17 results on '"Arne Olav Melleby"'
Search Results
2. Temporal expression and spatial distribution of the proteoglycan versican during cardiac fibrosis development
- Author
-
Athiramol Sasi, Andreas Romaine, Pugazendhi Murugan Erusappan, Arne Olav Melleby, Almira Hasic, Christen Peder Dahl, Kaspar Broch, Vibeke Marie Almaas, Rosa Doñate Puertas, H. Llewelyn Roderick, Ida Gjervold Lunde, Ivar Sjaastad, Maria Vistnes, and Geir Christensen
- Subjects
Versican ,Cardiac fibrosis ,Cardiomyopathy ,Pressure overload ,Extracellular matrix ,Biology (General) ,QH301-705.5 - Abstract
Cardiac fibrosis is a central pathological feature in several cardiac diseases, but the underlying molecular players are insufficiently understood. The extracellular matrix proteoglycan versican is elevated in heart failure and suggested to be a target for treatment. However, the temporal expression and spatial distribution of versican and the versican cleavage fragment containing the neoepitope DPEAAE in cardiac fibrosis remains to be elucidated. In this study, we have examined versican during cardiac fibrosis development in a murine pressure overload model and in patients with cardiomyopathies. We found that versican, mainly the V1 isoform, was expressed immediately after induction of pressure overload, preceding collagen accumulation, and versican protein levels extended from the perivascular region into the cardiac interstitium. In addition, we found increased production of versican by collagen expressing fibroblasts, and that it was deposited extensively in the fibrotic extracellular matrix during pressure overload. In cardiac cell cultures, the expression of versican was induced by the pro-fibrotic transforming growth factor beta and mechanical stretch. Furthermore, we observed that the proteolytic cleavage of versican (DPEAAE fragment) increased in the late phase of fibrosis development during pressure overload. In patients with hypertrophic and dilated cardiomyopathies, we found elevated levels of versican and a positive correlation between versican and collagen mRNA in the heart, as well as increased cleavage of full-length protein. Taken together, the temporal expression profile and the spatial distribution of both the full-length versican and the DPEAAE fragment observed in this study indicates a role for versican in development of cardiac fibrosis.
- Published
- 2023
- Full Text
- View/download PDF
3. Impact of delayed type hypersensitivity arthritis on development of heart failure by aortic constriction in mice.
- Author
-
Theis Christian Tønnessen, Arne Olav Melleby, Ida Marie Hauge-Iversen, Emil Knut Stenersen Espe, Mohammed Shakil Ahmed, Thor Ueland, Espen Andre Haavardsholm, Sara Marie Atkinson, Espen Melum, Håvard Attramadal, Ivar Sjaastad, and Leif Erik Vinge
- Subjects
Medicine ,Science - Abstract
AimsPatients with rheumatoid arthritis (RA) have increased risk of heart failure (HF). The mechanisms and cardiac prerequisites explaining this association remain unresolved. In this study, we sought to determine the potential cardiac impact of an experimental model of RA in mice subjected to HF by constriction of the ascending aorta.MethodsAorta was constricted via thoracotomy and placement of o-rings with inner diameter 0.55 mm or 0.66 mm, or sham operated. RA-like phenotype was instigated by delayed-type hypersensitivity arthritis (DTHA) two weeks after surgery and re-iterated after additional 18 days. Cardiac magnetic resonance imaging (MRI) was performed before surgery and at successive time points throughout the study. Six weeks after surgery the mice were euthanized, blood and tissue were collected, organ weights were documented, and expression levels of cardiac foetal genes were analysed. In a supplemental study, DTHA-mice were euthanized throughout 14 days after induction of arthritis, and blood was analysed for important markers and mediators of RA (SAP, TNF-α and IL-6). In order to put the latter findings into clinical context, the same molecules were analysed in serum from untreated RA patients and compared to healthy controls.ResultsSignificant elevations of inflammatory markers were found in both patient- and murine blood. Furthermore, the DTHA model appeared clinically relevant when compared to the inflammatory responses observed in three prespecified RA severity disease states. Two distinct trajectories of cardiac dysfunction and HF development were found using the two o-ring sizes. These differences were consistent by both MRI, organ weights and cardiac foetal gene expression levels. Still, no difference within the HF groups, nor within the sham groups, could be found when DTHA was induced.ConclusionDTHA mediated systemic inflammation did not cause, nor modify HF caused by aortic constriction. This indicates other prerequisites for RA-induced cardiac dysfunction.
- Published
- 2022
- Full Text
- View/download PDF
4. Generation of a novel mouse strain with fibroblast-specific expression of Cre recombinase
- Author
-
Jahedul Alam, Moses Musiime, Andreas Romaine, Mugdha Sawant, Arne Olav Melleby, Ning Lu, Beate Eckes, Geir Christensen, and Donald Gullberg
- Subjects
Integrin alpha11 ,Fibroblast ,Cre-recombinase ,Integrase ,Fibroblast-specific ,Biology (General) ,QH301-705.5 - Abstract
Cell-specific expression of genes offers the possibility to use their promoters to drive expression of Cre-recombinase, thereby allowing for detailed expression analysis using reporter gene systems, cell lineage tracing, conditional gene deletion, and cell ablation. In this context, current data suggest that the integrin α11 subunit has the potential to serve as a fibroblast biomarker in tissue regeneration and pathology, in particular in wound healing and in tissue- and tumor fibrosis. The mesenchyme-restricted expression pattern of integrin α11 thus prompted us to generate a novel ITGA11-driver Cre mouse strain using a ϕC31 integrase-mediated knock-in approach. In this transgenic mouse, the Cre recombinase is driven by regulatory promoter elements within the 3 kb segment of the human ITGA11 gene. β-Galactosidase staining of embryonic tissues obtained from a transgenic ITGA11-Cre mouse line crossed with Rosa 26R reporter mice (ITGA11-Cre;R26R) revealed ITGA11-driven Cre expression and activity in mesenchymal cells in a variety of mesenchymal tissues in a pattern reminiscent of endogenous α11 protein expression in mouse embryos. Interestingly, X-gal staining of mouse embryonic fibroblasts (MEFs) isolated from the ITGA11-Cre;R26R mice indicated heterogeneity in the MEF population. ITGA11-driven Cre activity was shown in approximately 60% of the MEFs, suggesting that the expression of integrin α11 could be exploited for isolation of different fibroblast populations. ITGA11-driven Cre expression was found to be low in adult mouse tissues but was induced in granulation tissue of excisional wounds and in fibrotic hearts following aortic banding. We predict that the ITGA11-Cre transgenic mouse strain described in this report will be a useful tool in matrix research for the deletion of genes in subsets of fibroblasts in the developing mouse and for determining the function of subsets of pro-fibrotic fibroblasts in tissue fibrosis and in different subsets of cancer-associated fibroblasts in the tumor microenvironment.
