CD45+ and CD3+ cells, or between the presence of PVB19 and fibrosis, in either control or AF patients. CONCLUSION: The presence of viral genomes is not increased in the atria of AF patients. These results do not support an important role for viral infection of the atria in the pathogenesis of AF.

2.
Clin Res Cardiol ; 109(10): 1271-1281, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32072262

ABSTRACT

OBJECTIVE: Inflammation of the atria is an important factor in the pathogenesis of atrial fibrillation (AF). Whether the extent of atrial inflammation relates with clinical risk factors of AF, however, is largely unknown. This we have studied comparing patients with paroxysmal and long-standing persistent/permanent AF. METHODS: Left atrial tissue was obtained from 50 AF patients (paroxysmal = 20, long-standing persistent/permanent = 30) that underwent a left atrial ablation procedure either or not in combination with coronary artery bypass grafting and/or valve surgery. Herein, the numbers of CD45+ and CD3+ inflammatory cells were quantified and correlated with the AF risk factors age, gender, diabetes, and blood CRP levels. RESULTS: The numbers of CD45+ and CD3+ cells were significantly higher in the adipose tissue of the atria compared with the myocardium in all AF patients but did not differ between AF subtypes. The numbers of CD45+ and CD3+ cells did not relate significantly to gender or diabetes in any of the AF subtypes. However, the inflammatory infiltrates as well as CK-MB and CRP blood levels increased significantly with increasing age in long-standing persistent/permanent AF and a moderate positive correlation was found between the extent of atrial inflammation and the CRP blood levels in both AF subtypes. CONCLUSION: The extent of left atrial inflammation in AF patients was not related to the AF risk factors, diabetes and gender, but was associated with increasing age in patients with long-standing persistent/permanent AF. This may be indicative for a role of inflammation in the progression to long-standing persistent/permanent AF with increasing age.


Subject(s)
Atrial Fibrillation/physiopathology , Heart Atria/physiopathology , Inflammation/physiopathology , Adult , Age Factors , Aged , Aged, 80 and over , Atrial Fibrillation/surgery , Catheter Ablation , Disease Progression , Female , Humans , Male , Middle Aged , Risk Factors
3.
Case Rep Cardiol ; 2019: 7916298, 2019.
Article in English | MEDLINE | ID: mdl-31016051

ABSTRACT

The incidence of heart valve hemangioma is very low and is mostly observed in the mitral and tricuspid valve. In 2006, two cases of aortic valve hemangioma were reported for the first time, including one with calcifying aortic valve stenosis. We now present a case of aortic valve hemangioma in a patient suffering from aortic valve insufficiency combined with atherosclerotic thickening.

4.
Stem Cell Res ; 17(1): 6-15, 2016 07.
Article in English | MEDLINE | ID: mdl-27186654

ABSTRACT

Successful stem cell therapy after acute myocardial infarction (AMI) is hindered by lack of engraftment of sufficient stem cells at the site of injury. We designed a novel technique to overcome this problem by assembling stem cell-microbubble complexes, named 'StemBells'. StemBells were assembled through binding of dual-targeted microbubbles (~3µm) to adipose-derived stem cells (ASCs) via a CD90 antibody. StemBells were targeted to the infarct area via an ICAM-1 antibody on the microbubbles. StemBells were characterized microscopically and by flow cytometry. The effect of ultrasound on directing StemBells towards the vessel wall was demonstrated in an in vitro flow model. In a rat AMI-reperfusion model, StemBells or ASCs were injected one week post-infarction. A pilot study demonstrated feasibility of intravenous StemBell injection, resulting in localization in ICAM-1-positive infarct area three hours post-injection. In a functional study five weeks after injection of StemBells cardiac function was significantly improved compared with controls, as monitored by 2D-echocardiography. This functional improvement neither coincided with a reduction in infarct size as determined by histochemical analysis, nor with a change in anti- and pro-inflammatory macrophages. In conclusion, the StemBell technique is a novel and feasible method, able to improve cardiac function post-AMI in rats.


