Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Jarkko Soronen is active.

Publication


Featured researches published by Jarkko Soronen.


The FASEB Journal | 2005

Stabilized HIF-1α is superior to VEGF for angiogenesis in skeletal muscle via adeno-associated virus gene transfer

Katri Pajusola; Jaana Künnapuu; Sanna Vuorikoski; Jarkko Soronen; Helder André; Teresa Pereira; Petra Korpisalo; Seppo Ylä-Herttuala; Lorenz Poellinger; Kari Alitalo

Therapeutic angiogenesis provides a potential alternative for the treatment of cardiovascular ischemic diseases. Vascular endothelial growth factor (VEGF) is an important component of the angiogenic response to ischemia. Here we used adeno‐associated virus (AAV) gene delivery to skeletal muscle to examine the effects of VEGF vs. a stabilized form of hypoxia‐inducible factor‐1α (HIF‐1α). The recombinant AAVs were injected into mouse tibialis anterior muscle, and their effects were analyzed by immunohistochemistry and functional assays. These analyses showed that stabilized HIF‐1 α markedly increase capillary sprouting and proliferation, whereas VEGF164 or VEGF120 induced only proliferation of endothelial cells without formation of proper capillary structures. The Evans Blue permeability assay indicated that, unlike VEGF, HIF‐1 α overexpression did not increase vascular leakiness in the transduced muscle. Doppler ultrasound imaging showed that vascular perfusion in the HIF‐1 α treated muscles was significantly enhanced when compared to the controls and not further improved by co‐expression of the arteriogenic growth factors angiopoietin‐1 or platelet‐derived growth factor‐B. Our results show that AAV‐mediated transduction of a stabilized form of HIF‐1 α can circumvent the problems associated with overexpression of individual angiogenic growth factors. HIF‐1 α should thus offer a potent alternative for pro‐angiogenic gene therapy.


Circulation | 2010

Vascular Endothelial Growth Factor-B Acts as a Coronary Growth Factor in Transgenic Rats Without Inducing Angiogenesis, Vascular Leak, or Inflammation

Maija Bry; Riikka Kivelä; Tanja Holopainen; Andrey Anisimov; Tuomas Tammela; Jarkko Soronen; Johanna M. U. Silvola; Antti Saraste; Michael Jeltsch; Petra Korpisalo; Peter Carmeliet; Karl B. Lemström; Masabumi Shibuya; Seppo Ylä-Herttuala; Leena Alhonen; Eero Mervaala; Leif C. Andersson; Juhani Knuuti; Kari Alitalo

Background— Vascular endothelial growth factor-B (VEGF-B) binds to VEGF receptor-1 and neuropilin-1 and is abundantly expressed in the heart, skeletal muscle, and brown fat. The biological function of VEGF-B is incompletely understood. Methods and Results— Unlike placenta growth factor, which binds to the same receptors, adeno-associated viral delivery of VEGF-B to mouse skeletal or heart muscle induced very little angiogenesis, vascular permeability, or inflammation. As previously reported for the VEGF-B167 isoform, transgenic mice and rats expressing both isoforms of VEGF-B in the myocardium developed cardiac hypertrophy yet maintained systolic function. Deletion of the VEGF receptor-1 tyrosine kinase domain or the arterial endothelial Bmx tyrosine kinase inhibited hypertrophy, whereas loss of VEGF-B interaction with neuropilin-1 had no effect. Surprisingly, in rats, the heart-specific VEGF-B transgene induced impressive growth of the epicardial coronary vessels and their branches, with large arteries also seen deep inside the subendocardial myocardium. However, VEGF-B, unlike other VEGF family members, did not induce significant capillary angiogenesis, increased permeability, or inflammatory cell recruitment. Conclusions— VEGF-B appears to be a coronary growth factor in rats but not in mice. The signals for the VEGF-B–induced cardiac hypertrophy are mediated at least in part via the endothelium. Because cardiomyocyte damage in myocardial ischemia begins in the subendocardial myocardium, the VEGF-B–induced increased arterial supply to this area could have therapeutic potential in ischemic heart disease.