- Published
- 2020
- Full Text
- View/download PDF
5. Syndecan‐4 Protects the Heart From the Profibrotic Effects of Thrombin‐Cleaved Osteopontin
- Author
-
Kate M. Herum, Andreas Romaine, Ariel Wang, Arne Olav Melleby, Mari E. Strand, Julian Pacheco, Bjørn Braathen, Pontus Dunér, Theis Tønnessen, Ida G. Lunde, Ivar Sjaastad, Cord Brakebusch, Andrew D. McCulloch, Maria F. Gomez, Cathrine R. Carlson, and Geir Christensen
- Subjects
aortic stenosis ,cardiac fibroblasts ,left ventricular fibrosis ,extracellular matrix ,mechanical ,myofibroblast ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
Background Pressure overload of the heart occurs in patients with hypertension or valvular stenosis and induces cardiac fibrosis because of excessive production of extracellular matrix by activated cardiac fibroblasts. This initially provides essential mechanical support to the heart, but eventually compromises function. Osteopontin is associated with fibrosis; however, the underlying signaling mechanisms are not well understood. Herein, we examine the effect of thrombin‐cleaved osteopontin on fibrosis in the heart and explore the role of syndecan‐4 in regulating cleavage of osteopontin. Methods and Results Osteopontin was upregulated and cleaved by thrombin in the pressure‐overloaded heart of mice subjected to aortic banding. Cleaved osteopontin was higher in plasma from patients with aortic stenosis receiving crystalloid compared with blood cardioplegia, likely because of less heparin‐induced inhibition of thrombin. Cleaved osteopontin and the specific osteopontin peptide sequence RGDSLAYGLR that is exposed after thrombin cleavage both induced collagen production in cardiac fibroblasts. Like osteopontin, the heparan sulfate proteoglycan syndecan‐4 was upregulated after aortic banding. Consistent with a heparan sulfate binding domain in the osteopontin cleavage site, syndecan‐4 was found to bind to osteopontin in left ventricles and cardiac fibroblasts and protected osteopontin from cleavage by thrombin. Shedding of the extracellular part of syndecan‐4 was more prominent at later remodeling phases, at which time levels of cleaved osteopontin were increased. Conclusions Thrombin‐cleaved osteopontin induces collagen production by cardiac fibroblasts. Syndecan‐4 protects osteopontin from cleavage by thrombin, but this protection is lost when syndecan‐4 is shed in later phases of remodeling, contributing to progression of cardiac fibrosis.
- Published
- 2020
- Full Text
- View/download PDF
6. Disruption of Phosphodiesterase 3A Binding to SERCA2 Increases SERCA2 Activity and Reduces Mortality in Mice With Chronic Heart Failure
- Author
-
Jonas Skogestad, Ingrid Albert, Karina Hougen, Gustav B. Lothe, Marianne Lunde, Olav Søvik Eken, Ioanni Veras, Ngoc Trang Thi Huynh, Mira Børstad, Serena Marshall, Xin Shen, William E. Louch, Emma Louise Robinson, Joseph C. Cleveland, Amrut V. Ambardekar, Jessica A. Schwisow, Eric Jonas, Ana I. Calejo, Jens Preben Morth, Kjetil Taskén, Arne Olav Melleby, Per Kristian Lunde, Ivar Sjaastad, Cathrine Rein Carlson, and Jan Magnus Aronsen
- Subjects
Physiology (medical) ,Cardiology and Cardiovascular Medicine - Abstract
Background: Increasing SERCA2 (sarco[endo]-plasmic reticulum Ca 2+ ATPase 2) activity is suggested to be beneficial in chronic heart failure, but no selective SERCA2-activating drugs are available. PDE3A (phosphodiesterase 3A) is proposed to be present in the SERCA2 interactome and limit SERCA2 activity. Disruption of PDE3A from SERCA2 might thus be a strategy to develop SERCA2 activators. Methods: Confocal microscopy, 2-color direct stochastic optical reconstruction microscopy, proximity ligation assays, immunoprecipitations, peptide arrays, and surface plasmon resonance were used to investigate colocalization between SERCA2 and PDE3A in cardiomyocytes, map the SERCA2/PDE3A interaction sites, and optimize disruptor peptides that release PDE3A from SERCA2. Functional experiments assessing the effect of PDE3A-binding to SERCA2 were performed in cardiomyocytes and HEK293 vesicles. The effect of SERCA2/PDE3A disruption by the disruptor peptide OptF (optimized peptide F) on cardiac mortality and function was evaluated during 20 weeks in 2 consecutive randomized, blinded, and controlled preclinical trials in a total of 148 mice injected with recombinant adeno-associated virus 9 (rAAV9)-OptF, rAAV9-control (Ctrl), or PBS, before undergoing aortic banding (AB) or sham surgery and subsequent phenotyping with serial echocardiography, cardiac magnetic resonance imaging, histology, and functional and molecular assays. Results: PDE3A colocalized with SERCA2 in human nonfailing, human failing, and rodent myocardium. Amino acids 277–402 of PDE3A bound directly to amino acids 169–216 within the actuator domain of SERCA2. Disruption of PDE3A from SERCA2 increased SERCA2 activity in normal and failing cardiomyocytes. SERCA2/PDE3A disruptor peptides increased SERCA2 activity also in the presence of protein kinase A inhibitors and in phospholamban-deficient mice, and had no effect in mice with cardiomyocyte-specific inactivation of SERCA2. Cotransfection of PDE3A reduced SERCA2 activity in HEK293 vesicles. Treatment with rAAV9-OptF reduced cardiac mortality compared with rAAV9-Ctrl (hazard ratio, 0.26 [95% CI, 0.11 to 0.63]) and PBS (hazard ratio, 0.28 [95% CI, 0.09 to 0.90]) 20 weeks after AB. Mice injected with rAAV9-OptF had improved contractility and no difference in cardiac remodeling compared with rAAV9-Ctrl after aortic banding. Conclusions: Our results suggest that PDE3A regulates SERCA2 activity through direct binding, independently of the catalytic activity of PDE3A. Targeting the SERCA2/PDE3A interaction prevented cardiac mortality after AB, most likely by improving cardiac contractility.