Subject(s)
Microbubbles , Myocardial Infarction/therapy , Stem Cell Transplantation/methods , Adipose Tissue/cytology , Adipose Tissue/metabolism , Administration, Intravenous , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Cell Survival , Cells, Cultured , Disease Models, Animal , Echocardiography , Heart/diagnostic imaging , Heart/physiopathology , Humans , Macrophages/metabolism , Macrophages/pathology , Male , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Pilot Projects , Rats , Rats, Wistar , Sonication , Stem Cells/cytology , Stem Cells/metabolism
5.
Cell Tissue Res ; 362(3): 623-32, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26202892

ABSTRACT

In most pre-clinical animal studies investigating stem cell therapy in acute myocardial infarction (AMI), the administered stem cells are isolated from healthy donors. In clinical practice, however, patients who suffer from AMI will receive autologous cells, for example using adipose-derived stem cells (ASC). During AMI, inflammation is induced and we hypothesized that this might affect characteristics of ASC. To investigate this, ASC were isolated from rat adipose tissue 1 day (1D group, n = 5) or 7 days (7D group, n = 6) post-AMI, and were compared with ASC from healthy control rats (Control group, n = 6) and sham-operated rats (Sham 1D group, n = 5). We found that significantly fewer ASC were present 1 day post-AMI in the stromal vascular fraction (SVF), determined by a colony-forming-unit assay (p < 0.001 vs. Control and 7D). These data were confirmed by flow cytometry, showing fewer CD90-positive cells in SVF of the 1D group. When cultured, no differences were found in proliferation rate and cell size between the groups in the first three passages. Also, no difference in the differentiation capacity of ASC was found. In conclusion, it was shown that significantly fewer stem cells were present in the SVF 1 day post-AMI; however, the stem cells that were present showed no functional differences.


Subject(s)
Adipose Tissue/cytology , Myocardial Infarction/pathology , Stem Cells/cytology , Animals , Cell Count , Cell Differentiation , Cell Lineage , Cells, Cultured , Male , Rats, Wistar , Stromal Cells/cytology
6.
Stem Cell Rev Rep ; 10(3): 389-98, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24577790

ABSTRACT

The majority of patients survive an acute myocardial infarction (AMI). Their outcome is negatively influenced by post-AMI events, such as loss of viable cardiomyocytes due to a post-AMI inflammatory response, eventually resulting in heart failure and/or death. Recent pre-clinical animal studies indicate that mesenchymal stem cells derived from adipose tissue (ASC) are new promising candidates that may facilitate cardiovascular regeneration in the infarcted myocardium. In this review we have compared all animal studies in which ASC were used as a therapy post-AMI and have focused on aspects that might be important for future successful clinical application of ASC.


Subject(s)
Mesenchymal Stem Cell Transplantation/methods , Myocardial Infarction/therapy , Adipose Tissue/cytology , Animals , Cells, Cultured , Disease Models, Animal , Humans , Mesenchymal Stem Cells/physiology
7.
Res Vet Sci ; 96(2): 377-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24445251

ABSTRACT

The Wistar rat is a commonly used strain for experimental animal models. Recently it was shown that results vary between studies using Wistar rats of different suppliers. Therefore we studied whether Wistar rats obtained from Harlan Laboratories (Ha, n=24) and Charles River (CR, n=22) had a different outcome in an acute myocardial infarction (AMI) model. AMI was induced in both Ha and CR Wistar rats by one operator. This resulted in a significantly higher survival rate for Ha (79.2±10.2%) compared with CR rats (54.2±10.2%, p<0.05). Furthermore, CR rats had lost significantly more weight after 7 days (-5.9±3.1%) compared with Ha rats (-0.8±1.7%; p<0.001), indicating a worse health status of the CR rats. Paradoxically, the induced infarct was smaller in CR rats (7.3±3.6% of the heart) compared with Ha rats (12.1±4.7%, p<0.05). This indicates that CR rats were less sensitive for the cardiomyocyte damage subsequent to AMI induction, but remarkably showed more clinical side effects indicating that Wistar rats from two suppliers had a different response within the same AMI model.


