68 results on '"van Nieuwenhoven FA"'
Search Results
2. P97Cyclic stretch induces connective tissue growth factor expression in isolated adult rabbit cardiomyocytes
- Author
-
Van Nieuwenhoven, FA, Blaauw, E, Lorenzen-Schmidt, I, Babiker, FA, Munts, C, Prinzen, FW, Snoeckx, LH, Van Bilsen, M, and Van Der Vusse, GJ
- Published
- 2014
- Full Text
- View/download PDF
3. Long-term severe diabetes only leads to mild cardiac diastolic dysfunction in Zucker diabetic fatty rats.
- Author
-
Daniels A, Linz D, van Bilsen M, Rütten H, Sadowski T, Ruf S, Juretschke HP, Neumann-Haefelin C, Munts C, van der Vusse GJ, and van Nieuwenhoven FA
- Published
- 2012
4. P97 Cyclic stretch induces connective tissue growth factor expression in isolated adult rabbit cardiomyocytes.
- Author
-
Van Nieuwenhoven, FA, Blaauw, E, Lorenzen-Schmidt, I, Babiker, FA, Munts, C, Prinzen, FW, Snoeckx, LH, Van Bilsen, M, and Van Der Vusse, GJ
- Subjects
- *
CELL cycle , *CONNECTIVE tissues , *GROWTH factors , *PROTEIN expression , *CARDIAC hypertrophy , *EXTRACELLULAR matrix - Abstract
Introduction: A chronic increase in cardiac workload leads to hypertrophic growth of cardiomyocytes, often accompanied by accumulation of extracellular matrix (fibrosis). This structural remodeling of the myocardial wall impairs cardiac function and may eventually lead to heart failure. Connective Tissue Growth Factor (CTGF, CCN2) is considered to play an important role in cardiac structural remodeling. We studied whether stretch is a primary stimulus to induce CTGF expression in vivo in rabbit heart, and in vitro in isolated adult rabbit cardiomyocytes and fibroblasts.Methods: Cardiac function, structural remodeling and CTGF expression were studied in a rabbit model of combined pressure and volume overload. Furthermore, CTGF expression was investigated in cardiomyocytes and fibroblasts isolated from adult rabbit hearts, cultured and subjected to 10% cyclic equibiaxial stretch (1Hz) using the Flexcell FX4000 strain unit for up to 48 h.Results: Twenty weeks of combined volume and pressure overload resulted in eccentric left ventricular (LV) hypertrophy, with increased LV internal diameter (+36%) and LV weight (+53%). In the overloaded animals, myocardial CTGF mRNA levels were increased 5-fold and Western blot analyses showed a substantial increase in myocardial CTGF protein levels. In isolated adult rabbit cardiomyocytes, cyclic stretch strongly induced CTGF mRNA expression (2.9-fold at 48h), whereas CTGF-induction in cardiac fibroblasts was transient and modest (1.4-fold after 4h). Conditioned medium from stretched fibroblasts induced CTGF mRNA expression in non-stretched cardiomyocytes (2.3-fold at 48h).Conclusion: Our results show that cyclic stretch is a primary and strong stimulus to increase CTGF expression in isolated cardiomyocytes, and indicate that the elevated expression of CTGF in the overloaded heart is derived from mechanically stimulated cardiomyocytes. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
5. Plasma Extracellular Vesicles as Liquid Biopsy to Unravel the Molecular Mechanisms of Cardiac Reverse Remodeling Following Resynchronization Therapy?
- Author
-
van Nieuwenhoven FA, Schroen B, Barile L, van Middendorp L, Prinzen FW, and Auricchio A
- Abstract
Cardiac resynchronization therapy (CRT) has become a valuable addition to the treatment options for heart failure, in particular for patients with disturbances in electrical conduction that lead to regionally different contraction patterns (dyssynchrony). Dyssynchronous hearts show extensive molecular and cellular remodeling, which has primarily been investigated in experimental animals. Evidence showing that at least several miRNAs play a role in this remodeling is increasing. A comparison of results from measurements in plasma and myocardial tissue suggests that plasma levels of miRNAs may reflect the expression of these miRNAs in the heart. Because many miRNAs released in the plasma are included in extracellular vesicles (EVs), which protect them from degradation, measurement of myocardium-derived miRNAs in peripheral blood EVs may open new avenues to investigate and monitor (reverse) remodeling in dyssynchronous and resynchronized hearts of patients.
- Published
- 2023
- Full Text
- View/download PDF
6. Culturing of Cardiac Fibroblasts in Engineered Heart Matrix Reduces Myofibroblast Differentiation but Maintains Their Response to Cyclic Stretch and Transforming Growth Factor β1.
- Author
-
Ploeg MC, Munts C, Seddiqi T, Ten Brink TJL, Breemhaar J, Moroni L, Prinzen FW, and van Nieuwenhoven FA
- Abstract
Isolation and culturing of cardiac fibroblasts (CF) induces rapid differentiation toward a myofibroblast phenotype, which is partly mediated by the high substrate stiffness of the culture plates. In the present study, a 3D model of Engineered Heart Matrix (EHM) of physiological stiffness (Youngs modulus ~15 kPa) was developed using primary adult rat CF and a natural hydrogel collagen type 1 matrix. CF were equally distributed, viable and quiescent for at least 13 days in EHM and the baseline gene expression of myofibroblast-markers alfa-smooth muscle actin (Acta2), and connective tissue growth factor (Ctgf) was significantly lower, compared to CF cultured in 2D monolayers. CF baseline gene expression of transforming growth factor-beta1 (Tgfβ1) and brain natriuretic peptide (Nppb) was higher in EHM-fibers compared to the monolayers. EHM stimulation by 10% cyclic stretch (1 Hz) increased the gene expression of Nppb (3.0-fold), Ctgf (2.1-fold) and Tgfβ1 (2.3-fold) after 24 h. Stimulation of EHM with TGFβ1 (1 ng/mL, 24 h) induced Tgfβ1 (1.6-fold) and Ctgf (1.6-fold). In conclusion, culturing CF in EHM of physiological stiffness reduced myofibroblast marker gene expression, while the CF response to stretch or TGFβ1 was maintained, indicating that our novel EHM structure provides a good physiological model to study CF function and myofibroblast differentiation.
- Published
- 2022
- Full Text
- View/download PDF
7. The Complex Relation between Atrial Cardiomyopathy and Thrombogenesis.
- Author
-
D'Alessandro E, Winters J, van Nieuwenhoven FA, Schotten U, and Verheule S
- Subjects
- Animals, Blood Coagulation Factors, Heart Atria metabolism, Atrial Fibrillation, Cardiomyopathies pathology, Stroke pathology
- Abstract
Heart disease, as well as systemic metabolic alterations, can leave a 'fingerprint' of structural and functional changes in the atrial myocardium, leading to the onset of atrial cardiomyopathy. As demonstrated in various animal models, some of these changes, such as fibrosis, cardiomyocyte hypertrophy and fatty infiltration, can increase vulnerability to atrial fibrillation (AF), the most relevant manifestation of atrial cardiomyopathy in clinical practice. Atrial cardiomyopathy accompanying AF is associated with thromboembolic events, such as stroke. The interaction between AF and stroke appears to be far more complicated than initially believed. AF and stroke share many risk factors whose underlying pathological processes can reinforce the development and progression of both cardiovascular conditions. In this review, we summarize the main mechanisms by which atrial cardiomyopathy, preceding AF, supports thrombogenic events within the atrial cavity and myocardial interstitial space. Moreover, we report the pleiotropic effects of activated coagulation factors on atrial remodeling, which may aggravate atrial cardiomyopathy. Finally, we address the complex association between AF and stroke, which can be explained by a multidirectional causal relation between atrial cardiomyopathy and hypercoagulability., Competing Interests: The authors declare no conflict of interest.
- Published
- 2022
- Full Text
- View/download PDF
8. Left atrial remodeling in mitral regurgitation: A combined experimental-computational study.
- Author
-
Bouwmeester S, van Loon T, Ploeg M, Mast TP, Verzaal NJ, van Middendorp LB, Strik M, van Nieuwenhoven FA, Dekker LR, Prinzen FW, Lumens J, and Houthuizen P
- Subjects
- Animals, Bundle-Branch Block, Dogs, Echocardiography, Fibrosis, Heart Atria, Atrial Remodeling, Mitral Valve Insufficiency diagnostic imaging
- Abstract
Aims: Progressive changes to left atrial (LA) structure and function following mitral regurgitation (MR) remain incompletely understood. This study aimed to demonstrate potential underlying mechanisms using experimental canine models and computer simulations., Methods: A canine model of MR was created by cauterization of mitral chordae followed by radiofrequency ablation-induced left bundle-branch block (LBBB) after 4 weeks (MR-LBBB group). Animals with LBBB alone served as control. Echocardiography was performed at baseline, acutely after MR induction, and at 4 and 20 weeks, and correlated with histology and computer simulations., Results: Acute MR augmented LA reservoir and contractile strain (40±4 to 53±6% and -11±5 to -22±9% respectively, p<0.05). LA fractional area change increased significantly (47±4 to 56±4%, p<0.05) while LA end-systolic area remained unchanged (7.2±1.1 versus 7.9±1.1 cm2 respectively, p = 0.08). LA strain 'pseudonormalized' after 4 weeks and decompensated at 20 weeks with both strains decreasing to 25±6% and -3±2% respectively (p<0.05) together with a progressive increase in LA end-systolic area (7.2±1.1 to 14.0±6.3 cm2, p<0.05). In the LBBB-group, LA remodeling was less pronounced. Histology showed a trend towards increased interstitial fibrosis in the LA of the MR-LBBB group. Computer simulations indicated that the progressive changes in LA structure and function are a combination of progressive eccentric remodeling and fibrosis., Conclusion: MR augmented LA strain acutely to supranormal values without significant LA dilation. However, over time, LA strain gradually decreases (pseudornormal and decompensated) with LA dilation. Histology and computer simulations indicated a correlation to a varying degree of LA eccentric remodeling and fibrosis., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2022
- Full Text
- View/download PDF
9. A Lumped Two-Compartment Model for Simulation of Ventricular Pump and Tissue Mechanics in Ischemic Heart Disease.
- Author
-
Koopsen T, Van Osta N, Van Loon T, Van Nieuwenhoven FA, Prinzen FW, Van Klarenbosch BR, Kirkels FP, Teske AJ, Vernooy K, Delhaas T, and Lumens J
- Abstract
Introduction: Computational modeling of cardiac mechanics and hemodynamics in ischemic heart disease (IHD) is important for a better understanding of the complex relations between ischemia-induced heterogeneity of myocardial tissue properties, regional tissue mechanics, and hemodynamic pump function. We validated and applied a lumped two-compartment modeling approach for IHD integrated into the CircAdapt model of the human heart and circulation. Methods: Ischemic contractile dysfunction was simulated by subdividing a left ventricular (LV) wall segment into a hypothetical contractile and noncontractile compartment, and dysfunction severity was determined by the noncontractile volume fraction ( N C V F ). Myocardial stiffness was determined by the zero-passive stress length ( L s 0 , p a s ) and nonlinearity ( k E C M ) of the passive stress-sarcomere length relation of the noncontractile compartment. Simulated end-systolic pressure volume relations (ESPVRs) for 20% acute ischemia were qualitatively compared between a two- and one-compartment simulation, and parameters of the two-compartment model were tuned to previously published canine data of regional myocardial deformation during acute and prolonged ischemia and reperfusion. In six patients with myocardial infarction (MI), the N C V F was automatically estimated using the echocardiographic LV strain and volume measurements obtained acutely and 6 months after MI. Estimated segmental N C V F values at the baseline and 6-month follow-up were compared with percentage late gadolinium enhancement (LGE) at 6-month follow-up. Results: Simulation of 20% of N C V F shifted the ESPVR rightward while moderately reducing the slope, while a one-compartment simulation caused a leftward shift with severe reduction in the slope. Through tuning of the N C V F , L s 0 , p a s , and k E C M , it was found that manipulation of the N C V F alone reproduced the deformation during acute ischemia and reperfusion, while additional manipulations of L s 0 , p a s and k E C M were required to reproduce deformation during prolonged ischemia and reperfusion. Out of all segments with LGE>25% at the follow-up, the majority (68%) had higher estimated N C V F at the baseline than at the follow-up. Furthermore, the baseline N C V F correlated better with percentage LGE than N C V F did at the follow-up. Conclusion: We successfully used a two-compartment model for simulation of the ventricular pump and tissue mechanics in IHD. Patient-specific optimizations using regional myocardial deformation estimated the N C V F in a small cohort of MI patients in the acute and chronic phase after MI, while estimated N C V F values closely approximated the extent of the myocardial scar at the follow-up. In future studies, this approach can facilitate deformation imaging-based estimation of myocardial tissue properties in patients with cardiovascular diseases., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Koopsen, Van Osta, Van Loon, Van Nieuwenhoven, Prinzen, Van Klarenbosch, Kirkels, Teske, Vernooy, Delhaas and Lumens.)