- Published
- 2023
- Full Text
- View/download PDF
7. Integrin α11β1 and syndecan-4 dual receptor ablation attenuate cardiac hypertrophy in the pressure overloaded heart
- Author
-
Andreas Romaine, Arne Olav Melleby, Jahedul Alam, Viola Hélène Lobert, Ning Lu, Francesca E. Lockwood, Almira Hasic, Ida G. Lunde, Ivar Sjaastad, Harald Stenmark, Kate M. Herum, Donald Gullberg, and Geir Christensen
- Subjects
Mice, Knockout ,Integrins ,Receptors, Collagen ,Isoflurane ,Physiology ,Cardiomegaly ,Oxygen ,Mice ,Physiology (medical) ,Animals ,Myocytes, Cardiac ,Syndecan-4 ,Cardiology and Cardiovascular Medicine ,Integrin alpha Chains - Abstract
Pathological myocardial hypertrophy in response to an increase in left ventricular (LV) afterload may ultimately lead to heart failure. Cell surface receptors bridge the interface between the cell and the extracellular matrix (ECM) in cardiac myocytes and cardiac fibroblasts and have been suggested to be important mediators of pathological myocardial hypertrophy. We identify for the first time that integrin α11 (α11) is preferentially upregulated among integrin β1 heterodimer-forming α-subunits in response to increased afterload induced by aortic banding (AB) in wild-type (WT) mice. Mice were anesthetized in a chamber with 4% isoflurane and 95% oxygen before being intubated and ventilated with 2.5% isoflurane and 97% oxygen. For pre- and postoperative analgesia, animals were administered 0.02-mL buprenorphine (0.3 mg/mL) subcutaneously. Surprisingly, mice lacking α11 develop myocardial hypertrophy following AB comparable to WT. In the mice lacking α11, we further show a compensatory increase in the expression of another mechanoreceptor, syndecan-4, following AB compared with WT AB mice, indicating that syndecan-4 compensated for lack of α11. Intriguingly, mice lacking mechanoreceptors α11 and syndecan-4 show ablated myocardial hypertrophy following AB compared with WT mice. Expression of the main cardiac collagen isoforms col1a2 and col3a1 was significantly reduced in AB mice lacking mechanoreceptors α11 and syndecan-4 compared with WT AB.
- Published
- 2022
- Full Text
- View/download PDF
8. Inhibition of the extracellular enzyme ADAMTS4 prevents cardiac fibrosis and dysfunction
- Author
-
Maria Vistnes, Pugazendhi Murugan Erusappan, Athiramol Sasi, Einar Sjaastad Nordén, Kaja Bergo, Andreas Romaine, Ida Gjervold Lunde, Lili Zhang, Maria Belland Olsen, Jonas Øgaard, Cathrine Rein Carlson, Christian Hjorth Wang, Jon Riise, Christen P Dahl, Arnt Eltvedt Fiane, IdaMarie Hauge-Iversen, Emil Espe, Arne Olav Melleby, Theis Tønnessen, Jan Magnus Aronsen, Ivar Sjaastad, and Geir Christensen
- Subjects
Physiology ,Physiology (medical) ,Cardiology and Cardiovascular Medicine - Abstract
Aims Heart failure is a condition with high mortality rates, and there is a lack of therapies that directly target maladaptive changes in the extracellular matrix (ECM), such as fibrosis. We investigated whether the ECM enzyme known as A disintegrin and metalloprotease with thrombospondin motif (ADAMTS) 4 might serve as a therapeutic target in treatment of heart failure and cardiac fibrosis. Methods and results The effects of pharmacological ADAMTS4 inhibition on cardiac function and fibrosis were examined in rats exposed to cardiac pressure overload. Disease mechanisms affected by the treatment were identified based on changes in the myocardial transcriptome. Following aortic banding (AB), rats receiving an ADAMTS inhibitor, with high inhibitory capacity for ADAMTS4, showed substantially better cardiac function than vehicle-treated rats, including ∼30 % reduction in E/e’ and left atrial diameter, indicating an improvement in diastolic function. ADAMTS inhibition also resulted in a marked reduction in myocardial collagen content and a downregulation of transforming growth factor (TGF) β target genes. The mechanism for the beneficial effects of ADAMTS inhibition was further studied in cultured human cardiac fibroblasts producing mature ECM. ADAMTS4 caused a 50% increase in the TGF-β levels in the medium. Simultaneously, ADAMTS4 elicited a not previously known cleavage of TGF-β-binding proteins, i.e. latent binding protein of TGF-β (LTBP1) and extra domain A (EDA)-fibronectin. These effects were abolished by the ADAMTS inhibitor. In failing human hearts, we observed a marked increase in ADAMTS4 expression and cleavage activity. Conclusion Inhibition of ADAMTS4 improves cardiac function and reduces collagen accumulation in rats with cardiac pressure overload, possibly through a not previously known cleavage of molecules that control TGF-β availability. Targeting ADAMTS4 may serve as a novel strategy in heart failure treatment, in particular in heart failure with fibrosis and diastolic dysfunction.