Subject(s)
Myocardial Infarction/veterinary , Myocytes, Cardiac/ultrastructure , Rats, Wistar/surgery , Animals , Disease Models, Animal , Histocytochemistry , Kaplan-Meier Estimate , Male , Myocardial Infarction/physiopathology , Rats
8.
Cell Tissue Res ; 348(1): 119-30, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22395775

ABSTRACT

Adipose-derived stromal cells (ASC) are promising candidates for cell therapy, for example to treat myocardial infarction. Commonly, fetal bovine serum (FBS) is used in ASC culturing. However, FBS has several disadvantages. Its effects differ between batches and, when applied clinically, transmission of pathogens and antibody development against FBS are possible. In this study, we investigated whether FBS can be substituted by human platelet lysate (PL) in ASC culture, without affecting functional capacities particularly important for cardiac repair application of ASC. We found that PL-cultured ASC had a significant 3-fold increased proliferation rate and a significantly higher attachment to tissue culture plastic as well as to endothelial cells compared with FBS-cultured ASC. PL-cultured ASC remained a significant 25% smaller than FBS-cultured ASC. Both showed a comparable surface marker profile, with the exception of significantly higher levels of CD73, CD90, and CD166 on PL-cultured ASC. PL-cultured ASC showed a significantly higher migration rate compared with FBS-cultured ASC in a transwell assay. Finally, FBS- and PL-cultured ASC had a similar high capacity to differentiate towards cardiomyocytes. In conclusion, this study showed that culturing ASC is more favorable in PL-supplemented medium compared with FBS-supplemented medium.


Subject(s)
Adipose Tissue/cytology , Blood Platelets/metabolism , Blood Substitutes/pharmacology , Cell Extracts/pharmacology , Myocardium/pathology , Serum/metabolism , Wound Healing/drug effects , Adult , Aged , Animals , Biomarkers/metabolism , Blood Platelets/drug effects , Cattle , Cell Adhesion/drug effects , Cell Culture Techniques , Cell Differentiation/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Size/drug effects , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Flow Cytometry , Humans , Middle Aged , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Stromal Cells/cytology , Stromal Cells/drug effects , Stromal Cells/metabolism
9.
Eur J Clin Invest ; 40(1): 4-10, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19843156

ABSTRACT

BACKGROUND: Recent studies indicate a role for complement in the pathogenesis of aortic valve disease. However, the role of naturally occurring anti-complement mediators in this context is unknown. In this study, we have analysed this in three different pathological conditions of the aortic valve: degeneration, atherosclerosis and bacterial endocarditis. MATERIALS AND METHODS: Human aortic valves were obtained at autopsy (n = 30): 5 control valves, 10 aortic valves with atherosclerotic changes, 10 aortic valves with degenerative changes and 5 degenerative changed aortic valves with bacterial infection. These valves were analysed immunohistochemically for the presence of activated complement (C3d and C5b9) and the complement inhibitors C1-inh and clusterin. Areas of positivity were then quantified. RESULTS: C3d, C5b9 and the complement inhibitors C1-inh and clusterin depositions were mainly found in the endothelium and extracellular matrix in aortic valves. All these mediators were already present in control valves, but the area of positivity increased significantly in response to the different diseases, with the highest increase in response to bacterial endocarditis. Interestingly, in all three aortic diseases, the depositions of complement were significantly more widespread than that of their inhibitors. CONCLUSIONS: Our study indicates that anti-complement mediators (C1-inh and clusterin) are deposited in diseased aortic valves together with activated complement, indicating an existing counter response against complement locally in the valve. However, deposition of activated complement is significantly more widespread than that of its inhibitors, which could explain ongoing inflammation in those diseased aortic valves.


Subject(s)
Aortic Valve/immunology , Atherosclerosis/immunology , Complement System Proteins/metabolism , Inflammation , Adult , Aged , Aged, 80 and over , Aortic Valve/metabolism , Aortic Valve/pathology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Clusterin/analysis , Complement C1 Inactivator Proteins/analysis , Complement C1 Inhibitor Protein , Complement C3d/analysis , Complement Membrane Attack Complex/analysis , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Extracellular Matrix/metabolism , Female , Humans , Immunohistochemistry , Male , Middle Aged
10.
Eur J Clin Invest ; 38(4): 205-10, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18339001