- Published
- 2022
- Full Text
- View/download PDF
10. Thrombin generation by calibrated automated thrombography in goat plasma: Optimization of an assay.
- Author
-
D'Alessandro E, Scaf B, van Oerle R, van Nieuwenhoven FA, van Hunnik A, Verheule S, Schotten U, Ten Cate H, and Spronk HMH
- Abstract
The goat model of atrial fibrillation (AF) allows investigation of the effect of AF on coagulation. However, assays for goat plasma are not available from commercial sources. Calibrated automated thrombography (CAT) provides a global view of the coagulation profile by assessing in vitro thrombin generation (TG). We describe the customization of the CAT assay in goat platelet-poor plasma (PPP) and in factor Xa (FXa)-inhibitor-anticoagulated PPP. TG was initiated in the presence of phospholipids and either (a) PPP reagent, reagent low, or reagent high; (b) goat brain protein extraction (GBP); or (c) Russell's viper venom-factor X activator (RVV-X). Contact activation was assessed by adding corn trypsin inhibitor. Different concentrations of prothrombin complex concentrate (PCC) were used to determine the sensitivity of both the GBP and RVV-X method. To obtain FXa-inhibitor anticoagulated plasma, rivaroxaban was added to plasma. TG settings with human reagents were not suitable for goat plasma. TG triggered with GBP increased peak height and ETP values. Similarly, the RVV-X method produced comparable TG curves and was more sensitive to PCC titration. Finally, both methods were able to detect the decrease in clotting potential induced by FXa inhibition. This is the first study that reports the customization of the CAT assay for goats. The GBP and RVV-X methods were comparable in triggering TG in goat plasma. The RVV-X method seemed to better discriminate changes in TG curves due to increases in clotting potential as well as to FXa inhibition by rivaroxaban in goat plasma., (© 2021 The Authors. Research and Practice in Thrombosis and Haemostasis published by Wiley Periodicals LLC on behalf of International Society on Thrombosis and Haemostasis (ISTH).)
- Published
- 2021
- Full Text
- View/download PDF
11. Specific amino acid supplementation rescues the heart from lipid overload-induced insulin resistance and contractile dysfunction by targeting the endosomal mTOR-v-ATPase axis.
- Author
-
Wang S, Schianchi F, Neumann D, Wong LY, Sun A, van Nieuwenhoven FA, Zeegers MP, Strzelecka A, Col U, Glatz JFC, Nabben M, and Luiken JJFP
- Subjects
- Amino Acids administration & dosage, Animals, Diet, High-Fat, Dietary Supplements, Endosomes drug effects, Endosomes metabolism, Insulin Resistance, Lipids adverse effects, Male, Myocardial Contraction drug effects, Myocytes, Cardiac metabolism, Rats, Rats, Inbred Lew, Amino Acids metabolism, Myocytes, Cardiac drug effects, TOR Serine-Threonine Kinases metabolism, Vacuolar Proton-Translocating ATPases metabolism
- Abstract
Objective: The diabetic heart is characterized by extensive lipid accumulation which often leads to cardiac contractile dysfunction. The underlying mechanism involves a pivotal role for vacuolar-type H
+ -ATPase (v-ATPase, functioning as endosomal/lysosomal proton pump). Specifically, lipid oversupply to the heart causes disassembly of v-ATPase and endosomal deacidification. Endosomes are storage compartments for lipid transporter CD36. However, upon endosomal deacidification, CD36 is expelled to translocate to the sarcolemma, thereby inducing myocardial lipid accumulation, insulin resistance, and contractile dysfunction. Hence, the v-ATPase assembly may be a suitable target for ameliorating diabetic cardiomyopathy. Another function of v-ATPase involves the binding of anabolic master-regulator mTORC1 to endosomes, a prerequisite for the activation of mTORC1 by amino acids (AAs). We examined whether the relationship between v-ATPase and mTORC1 also operates reciprocally; specifically, whether AA induces v-ATPase reassembly in a mTORC1-dependent manner to prevent excess lipids from entering and damaging the heart., Methods: Lipid overexposed rodent/human cardiomyocytes and high-fat diet-fed rats were treated with a specific cocktail of AAs (lysine/leucine/arginine). Then, v-ATPase assembly status/activity, cell surface CD36 content, myocellular lipid uptake/accumulation, insulin sensitivity, and contractile function were measured. To elucidate underlying mechanisms, specific gene knockdown was employed, followed by subcellular fractionation, and coimmunoprecipitation., Results: In lipid-overexposed cardiomyocytes, lysine/leucine/arginine reinternalized CD36 to the endosomes, prevented/reversed lipid accumulation, preserved/restored insulin sensitivity, and contractile function. These beneficial AA actions required the mTORC1-v-ATPase axis, adaptor protein Ragulator, and endosomal/lysosomal AA transporter SLC38A9, indicating an endosome-centric inside-out AA sensing mechanism. In high-fat diet-fed rats, lysine/leucine/arginine had similar beneficial actions at the myocellular level as in vitro in lipid-overexposed cardiomyocytes and partially reversed cardiac hypertrophy., Conclusion: Specific AAs acting through v-ATPase reassembly reduce cardiac lipid uptake raising the possibility for treatment in situations of lipid overload and associated insulin resistance., (Copyright © 2021 The Author(s). Published by Elsevier GmbH.. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
12. Coagulation Factor Xa Induces Proinflammatory Responses in Cardiac Fibroblasts via Activation of Protease-Activated Receptor-1.
- Author
-
D'Alessandro E, Scaf B, Munts C, van Hunnik A, Trevelyan CJ, Verheule S, Spronk HMH, Turner NA, Ten Cate H, Schotten U, and van Nieuwenhoven FA
- Subjects
- Adult, Animals, Cattle, Cell Proliferation, Chemokine CCL2 genetics, Chemokine CCL2 metabolism, Fibroblasts metabolism, Heart Atria pathology, Humans, Interleukin-6 genetics, Interleukin-6 metabolism, Rats, Wistar, Receptor, PAR-1 agonists, Receptor, PAR-1 genetics, Thrombin metabolism, Up-Regulation genetics, Rats, Factor Xa metabolism, Fibroblasts pathology, Inflammation pathology, Myocardium metabolism, Receptor, PAR-1 metabolism
- Abstract
Coagulation factor (F) Xa induces proinflammatory responses through activation of protease-activated receptors (PARs). However, the effect of FXa on cardiac fibroblasts (CFs) and the contribution of PARs in FXa-induced cellular signalling in CF has not been fully characterised. To answer these questions, human and rat CFs were incubated with FXa (or TRAP-14, PAR-1 agonist). Gene expression of pro-fibrotic and proinflammatory markers was determined by qRT-PCR after 4 and 24 h. Gene silencing of F2R (PAR-1) and F2RL1 (PAR-2) was achieved using siRNA. MCP-1 protein levels were measured by ELISA of FXa-conditioned media at 24 h. Cell proliferation was assessed after 24 h of incubation with FXa ± SCH79797 (PAR-1 antagonist). In rat CFs, FXa induced upregulation of Ccl2 (MCP-1; >30-fold at 4 h in atrial and ventricular CF) and Il6 (IL-6; ±7-fold at 4 h in ventricular CF). Increased MCP-1 protein levels were detected in FXa-conditioned media at 24 h. In human CF, FXa upregulated the gene expression of CCL2 (>3-fold) and IL6 (>4-fold) at 4 h. Silencing of F2R (PAR-1 gene), but not F2RL1 (PAR-2 gene), downregulated this effect. Selective activation of PAR-1 by TRAP-14 increased CCL2 and IL6 gene expression; this was prevented by F2R (PAR-1 gene) knockdown. Moreover, SCH79797 decreased FXa-induced proliferation after 24 h. In conclusion, our study shows that FXa induces overexpression of proinflammatory genes in human CFs via PAR-1, which was found to be the most abundant PARs isoform in this cell type.
- Published
- 2021
- Full Text
- View/download PDF
13. Piezo1 Mechanosensitive Ion Channel Mediates Stretch-Induced Nppb Expression in Adult Rat Cardiac Fibroblasts.
- Author
-
Ploeg MC, Munts C, Prinzen FW, Turner NA, van Bilsen M, and van Nieuwenhoven FA
- Subjects
- Animals, Fibroblasts drug effects, Fibrosis, Membrane Proteins genetics, Myocytes, Cardiac drug effects, Myocytes, Cardiac metabolism, RNA, Messenger genetics, RNA, Messenger metabolism, Rats, Receptors, Atrial Natriuretic Factor metabolism, Recombinant Proteins pharmacology, Fibroblasts metabolism, Gene Expression Regulation drug effects, Membrane Proteins metabolism, Myocardium cytology, Receptors, Atrial Natriuretic Factor genetics, Stress, Mechanical
- Abstract
In response to stretch, cardiac tissue produces natriuretic peptides, which have been suggested to have beneficial effects in heart failure patients. In the present study, we explored the mechanism of stretch-induced brain natriuretic peptide (Nppb) expression in cardiac fibroblasts. Primary adult rat cardiac fibroblasts subjected to 4 h or 24 h of cyclic stretch (10% 1 Hz) showed a 6.6-fold or 3.2-fold ( p < 0.05) increased mRNA expression of Nppb, as well as induction of genes related to myofibroblast differentiation. Moreover, BNP protein secretion was upregulated 5.3-fold in stretched cardiac fibroblasts. Recombinant BNP inhibited TGFβ1-induced Acta2 expression. Nppb expression was >20-fold higher in cardiomyocytes than in cardiac fibroblasts, indicating that cardiac fibroblasts were not the main source of Nppb in the healthy heart. Yoda1, an agonist of the Piezo1 mechanosensitive ion channel, increased Nppb expression 2.1-fold ( p < 0.05) and significantly induced other extracellular matrix (ECM) remodeling genes. Silencing of Piezo1 reduced the stretch-induced Nppb and Tgfb1 expression in cardiac fibroblasts. In conclusion, our study identifies Piezo1 as mediator of stretch-induced Nppb expression, as well as other remodeling genes, in cardiac fibroblasts.
- Published
- 2021
- Full Text
- View/download PDF
14. Mechanically activated Piezo1 channels of cardiac fibroblasts stimulate p38 mitogen-activated protein kinase activity and interleukin-6 secretion.
- Author
-
Blythe NM, Muraki K, Ludlow MJ, Stylianidis V, Gilbert HTJ, Evans EL, Cuthbertson K, Foster R, Swift J, Li J, Drinkhill MJ, van Nieuwenhoven FA, Porter KE, Beech DJ, and Turner NA
- Subjects
- Animals, Calcium Signaling genetics, Endopeptidases genetics, Endothelial Cells chemistry, Endothelial Cells metabolism, Fibroblasts metabolism, Gene Expression Regulation genetics, Gene Knockdown Techniques, Humans, Interleukin-6 chemistry, Ion Channels chemistry, MAP Kinase Signaling System genetics, Mechanotransduction, Cellular genetics, Mice, Myocardium chemistry, Phosphorylation genetics, RNA, Messenger genetics, RNA, Small Interfering genetics, Signal Transduction genetics, Thiolester Hydrolases genetics, p38 Mitogen-Activated Protein Kinases chemistry, Interleukin-6 genetics, Ion Channels genetics, Myocardium metabolism, p38 Mitogen-Activated Protein Kinases genetics
- Abstract
Piezo1 is a mechanosensitive cation channel with widespread physiological importance; however, its role in the heart is poorly understood. Cardiac fibroblasts help preserve myocardial integrity and play a key role in regulating its repair and remodeling following stress or injury. Here we investigated Piezo1 expression and function in cultured human and mouse cardiac fibroblasts. RT-PCR experiments confirmed that Piezo1 mRNA in cardiac fibroblasts is expressed at levels similar to those in endothelial cells. The results of a Fura-2 intracellular Ca
2+ assay validated Piezo1 as a functional ion channel that is activated by its agonist, Yoda1. Yoda1-induced Ca2+ entry was inhibited by Piezo1 blockers (gadolinium and ruthenium red) and was reduced proportionally by siRNA-mediated Piezo1 knockdown or in murine Piezo1+/- cells. Results from cell-attached patch clamp recordings on human cardiac fibroblasts established that they contain mechanically activated ion channels and that their pressure responses are reduced by Piezo1 knockdown. Investigation of Yoda1 effects on selected remodeling genes indicated that Piezo1 activation increases both mRNA levels and protein secretion of IL-6, a pro-hypertrophic and profibrotic cytokine, in a Piezo1-dependent manner. Moreover, Piezo1 knockdown reduced basal IL-6 expression from cells cultured on softer collagen-coated substrates. Multiplex kinase activity profiling combined with kinase inhibitor experiments and phosphospecific immunoblotting established that Piezo1 activation stimulates IL-6 secretion via the p38 mitogen-activated protein kinase downstream of Ca2+ entry. In summary, cardiac fibroblasts express mechanically activated Piezo1 channels coupled to secretion of the paracrine signaling molecule IL-6. Piezo1 may therefore be important in regulating cardiac remodeling., (© 2019 Blythe et al.)- Published
- 2019
- Full Text
- View/download PDF
15. Pathobiology of cardiac dyssynchrony and resynchronization therapy.
- Author
-
Nguyên UC, Verzaal NJ, van Nieuwenhoven FA, Vernooy K, and Prinzen FW
- Subjects
- Action Potentials, Animals, Arrhythmias, Cardiac physiopathology, Heart Conduction System physiopathology, Heart Failure physiopathology, Heart Rate, Humans, Recovery of Function, Time Factors, Treatment Outcome, Ventricular Dysfunction, Left physiopathology, Arrhythmias, Cardiac therapy, Cardiac Resynchronization Therapy adverse effects, Heart Failure therapy, Myocardial Contraction, Ventricular Dysfunction, Left therapy, Ventricular Function, Left, Ventricular Remodeling
- Abstract
Synchronous ventricular electrical activation is a prerequisite for adequate left ventricular (LV) systolic function. Conduction abnormalities such as left bundle branch block, and ventricular pacing lead to a dyssynchronous electrical activation sequence, which may have deleterious consequences. The present review attempts to connect the various processes involved in the development of 'dyssynchronopathy', and its correction by cardiac resynchronization therapy (CRT). Abnormal electrical impulse conduction leads to abnormal contraction, characterized by regional differences in timing as well as shortening patterns and amount of external work performed. Early activated regions may show 'wasted work', which leads to inefficient action of the entire left ventricle. Moreover, both the development of heart failure (HF) in general and the regional differences in mechanical load lead to structural, electrical, and contractile remodelling processes. These have been demonstrated at the level of the myocardium (asymmetric hypertrophy, fibrosis, prolongation of activation and reduction in repolarization forces, decrease in LV ejection fraction), cell (gap junctional remodelling, derangement of the T-tubular structure), and molecule (under or overexpression of ion channels and contractile proteins subtypes and abnormal calcium handling). The myocardial adaptations to dyssynchrony are 'maladaptive'. This also explains why CRT, unlike most pharmacological treatments, continues to increase its therapeutic effect over time. Finally, better understanding of all processes involved in dyssynchrony and CRT may also lead to new pharmacological agents for treating HF and to novel pacing strategies.