- Published
- 2023
- Full Text
- View/download PDF
9. CNP regulates cardiac contractility and increases cGMP near both SERCA and TnI: difference from BNP visualized by targeted cGMP biosensors
- Author
-
Nicoletta C. Surdo, Finn Olav Levy, Ornella Manfra, Kjetil Wessel Andressen, Monica Aasrum, Manuela Zaccolo, Arne Olav Melleby, Dulasi Arunthavarajah, Gaia Calamera, Jan Magnus Aronsen, Lise Román Moltzau, Alexander Froese, Viacheslav O. Nikolaev, and Silja Meier
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Lusitropy ,Physiology ,medicine.drug_class ,Phosphodiesterase 3 ,Biosensing Techniques ,030204 cardiovascular system & hematology ,Endoplasmic Reticulum ,Contractility ,03 medical and health sciences ,0302 clinical medicine ,Physiology (medical) ,Internal medicine ,Natriuretic Peptide, Brain ,Troponin I ,medicine ,Natriuretic peptide ,Animals ,Myocytes, Cardiac ,Cyclic GMP ,Chemistry ,Phosphodiesterase ,Natriuretic Peptide, C-Type ,Brain natriuretic peptide ,Myocardial Contraction ,Rats ,Phospholamban ,030104 developmental biology ,Endocrinology ,Guanylate Cyclase ,cardiovascular system ,Cardiology and Cardiovascular Medicine ,Receptors, Atrial Natriuretic Factor ,Atrial Natriuretic Factor - Abstract
Aims Guanylyl cyclase-B (GC-B; natriuretic peptide receptor-B, NPR-B) stimulation by C-type natriuretic peptide (CNP) increases cGMP and causes a lusitropic and negative inotropic response in adult myocardium. These effects are not mimicked by NPR-A (GC-A) stimulation by brain natriuretic peptide (BNP), despite similar cGMP increase. More refined methods are needed to better understand the mechanisms of the differential cGMP signaling and compartmentation. The aim of this work was to measure cGMP near proteins involved in regulating contractility to understand compartmentation of cGMP signaling in adult cardiomyocytes. Methods and results We constructed several fluorescence resonance energy transfer (FRET)-based biosensors for cGMP subcellularly targeted to phospholamban (PLB) and troponin I (TnI). CNP stimulation of adult rat cardiomyocytes increased cGMP near PLB and TnI, whereas BNP stimulation increased cGMP near PLB, but not TnI. The phosphodiesterases PDE2 and PDE3 constrained cGMP in both compartments. Local receptor stimulation aided by scanning ion conductance microscopy (SICM) combined with FRET revealed that CNP stimulation both in the t-tubules and on the cell crests increases cGMP similarly near both TnI and PLB. In ventricular strips, CNP stimulation, but not BNP, induced a lusitropic response, enhanced by inhibition of either PDE2 or PDE3, and a negative inotropic response. In cardiomyocytes from heart failure rats, CNP increased cGMP near PLB and TnI more pronounced than in cells from sham-operated animals. Conclusions These targeted biosensors demonstrate that CNP, but not BNP, increases cGMP near TnI in addition to PLB, explaining how CNP, but not BNP is able to induce lusitropic and negative inotropic responses. Translational perspective Although best known as heart failure biomarkers, natriuretic peptides (ANP, BNP and CNP) are important signaling molecules in the heart and other organs through increasing cyclic GMP (cGMP). Treatment preventing their degradation improves heart failure prognosis. To better understand their cardiac signaling, we employed fluorescent cGMP biosensors targeted to troponin I and phospholamban and found that BNP and CNP increase cGMP differently around these proteins in both normal and failing cardiomyocytes. This may explain the different effects of BNP and CNP on cardiac contractility and relaxation, with possible implications for understanding and treatment of heart failure.
