Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
1.
Eur J Heart Fail ; 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38899626

ABSTRACT

AIMS: Haemodynamic monitoring using implantable pressure sensors reduces the risk of heart failure (HF) hospitalizations. Patient self-management (PSM) of haemodynamics in HF has the potential to personalize treatment, increase adherence, and reduce the risk of worsening HF, while lowering clinicians' burden. METHODS AND RESULTS: The VECTOR-HF I and IIa studies are prospective, single-arm, open-label clinical trials assessing safety, usability and performance of left atrial pressure (LAP)-guided HF management using PSM in New York Heart Association class II and III HF patients. Physician-prescribed LAP thresholds trigger patient self-adjustment of diuretics. Primary endpoints include the ability to perform LAP measurements and transmit data to the healthcare provider (HCP) interface and the patient guidance application, and safety outcomes. This is an interim analysis of 13 patients using the PSM approach. Over 12 months, no procedure- or device-related major adverse cardiovascular or neurological events were observed, and there were no failures to obtain measurements from the sensor and transmit the data to the HCP interface and the patient guidance application. Patient adherence was 91.4%. Using PSM, annualized HF hospitalization rate significantly decreased compared to a similar period prior to PSM utilization (0 admissions vs. 0.69 admissions over 11.84 months, p = 0.004). At 6 months, 6-min walk test distance and the Kansas City Cardiomyopathy Questionnaire overall summary score demonstrated significant improvement. CONCLUSIONS: Interim findings suggest that PSM using a LAP monitoring system is feasible and safe. PSM is associated with high patient adherence, potentially improving HF patients' functional status, quality of life, and limiting HF hospitalizations.

2.
J Am Heart Assoc ; 2(6): e000413, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-24231658

ABSTRACT

BACKGROUND: One of the most daunting complications of cardiac catheterization is a cerebrovascular event (CVE). We aimed to assess the real-life incidence, etiology, and risk factors of cardiac catheterization-related acute CVEs in a large cohort of patients treated in a single center. METHODS AND RESULTS: We undertook a retrospective analysis of 43,350 coronary procedures performed on 30,907 procedure days over the period 1992-2011 and compared patient and procedural characteristics of procedures complicated by CVEs with the remaining cohort. CVEs occurred in 47 cases: 43 were ischemic, 3 intracerebral hemorrhages, and 1 undetermined. The overall CVE rate was 0.15%, with percutaneous coronary intervention (PCI) and diagnostic coronary angiography rates 0.23% and 0.09%, respectively. Using a forward stepwise multivariate logistic regression model including patient demographic and procedural characteristics, a total of 5 significant predictors were defined: prior stroke (OR=15.09, 95% CI [8.11 to 28.08], P<0.0001), presence of coronary arterial thrombus (OR=2.79, 95% CI [1.25 to 6.22], P=0.012), age >75 years (OR=3.33, 95% CI [1.79 to 6.19], P<0.0001), triple vessel disease (OR=2.24, 95% CI [1.20 to 4.18], P=0.011), and performance of intervention (OR=2.21, 95% CI [1.12 to 4.33], P=0.021). An additional analysis excluded any temporal change of CVE rates but demonstrated a significant increase of all high-risk patient features. CONCLUSION: In a single-center, retrospective assessment over nearly 20 years, cardiac catheterization-related CVEs were very rare and nearly exclusively ischemic. The independent predictors for these events were found to be the performance of an intervention and those associated with increased atherosclerotic burden, specifically older age, triple vessel disease, and prior stroke. The presence of intracoronary thrombus appears also to raise the risk of procedure-related CVE.