- Published
- 2018
- Full Text
- View/download PDF
16. MicroRNA-221/222 Family Counteracts Myocardial Fibrosis in Pressure Overload-Induced Heart Failure.
- Author
-
Verjans R, Peters T, Beaumont FJ, van Leeuwen R, van Herwaarden T, Verhesen W, Munts C, Bijnen M, Henkens M, Diez J, de Windt LJ, van Nieuwenhoven FA, van Bilsen M, Goumans MJ, Heymans S, González A, and Schroen B
- Subjects
- Animals, Aortic Valve Stenosis complications, Aortic Valve Stenosis metabolism, Cardiomyopathies metabolism, Fibroblasts metabolism, Fibrosis metabolism, Humans, Male, Mice, Mice, Inbred C57BL, Myocardium pathology, Proto-Oncogene Mas, Rats, Signal Transduction, Transforming Growth Factor beta metabolism, Heart Failure metabolism, MicroRNAs metabolism, Myocardium metabolism
- Abstract
Pressure overload causes cardiac fibroblast activation and transdifferentiation, leading to increased interstitial fibrosis formation and subsequently myocardial stiffness, diastolic and systolic dysfunction, and eventually heart failure. A better understanding of the molecular mechanisms underlying pressure overload-induced cardiac remodeling and fibrosis will have implications for heart failure treatment strategies. The microRNA (miRNA)-221/222 family, consisting of miR-221-3p and miR-222-3p, is differentially regulated in mouse and human cardiac pathology and inversely associated with kidney and liver fibrosis. We investigated the role of this miRNA family during pressure overload-induced cardiac remodeling. In myocardial biopsies of patients with severe fibrosis and dilated cardiomyopathy or aortic stenosis, we found significantly lower miRNA-221/222 levels as compared to matched patients with nonsevere fibrosis. In addition, miRNA-221/222 levels in aortic stenosis patients correlated negatively with the extent of myocardial fibrosis and with left ventricular stiffness. Inhibition of both miRNAs during AngII (angiotensin II)-mediated pressure overload in mice led to increased fibrosis and aggravated left ventricular dilation and dysfunction. In rat cardiac fibroblasts, inhibition of miRNA-221/222 derepressed TGF-β (transforming growth factor-β)-mediated profibrotic SMAD2 (mothers against decapentaplegic homolog 2) signaling and downstream gene expression, whereas overexpression of both miRNAs blunted TGF-β-induced profibrotic signaling. We found that the miRNA-221/222 family may target several genes involved in TGF-β signaling, including JNK1 (c-Jun N-terminal kinase 1), TGF-β receptor 1 and TGF-β receptor 2, and ETS-1 (ETS proto-oncogene 1). Our findings show that heart failure-associated downregulation of the miRNA-221/222 family enables profibrotic signaling in the pressure-overloaded heart., (© 2017 American Heart Association, Inc.)
- Published
- 2018
- Full Text
- View/download PDF
17. Cartilage intermediate layer protein 1 (CILP1): A novel mediator of cardiac extracellular matrix remodelling.
- Author
-
van Nieuwenhoven FA, Munts C, Op't Veld RC, González A, Díez J, Heymans S, Schroen B, and van Bilsen M
- Subjects
- Animals, Extracellular Matrix Proteins genetics, Fibroblasts cytology, Fibroblasts metabolism, HEK293 Cells, Humans, Mice, Mice, Inbred C57BL, Myocardium pathology, Pyrophosphatases genetics, Transforming Growth Factor beta metabolism, Extracellular Matrix metabolism, Extracellular Matrix Proteins metabolism, Myocardium metabolism, Pyrophosphatases metabolism
- Abstract
Heart failure is accompanied by extracellular matrix (ECM) remodelling, often leading to cardiac fibrosis. In the present study we explored the significance of cartilage intermediate layer protein 1 (CILP1) as a novel mediator of cardiac ECM remodelling. Whole genome transcriptional analysis of human cardiac tissue samples revealed a strong association of CILP1 with many structural (e.g. COL1A2 r
2 = 0.83) and non-structural (e.g. TGFB3 r2 = 0.75) ECM proteins. Gene enrichment analysis further underscored the involvement of CILP1 in human cardiac ECM remodelling and TGFβ signalling. Myocardial CILP1 protein levels were significantly elevated in human infarct tissue and in aortic valve stenosis patients. CILP1 mRNA levels markedly increased in mouse heart after myocardial infarction, transverse aortic constriction, and angiotensin II treatment. Cardiac fibroblasts were found to be the primary source of cardiac CILP1 expression. Recombinant CILP1 inhibited TGFβ-induced αSMA gene and protein expression in cardiac fibroblasts. In addition, CILP1 overexpression in HEK293 cells strongly (5-fold p < 0.05) inhibited TGFβ signalling activity. In conclusion, our study identifies CILP1 as a new cardiac matricellular protein interfering with pro-fibrotic TGFβ signalling, and as a novel sensitive marker for cardiac fibrosis.- Published
- 2017
- Full Text
- View/download PDF
18. Local microRNA-133a downregulation is associated with hypertrophy in the dyssynchronous heart.
- Author
-
van Middendorp LB, Kuiper M, Munts C, Wouters P, Maessen JG, van Nieuwenhoven FA, and Prinzen FW
- Abstract
Aims: Left bundle branch block (LBBB) creates considerable regional differences in mechanical load within the left ventricle (LV). We investigated expression of selected microRNAs (miRs) in relation to regional hypertrophy and fibrosis in LBBB hearts and their reversibility upon cardiac resynchronization therapy (CRT)., Methods and Results: Eighteen dogs were followed for 4 months after induction of LBBB, 10 of which received CRT after 2 months. Five additional dogs served as control. LV geometric changes were determined by echocardiography and myocardial strain by magnetic resonance imaging tagging. Expression levels of miRs, their target genes: connective tissue growth factor (CTGF), serum response factor (SRF), nuclear factor of activated T cells (NFATc4), and cardiomyocyte diameter and collagen deposition were measured in the septum and LV free wall (LVfw). In LBBB hearts, LVfw and septal systolic circumferential strain were 200% and 50% of control, respectively. This coincided with local hypertrophy in the LVfw. MiR-133a expression was reduced by 33% in the LVfw, which corresponded with a selective increase of CTGF expression in the LVfw (279% of control). By contrast, no change was observed in SRF and NFATc4 expression was decreased in LBBB hearts. CRT normalized strain patterns and reversed miR-133a and CTGF expression towards normal, expression of other miRs, related to remodelling, such as miR-199b and miR-155f, were not affected., Conclusions: In the clinically relevant large animal model of LBBB, a close inverse relation exists between local hypertrophy and miR-133a. Reduced miR-133a correlated with increased CTGF levels but not with SRF and NFATc4., (© 2017 The Authors ESC Heart Failure published by John Wiley & Sons Ltd on behalf of European Society of Cardiology.)
- Published
- 2017
- Full Text
- View/download PDF
19. Hypertension is a conditional factor for the development of cardiac hypertrophy in type 2 diabetic mice.
- Author
-
van Bilsen M, Daniels A, Brouwers O, Janssen BJ, Derks WJ, Brouns AE, Munts C, Schalkwijk CG, van der Vusse GJ, and van Nieuwenhoven FA
- Subjects
- AMP-Activated Protein Kinases genetics, AMP-Activated Protein Kinases metabolism, Animals, Blood Pressure drug effects, Cell Size, Diabetes Mellitus, Type 2 diagnostic imaging, Diabetes Mellitus, Type 2 metabolism, Dobutamine pharmacology, Gene Expression, Glycation End Products, Advanced metabolism, Hypertension chemically induced, Hypertension diagnostic imaging, Hypertension metabolism, Hypertrophy, Left Ventricular diagnostic imaging, Hypertrophy, Left Ventricular metabolism, Male, Mice, Myocardium metabolism, Myocardium pathology, Myocytes, Cardiac drug effects, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology, Time Factors, Ultrasonography, Ventricular Function, Left drug effects, Ventricular Remodeling drug effects, Angiotensin II adverse effects, Diabetes Mellitus, Type 2 pathology, Hypertension pathology, Hypertrophy, Left Ventricular pathology
- Abstract
Background: Type 2 diabetes is frequently associated with co-morbidities, including hypertension. Here we investigated if hypertension is a critical factor in myocardial remodeling and the development of cardiac dysfunction in type 2 diabetic db/db mice., Methods: Thereto, 14-wks-old male db/db mice and non-diabetic db/+ mice received vehicle or angiotensin II (AngII) for 4 wks to induce mild hypertension (n = 9-10 per group). Left ventricular (LV) function was assessed by serial echocardiography and during a dobutamine stress test. LV tissue was subjected to molecular and (immuno)histochemical analysis to assess effects on hypertrophy, fibrosis and inflammation., Results: Vehicle-treated diabetic mice neither displayed marked myocardial structural remodeling nor cardiac dysfunction. AngII-treatment did not affect body weight and fasting glucose levels, and induced a comparable increase in blood pressure in diabetic and control mice. Nonetheless, AngII-induced LV hypertrophy was significantly more pronounced in diabetic than in control mice as assessed by LV mass (increase +51% and +34%, respectively, p<0.01) and cardiomyocyte size (+53% and +31%, p<0.001). This was associated with enhanced LV mRNA expression of markers of hypertrophy and fibrosis and reduced activation of AMP-activated protein kinase (AMPK), while accumulation of Advanced Glycation End products (AGEs) and the expression levels of markers of inflammation were not altered. Moreover, AngII-treatment reduced LV fractional shortening and contractility in diabetic mice, but not in control mice., Conclusions: Collectively, the present findings indicate that type 2 diabetes in its early stage is not yet associated with adverse cardiac structural changes, but already renders the heart more susceptible to hypertension-induced hypertrophic remodeling.
- Published
- 2014
- Full Text
- View/download PDF
20. Combined effects of interleukin-1α and transforming growth factor-β1 on modulation of human cardiac fibroblast function.