- Published
- 2021
- Full Text
- View/download PDF
10. Cardiac fibroblasts acquire properties of matrifibrocytes in vitro and in mice with pressure overload-induced congestive heart failure
- Author
-
Andreas Romaine, Geir Christensen, Cord Brakebusch, Andrew D. McCulloch, Arne Olav Melleby, Kate M. Herum, and George K. Gilles
- Subjects
Pressure overload ,medicine.medical_specialty ,business.industry ,Internal medicine ,Heart failure ,Cardiology ,Medicine ,Cardiology and Cardiovascular Medicine ,business ,medicine.disease ,In vitro - Abstract
Introduction Activation of cardiac fibroblasts (CFB) is a key step in development of fibrosis in the heart. It was recently shown that, in addition to the well-studied myofibroblast (myoFB) phenotype, activated cardiac fibroblasts can adopt a newly defined matrifibrocyte phenotype, characterized by expression of extracellular matrix (ECM) genes associated with bone, cartilage and tendon development. However, it is unknown whether matrifibrocytes exists in the pressure-overloaded fibrotic and failing heart, and whether substrate stiffness drives differentiation. Hypothesis Matrifibrocyte differentiation occurs in vitro during culturing of primary cardiac fibroblasts, and in vivo in response to left ventricular pressure overload. Methods Left ventricular pressure overload induced by o-ring aortic banding (ORAB) induced cardiac phenotypes of concentric hypertrophic remodelling and congestive heart failure. Primary CFB from adult mice were cultured on plastic or soft polyacrylamide hydrogels (4.5 kPa) for various times. mRNA expression of phenotypic markers were measured by RT-PCR. Presence of smooth muscle α-actin (SMA) fibers was determined by immunocytochemistry. Results ECM genes normally expressed in bone and cartilage (COMP, CILP-2, OPG and SCX) were upregulated in hypertrophic left ventricles of mice with congestive heart failure. The myoFB marker acta2 was increased 2 weeks after ORAB, returned to baseline at 4 weeks and increased again at 20 weeks when the left ventricle was dilating and failing, indicating that the myoFB phenotype is not permanent. In vitro, primary CFB upregulated bone/cartilage-associated ECM genes after 12 days of culturing on plastic. Acta2 mRNA and SMA protein levels peaked after 9 days in culture whereafter they declined, indicating a shift in phenotype. Culturing primary CFB on soft (4.5 kPa) hydrogels delayed, but did not prevent, myoFB differentiation while expression of bone/cartilage ECM genes was absent or low, indicating that high stiffness is a driver of the matrifibrocyte phenotype. Blockers of mechanotransduction, SB431542 (TGFβRI inhibitor), Y27623 (ROCK inhibitor) and cyclosporine A (calcineurin inhibitor), completely inhibited myoFB differentiation but upregulated several matrifibrocyte markers, indicating that distinct signaling pathways regulate myoFB and matrifibrocyte differentiation. Removing inhibitors re-induced myofibroblast markers in cells on plastic but not on soft gels consistent with high stiffness promoting myofibroblast differentiation. Conclusion Primary cardiac fibroblasts acquire characteristics of matrifibrocytes in vitro when cultured for long time on plastic and in vivo in left ventricles of mice with pressure overload-induced congestive heart failure. Funding Acknowledgement Type of funding source: Public grant(s) – EU funding. Main funding source(s): Marie Sklodowska-Curie Individual Fellowship
- Published
- 2020
- Full Text
- View/download PDF
11. Generation of a novel mouse strain with fibroblast-specific expression of Cre recombinase
- Author
-
Ning Lu, Moses Musiime, Andreas Romaine, Geir Christensen, Beate Eckes, Jahedul Alam, Arne Olav Melleby, Mugdha Sawant, and Donald Gullberg
- Subjects
Genetically modified mouse ,Histology ,Transgene ,Population ,Biophysics ,Cre recombinase ,Fibroblast-specific ,Biology ,Integrase ,Biochemistry ,Article ,Cre-recombinase ,Genetics ,medicine ,education ,Fibroblast ,Molecular Biology ,lcsh:QH301-705.5 ,education.field_of_study ,Tumor microenvironment ,Reporter gene ,Cell Biology ,Integrin alpha11 ,Embryonic stem cell ,Cell biology ,medicine.anatomical_structure ,lcsh:Biology (General) - Abstract
Cell-specific expression of genes offers the possibility to use their promoters to drive expression of Cre-recombinase, thereby allowing for detailed expression analysis using reporter gene systems, cell lineage tracing, conditional gene deletion, and cell ablation. In this context, current data suggest that the integrin α11 subunit has the potential to serve as a fibroblast biomarker in tissue regeneration and pathology, in particular in wound healing and in tissue- and tumor fibrosis. The mesenchyme-restricted expression pattern of integrin α11 thus prompted us to generate a novel ITGA11-driver Cre mouse strain using a ϕC31 integrase-mediated knock-in approach. In this transgenic mouse, the Cre recombinase is driven by regulatory promoter elements within the 3 kb segment of the human ITGA11 gene. β-Galactosidase staining of embryonic tissues obtained from a transgenic ITGA11-Cre mouse line crossed with Rosa 26R reporter mice (ITGA11-Cre;R26R) revealed ITGA11-driven Cre expression and activity in mesenchymal cells in a variety of mesenchymal tissues in a pattern reminiscent of endogenous α11 protein expression in mouse embryos. Interestingly, X-gal staining of mouse embryonic fibroblasts (MEFs) isolated from the ITGA11-Cre;R26R mice indicated heterogeneity in the MEF population. ITGA11-driven Cre activity was shown in approximately 60% of the MEFs, suggesting that the expression of integrin α11 could be exploited for isolation of different fibroblast populations. ITGA11-driven Cre expression was found to be low in adult mouse tissues but was induced in granulation tissue of excisional wounds and in fibrotic hearts following aortic banding. We predict that the ITGA11-Cre transgenic mouse strain described in this report will be a useful tool in matrix research for the deletion of genes in subsets of fibroblasts in the developing mouse and for determining the function of subsets of pro-fibrotic fibroblasts in tissue fibrosis and in different subsets of cancer-associated fibroblasts in the tumor microenvironment., Highlights • A mouse strain with Cre-recombinase driven by the human integrin α11 promoter has been generated. • Cre-recombinase expression in this strain has been characterized using the Rosa26R reporter mouse. • ITGA11-Cre is restricted to fibroblast subsets in mouse embryos, skin wounds and fibrotic hearts. • This Cre-driver strain will be a useful tool to study role fibroblasts in fibrosis and tumors.