Subject(s)
Cardiac Catheterization/adverse effects , Cerebrovascular Disorders/epidemiology , Coronary Angiography/adverse effects , Percutaneous Coronary Intervention/adverse effects , Aged , Aged, 80 and over , Cerebrovascular Disorders/diagnosis , Chi-Square Distribution , Female , Humans , Incidence , Israel/epidemiology , Logistic Models , Male , Middle Aged , Multivariate Analysis , Odds Ratio , Retrospective Studies , Risk Factors , Time Factors
3.
Arterioscler Thromb Vasc Biol ; 32(10): 2468-74, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22879580

ABSTRACT

OBJECTIVE: Thromboxane A(2) and prostacyclin are thromboregulatory prostaglandins. The inflammatory C-reactive protein (CRP) promotes thrombosis after vascular injury, presumably via potentiation of thromboxane activity. Using a genetic approach, we investigated the role of thromboxane receptor (TP) pathway in CRP-induced thrombosis. METHODS AND RESULTS: Four genetically engineered mice strains were used: C57BL/6 wild-type, human CRP transgenic (CRPtg), thromboxane receptor-deficient (Tp(-/-)), and CRPtgTp(-/-) mice. CRP and TP expression were correlated, and suppression of CRP expression using small interfering RNA/CRP led to reduction in TP expression. Platelet-endothelial adherence was increased in CRPtg and suppressed in CRPtgTP(-/-)and CRPtg cells that were suppressed with TP small interfering RNA. TP deficiency in both platelets and endothelial cells was synergistic in affecting platelet-endothelial interactions. Time until arterial occlusion, measured after photochemical injury, was significantly shorter in CRPtg and prolonged in CRPtgTp(-/-) compared with controls (n=10-15, 35±3.4, 136±13.8, and 67±8.9 minutes, respectively; P<0.05). CONCLUSIONS: TP pathway is of major importance in CRP-induced thrombosis. The expression of TP is increased in CRPtg endothelial cells, and its blockade significantly suppresses the prothrombotic effect of CRP.


Subject(s)
C-Reactive Protein/physiology , Receptors, Thromboxane/physiology , Signal Transduction/physiology , Thrombosis/physiopathology , Adult , Animals , Blood Platelets/pathology , Blood Platelets/physiology , C-Reactive Protein/deficiency , C-Reactive Protein/genetics , Cell Adhesion/physiology , Disease Models, Animal , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , RNA, Small Interfering/pharmacology , Receptors, Thromboxane/deficiency , Receptors, Thromboxane/drug effects , Thrombosis/pathology , Transfection
4.
Am J Physiol Heart Circ Physiol ; 301(3): H730-6, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21685272

ABSTRACT

While data regarding the pathogenetic role of C-reactive protein (CRP) in atherothrombosis are accumulating, it is still controversial whether local CRP secretion is of any pathobiological significance. The present study examined whether endothelial-derived CRP modulates autocrine prothrombotic activity. Endothelial cells were isolated from hearts of mice transgenic to human CRP and grown in primary cultures. Human CRP expression was confirmed in these cells compared with no expression in cultures derived from wild-type congenes. Adhesion of human platelets to endothelial cells was studied in the "cone and plate" flow system. Platelet adhesion to cells expressing CRP was significantly increased compared with that in controls (n = 6, P < 0.01). The proadhesive effect of CRP was significantly suppressed in mouse heart endothelial cells and in human umbilical vein endothelial cells following treatment with small interfering RNA for human CRP. Adhesion was modulated by an increase in P-selectin. P-selectin expression correlated with a proadhesive phenotype, and blocking P-selectin with neutralizing antibody significantly decreased the adhesion of platelets to CRP-expressing cells (40.4 ± 10.5 to 9.4 ± 6.9 platelets/high-power field, n = 5 to 6, P < 0.01). In conclusion, human CRP that is locally produced in endothelial cells increases platelet adhesion to endothelial cells under normal shear flow conditions. These findings indicate that CRP exerts a local effect on endothelial cells via P-selectin expression, which promotes platelet adhesion and subsequent thrombus formation.


Subject(s)
C-Reactive Protein/metabolism , Endothelial Cells/metabolism , Platelet Adhesiveness , Thrombosis/metabolism , Analysis of Variance , Animals , Antibodies, Neutralizing/pharmacology , Autocrine Communication , C-Reactive Protein/genetics , Cells, Cultured , Endothelial Cells/drug effects , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , P-Selectin/antagonists & inhibitors , P-Selectin/immunology , P-Selectin/metabolism , RNA Interference , Regional Blood Flow , Thrombosis/blood , Thrombosis/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...