- Author
-
van Nieuwenhoven FA, Hemmings KE, Porter KE, and Turner NA
- Subjects
- Blotting, Western, Collagen Type III metabolism, Fibroblasts metabolism, Humans, Interleukin-1alpha genetics, Interleukin-6 metabolism, Interleukin-8 metabolism, Matrix Metalloproteinase 3 metabolism, Myocardium metabolism, Real-Time Polymerase Chain Reaction, Reverse Transcriptase Polymerase Chain Reaction, Transforming Growth Factor beta1 genetics, Fibroblasts physiology, Gene Expression Regulation genetics, Interleukin-1alpha metabolism, Myocardium cytology, Signal Transduction physiology, Transforming Growth Factor beta1 metabolism
- Abstract
During cardiac remodeling, cardiac fibroblasts (CF) are influenced by increased levels of interleukin-1α (IL-1α) and transforming growth factor-β1 (TGFβ1). The present study investigated the interaction between these two important cytokines on function of human CF and their differentiation to myofibroblasts (CMF). CF were isolated from human atrial appendage and exposed to IL-1α and/or TGFβ1 (both 0.1 ng/ml). mRNA expression levels of selected genes were determined after 6-24h by real-time RT-PCR, while protein levels were analyzed at 24-48 h by ELISA or western blot. Activation of canonical signaling pathways (NFκB, Smad3, p38 MAPK) was determined by western blotting. Differentiation to CMF was examined by collagen gel contraction assays. Exposure of CF to IL-1α alone enhanced levels of IL-6, IL-8, matrix metalloproteinase-3 (MMP3) and collagen III (COL3A1), but reduced the CMF markers α-smooth muscle actin (αSMA) and connective tissue growth factor (CTGF/CCN2). By contrast, TGFβ1 alone had minor effects on IL-6, IL-8 and MMP3 levels, but significantly increased levels of the CMF markers αSMA, CTGF, COL1A1 and COL3A1. Co-stimulation with both IL-1α and TGFβ1 increased MMP3 expression synergistically. Furthermore, while TGFβ1 had no effect on IL-1α-induced IL-6 or IL-8 levels, co-stimulation inhibited the TGFβ1-induced increase in αSMA and blocked the gel contraction caused by TGFβ1. Combining IL-1α and TGFβ1 had no apparent effect on their canonical signaling pathways. In conclusion, IL-1α and TGFβ1 act synergistically to stimulate MMP3 expression in CF. Moreover, IL-1α has a dominant inhibitory effect on the phenotypic switch of CF to CMF induced by TGFβ1., (Copyright © 2013 International Society of Matrix Biology. Published by Elsevier B.V. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
21. Stretch-induced upregulation of connective tissue growth factor in rabbit cardiomyocytes.
- Author
-
Blaauw E, Lorenzen-Schmidt I, Babiker FA, Munts C, Prinzen FW, Snoeckx LH, van Bilsen M, van der Vusse GJ, and van Nieuwenhoven FA
- Subjects
- Animals, Cells, Cultured, Connective Tissue Growth Factor genetics, Culture Media, Conditioned metabolism, Disease Models, Animal, Female, Fibroblasts metabolism, Hemodynamics, Hypertrophy, Left Ventricular pathology, Hypertrophy, Left Ventricular physiopathology, Myocytes, Cardiac pathology, RNA, Messenger metabolism, Rabbits, Time Factors, Transforming Growth Factor beta1 metabolism, Up-Regulation, Ventricular Function, Left, Ventricular Pressure, Connective Tissue Growth Factor metabolism, Hypertrophy, Left Ventricular metabolism, Mechanotransduction, Cellular, Myocytes, Cardiac metabolism, Ventricular Remodeling
- Abstract
Connective Tissue Growth Factor (CTGF, CCN2) is considered to play an important role in cardiac remodelling. We studied whether stretch is a primary stimulus to induce CTGF expression in vivo in rabbit heart, and in vitro in isolated cardiomyocytes and fibroblasts. Twenty weeks of combined volume and pressure overload resulted in eccentric left ventricular (LV) hypertrophy, with increased LV internal diameter (+36 %) and LV weight (+53 %). Myocardial CTGF mRNA and protein levels were substantially increased in the overloaded animals. In isolated adult rabbit cardiomyocytes, cyclic stretch strongly induced CTGF mRNA expression (2.9-fold at 48 h), whereas in cardiac fibroblasts CTGF-induction was transient and modest (1.4-fold after 4 h). Conditioned medium from stretched fibroblasts induced CTGF mRNA expression in non-stretched cardiomyocytes (2.3-fold at 48 h). Our findings indicate that stretch is an important primary trigger for CTGF-induction in the overloaded heart.
- Published
- 2013
- Full Text
- View/download PDF
22. CCN2/CTGF is required for matrix organization and to protect growth plate chondrocytes from cellular stress.
- Author
-
Hall-Glenn F, Aivazi A, Akopyan L, Ong JR, Baxter RR, Benya PD, Goldschmeding R, van Nieuwenhoven FA, Hunziker EB, and Lyons KM
- Abstract
CCN2 (connective tissue growth factor (CTGF/CCN2)) is a matricellular protein that utilizes integrins to regulate cell proliferation, migration and survival. The loss of CCN2 leads to perinatal lethality resulting from a severe chondrodysplasia. Upon closer inspection of Ccn2 mutant mice, we observed defects in extracellular matrix (ECM) organization and hypothesized that the severe chondrodysplasia caused by loss of CCN2 might be associated with defective chondrocyte survival. Ccn2 mutant growth plate chondrocytes exhibited enlarged endoplasmic reticula (ER), suggesting cellular stress. Immunofluorescence analysis confirmed elevated stress in Ccn2 mutants, with reduced stress observed in Ccn2 overexpressing transgenic mice. In vitro studies revealed that Ccn2 is a stress responsive gene in chondrocytes. The elevated stress observed in Ccn2-/- chondrocytes is direct and mediated in part through integrin α5. The expression of the survival marker NFκB and components of the autophagy pathway were decreased in Ccn2 mutant growth plates, suggesting that CCN2 may be involved in mediating chondrocyte survival. These data demonstrate that absence of a matricellular protein can result in increased cellular stress and highlight a novel protective role for CCN2 in chondrocyte survival. The severe chondrodysplasia caused by the loss of CCN2 may be due to increased chondrocyte stress and defective activation of autophagy pathways, leading to decreased cellular survival. These effects may be mediated through nuclear factor κB (NFκB) as part of a CCN2/integrin/NFκB signaling cascade.
- Published
- 2013
- Full Text
- View/download PDF
23. The role of cardiac fibroblasts in the transition from inflammation to fibrosis following myocardial infarction.
- Author
-
van Nieuwenhoven FA and Turner NA
- Subjects
- Animals, Chemokines metabolism, Cytokines metabolism, Fibrosis pathology, Humans, Intercellular Signaling Peptides and Proteins metabolism, Myocardium metabolism, Myocardium pathology, Myofibroblasts metabolism, Neovascularization, Physiologic, Ventricular Remodeling, Fibroblasts metabolism, Inflammation pathology, Myocardial Infarction physiopathology
- Abstract
Cardiac fibroblasts (CF) play a pivotal role in the repair and remodeling of the heart that occur following myocardial infarction (MI). The transition through the inflammatory, granulation and maturation phases of infarct healing is driven by cellular responses to local levels of cytokines, chemokines and growth factors that fluctuate in a temporal and spatial manner. In the acute inflammatory phase early after MI, CF contribute to the inflammatory milieu through increased secretion of proinflammatory cytokines and chemokines, and they promote extracellular matrix (ECM) degradation by increasing matrix metalloproteinase (MMP) expression and activity. In the granulation phase, CF migrate into the infarct zone, proliferate and produce MMPs and pro-angiogenic molecules to facilitate revascularization. Fibroblasts also undergo a phenotypic change to become myofibroblasts. In the maturation phase, inflammation is reduced by anti-inflammatory cytokines, and increased levels of profibrotic stimuli induce myofibroblasts to synthesize new ECM to form a scar. The scar is contracted through the mechanical force generated by myofibroblasts, preventing cardiac dilation. In this review we discuss the transition from myocardial inflammation to fibrosis with particular focus on how CF respond to alterations in proinflammatory and profibrotic signals. By furthering our understanding of these events, it is hoped that new therapeutic interventions will be developed that selectively reduce adverse myocardial remodeling post-MI, while sparing essential repair mechanisms., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
24. PPARs as therapeutic targets in cardiovascular disease.
- Author
-
van Bilsen M and van Nieuwenhoven FA
- Subjects
- Animals, Atherosclerosis physiopathology, Diabetes Mellitus, Type 2 drug therapy, Diabetes Mellitus, Type 2 physiopathology, Heart Diseases physiopathology, Humans, Inflammation metabolism, Ligands, Myocardial Ischemia drug therapy, Myocardial Ischemia physiopathology, PPAR alpha agonists, PPAR alpha metabolism, PPAR delta agonists, PPAR delta metabolism, PPAR gamma agonists, PPAR gamma metabolism, Peroxisome Proliferator-Activated Receptors chemistry, Peroxisome Proliferator-Activated Receptors metabolism, Transcription Factors agonists, Transcription Factors metabolism, Atherosclerosis drug therapy, Heart Diseases drug therapy, Peroxisome Proliferator-Activated Receptors agonists
- Abstract
Importance of the Field: The role of peroxisome proliferator-activated receptors PPARα, PPARδ and PPARγ in cardiovascular disease is receiving widespread attention. As ligand-activated nuclear receptors, they play a role in regulation of lipid and glucose metabolism. This feature of the PPARs has been successfully exploited to treat systemic metabolic diseases, like hyperlipidemia and type-2 diabetes. Indirectly, their lipid lowering effect also leads to a reduction of the risk for cardiovascular diseases, primarily atherosclerosis., Areas Covered in This Review: The pleiotropic effects of each of the PPAR isotypes on vascular and cardiac disease are discussed, with special emphasis on the molecular mechanism of action and on preclinical observations. The mechanism underlying the beneficial effect of PPARs is not confined to whole body metabolism, but also includes modulation of other vital processes, such as inflammation and cell fate (proliferation, differentiation, apoptosis)., What the Reader Will Gain: A large body of preclinical studies indicates that, in addition to their effect on atherogenesis, PPAR ligands also impact on ischemic heart disease and the development of cardiac failure. It remains to be established to what extent these intriguing observations can be translated into clinical practice., Take Home Message: The versatile mechanism of action extends the potential therapeutic profile of the PPARs enormously. Conversely, this versatility makes it harder to attain a specific therapeutic effect, without increasing the risk of undesirable side effects. The future challenge will be to design PPAR-based therapeutic strategies that minimize the detrimental side effects.
- Published
- 2010
- Full Text
- View/download PDF
25. Impaired cardiac functional reserve in type 2 diabetic db/db mice is associated with metabolic, but not structural, remodelling.
- Author
-
Daniels A, van Bilsen M, Janssen BJ, Brouns AE, Cleutjens JP, Roemen TH, Schaart G, van der Velden J, van der Vusse GJ, and van Nieuwenhoven FA
- Subjects
- Adrenergic beta-Agonists, Age Factors, Animals, Cardiomyopathies diagnostic imaging, Cardiomyopathies genetics, Cardiomyopathies metabolism, Cardiomyopathies physiopathology, Diabetes Mellitus, Type 2 diagnostic imaging, Diabetes Mellitus, Type 2 genetics, Diabetes Mellitus, Type 2 metabolism, Diabetes Mellitus, Type 2 physiopathology, Disease Models, Animal, Dobutamine, Echocardiography, Doppler, Female, Gene Expression Regulation, Male, Mice, Myocardium pathology, RNA, Messenger metabolism, Ventricular Pressure, Cardiomyopathies etiology, Diabetes Mellitus, Type 2 complications, Energy Metabolism genetics, Myocardial Contraction genetics, Myocardium metabolism, Ventricular Function, Left genetics, Ventricular Remodeling genetics
- Abstract
Aim: To identify the initial alterations in myocardial tissue associated with the early signs of diabetic cardiac haemodynamic dysfunction, we monitored changes in cardiac function, structural remodelling and gene expression in hearts of type 2 diabetic db/db mice., Methods: Cardiac dimensions and function were determined echocardiographically at 8, 12, 16 and 18 weeks of age. Left ventricular pressure characteristics were measured at 18 weeks under baseline conditions and upon dobutamine infusion., Results: The db/db mice were severely diabetic already at 8 weeks after birth, showing elevated fasting blood glucose levels and albuminuria. Nevertheless, echocardiography revealed no significant changes in cardiac function up to 18 weeks of age. At 18 weeks of age, left ventricular pressure characteristics were not significantly different at baseline between diabetic and control mice. However, dobutamine stress test revealed significantly attenuated cardiac inotropic and lusitropic responses in db/db mice. Post-mortem cardiac tissue analyses showed minor structural remodelling and no significant changes in gene expression levels of the sarcoplasmic reticulum calcium ATPase (SERCA2a) or beta1-adrenoceptor (beta1-AR). Moreover, the phosphorylation state of known contractile protein targets of protein kinase A (PKA) was not altered, indicating unaffected cardiac beta-adrenergic signalling activity in diabetic animals. By contrast, the substantially increased expression of uncoupling protein-3 (UCP3) and angiopoietin-like-4 (Angptl4), along with decreased phosphorylation of AMP-activated protein kinase (AMPK) in the diabetic heart, is indicative of marked changes in cardiac metabolism., Conclusion: db/db mice show impaired cardiac functional reserve capacity during maximal beta-adrenergic stimulation which is associated with unfavourable changes in cardiac energy metabolism.
- Published
- 2010
- Full Text
- View/download PDF
26. Stretch-induced hypertrophy of isolated adult rabbit cardiomyocytes.
- Author
-
Blaauw E, van Nieuwenhoven FA, Willemsen P, Delhaas T, Prinzen FW, Snoeckx LH, van Bilsen M, and van der Vusse GJ
- Subjects
- Angiotensin II genetics, Angiotensin II metabolism, Angiotensin II pharmacology, Animals, Cell Size, Cells, Cultured, Culture Media, Conditioned, Insulin-Like Growth Factor I genetics, Insulin-Like Growth Factor I metabolism, Insulin-Like Growth Factor I pharmacology, Male, Myocytes, Cardiac drug effects, Natriuretic Peptide, Brain genetics, Natriuretic Peptide, Brain metabolism, RNA, Messenger genetics, RNA, Messenger metabolism, Rabbits, Reverse Transcriptase Polymerase Chain Reaction, Transforming Growth Factor beta genetics, Transforming Growth Factor beta metabolism, Transforming Growth Factor beta pharmacology, Cell Enlargement, Myocytes, Cardiac metabolism, Stress, Mechanical
- Abstract
Both mechanical and humoral triggers have been put forward to explain the hypertrophic response of the challenged cardiomyocyte. The aim of the present study was to investigate whether cyclic equibiaxial stretch is a direct stimulus for isolated adult rabbit cardiomyocytes to develop hypertrophy and to explore the potential involvement of the autocrine/paracrine factors ANG II, transforming growth factor (TGF)-beta(1), and IGF-I in this process. Isolated cardiomyocytes were exposed to 10% cyclic equibiaxial stretch (1 Hz) for up to 48 h or treated with ANG II (100 nM), TGF-beta(1) (5 ng/ml), IGF-I (100 ng/ml), ANG II type 1 (AT(1)) receptor blockers, or conditioned medium of stretched fibroblasts. Cyclic stretch significantly increased cell surface area (+3.1%), protein synthesis (+21%), and brain natriuretic peptide (BNP) mRNA expression (6-fold) in cardiomyocytes. TGF-beta(1) expression increased (+42%) transiently at 4 h, whereas cardiomyocyte IGF-I expression was not detectable under all experimental conditions. The AT(1) receptor blockers candesartan and irbesartan (100 nM) did not prevent the stretch-induced hypertrophic response. Direct exposure to ANG II, TGF-beta(1), or IGF-I did not enhance cardiomyocyte BNP expression. In cardiac fibroblasts, stretch elicited a significant approximately twofold increase in TGF-beta(1) and IGF-I expression. Conditioned medium of stretched fibroblasts increased BNP expression in cardiomyocytes ( approximately 2-fold, P = 0.07). This study clearly indicates that cyclic stretch is a strong, direct trigger to induce hypertrophy in fully differentiated rabbit cardiomyocytes. The present findings do not support the notion that stretch-mediated hypertrophy of adult rabbit cardiomyocytes involves autocrine/paracrine actions of ANG II, TGF-beta(1), or IGF-I.