- Published
- 2020
12. Moderate Loss of the Extracellular Matrix Proteoglycan Lumican Attenuates Cardiac Fibrosis in Mice Subjected to Pressure Overload
- Author
-
Ivar Sjaastad, Geir Christensen, Arne Olav Melleby, Theis Tønnessen, Kristin V.T. Engebretsen, Naiyereh Mohammadzadeh, Ida G. Lunde, Sheryl Palmero, and Shukti Chakravarti
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,Lumican ,Cardiac fibrosis ,Heart Ventricles ,Diastole ,030204 cardiovascular system & hematology ,Periostin ,Article ,Extracellular matrix ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Fibrosis ,Internal medicine ,Medicine ,Animals ,Pharmacology (medical) ,Myofibroblasts ,Pressure overload ,Heart Failure ,Mice, Knockout ,Ventricular Remodeling ,business.industry ,medicine.disease ,Extracellular Matrix ,Disease Models, Animal ,030104 developmental biology ,Endocrinology ,Echocardiography ,Heart failure ,Hypertrophy, Left Ventricular ,Collagen ,Cardiology and Cardiovascular Medicine ,business - Abstract
Introduction: The heart undergoes myocardial remodeling during progression to heart failure following pressure overload. Myocardial remodeling is associated with structural and functional changes in cardiac myocytes, fibroblasts, and the extracellular matrix (ECM) and is accompanied by inflammation. Cardiac fibrosis, the accumulation of ECM molecules including collagens and collagen cross-linking, contributes both to impaired systolic and diastolic function. Insufficient mechanistic insight into what regulates cardiac fibrosis during pathological conditions has hampered therapeutic solutions. Lumican (LUM) is an ECM-secreted proteoglycan known to regulate collagen fibrillogenesis. Its expression in the heart is increased in clinical and experimental heart failure. Furthermore, LUM is important for survival and cardiac remodeling following pressure overload. We have recently reported that total lack of LUM increased mortality and left ventricular dilatation, and reduced collagen expression and cross-linking in LUM knockout mice after aortic banding (AB). Here, we examined the effect of LUM on myocardial remodeling and function following pressure overload in a less extreme mouse model, where cardiac LUM level was reduced to 50% (i.e., moderate loss of LUM). Methods and Results: mRNA and protein levels of LUM were reduced to 50% in heterozygous LUM (LUM+/–) hearts compared to wild-type (WT) controls. LUM+/– mice were subjected to AB. There was no difference in survival between LUM+/– and WT mice post-AB. Echocardiography revealed no striking differences in cardiac geometry between LUM+/– and WT mice 2, 4, and 6 weeks post-AB, although markers of diastolic dysfunction indicated better function in LUM+/– mice. LUM+/– hearts revealed reduced cardiac fibrosis assessed by histology. In accordance, the expression of collagen I and III, the main fibrillar collagens in the heart, and other ECM molecules central to fibrosis, i.e. including periostin and fibronectin, was reduced in the hearts of LUM+/– compared to WT 6 weeks post-AB. We found no differences in collagen cross-linking between LUM+/– and WT mice post-AB, as assessed by histology and qPCR. Conclusions: Moderate lack of LUM attenuated cardiac fibrosis and improved diastolic dysfunction following pressure overload in mice, adding to the growing body of evidence suggesting that LUM is a central profibrotic molecule in the heart that could serve as a potential therapeutic target.
- Published
- 2019
13. H2S-producing enzymes in anoxia-tolerant vertebrates: Effects of cold acclimation, anoxia exposure and reoxygenation on gene and protein expression
- Author
-
Göran E. Nilsson, Arne Olav Melleby, Jonathan A. W. Stecyk, Guro K. Sandvik, and Christine S. Couturier
- Subjects
inorganic chemicals ,0301 basic medicine ,chemistry.chemical_classification ,biology ,Physiology ,Sulfurtransferase ,musculoskeletal system ,biology.organism_classification ,Biochemistry ,Cystathionine beta synthase ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Enzyme ,chemistry ,Downregulation and upregulation ,Gene expression ,Crucian carp ,biology.protein ,Cold acclimation ,Molecular Biology ,Gene ,030217 neurology & neurosurgery - Abstract
To lend insight into the potential role of the gasotransmitter hydrogen sulfide (H2S) in facilitating anoxia survival of anoxia-tolerant vertebrates, we quantified the gene expression of the primary H2S-synthesizing enzymes, 3-mercaptopyruvate sulfurtransferase (3MST), cystathionine γ-lyase (CSE) and cystathionine β-synthase (CBS), in ventricle and brain of normoxic, anoxic and reoxygenated 21 °C- and 5 °C-acclimated freshwater turtles (Trachemys scripta) and 10 °C-acclimated crucian carp (Carassius carassius). Semi-quantitative Western blotting analysis was also conducted to assess 3MST and CBS protein abundance in ventricle and brain of 5 °C turtles and 10 °C crucian carp subjected to normoxia, anoxia and reoxygenation. We hypothesized that if H2S was advantageous for anoxia survival, expression levels would remain unchanged or be upregulated with anoxia and/or reoxygenation. Indeed, for both species, gene and protein expression were largely maintained with anoxia exposure (24 h, 21 °C; 5 d, 10 °C; 14 d, 5 °C). With reoxygenation, 3MST expression was increased in turtle and crucian carp brain at the protein and gene level, respectively. Additionally, the effect of cold acclimation on gene expression was assessed in several tissues of the turtle. Expression levels were maintained in most tissues, but decreased in others. The maintenance of gene and protein expression of the H2S-producing enzymes with anoxia exposure and the up-regulation of 3MST with reoxygenation suggests that H2S may facilitate anoxic survival of the two champions of vertebrate anoxia survival. The differential effects of cold acclimation on H2S enzyme expression may influence blood flow to different tissues during winter anoxia.