ABSTRACT

BACKGROUND: Secretory type-II phospholipase A(2) (sPLA(2)-II) is a cardiovascular risk marker since higher levels of this acute phase protein imply an increased risk for coronary artery disease. Moreover, it is hypothesized that local activity of sPLA(2)-II in the atherosclerotic plaque facilitates an inflammatory response to induce plaque instability or rupture. We have studied the presence of sPLA(2)-II in culprit lesions in the coronary arteries of patients with acute myocardial infarction (AMI) or angina pectoris. MATERIALS AND METHODS: We performed a histological examination of culprit lesions in 41 patients with stable (SAP) or unstable angina pectoris (UAP), or AMI using directed coronary atherectomy (DCA). Frozen slides were analysed immuno-histochemically for the presence of sPLA(2)-II, macrophages and smooth muscle cells. Immunopositive areas were calculated as a percentage of the total tissue area using image analysis software. RESULTS: Intracellular sPLA(2)-II was found in atherosclerotic lesions in the macrophages of the intima as well as in vascular smooth muscle cells. Next to this, extracellular sPLA(2)-II depositions were also found. These depositions were significantly more extensive in patients with AMI, i.e. 26%(median)[6%(25th(percentile))-44%(75th(percentile))] of the intima area, than in patients with SAP 0%(median) (0%(25th)-10%(75th); P = 0.013) or UAP 0%(median) (0%(25th)-0%(75th); P = 0.04). CONCLUSIONS: Extracellular sPLA(2)-II is more abundantly present in atherosclerotic culprit lesions that have led to myocardial infarction. This suggests a role for extracellular sPLA(2)-II in the development of complications of atherosclerotic lesions in coronary arteries.


Subject(s)
Angina Pectoris/diagnosis , Coronary Artery Disease/diagnosis , Myocardial Infarction/diagnosis , Phospholipases A2/analysis , Actins/metabolism , Adult , Aged , Biomarkers/analysis , Female , Humans , Immunohistochemistry , Macrophages/pathology , Male , Middle Aged , Myocytes, Smooth Muscle/pathology
11.
Arterioscler Thromb Vasc Biol ; 26(11): 2497-503, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16973974

ABSTRACT

OBJECTIVE: Advanced glycation end products (AGEs), such as N(epsilon)-(carboxymethyl)lysine (CML), are implicated in vascular disease. We previously reported increased CML accumulation in small intramyocardial blood vessels in diabetes patients. Diabetes patients have an increased risk for acute myocardial infarction (AMI). Here, we examined a putative relationship between CML and AMI. METHODS AND RESULTS: Heart tissue was stained for CML, myeloperoxidase, and E-selectin in AMI patients (n=26), myocarditis patients (n=17), and control patients (n=15). In AMI patients, CML depositions were 3-fold increased compared with controls in the small intramyocardial blood vessels and predominantly colocalized with activated endothelium (E-selectin-positive) both in infarction and noninfarction areas. A trend of increased CML positivity of the intima of epicardial coronary arteries did not reach significance in AMI patients. In the rat heart AMI model, CML depositions were undetectable after 24 hours of reperfusion, but became clearly visible after 5 days of reperfusion. In line with an inflammatory contribution, human myocarditis was also accompanied by accumulation of CML on the endothelium of intramyocardial blood vessels. CONCLUSIONS: CML, present predominantly on activated endothelium in small intramyocardial blood vessels in patients with AMI, might reflect an increased risk for AMI rather than being a result of AMI.


Subject(s)
Coronary Vessels/metabolism , Lysine/analogs & derivatives , Myocardial Infarction/metabolism , Aged , Animals , E-Selectin/metabolism , Endothelial Cells/metabolism , Female , Humans , Immunohistochemistry , Lysine/biosynthesis , Lysine/metabolism , Male , Middle Aged , Myocardial Infarction/etiology , Myocardial Reperfusion , Myocarditis/metabolism , Oxidative Stress , Peroxidase/metabolism , Prognosis , Rats , Risk Factors , Time Factors
12.
Apoptosis ; 11(6): 913-21, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16544099

ABSTRACT

In previous work we have demonstrated increased expression of NOX2 in cardiomyocytes of infarcted human hearts. In the present manuscript we investigated the functional role of NOX2 in ischemically challenged H9c2 cells, a rat cardiomyoblast cell line, and adult rat cardiomyocytes. Expression of NOX2 in H9c2 cells was confirmed by RT-PCR. In Western-blot experiments, increased NOX2 expression was detected during ischemia, which was inhibited by transcription and translation inhibitors. Surprisingly, under ischemia, in addition to an increased cytosolic expression, NOX2 was localized mainly in the nucleus of apoptotic cardiomyocytes, where it colocalized with nitrotyrosine residues and activated caspase 3. Inhibition of reactive-oxygen-species generation with the flavoenzyme inhibitor diphenylene iodonium (DPI) and the NADPH-oxidase inhibitor apocynin led to a significantly decreased induction of apoptosis as assessed by quantification of caspase-3 activity and by TUNEL analysis. These results demonstrate that NOX2 is expressed in the nucleus of cardiomyocytes during apoptosis and that it likely participates in proapoptotic signaling. To the best of our knowledge, this is the first demonstration of nuclear NOX2 expression and its involvement in cardiomyocyte apoptosis.