- Published
- 2010
- Full Text
- View/download PDF
27. Connective tissue growth factor and cardiac fibrosis.
- Author
-
Daniels A, van Bilsen M, Goldschmeding R, van der Vusse GJ, and van Nieuwenhoven FA
- Subjects
- Animals, Biomarkers metabolism, Diabetic Angiopathies metabolism, Fibrosis metabolism, Humans, Hypertension metabolism, Mice, Myocardial Ischemia metabolism, Signal Transduction physiology, Connective Tissue Growth Factor physiology, Myocardium pathology
- Abstract
Cardiac fibrosis is a major pathogenic factor in a variety of cardiovascular diseases and refers to an excessive deposition of extracellular matrix components in the heart, which leads to cardiac dysfunction and eventually overt heart failure. Evidence is accumulating for a crucial role of connective tissue growth factor (CTGF) in fibrotic processes in several tissues including the heart. CTGF orchestrates the actions of important local factors evoking cardiac fibrosis. The central role of CTGF as a matricellular protein modulating the fibrotic process in cardiac remodelling makes it a possible biomarker for cardiac fibrosis and a potential candidate for therapeutic intervention to mitigate fibrosis in the heart.
- Published
- 2009
- Full Text
- View/download PDF
28. Metabolic remodelling of the failing heart: beneficial or detrimental?
- Author
-
van Bilsen M, van Nieuwenhoven FA, and van der Vusse GJ
- Subjects
- Adaptation, Physiological, Adenosine Triphosphate metabolism, Animals, Cardiomegaly metabolism, Cardiomegaly physiopathology, Fatty Acids metabolism, Glucose metabolism, Heart Failure physiopathology, Heart Failure therapy, Humans, Mitochondria, Heart metabolism, Oxidation-Reduction, Energy Metabolism, Heart Failure metabolism, Myocardial Contraction, Myocardium metabolism, Ventricular Remodeling
- Abstract
The failing heart is characterized by alterations in energy metabolism, including mitochondrial dysfunction and a reduction in fatty acid (FA) oxidation rate, which is partially compensated by an increase in glucose utilization. Together, these changes lead to an impaired capacity to convert chemical energy into mechanical work. This has led to the concept that supporting cardiac energy conversion through metabolic interventions provides an important adjuvant therapy for heart failure. The potential success of such a therapy depends on whether the shift from FA towards glucose utilization should be considered beneficial or detrimental, a question still incompletely resolved. In this review, the current status of the literature is evaluated and possible causes of observed discrepancies are discussed. It is cautiously concluded that for the failing heart, from a therapeutic point of view, it is preferable to further stimulate glucose oxidation rather than to normalize substrate metabolism by stimulating FA utilization. Whether this also applies to the pre-stages of cardiac failure remains to be established.
- Published
- 2009
- Full Text
- View/download PDF
29. CTGF inhibits BMP-7 signaling in diabetic nephropathy.
- Author
-
Nguyen TQ, Roestenberg P, van Nieuwenhoven FA, Bovenschen N, Li Z, Xu L, Oliver N, Aten J, Joles JA, Vial C, Brandan E, Lyons KM, and Goldschmeding R
- Subjects
- Animals, Bone Morphogenetic Protein 7, Bone Morphogenetic Proteins deficiency, Bone Morphogenetic Proteins genetics, Connective Tissue Growth Factor, Diabetes Mellitus, Experimental genetics, Diabetes Mellitus, Experimental pathology, Diabetes Mellitus, Experimental physiopathology, Diabetic Nephropathies genetics, Diabetic Nephropathies pathology, Female, Gene Expression, Immediate-Early Proteins deficiency, Immediate-Early Proteins genetics, Immediate-Early Proteins pharmacology, Inhibitor of Differentiation Protein 1 genetics, Intercellular Signaling Peptides and Proteins deficiency, Intercellular Signaling Peptides and Proteins genetics, Intercellular Signaling Peptides and Proteins pharmacology, Kidney metabolism, Kidney pathology, Male, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Knockout, Protein Binding, RNA, Messenger genetics, RNA, Messenger metabolism, Recombinant Proteins pharmacology, Signal Transduction, Smad1 Protein metabolism, Smad5 Protein metabolism, Transforming Growth Factor beta deficiency, Transforming Growth Factor beta genetics, Bone Morphogenetic Proteins physiology, Diabetic Nephropathies physiopathology, Immediate-Early Proteins physiology, Intercellular Signaling Peptides and Proteins physiology, Transforming Growth Factor beta physiology
- Abstract
In diabetic nephropathy, connective tissue growth factor (CTGF) is upregulated and bone morphogenetic protein 7 (BMP-7) is downregulated. CTGF is known to inhibit BMP-4, but similar cross-talk between BMP-7 and CTGF has not been studied. In this study, it was hypothesized that CTGF acts as an inhibitor of BMP-7 signaling activity in diabetic nephropathy. Compared with diabetic wild-type CTGF(+/+) mice, diabetic CTGF(+/-) mice had approximately 50% lower CTGF mRNA and protein, less severe albuminuria, no thickening of the glomerular basement membrane, and preserved matrix metalloproteinase (MMP) activity. Although the amount of BMP-7 mRNA was similar in the kidneys of diabetic CTGF(+/+) and CTGF(+/-) mice, phosphorylation of the BMP signal transduction protein Smad1/5 and expression of the BMP target gene Id1 were lower in diabetic CTGF(+/+) mice. Moreover, renal Id1 mRNA expression correlated with albuminuria (R = -0.86) and MMP activity (R = 0.76). In normoglycemic mice, intraperitoneal injection of CTGF led to a decrease of pSmad1/5 in the renal cortex. In cultured renal glomerular and tubulointerstitial cells, CTGF diminished BMP-7 signaling activity, evidenced by lower levels of pSmad1/5, Id1 mRNA, and BMP-responsive element-luciferase activity. Co-immunoprecipitation, solid-phase binding assay, and surface plasmon resonance analysis showed that CTGF binds BMP-7 with high affinity (Kd approximately 14 nM). In conclusion, upregulation of CTGF inhibits BMP-7 signal transduction in the diabetic kidney and contributes to altered gene transcription, reduced MMP activity, glomerular basement membrane thickening, and albuminuria, all of which are hallmarks of diabetic nephropathy.
- Published
- 2008
- Full Text
- View/download PDF
30. The angio-fibrotic switch of VEGF and CTGF in proliferative diabetic retinopathy.
- Author
-
Kuiper EJ, Van Nieuwenhoven FA, de Smet MD, van Meurs JC, Tanck MW, Oliver N, Klaassen I, Van Noorden CJ, Goldschmeding R, and Schlingemann RO
- Subjects
- Adult, Aged, Cell Proliferation, Connective Tissue Growth Factor, Enzyme-Linked Immunosorbent Assay methods, Eye metabolism, Female, Fibrosis, Humans, Male, Middle Aged, Neovascularization, Pathologic, Treatment Outcome, Diabetic Retinopathy metabolism, Immediate-Early Proteins metabolism, Intercellular Signaling Peptides and Proteins metabolism, Vascular Endothelial Growth Factor A metabolism
- Abstract
Background: In proliferative diabetic retinopathy (PDR), vascular endothelial growth factor (VEGF) and connective tissue growth factor (CTGF) cause blindness by neovascularization and subsequent fibrosis, but their relative contribution to both processes is unknown. We hypothesize that the balance between levels of pro-angiogenic VEGF and pro-fibrotic CTGF regulates angiogenesis, the angio-fibrotic switch, and the resulting fibrosis and scarring., Methods/principal Findings: VEGF and CTGF were measured by ELISA in 68 vitreous samples of patients with proliferative DR (PDR, N = 32), macular hole (N = 13) or macular pucker (N = 23) and were related to clinical data, including degree of intra-ocular neovascularization and fibrosis. In addition, clinical cases of PDR (n = 4) were studied before and after pan-retinal photocoagulation and intra-vitreal injections with bevacizumab, an antibody against VEGF. Neovascularization and fibrosis in various degrees occurred almost exclusively in PDR patients. In PDR patients, vitreous CTGF levels were significantly associated with degree of fibrosis and with VEGF levels, but not with neovascularization, whereas VEGF levels were associated only with neovascularization. The ratio of CTGF and VEGF was the strongest predictor of degree of fibrosis. As predicted by these findings, patients with PDR demonstrated a temporary increase in intra-ocular fibrosis after anti-VEGF treatment or laser treatment., Conclusions/significance: CTGF is primarily a pro-fibrotic factor in the eye, and a shift in the balance between CTGF and VEGF is associated with the switch from angiogenesis to fibrosis in proliferative retinopathy.
- Published
- 2008
- Full Text
- View/download PDF
31. Plasma connective tissue growth factor is an independent predictor of end-stage renal disease and mortality in type 1 diabetic nephropathy.
- Author
-
Nguyen TQ, Tarnow L, Jorsal A, Oliver N, Roestenberg P, Ito Y, Parving HH, Rossing P, van Nieuwenhoven FA, and Goldschmeding R
- Subjects
- Adult, Biomarkers blood, Connective Tissue Growth Factor, Diabetic Nephropathies mortality, Diabetic Retinopathy epidemiology, Female, Glomerular Filtration Rate, Humans, Kidney Function Tests, Male, Middle Aged, Predictive Value of Tests, Proportional Hazards Models, Risk Factors, Diabetic Nephropathies complications, Immediate-Early Proteins blood, Intercellular Signaling Peptides and Proteins blood, Kidney Failure, Chronic epidemiology, Kidney Failure, Chronic mortality
- Abstract
Objective: We evaluated the predictive value of baseline plasma connective tissue growth factor (CTGF) in a prospective study of patients with type 1 diabetes., Research Design and Methods: Subjects were 198 type 1 diabetic patients with established diabetic nephropathy and 188 type 1 diabetic patients with persistent normoalbuminuria. Follow-up time was 12.8 years. Prediction of end-stage renal disease (ESRD) and mortality by plasma CTGF was analyzed in conjunction with conventional risk factors., Results: Plasma CTGF was higher in patients with nephropathy than in patients with normoalbuminuria (median 381 [interquartile range 270-630] vs. 235 [168-353] pmol/l). In patients with nephropathy, elevated plasma CTGF was an independent predictor of ESRD (covariate-adjusted hazard ratio [HR] 1.6 [95% CI 1.1-2.5]) and correlated with the rate of decline in glomerular filtration rate (GFR) (cumulative R = 0.46). Area under the receiver operating characteristic curve for prediction of ESRD was 0.72. Plasma CTGF above a cutoff level of 413 pmol/l predicted ESRD with a sensitivity of 73% and a specificity of 63% and was associated with a higher rate of decline in GFR (mean +/- SD 5.4 +/- 4.9 vs. 3.3 +/- 3.5 ml/min per 1.73 m(2) per year). Moreover, in patients with nephrotic range albuminuria (>3 g/day), plasma CTGF was the only predictor of ESRD (covariate-adjusted HR 4.5 [2.0-10.4]). Plasma CTGF was an independent predictor also of overall mortality (covariate-adjusted HR 1.4 [1.1-1.7]). In contrast, in normoalbuminuric patients, plasma CTGF did not correlate with clinical parameters and did not predict outcome., Conclusions: Plasma CTGF contributes significantly to prediction of ESRD and mortality in patients with type 1 diabetic nephropathy.