- Published
- 2020
- Full Text
- View/download PDF
14. A novel method for high precision aortic constriction that allows for generation of specific cardiac phenotypes in mice
- Author
-
Ida G. Lunde, Geir Christensen, Andreas Romaine, Ivar Sjaastad, Ioanni Veras, Lili Zhang, Arne Olav Melleby, and Jan Magnus Aronsen
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,Time Factors ,Physiology ,Mice, Transgenic ,030204 cardiovascular system & hematology ,Ventricular Function, Left ,Constriction ,Muscle hypertrophy ,03 medical and health sciences ,Ventricular Dysfunction, Left ,0302 clinical medicine ,Afterload ,Physiology (medical) ,Internal medicine ,medicine.artery ,Ascending aorta ,medicine ,Animals ,Ventricular remodeling ,Ligation ,Aorta ,Ejection fraction ,NFATC Transcription Factors ,Ventricular Remodeling ,business.industry ,Myocardium ,Stroke Volume ,medicine.disease ,Fibrosis ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Phenotype ,Gene Expression Regulation ,Heart failure ,Cardiology ,cardiovascular system ,Disease Progression ,Hypertrophy, Left Ventricular ,Inflammation Mediators ,Mitogen-Activated Protein Kinases ,Cardiology and Cardiovascular Medicine ,business ,Signal Transduction - Abstract
Aims Generation of reproducible cardiac disease phenotypes in mice is instrumental for investigating mechanisms leading to heart failure (HF). For decades, suture-based thoracic aortic constriction has been the preferred method for increasing left ventricular (LV) afterload in rodents, but the degree of stenosis resulting from this method is variable. In an effort to improve this methodology, we subjected mice to constriction of the ascending aorta using o-rings with fixed inner diameters (IDs). Methods and results Mice of C57BL/6J and FVB/N background were subjected to constriction of the ascending aorta using o-rings with fixed IDs of 0.71, 0.66, and 0.61 mm. O-ring aortic banding resulted in 98.7% survival 2 weeks post-surgery, with very low intra- and inter-surgeon variation. When using the narrowest o-ring (0.61 mm), mice developed hypertrophy within 1 week. Over 20 weeks, the mice gradually developed reduced LV ejection fraction (LVEF) and dilatation with increased left atrial dimensions and lung weight, indicating congestion. When using o-rings with IDs of 0.66 mm and 0.71 mm, the mice developed hypertrophy, but maintained a compensated state with stabilized LVEF 8-20 weeks post-surgery. The up-regulation of signature genes associated with HF, hypertrophy, fibrosis, and the level of activation of MAPK and NFAT signalling pathways corresponded to the degree of stenosis. Conclusion Here, we introduce a novel method for high precision aortic constriction in mice with high intra- and inter-surgeon reproducibility and low post-operative mortality that allows generation of specific cardiac disease phenotypes.
- Published
- 2018
15. Abstract 417: Syndecan-4 Regulates the Effect of Osteopontin on Cardiac Fibroblast Function and Phenotype
- Author
-
Kate M Herum, Andreas Romaine, Arne Olav Melleby, Mari E Strand, Bjørn Braathen, Andrew D McCulloch, Cathrine R Carlson, and Geir Christensen
- Subjects
carbohydrates (lipids) ,Physiology ,Cardiology and Cardiovascular Medicine - Abstract
Pressure overload of the heart induces cardiac fibrosis due to excessive production of extracellular matrix by activated cardiac fibroblasts. This compromises cardiac function and may lead to heart failure. The transmembrane proteoglycan syndecan-4 is upregulated in response to pressure overload and can bind extracellular signaling proteins via its heparan sulfate glycosaminoglycan chains. The matricellular protein osteopontin is associated with fibrosis and contains a heparan binding domain that overlaps with its thrombin cleavage site. We here examine the role of the extracellular part of syndecan-4 in regulating proteolytic cleavage and pro-fibrotic activity of the matricellular protein osteopontin. Syndecan-4 and osteopontin mRNA displayed similar regulation patterns in response to left ventricular pressure overload induced by aortic banding in mice. Syndecan-4 and osteopontin bound to each other in left ventricles and fibroblasts as shown by native western blots, immunoprecipitation and immunofluorescent staining. Osteopontin was protected from cleavage by thrombin when pre-incubated with the extracellular domain of syndecan-4. Thrombin-cleaved osteopontin induced a pro-fibrotic phenotype when given to fibroblasts, whereas full-length osteopontin favored a more quiescent phenotype. Thus, our data suggest that the extracellular domain of syndecan-4 protects against the pro-fibrotic effects of osteopontin. Following the initial stage of extracellular remodeling, the extracellular part of syndecan-4 is shed from the cell surface. Accordingly, increased cleaved osteopontin was detected in hypertrophic pressure-overloaded left ventricles three days after aortic banding. Cleaved osteopontin was also detected in plasma from patients with aortic stenosis and displayed a higher cleaved/full-length ratio in the coronary sinus than the radial artery, suggestion that osteopontin is in fact cleaved locally in the heart. Our results indicate an anti-fibrotic effect of the extracellular part of syndecan-4 when it is still attached to the cell surface. Shedding of syndecan-4 from the cell surface at later stages of remodeling exposes osteopontin to cleavage by thrombin, enabling pro-fibrotic effects on cardiac fibroblasts.