Subject(s)
Apoptosis/physiology , Cell Nucleus/physiology , Ischemia/physiopathology , Membrane Glycoproteins/genetics , Myocytes, Cardiac/physiology , NADPH Oxidases/genetics , Animals , Cell Line , Membrane Glycoproteins/biosynthesis , NADPH Oxidase 2 , NADPH Oxidases/biosynthesis , Rats , Rats, Wistar , Signal Transduction/physiology
13.
Am J Physiol Heart Circ Physiol ; 289(5): H2193-202, 2005 Nov.
Article in English | MEDLINE | ID: mdl-15994859

ABSTRACT

We examined the relationship between clusterin and activated complement in human heart infarction and evaluated the effect of this protein on ischemic rat neonatal cardiomyoblasts (H9c2) and isolated adult ventricular rat cardiomyocytes as in vitro models of acute myocardial infarction. Clusterin protects cells by inhibiting complement and colocalizes with complement on jeopardized human cardiomyocytes after infarction. The distribution of clusterin and complement factor C3d was evaluated in the infarcted human heart. We also analyzed the protein expression of clusterin in ischemic H9c2 cells. The binding of endogenous and purified human clusterin on H9c2 cells was analyzed by flow cytometry. Furthermore, the effect of clusterin on the viability of ischemically challenged H9c2 cells and isolated adult ventricular rat cardiomyocytes was analyzed. In human myocardial infarcts, clusterin was found on scattered, morphologically viable cardiomyocytes within the infarcted area that were negative for complement. In H9c2 cells, clusterin was rapidly expressed after ischemia. Its expression was reduced after reperfusion. Clusterin bound to single annexin V-positive or annexin V and propidium iodide-positive H9c2 cells. Clusterin inhibited ischemia-induced death in H9c2 cells as well as in isolated adult ventricular rat cardiomyocytes in the absence of complement. We conclude that ischemia induces the upregulation of clusterin in ischemically challenged, but viable, cardiomyocytes. Our data suggest that clusterin protects cardiomyocytes against ischemic cell death via a complement-independent pathway.


Subject(s)
Heart/physiology , Myocardial Ischemia/physiopathology , Myocytes, Cardiac/physiology , Animals , Animals, Newborn , Antibodies, Monoclonal , Blotting, Western , Cells, Cultured , Flow Cytometry , Fluorescein-5-isothiocyanate , Fluorescent Dyes , Humans , Immunohistochemistry , In Vitro Techniques , Myoblasts/physiology , Myocardial Infarction/physiopathology , Myocardium/metabolism , Rats
14.
J Clin Pathol ; 58(4): 382-8, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15790702

ABSTRACT

AIMS: Reperfusion of ischaemic myocardium after acute myocardial infarction (AMI) can induce ischaemia/reperfusion (I/R) injury, as a result of local activation of the complement system. C reactive protein (CRP) is involved in this activation. This study analysed the potential role of IgM in complement activation in the infarcted human myocardium. METHODS: Immunochemical analysis was carried out on heart specimens from 59 patients who died from AMI. Serial slides of frozen tissue from the infarction site were stained for IgM, complement factors C3d and C5b-9 (membrane attack complex), and CRP. RESULTS: IgM deposits were found on the plasma membrane, cross striations, and in the cytoplasm of jeopardised cardiomyocytes in infarcts of one to five days duration. IgM depositions were remarkably similar to those of CRP and both complement factors. The relative staining intensities of IgM and CRP varied greatly among patients. CONCLUSIONS: Similar to CRP, IgM targets complement locally to jeopardised cardiomyocytes in the human heart after AMI. Localisation patterns and relative staining intensities suggest that IgM and CRP recognise similar epitopes in the ischaemic heart, but that the relative contribution of each protein to complement activation in the ischaemic myocardium differs among patients.