- Published
- 2008
- Full Text
- View/download PDF
32. Cardiac hypertrophy is enhanced in PPAR alpha-/- mice in response to chronic pressure overload.
- Author
-
Smeets PJ, Teunissen BE, Willemsen PH, van Nieuwenhoven FA, Brouns AE, Janssen BJ, Cleutjens JP, Staels B, van der Vusse GJ, and van Bilsen M
- Subjects
- 3-Hydroxyacyl CoA Dehydrogenases metabolism, Actins metabolism, Animals, Aorta, Thoracic surgery, Atrial Natriuretic Factor metabolism, Coenzyme A Ligases metabolism, Collagen Type I metabolism, Cyclooxygenase 2 metabolism, Disease Models, Animal, Fibrosis, Hypertrophy, Left Ventricular diagnostic imaging, Hypertrophy, Left Ventricular genetics, Hypertrophy, Left Ventricular physiopathology, Interleukin-6 metabolism, Ligation, Male, Matrix Metalloproteinase 2 metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Myocardial Contraction, Myocardium enzymology, Myocardium pathology, PPAR alpha deficiency, PPAR alpha genetics, RNA, Messenger metabolism, Stroke Volume, Tumor Necrosis Factor-alpha metabolism, Ultrasonography, Ventricular Function, Left, Hypertrophy, Left Ventricular metabolism, Myocardium metabolism, PPAR alpha metabolism, Ventricular Remodeling genetics
- Abstract
Aims: Peroxisome proliferator-activated receptor-alpha (PPARalpha) is a nuclear receptor regulating cardiac metabolism that also has anti-inflammatory properties. Since the activation of inflammatory signalling pathways is considered to be important in cardiac hypertrophy and fibrosis, it is anticipated that PPARalpha modulates cardiac remodelling. Accordingly, in this study the hypothesis was tested that the absence of PPARalpha aggravates the cardiac hypertrophic response to pressure overload., Methods and Results: Male PPARalpha-/- and wild-type mice were subjected to transverse aortic constriction (TAC) for 28 days. TAC resulted in a more pronounced increase in ventricular weight and left ventricular (LV) wall thickness in PPARalpha-/- than in wild-type mice. Compared with sham-operated mice, TAC did not affect cardiac function in wild-type mice, but significantly depressed LV ejection fraction and LV contractility in PPARalpha-/- mice. Moreover, after TAC mRNA levels of hypertrophic (atrial natriuretic factor, alpha-skeletal actin), fibrotic (collagen 1, matrix metalloproteinase-2), and inflammatory (interleukin-6, tumour necrosis factor-alpha, cyclo-oxygenase-2) marker genes were higher in PPARalpha-/- than in wild-type mice. The mRNA levels of genes involved in fatty acid metabolism (long-chain acyl-CoA synthetase, hydroxyacyl-CoA dehydrogenase) were decreased in PPARalpha-/- mice, but were not further compromised by TAC., Conclusion: The present findings show that the absence of PPARalpha results in a more pronounced hypertrophic growth response and cardiac dysfunction that are associated with an enhanced expression of markers of inflammation and extracellular matrix remodelling. These findings indicate that PPARalpha exerts salutary effects during cardiac hypertrophy.
- Published
- 2008
- Full Text
- View/download PDF
33. Association of connective tissue growth factor with fibrosis in vitreoretinal disorders in the human eye.
- Author
-
Kuiper EJ, de Smet MD, van Meurs JC, Tan HS, Tanck MW, Oliver N, van Nieuwenhoven FA, Goldschmeding R, and Schlingemann RO
- Subjects
- Connective Tissue Growth Factor, Diabetic Retinopathy pathology, Enzyme-Linked Immunosorbent Assay, Epiretinal Membrane pathology, Female, Fibrosis metabolism, Humans, Male, Middle Aged, Retina pathology, Retinal Perforations pathology, Vitreoretinopathy, Proliferative pathology, Vitreous Body pathology, Diabetic Retinopathy metabolism, Epiretinal Membrane metabolism, Immediate-Early Proteins metabolism, Intercellular Signaling Peptides and Proteins metabolism, Retinal Perforations metabolism, Vitreoretinopathy, Proliferative metabolism, Vitreous Body metabolism
- Abstract
Objective: To investigate the expression of the profibrotic connective tissue growth factor (CTGF) in relation to severity of intraocular fibrosis and neovascularization in human vitreoretinal disorders for the identification of potential therapeutic targets to prevent fibrosis., Methods: Concentrations of CTGF were measured by enzyme-linked immunosorbent assay in 119 vitreous samples from patients with proliferative diabetic retinopathy, proliferative vitreoretinopathy, epiretinal membrane, and macular hole. Clinical data, including degree of intraocular fibrosis and neovascularization, were collected using standardized forms., Results: Multifactorial analysis revealed that only CTGF levels correlated highly significantly with degree of fibrosis in the various vitreoretinal disorders studied (P<.001; R2= 47.7%). Likewise, variation in degree of fibrosis was best predicted by CTGF levels (P<.001)., Conclusion: The strong correlation between CTGF levels and degree of fibrosis in vitreoretinal disorders suggests that CTGF is an important factor in ocular fibrosis, similar to its role in pathologic fibrosis in other organs., Clinical Relevance: Connective tissue growth factor may be a therapeutic target for prevention of sight-threatening vitreoretinal scarring in the eye.
- Published
- 2006
- Full Text
- View/download PDF
34. Temporal expression profile and distribution pattern indicate a role of connective tissue growth factor (CTGF/CCN-2) in diabetic nephropathy in mice.
- Author
-
Roestenberg P, van Nieuwenhoven FA, Joles JA, Trischberger C, Martens PP, Oliver N, Aten J, Höppener JW, and Goldschmeding R
- Subjects
- Amyloid genetics, Animals, Connective Tissue Growth Factor, Crosses, Genetic, Diabetes Mellitus, Experimental complications, Diabetes Mellitus, Experimental genetics, Diabetic Nephropathies metabolism, Enzyme-Linked Immunosorbent Assay, Female, Humans, Immediate-Early Proteins physiology, Intercellular Signaling Peptides and Proteins physiology, Leptin deficiency, Leptin genetics, Male, Mice, Mice, Inbred C57BL, Mice, Transgenic, RNA, Messenger analysis, Reverse Transcriptase Polymerase Chain Reaction, Diabetic Nephropathies etiology, Gene Expression, Immediate-Early Proteins analysis, Immediate-Early Proteins genetics, Intercellular Signaling Peptides and Proteins analysis, Intercellular Signaling Peptides and Proteins genetics, Kidney chemistry
- Abstract
Connective tissue growth factor (CTGF) is overexpressed in diabetic nephropathy (DN) and has therefore been implicated in its pathogenesis. The objective of the present study was to determine the tissue distribution of increased CTGF expression and the relationship of plasma, urinary, and renal CTGF levels to the development and severity of DN. We studied the relationship between CTGF and renal pathology in streptozotocin (STZ)-induced diabetes in C57BL/6J mice. Diabetic and age-matched control mice were killed after 1, 2, 4, and 9 wk of diabetes. In addition, key parameters of diabetes and DN were analyzed in 10-mo-old diabetic ob/ob mice and their ob/+ littermates. STZ-induced diabetic mice showed a significantly increased urinary albumin excretion after 1 wk and increased mesangial matrix score after 2 wk. Increased renal fibronectin, fibronectin ED-A, and collagen IValpha1 expression, as well as elevated plasma creatinine levels, were observed after 9 wk. After 2 wk, CTGF mRNA was upregulated threefold in the renal cortex. By 9 wk, CTGF mRNA was also increased in the heart and liver. In contrast, transforming growth factor-beta1 mRNA content was significantly increased only in the kidney by 9 wk. Renal CTGF expression was mainly localized in podocytes and parietal glomerular epithelial cells, and less prominent in mesangial cells. In addition, plasma CTGF levels and urinary CTGF excretion were increased in diabetic mice. Moreover, albuminuria strongly correlated with urinary CTGF excretion (R = 0.83, P < 0.0001). Increased CTGF expression was also demonstrated in type 2 diabetic ob/ob mice, which points to a causal relationship between diabetes and CTGF and thus argues against a role of STZ in this process. The observed relationship of podocyte and urinary CTGF to markers of DN suggests a pathogenic role of CTGF in the development of DN.
- Published
- 2006
- Full Text
- View/download PDF
35. Myofibroblast progenitor cells are increased in number in patients with type 1 diabetes and express less bone morphogenetic protein 6: a novel clue to adverse tissue remodelling?
- Author
-
Nguyen TQ, Chon H, van Nieuwenhoven FA, Braam B, Verhaar MC, and Goldschmeding R
- Subjects
- Bone Morphogenetic Protein 6, Bone Morphogenetic Proteins deficiency, Cell Culture Techniques, Connective Tissue Growth Factor, Fibroblasts pathology, Humans, Immediate-Early Proteins genetics, Intercellular Signaling Peptides and Proteins genetics, Kinetics, Lymphotoxin-alpha pharmacology, Phenotype, Plasminogen Activator Inhibitor 1 genetics, Reference Values, Stem Cells pathology, Bone Morphogenetic Proteins genetics, Diabetes Mellitus, Type 1 genetics, Diabetes Mellitus, Type 1 pathology, Fibroblasts cytology, Myoblasts cytology, Stem Cells cytology
- Abstract
Aims/hypothesis: Growth factor imbalance and endothelial progenitor cell dysfunction are well-known elements of the inappropriate response to injury in human and experimental diabetes. We hypothesised that in diabetes the outgrowth of myofibroblast progenitor cells (MFPCs) is also altered and that this relates to aberrant gene expression of growth factors involving members of the TGF-beta/bone morphogenetic protein (BMP) superfamily., Subjects and Methods: MFPCs were cultured from peripheral blood mononuclear cells of patients with type 1 diabetes and control subjects. Microarray analysis, quantitative PCR and ELISA were used to identify differentially regulated TGF-beta/BMP superfamily genes in diabetes- and control-derived MFPC. Possible effects of BMP6 on TGF-beta-induced gene expression were examined in cultured renal fibroblasts (TK173 cells)., Results: Blood from diabetic patients yielded higher numbers of MFPCs than blood from control subjects (1.6-fold increase, p<0.05), involving increased proliferation and decreased apoptosis. BMP6 mRNA and protein were downregulated in MFPCs derived from patients with diabetes (3.9- and 1.8-fold decrease, respectively, p<0.05). Furthermore, an inverse correlation was observed between BMP6 mRNA level and the number of MFPCs in patients with diabetes (r=-0.85, p<0.05). In TK173 cells, BMP6 antagonised the TGF-beta-induced expression of the genes encoding plasminogen activator inhibitor-1 and connective tissue growth factor (70 and 50% reduction, respectively)., Conclusions/interpretation: Considering the importance of BMP6 in processes such as angiogenesis and its novel anti-TGF-beta effects, we propose that the excess numbers of BMP6-deficient MFPCs may favour adverse tissue remodelling in patients with diabetes, both numerically and by inappropriate orchestration of their microenvironment.
- Published
- 2006
- Full Text
- View/download PDF
36. Purification, immunochemical quantification and localization in rat heart of putative fatty acid translocase (FAT/CD36).
- Author
-
Brinkmann JF, Pelsers MM, van Nieuwenhoven FA, Tandon NN, van der Vusse GJ, and Glatz JF
- Subjects
- Animals, CD36 Antigens isolation & purification, Endothelial Cells enzymology, Fluorescent Antibody Technique, Humans, In Vitro Techniques, Male, Myocytes, Cardiac enzymology, Rats, Rats, Inbred Lew, CD36 Antigens metabolism, Myocardium enzymology
- Abstract
Evidence is accumulating that the heavily glycosylated integral membrane protein fatty acid translocase (FAT/CD36) is involved in the transport of long-chain fatty acids across the sarcolemma of heart muscle cells. The aim of this study was to analyse the distribution between FAT/CD36 present in cardiac myocytes and endothelial cells. We therefore developed a method to purify FAT/CD36 from total rat heart and isolated cardiomyocytes, and used the proteins as standards in an immunochemical assay. Two steps, chromatography on wheat germ agglutinin-agarose and anion-exchange chromatography on Q-Sepharose fast flow, were sufficient for obtaining the protein in a > 95% pure form. When used to isolate FAT/CD36 from total heart tissue, the FAT/CD36 yield of the method was 9% and the purification factor was 64. Purifying FAT/CD36 from isolated cardiomyocytes yielded the same 88 kDa protein band on SDS-PAGE gels and reactivity of this band on western blots was comparable to that of the FAT/CD36 isolated from total hearts. Quantifying FAT/CD36 contents by western blotting showed that the amounts of FAT/CD36 that are present in isolated cardiomyocytes (10 +/- 3 microg/mg protein) and total hearts (14 +/- 4 microg/mg protein) are of comparable magnitude. Immunofluorescence labelling showed that at least a part of the FAT/CD36 present in the cardiomyocyte is associated with the sarcolemma. This study established that FAT/CD36 is a relatively abundant protein in the cardiomyocyte. In addition, the further developed purification procedure is the first method for isolating FAT/CD36 from rat heart and cardiomyocyte FAT/CD36.
- Published
- 2006
- Full Text
- View/download PDF
37. Urinary connective tissue growth factor excretion correlates with clinical markers of renal disease in a large population of type 1 diabetic patients with diabetic nephropathy.