- Published
- 2017
- Full Text
- View/download PDF
16. The Heparan Sulfate Proteoglycan Glypican-6 Is Upregulated in the Failing Heart, and Regulates Cardiomyocyte Growth through ERK1/2 Signaling
- Author
-
Geir Christensen, Ida G. Lunde, Biljana Skrbic, Mari E. Strand, Arnt E. Fiane, Jorge Filmus, Andreas Romaine, Kate M. Herum, Ivar Sjaastad, Christen P. Dahl, and Arne Olav Melleby
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,Cell signaling ,Glypican ,Glycobiology ,lcsh:Medicine ,Protein Synthesis ,030204 cardiovascular system & hematology ,Signal transduction ,Biochemistry ,Muscle hypertrophy ,Mice ,0302 clinical medicine ,Animal Cells ,Medicine and Health Sciences ,Myocyte ,Myocytes, Cardiac ,lcsh:Science ,Connective Tissue Cells ,Multidisciplinary ,Ejection fraction ,Sulfates ,Signaling cascades ,Chemical Synthesis ,Heart ,Up-Regulation ,Chemistry ,Connective Tissue ,Physical Sciences ,Proteoglycans ,Cellular Types ,Anatomy ,Research Article ,medicine.medical_specialty ,MAPK signaling cascades ,Biosynthetic Techniques ,MAP Kinase Signaling System ,Immunoblotting ,Cardiology ,Molecular Probe Techniques ,Biology ,Research and Analysis Methods ,03 medical and health sciences ,Downregulation and upregulation ,Glypicans ,Internal medicine ,medicine ,Animals ,Humans ,Rats, Wistar ,Molecular Biology Techniques ,Molecular Biology ,Pressure overload ,Heart Failure ,lcsh:R ,Chemical Compounds ,Biology and Life Sciences ,Proteins ,Cell Biology ,Fibroblasts ,medicine.disease ,Rats ,Mice, Inbred C57BL ,030104 developmental biology ,Endocrinology ,Biological Tissue ,HEK293 Cells ,Heart failure ,Cardiovascular Anatomy ,NIH 3T3 Cells ,Salts ,lcsh:Q - Abstract
Pressure overload is a frequent cause of heart failure. Heart failure affects millions of patients worldwide and is a major cause of morbidity and mortality. Cell surface proteoglycans are emerging as molecular players in cardiac remodeling, and increased knowledge about their regulation and function is needed for improved understanding of cardiac pathogenesis. Here we investigated glypicans (GPC1-6), a family of evolutionary conserved heparan sulfate proteoglycans anchored to the extracellular leaflet of the cell membrane, in experimental and clinical heart failure, and explored the function of glypican-6 in cardiac cells in vitro. In mice subjected to pressure overload by aortic banding (AB), we observed elevated glypican-6 levels during hypertrophic remodeling and dilated, end-stage heart failure. Consistently, glypican-6 mRNA was elevated in left ventricular myocardium from explanted hearts of patients with end-stage, dilated heart failure with reduced ejection fraction. Glypican-6 levels correlated negatively with left ventricular ejection fraction in patients, and positively with lung weight after AB in mice. Glypican-6 mRNA was expressed in both cardiac fibroblasts and cardiomyocytes, and the corresponding protein displayed different sizes in the two cell types due to tissue-specific glycanation. Importantly, adenoviral overexpression of glypican-6 in cultured cardiomyocytes increased protein synthesis and induced mRNA levels of the pro-hypertrophic signature gene ACTA1 and the hypertrophy and heart failure signature genes encoding natriuretic peptides, NPPA and NPPB. Overexpression of GPC6 induced ERK1/2 phosphorylation, and co-treatment with the ERK inhibitor U0126 attenuated the GPC6-induced increase in NPPA, NPPB and protein synthesis. In conclusion, our data suggests that glypican-6 plays a role in clinical and experimental heart failure progression by regulating cardiomyocyte growth through ERK signaling.
- Published
- 2016
17. Overexpression of integrinα11 induces cardiac fibrosis in mice
- Author
-
Lili Zhang, Bård Andre Bendiksen, Arne Olav Melleby, Ivar Sjaastad, Cédric Zeltz, Pugazendhi Erusappan, Ning Lu, Geir Christensen, I. W. Sørensen, Andreas Romaine, Donald Gullberg, Emma L. Robinson, Håvard E. Danielsen, Kate M. Herum, and Jan Magnus Aronsen
- Subjects
Male ,0301 basic medicine ,Genetically modified mouse ,Myocardium/metabolism ,Physiology ,Cardiac fibrosis ,Left Ventricular/metabolism ,Integrin ,Mice, Transgenic ,Left ventricular hypertrophy ,Transgenic ,Muscle hypertrophy ,Extracellular matrix ,Masson's trichrome stain ,Hypertrophy, Left Ventricular/metabolism ,Mice ,03 medical and health sciences ,Fibrosis ,medicine ,Animals ,biology ,Chemistry ,Myocardium ,Hypertrophy ,Integrin alpha Chains/metabolism ,medicine.disease ,Molecular biology ,030104 developmental biology ,biology.protein ,Hypertrophy, Left Ventricular ,Integrin alpha Chains - Abstract
AIM: To understand the role of the collagen-binding integrin α11 in vivo, we have used a classical approach of creating a mouse strain overexpressing integrin α11. A transgenic mouse strain overexpressing α11 in muscle tissues was analysed in the current study with special reference to the heart tissue.METHODS: We generated and phenotyped integrin α11 transgenic (TG) mice by echocardiography, magnetic resonance imaging and histology. Wild-type (WT) mice were subjected to aortic banding (AB) and the expression of integrin α11 was measured in flow cytometry-sorted cardiomyocytes and non-myocytes.RESULTS: TG mice developed left ventricular concentric hypertrophy by 6 months, with increased collagen deposition and reactivation of mRNA encoding foetal genes associated with cardiovascular pathological remodelling compared to WT mice. Masson's trichrome staining revealed interstitial fibrosis, confirmed additionally by magnetic resonance imaging and was found to be most prominent in the cardiac septum of TG but not WT mice. TG hearts expressed increased levels of transforming growth factor-β2 and transforming growth factor-β3 and upregulated smooth muscle actin. Macrophage infiltration coincided with increased NF-κB signalling in TG but not WT hearts. Integrin α11 expression was increased in both cardiomyocytes and non-myocyte cells from WT AB hearts compared to sham-operated animals.CONCLUSION: We report for the first time that overexpression of integrin α11 induces cardiac fibrosis and left ventricular hypertrophy. This is a result of changes in intracellular hypertrophic signalling and secretion of soluble factors that increase collagen production in the heart.
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.