Subject(s)
C-Reactive Protein/analysis , Complement Activation/immunology , Immunoglobulin M/analysis , Myocardial Infarction/immunology , Adult , Aged , Aged, 80 and over , C-Reactive Protein/immunology , Complement System Proteins/analysis , Complement System Proteins/immunology , Female , Humans , Immunohistochemistry/methods , Male , Middle Aged , Myocardial Infarction/pathology , Myocardium/immunology , Myocardium/pathology , Myocytes, Cardiac/immunology , Myocytes, Cardiac/pathology
15.
Eur J Clin Invest ; 34(12): 803-10, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15606722

ABSTRACT

BACKGROUND: Impaired perfusion of the heart induces a local inflammatory response, which involves deposition of C-reactive protein and complement activation products C3d and C5b-9. We investigated whether reperfusion or reinfarction enhances these phenomena in humans. MATERIALS AND METHODS: Depositions of C-reactive protein and complement were quantified in tissue samples of infarcted myocardium from 76 patients who had died after acute myocardial infarction. The extent of depositions in patients treated with reperfusion or suffering from reinfarction was compared with that in patients who had no reperfusion or reinfarction. RESULTS: Patients with reinfarction had significantly more extensive depositions of C-reactive protein and complement (C3d and C5b-9) in the infarcted myocardium than patients without reinfarction. Similarly, patients who received reperfusion therapy had more extensive depositions also than those who had not received this therapy. CONCLUSIONS: Both reinfarction and reperfusion therapy significantly increase the extent of C-reactive protein and complement depositions in human myocardial infarcts.


Subject(s)
C-Reactive Protein/metabolism , Complement System Proteins/metabolism , Myocardial Infarction/metabolism , Myocardial Reperfusion , Aged , Aged, 80 and over , Complement C3d/metabolism , Complement Membrane Attack Complex/metabolism , Female , Humans , Inflammation Mediators/metabolism , Male , Middle Aged , Myocardial Infarction/pathology , Myocardial Infarction/therapy , Recurrence
17.
J Clin Pathol ; 56(3): 194-9, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12610097

ABSTRACT

BACKGROUND/AIMS: Recent studies indicate the presence of reactive oxygen species (ROS) producing homologues of the enzymatic subunit (Nox2) of phagocytic NADPH oxidase in non-phagocytic cells. Interestingly, in these cells, ROS produced by the Nox2 homologue(s) was shown to play a role in various regulatory processes, including cell death, proliferation, and aging. The purpose of this study was to investigate whether human cardiomyocytes express Nox2. METHODS: The expression of Nox2 was studied in human cardiomyocytes using western blot and immunohistochemical analysis. To analyse the putative expression of Nox2 in human heart disease, cardiac samples from patients who had died subsequent to acute myocardial infarction (AMI) were studied. RESULTS: Both in western blot and immunohistochemical studies, Nox2 expression was found in normal human cardiomyocytes. In patients with AMI, a significant increase in Nox2 expression was found both in viable and in jeopardised cardiomyocytes in the infarcted area. In addition, in the "remote from infarction" area, Nox2 expression was present in cardiomyocytes, but was not increased. CONCLUSIONS: Nox2 or its homologue(s) is expressed in normal and jeopardised human cardiomyocytes. This expression is increased in patients with AMI, suggesting a role for this ROS producing Nox2 homologue(s) in the human heart after AMI.


Subject(s)
Bacterial Proteins , Myocardial Infarction/enzymology , NADH, NADPH Oxidoreductases/metabolism , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/immunology , Antigens, Neoplasm/metabolism , Blotting, Western , Cell Adhesion Molecules/metabolism , Complement System Proteins/analysis , Female , Humans , Male , Middle Aged , Myocardial Infarction/pathology , Myocardium/enzymology , Myocardium/pathology , NADH, NADPH Oxidoreductases/immunology , Specimen Handling/methods , Up-Regulation
18.
J Clin Pathol ; 55(11): 801-11, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12401816

ABSTRACT

Recent studies indicate that, in addition to necrosis, apoptosis also plays a role in the process of tissue damage after myocardial infarction, which has pathological and therapeutic implications. This review article will discuss studies in which the role and mechanisms of apoptosis in myocardial infarction were analysed in vivo and in vitro in humans and in animals.


Subject(s)
Apoptosis/physiology , Myocardial Ischemia/pathology , Cell Culture Techniques , Disease Models, Animal , Humans , Myocardial Infarction/pathology , Myocytes, Cardiac/pathology , Organ Culture Techniques , Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...