- Author
-
Nguyen TQ, Tarnow L, Andersen S, Hovind P, Parving HH, Goldschmeding R, and van Nieuwenhoven FA
- Subjects
- Adult, Biomarkers urine, Blood Glucose metabolism, Connective Tissue Growth Factor, Cross-Sectional Studies, Diabetic Nephropathies epidemiology, Enzyme-Linked Immunosorbent Assay, Female, Glomerular Filtration Rate, Glycated Hemoglobin analysis, Humans, Male, Middle Aged, Reference Values, Regression Analysis, Diabetes Mellitus, Type 1 urine, Diabetic Nephropathies urine, Immediate-Early Proteins urine, Intercellular Signaling Peptides and Proteins urine
- Abstract
Objective: Levels of connective tissue growth factor (CTGF; CCN-2) in plasma are increased in various fibrotic disorders, including diabetic nephropathy. Recently, several articles have reported a strong increase of urinary CTGF excretion (U-CTGF) in patients with diabetic nephropathy. However, these studies addressed too small a number of patients to allow general conclusions to be drawn. Therefore, we evaluated U-CTGF in a large cross-sectional study of patients with type 1 diabetes., Research Design and Methods: Subjects were 318 type 1 diabetic patients and 29 normoglycemic control subjects. U-CTGF was measured by sandwich enzyme-linked immunosorbent assay. Groups were compared by Kruskal-Wallis and Mann-Whitney analysis. The relation between U-CTGF and markers of diabetic nephropathy was determined by regression analysis., Results: U-CTGF in patients with diabetic nephropathy (n = 89, median 155 pmol/24 h [interquartile range 96-258]) was significantly higher than in microalbuminuric (n = 79, 100 [65-78]) and normoalbuminuric (n = 150, 85 [48-127]) patients and control subjects (n = 29, 100 [78-114]). U-CTGF correlated with urinary albumin excretion (UAE) (R = 0.31) and glomerular filtration rate (R = -0.38) in patients with diabetic nephropathy. A standardized increase in U-CTGF was associated with diabetic nephropathy (odds ratio 2.3 [95% CI 1.7-3.1]), which was comparable with the odds ratios for diabetic nephropathy of increased HbA(1c) (2.0 [1.5-2.7]), and blood pressure (2.0 [1.5-2.6])., Conclusions: This is the first large cross-sectional study addressing U-CTGF in human type 1 diabetes. The observed association of U-CTGF with UAE and glomerular filtration rate might reflect a role of CTGF as progression promoter in diabetic nephropathy.
- Published
- 2006
- Full Text
- View/download PDF
38. Reduction of urinary connective tissue growth factor by Losartan in type 1 patients with diabetic nephropathy.
- Author
-
Andersen S, van Nieuwenhoven FA, Tarnow L, Rossing P, Rossing K, Wieten L, Goldschmeding R, and Parving HH
- Subjects
- Adult, Connective Tissue Growth Factor, Diabetes Mellitus, Type 1 complications, Female, Glomerular Filtration Rate, Humans, Immediate-Early Proteins blood, Intercellular Signaling Peptides and Proteins blood, Male, Middle Aged, Prospective Studies, Diabetes Mellitus, Type 1 urine, Diabetic Nephropathies urine, Immediate-Early Proteins urine, Intercellular Signaling Peptides and Proteins urine, Losartan pharmacology
- Abstract
Background: Connective tissue growth factor (CTGF) is an important profibrotic cytokine implicated in development of diabetic glomerulosclerosis. Urinary CTGF is reported to be significantly increased in patients with diabetic nephropathy. The present study aimed to investigate the short- and long term effects of angiotensin II receptor blockade by Losartan on urinary CTGF levels in hypertensive type 1 diabetic patients with diabetic nephropathy., Methods: Seventy-one hypertensive type 1 diabetic patients with diabetic nephropathy were included in the study. After a washout period of 4 weeks, the patients received Losartan 50 mg, 100 mg, and 150 mg once daily in treatment periods each lasting 2 months. Thereafter, patients were followed prospectively during treatment with Losartan 100 mg o.d. with a total mean follow-up time of 36 months. At baseline, after 2, 4, and 6 months and then biannually, urinary and plasma CTGF levels [enzyme linked immunosorbent assay (ELISA) fibroGen], albuminuria (Turbidimetry), glomerular filtration rate (GFR) [51-creatinine ethylenediaminetetraacetic acid ((51)Cr-EDTA plasma clearance)] and 24 hours blood pressure (TM2420)) were determined., Results: Baseline levels of urinary and plasma CTGF were 7076 (5708 to 8770) ng/24 hours [geometric mean (95% CI)] and 12.7 (7.3) ng/mL [mean (SD)], respectively. Albuminuria, GFR, and arterial blood pressure at baseline were 1152 (937 to 1416) mg/24 hours, 88 (24) mL/min/1.73 m(2), and 153/80 (17/9) mm Hg, respectively. Losartan significantly reduced urinary CTGF by 21% (9 to 31) (95% CI) initially (P < 0.05 vs. baseline), with no further reduction after increasing dose. The sustained reduction in urinary CTGF was 22% (12 to 32) (P < 0.05 vs. baseline). Rate of decline in GFR during the study was 3.2 (-1.6 to 15.9) mL/min/year [median (range)]. Reduction in urinary CTGF was correlated with a lower rate of decline in GFR (r= 0.23, P= 0.05). Plasma CTGF remained unchanged throughout the study., Conclusion: Our 3-year study demonstrates that Losartan persistently reduces urinary CTGF excretion, which is associated with a slower rate of decline in GFR.
- Published
- 2005
- Full Text
- View/download PDF
39. Imbalance of growth factor signalling in diabetic kidney disease: is connective tissue growth factor (CTGF, CCN2) the perfect intervention point?
- Author
-
van Nieuwenhoven FA, Jensen LJ, Flyvbjerg A, and Goldschmeding R
- Subjects
- Biomarkers metabolism, Connective Tissue Growth Factor, Diabetic Nephropathies blood, Disease Progression, Female, Humans, Immediate-Early Proteins analysis, Male, Prognosis, Sensitivity and Specificity, Severity of Illness Index, Transforming Growth Factor beta analysis, Diabetic Nephropathies diagnosis, Immediate-Early Proteins metabolism, Intercellular Signaling Peptides and Proteins analysis, Intercellular Signaling Peptides and Proteins metabolism, Transforming Growth Factor beta metabolism
- Published
- 2005
- Full Text
- View/download PDF
40. Connective tissue growth factor is increased in plasma of type 1 diabetic patients with nephropathy.
- Author
-
Roestenberg P, van Nieuwenhoven FA, Wieten L, Boer P, Diekman T, Tiller AM, Wiersinga WM, Oliver N, Usinger W, Weitz S, Schlingemann RO, and Goldschmeding R
- Subjects
- Adult, Biomarkers blood, Connective Tissue Growth Factor, Creatinine metabolism, Female, Humans, Male, Middle Aged, Proteinuria, Reference Values, Regression Analysis, Transforming Growth Factor beta blood, Diabetes Mellitus, Type 1 blood, Diabetic Nephropathies blood, Immediate-Early Proteins blood, Intercellular Signaling Peptides and Proteins blood
- Abstract
Objective: Connective tissue growth factor (CTGF) is strongly upregulated in fibrotic disorders and has been hypothesized to play a role in the development and progression of diabetes complications. The aim of the present study was to investigate the possible association of plasma CTGF levels in type 1 diabetic patients with markers relevant to development of diabetes complications., Research Design and Methods: Plasma CTGF levels (full-length and NH2-terminal fragments) were determined in 62 well-characterized patients with type 1 diabetes and in 21 healthy control subjects. Correlations of these plasma CTGF levels with markers of glycemic control, platelet activation, endothelial activation, nephropathy, and retinopathy were investigated., Results: -Elevated plasma NH2-terminal fragment of CTGF (CTGF-N) levels were detected in a subpopulation of type 1 diabetic patients and were associated with diabetic nephropathy. Stepwise regression analysis revealed contribution of albuminuria, creatinine clearance, and duration of diabetes as predictors of plasma CTGF-N level. Elevation of plasma CTGF-N levels in patients with retinopathy was probably due to renal comorbidity., Conclusions: Plasma CTGF-N levels are elevated in type 1 diabetic patients with nephropathy and appear to be correlated with proteinuria and creatinine clearance. Further studies will be needed to determine the relevance of plasma CTGF as a clinical marker and/or pathogenic factor in diabetic nephropathy.
- Published
- 2004
- Full Text
- View/download PDF
41. Connective tissue growth factor and igf-I are produced by human renal fibroblasts and cooperate in the induction of collagen production by high glucose.
- Author
-
Lam S, van der Geest RN, Verhagen NA, van Nieuwenhoven FA, Blom IE, Aten J, Goldschmeding R, Daha MR, and van Kooten C
- Subjects
- Cells, Cultured, Connective Tissue Growth Factor, Dose-Response Relationship, Drug, Drug Synergism, Fibroblasts drug effects, Humans, Immediate-Early Proteins biosynthesis, Immediate-Early Proteins pharmacology, Insulin-Like Growth Factor I biosynthesis, Insulin-Like Growth Factor I pharmacology, Intercellular Signaling Peptides and Proteins biosynthesis, Intercellular Signaling Peptides and Proteins pharmacology, Recombinant Proteins pharmacology, Transforming Growth Factor beta pharmacology, Transforming Growth Factor beta1, Collagen Type I biosynthesis, Collagen Type III biosynthesis, Fibroblasts metabolism, Glucose administration & dosage, Immediate-Early Proteins physiology, Insulin-Like Growth Factor I physiology, Intercellular Signaling Peptides and Proteins physiology, Kidney metabolism
- Abstract
Tubulointerstitial fibrosis is an important component in the development of diabetic nephropathy. Various renal cell types, including fibroblasts, contribute to the excessive matrix deposition in the kidney. Although transforming growth factor-beta (TGF-beta) has been thought to play a major role during fibrosis, other growth factors are also involved. Here we examined the effects of connective tissue growth factor (CTGF) and IGF-I on collagen type I and III production by human renal fibroblasts and their involvement in glucose-induced matrix accumulation. We have demonstrated that both CTGF and IGF-I expressions were increased in renal fibroblasts under hyperglycemic conditions, also in the absence of TGF-beta signaling. Although CTGF alone had no effect on collagen secretion, combined stimulation with IGF-I enhanced collagen accumulation. Furthermore, IGF-I also had a synergistic effect with glucose on the induction of collagens. Moreover, we observed a partial inhibition in glucose-induced collagen secretion with neutralizing anti-CTGF antibodies, thereby demonstrating for the first time the involvement of endogenous CTGF in glucose-induced effects in human renal fibroblasts. Therefore, the cooperation between CTGF and IGF-I might be involved in glucose-induced matrix accumulation in tubulointerstitial fibrosis and might contribute to the pathogenesis of diabetic nephropathy.
- Published
- 2003
- Full Text
- View/download PDF
42. Presence of heat shock protein 72 in cardiomyocytes after heat stress.
- Author
-
Cornelussen RN, van Nieuwenhoven FA, Snoeckx LH, and Knowlton AA
- Subjects
- Animals, Endothelium, Vascular cytology, Endothelium, Vascular metabolism, HSP27 Heat-Shock Proteins, HSP72 Heat-Shock Proteins, Myocardial Ischemia metabolism, Myocardial Reperfusion, Myocardium cytology, Neoplasm Proteins metabolism, Rats, Research Design, Heat-Shock Proteins metabolism, Heat-Shock Response physiology, Myocardium metabolism
- Published
- 2001
43. Heat shock proteins and cardiovascular pathophysiology.
- Author
-
Snoeckx LH, Cornelussen RN, Van Nieuwenhoven FA, Reneman RS, and Van Der Vusse GJ
- Subjects
- Animals, DNA-Binding Proteins metabolism, Gene Expression Regulation, Heat Shock Transcription Factors, Heat-Shock Proteins genetics, Humans, Ischemic Preconditioning, Myocardial, Signal Transduction physiology, Transcription Factors, Transcription, Genetic physiology, Cardiovascular Diseases physiopathology, Cardiovascular Physiological Phenomena, Cardiovascular System metabolism, Cardiovascular System physiopathology, Heat-Shock Proteins physiology
- Abstract
In the eukaryotic cell an intrinsic mechanism is present providing the ability to defend itself against external stressors from various sources. This defense mechanism probably evolved from the presence of a group of chaperones, playing a crucial role in governing proper protein assembly, folding, and transport. Upregulation of the synthesis of a number of these proteins upon environmental stress establishes a unique defense system to maintain cellular protein homeostasis and to ensure survival of the cell. In the cardiovascular system this enhanced protein synthesis leads to a transient but powerful increase in tolerance to such endangering situations as ischemia, hypoxia, oxidative injury, and endotoxemia. These so-called heat shock proteins interfere with several physiological processes within several cell organelles and, for proper functioning, are translocated to different compartments following stress-induced synthesis. In this review we describe the physiological role of heat shock proteins and discuss their protective potential against various stress agents in the cardiovascular system.
- Published
- 2001
- Full Text
- View/download PDF
44. HSP70-mediated acceleration of translational recovery after stress is independent of ribosomal RNA synthesis.
- Author
-
Van Nieuwenhoven FA, Martin X, Heijnen VV, Cornelussen RN, and Snoeckx LH
- Subjects
- Adenoviridae genetics, Adenoviridae metabolism, Animals, Cell Line, Gene Expression Regulation, Hot Temperature, Rats, Recombinant Fusion Proteins metabolism, HSP70 Heat-Shock Proteins metabolism, Protein Biosynthesis, RNA, Ribosomal biosynthesis, Transcription, Genetic
- Abstract
HSP70 is known to protect cells against stressful events. In the present study, the hypothesis was investigated that elevated HSP70 levels protect RNA polymerase I during stress, leading to decreased inhibition of ribosomal RNA (rRNA) synthesis and accelerated recovery of protein translation after stress. To this end, transcriptional and translational activity was studied in H9c2 cells during recovery after a severe heat treatment (SHT, 1 h 45 degrees C) in the presence of elevated HSP70 levels. The latter was achieved by heat pretreatment or by adenovirus-mediated hsp70 gene transfer. Rates of transcription and translation were determined by measuring cellular 3H-labelled uridine and leucine incorporation, respectively. The two types of pretreatment did not affect basal rates of transcription and translation, immediately before SHT. During SHT, both transcriptional and translational rates dropped to less than 10% of basal levels in pretreated as well as non-pretreated cells. Two and four h after SHT, both transcriptional and translational rates were significantly higher in HSP70-overexpressing cells compared to non-pretreated cells. However, immediately after SHT, transcription rates were similarly depressed in non-pretreated and pretreated cells, showing that increased levels of HSP70 did not protect RNA polymerase I activity during SHT. Thus, the HSP70-mediated acceleration of translational recovery is not preceded in time by an enhanced recovery of rRNA synthesis. Therefore, the HSP70-mediated early recovery of protein synthesis after heat stress is independent of rRNA synthesis.
- Published
- 2001
- Full Text
- View/download PDF
45. Selection-dominant and nonaccessible epitopes on cell-surface receptors revealed by cell-panning with a large phage antibody library.
- Author
-
Hoogenboom HR, Lutgerink JT, Pelsers MM, Rousch MJ, Coote J, Van Neer N, De Bruïne A, Van Nieuwenhoven FA, Glatz JF, and Arends JW
- Subjects
- Animals, Antibodies, Viral immunology, Antibody Specificity, Bacteriophage M13 immunology, CHO Cells, Cricetinae, Humans, Peptide Library, Rats, CD36 Antigens immunology, Immunodominant Epitopes immunology, Receptors, Somatostatin immunology
- Abstract
To generate antibodies to defined cell-surface antigens, we used a large phage antibody fragment library to select on cell transfectants expressing one of three chosen receptors. First, in vitro panning procedures and phage antibody screening ELISAs were developed using whole live cells stably expressing the antigen of interest. When these methodologies were applied to Chinese hamster ovary (CHO) cells expressing one of the receptors for a neuropeptide, somatostatin, using either direct cell panning or a strategy of depletion or ligand-directed elution, many different pan-CHO-cell binders were selected, but none was receptor specific. However, when using direct panning on CHO-cells expressing the human membrane protein CD36, an extraordinary high frequency of antigen-specific phage antibodies was found. Panning on myoblasts expressing the rat homologue of CD36 revealed a similar selection dominance for anti-(CD36). Binding of all selected 20 different anti-(CD36) phage was surprisingly inhibited by one anti-(CD36) mAb CLB-IVC7, which recognizes a functional epitope that is also immunodominant in vivo. Similar inhibition was found for seven anti-(rat) CD36 that cross-reacted with human CD36. Our results show that, although cells can be used as antigen carriers to select and screen phage antibodies, the nature of the antigen target has a profound effect on the outcome of the selection.
- Published
- 1999
- Full Text
- View/download PDF
46. Co-expression in rat heart and skeletal muscle of four genes coding for proteins implicated in long-chain fatty acid uptake.
- Author
-
Van Nieuwenhoven FA, Willemsen PH, Van der Vusse GJ, and Glatz JF
- Subjects
- Age Factors, Animals, Blotting, Northern, Carrier Proteins metabolism, Cells, Cultured, Diabetes Mellitus, Experimental metabolism, Fatty Acid Transport Proteins, Fatty Acid-Binding Protein 7, Fatty Acid-Binding Proteins, Heart physiology, Male, Membrane Proteins metabolism, Myelin P2 Protein metabolism, RNA, Messenger metabolism, Rats, Rats, Wistar, Tissue Distribution, Cadherins, Fatty Acids metabolism, Membrane Transport Proteins, Muscle, Skeletal metabolism, Myocardium metabolism, Neoplasm Proteins, Nerve Tissue Proteins
- Abstract
It has been suggested that specific membrane-associated and cytoplasmic proteins cooperate in the uptake of long-chain fatty acids by cardiac and skeletal muscle cells. A prerequisite for this hypothesis would be the co-occurrence of these proteins in muscle. Thus, we studied the possible co-expression in rat muscles of the genes coding for the integral membrane proteins fatty acid transport protein (FATP) and fatty acid translocase (FAT), the membrane-associated plasmalemmal fatty acid-binding protein (FABPpm) and the cytoplasmic heart-type fatty acid-binding protein (H-FABPc). The transcripts of the four proteins were assessed in heart and skeletal muscles of adult Wistar rats, in isolated cells and cell lines from rat heart and also in rat heart during development and upon streptozotocin-induced diabetes. All four genes showed high expression levels in heart, somewhat lower in red skeletal muscle (soleus) and appreciably lower in white skeletal muscle (extensor digitorum longus). FATP, FAT and H-FABPc showed a 3- to 5-fold increase in mRNA expression during maturational growth of the heart, while the FABPpm expression remained virtually constant. In the heart, streptozotocin-diabetes induced a slight, but statistically not significant, increase in the expression of all four genes. In conclusion, this study shows the co-expression of FATP, FAT, FABPpm and H-FABPc in rat muscles. This finding supports the possible cooperation of these proteins in the uptake of long-chain fatty acids by muscle cells.
- Published
- 1999
- Full Text
- View/download PDF
47. Cellular fatty acid transport in heart and skeletal muscle as facilitated by proteins.
- Author
-
Luiken JJ, Schaap FG, van Nieuwenhoven FA, van der Vusse GJ, Bonen A, and Glatz JF
- Subjects
- Animals, Biological Transport, Cell Membrane metabolism, Fatty Acid-Binding Protein 7, Fatty Acid-Binding Proteins, Humans, Models, Biological, Sarcolemma metabolism, Carrier Proteins metabolism, Fatty Acids metabolism, Muscle, Skeletal metabolism, Myelin P2 Protein metabolism, Myocardium metabolism, Neoplasm Proteins, Tumor Suppressor Proteins
- Abstract
Despite the importance of long-chain fatty acids (FA) as fuels for heart and skeletal muscles, the mechanism of their cellular uptake has not yet been clarified. There is dispute as to whether FA are taken up by the muscle cells via passive diffusion and/or carrier-mediated transport. Kinetic studies of FA uptake by cardiac myocytes and the use of membrane protein-modifying agents have suggested the bulk of FA uptake is due to a protein component. Three membrane-associated FA-binding proteins were proposed to play a role in FA uptake, a 40-kDa plasma membrane FA-binding protein (FABPpm), an 88-kDa FA translocase (FAT/CD36), and a 60-kDa FA transport protein (FATP). In cardiac and skeletal myocytes the intracellular carrier for FA is cytoplasmic heart-type FA-binding protein (H-FABP), which likely transports FA from the sarcolemma to their intracellular sites of metabolism. A scenario is discussed in which FABPpm, FAT/CD36, and H-FABP, probably assisted by an albumin-binding protein, cooperate in the translocation of FA across the sarcolemma.
- Published
- 1999
- Full Text
- View/download PDF
48. Stable transfection of fatty acid translocase (CD36) in a rat heart muscle cell line (H9c2).
- Author
-
Van Nieuwenhoven FA, Luiken JJ, De Jong YF, Grimaldi PA, Van der Vusse GJ, and Glatz JF
- Subjects
- Animals, Blotting, Northern, Blotting, Southern, Blotting, Western, CD36 Antigens, Cell Line, Diffusion, Fluorescent Antibody Technique, Kinetics, Palmitic Acid metabolism, Rats, Gene Expression, Membrane Glycoproteins genetics, Myocardium enzymology, Organic Anion Transporters, Transfection
- Abstract
Fatty acid translocase (FAT/CD36) is a membrane protein putatively involved in the transmembrane transport of long-chain fatty acids. We tested the hypothesis that expression of this protein in H9c2, a rat heart cell line normally not expressing FAT, would increase cellular palmitate uptake. We were able to stably transfect H9c2 cells with FAT, yielding 15 cell lines showing varying levels of FAT expression. The uptake and metabolism of palmitate was first studied in the non-transfected H9c2 cells and in two FAT-transfected cell lines. In each case, uptake of palmitate was found to be linear in time for at least 30 min and the uptake rate was saturable with increasing palmitate concentrations. Using conditions under which the maximal capacity of intracellular palmitate handling was not fully utilized, we tested 7 out of 15 FAT-transfected cell lines with varying FAT expression levels. No significant correlation was found between the level of FAT expression and the rate of palmitate uptake. In conclusion, we found that palmitate uptake by H9c2 cells occurs mainly by passive diffusion. Fatty acid translocase (FAT) transfection did not significantly increase the palmitate uptake rate, raising the possibility that H9c2 cells lack a protein (or set of proteins) that acts as an obligatory partner of FAT in long-chain fatty acid transport from the extracellular compartment to the cytoplasm.
- Published
- 1998
49. Transport of long-chain fatty acids across the muscular endothelium.
- Author
-
Van der Vusse GJ, Glatz JF, Van Nieuwenhoven FA, Reneman RS, and Bassingthwaighte JB
- Subjects
- Animals, Biological Transport, Humans, Muscle, Skeletal blood supply, Endothelium, Vascular metabolism, Fatty Acids metabolism, Muscle, Skeletal metabolism
- Abstract
Both skeletal and cardiac muscle cells rely heavily on the oxidation of long-chain fatty acids to utilize chemically stored energy for contractile work. Under normal conditions fatty acids are continuously supplied from the microvascular compartment to the contracting myocytes. Exogenous fatty acids are transported to muscle tissue via the blood either complexed to albumin or covalently bound in triacylglycerols forming the neutral lipid core of circulating lipoproteins such as chylomicrons or very low-density lipoproteins. The first barrier met by fatty acids on their way from the vascular compartment to the myocytes is the endothelium constituting the capillary wall. After dissociation of the albumin-fatty acid complex or release from the triacylglycerol core of lipoproteins, fatty acids most likely transverse the endothelium by crossing the luminal membrane, the cytosol, and subsequently the abluminal wall of the endothelial cell. Transfer through the interendothelial clefts or lateral diffusion within the phospholipid bilayer of the endothelial plasmalemma should be considered as inconsequential. The mechanism responsible for transmembrane movement of fatty acids is incompletely understood, although recent findings suggest the involvement of a number of membrane-associated proteins. Kinetic studies have revealed that interaction of the albumin-fatty acid complex with the endothelial membrane may accelerate the dissociation of the complex, which facilitates the uptake of fatty acids by the endothelium. Albumin-binding proteins (ABP) might be instrumental in this interaction. Moreover, plasmalemmal fatty acid-binding protein (FABPpm), fatty acid translocase (FAT) and fatty acid-transport protein (FATP) are putatively involved in transmembrane movement of the fatty acid molecules. Diffusion through the endothelial cytosol might be facilitated by a cytoplasmic fatty acid-binding protein, the type of which may be related to the epithelial fatty acid-binding protein (E-FAPBc).
- Published
- 1998
- Full Text
- View/download PDF
50. Role of membrane-associated and cytoplasmic fatty acid-binding proteins in cellular fatty acid metabolism.
- Author
-
Glatz JF, van Nieuwenhoven FA, Luiken JJ, Schaap FG, and van der Vusse GJ
- Subjects
- Animals, Cytoplasm metabolism, Endothelium metabolism, Fatty Acid-Binding Protein 7, Fatty Acid-Binding Proteins, Rats, Carrier Proteins metabolism, Fatty Acids metabolism, Membrane Proteins metabolism, Myelin P2 Protein metabolism, Neoplasm Proteins, Nerve Tissue Proteins
- Abstract
A number of membrane-associated and cytoplasmic fatty acid-binding proteins (FABPs) are now being implicated in the cellular uptake and intracellular transport of long-chain fatty acids (FA). These proteins each have the capacity of non-covalent binding of FA, are present in tissues actively involved in FA metabolism, and are upregulated in conditions of increased cellular FA metabolism. To date, five distinct membrane FABPs have been described, ranging in mass from 22 to 88 kDa and each showing a characteristic tissue distribution. Evidence for involvement in cellular fatty acid uptake has been provided for several of them, because it was recently found that isolated cell lines transfected with 88-kDa putative fatty acid translocase (FAT; homologous to CD36) or with 63-kDa fatty acid-transport protein show an increased rate of FA uptake. The (at least nine) FABPs of cytoplasmic origin belong to a family of small (14-15 kDa) lipid binding proteins, all having a similar tertiairy structure but differing in binding properties and in tissue occurrence. The biological functions of the various FABPs, possibly exerted in a concerted action among them, comprise solubilization and compartmentalization of FA, facilitation of the cellular uptake and intracellular trafficking of FA, and modulation of mitosis, cell growth, and cell differentiation. In addition, the FABPs have been suggested to participate in and/or modulate FA-mediated signal transduction pathways and FA regulation of gene expression, and to prevent local high FA concentrations thereby contributing to the protection of cells against the toxic effects of FA. In conclusion, long-chain fatty acids are subject to continuous interaction with multiple proteins, which interplay influences their cellular metabolism.
- Published
- 1997
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.