Circulation Research | 2009

Activated Forms of VEGF-C and VEGF-D Provide Improved Vascular Function in Skeletal Muscle

Andrey Anisimov; Annamari Alitalo; Petra Korpisalo; Jarkko Soronen; Seppo Kaijalainen; Veli-Matti Leppänen; Michael Jeltsch; Seppo Ylä-Herttuala; Kari Alitalo

The therapeutic potential of vascular endothelial growth factor (VEGF)-C and VEGF-D in skeletal muscle has been of considerable interest as these factors have both angiogenic and lymphangiogenic activities. Previous studies have mainly used adenoviral gene delivery for short-term expression of VEGF-C and VEGF-D in pig, rabbit, and mouse skeletal muscles. Here we have used the activated mature forms of VEGF-C and VEGF-D expressed via recombinant adeno-associated virus (rAAV), which provides stable, long-lasting transgene expression in various tissues including skeletal muscle. Mouse tibialis anterior muscle was transduced with rAAV encoding human or mouse VEGF-C or VEGF-D. Two weeks later, immunohistochemical analysis showed increased numbers of both blood and lymph vessels, and Doppler ultrasound analysis indicated increased blood vessel perfusion. The lymphatic vessels further increased at the 4-week time point were functional, as shown by FITC-lectin uptake and transport. Furthermore, receptor activation and arteriogenic activity were increased by an alanine substitution mutant of human VEGF-C (C137A) having an increased dimer stability and by a chimeric CAC growth factor that contained the VEGF receptor-binding domain flanked by VEGF-C propeptides, but only the latter promoted significantly more blood vessel perfusion when compared to the other growth factors studied. We conclude that long-term expression of VEGF-C and VEGF-D in skeletal muscle results in the generation of new functional blood and lymphatic vessels. The therapeutic value of intramuscular lymph vessels in draining tissue edema and lymphedema can now be evaluated using this model system.


BMC Medical Genomics | 2012

Adipose tissue gene expression analysis reveals changes in inflammatory, mitochondrial respiratory and lipid metabolic pathways in obese insulin-resistant subjects

Jarkko Soronen; Pirkka-Pekka Laurila; Jussi Naukkarinen; Ida Surakka; Samuli Ripatti; Matti Jauhiainen; Vesa M. Olkkonen; Hannele Yki-Järvinen

BackgroundTo get insight into molecular mechanisms underlying insulin resistance, we compared acute in vivo effects of insulin on adipose tissue transcriptional profiles between obese insulin-resistant and lean insulin-sensitive women.MethodsSubcutaneous adipose tissue biopsies were obtained before and after 3 and 6 hours of intravenously maintained euglycemic hyperinsulinemia from 9 insulin-resistant and 11 insulin-sensitive females. Gene expression was measured using Affymetrix HG U133 Plus 2 microarrays and qRT-PCR. Microarray data and pathway analyses were performed with Chipster v1.4.2 and by using in-house developed nonparametric pathway analysis software.ResultsThe most prominent difference in gene expression of the insulin-resistant group during hyperinsulinemia was reduced transcription of nuclear genes involved in mitochondrial respiration (mitochondrial respiratory chain, GO:0001934). Inflammatory pathways with complement components (inflammatory response, GO:0006954) and cytokines (chemotaxis, GO:0042330) were strongly up-regulated in insulin-resistant as compared to insulin-sensitive subjects both before and during hyperinsulinemia. Furthermore, differences were observed in genes contributing to fatty acid, cholesterol and triglyceride metabolism (FATP2, ELOVL6, PNPLA3, SREBF1) and in genes involved in regulating lipolysis (ANGPTL4) between the insulin-resistant and -sensitive subjects especially during hyperinsulinemia.ConclusionsThe major finding of this study was lower expression of mitochondrial respiratory pathway and defective induction of lipid metabolism pathways by insulin in insulin-resistant subjects. Moreover, the study reveals several novel genes whose aberrant regulation is associated with the obese insulin-resistant phenotype.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2009

PDGF-A, -C, and -D but not PDGF-B Increase TGF-β1 and Chronic Rejection in Rat Cardiac Allografts

Raimo Tuuminen; Antti I. Nykänen; R. Krebs; Jarkko Soronen; Katri Pajusola; Mikko A.I. Keränen; Petri K. Koskinen; Kari Alitalo; Karl B. Lemström

Objective—Chronic rejection is the main reason for the poor long-term survival of heart transplant recipients and is characterized by cardiac allograft inflammation, fibrosis, and arteriosclerosis. We examined the specific roles of different platelet-derived growth factor (PDGF) ligands (A–D)—potent mesenchymal cell mitogens—in rat cardiac allografts. Methods and Results—PDGFR-&agr; mRNA was upregulated in acutely-rejecting, and PDGF-A and PDGF-C mRNA in chronically-rejecting cardiac¢hatn allografts. In acute rejection, PDGFR-&agr; immunoreactivity increased in the media of arteries. In chronically-rejecting allografts, immunoreactivity of all PDGF ligands and receptors—except that of PDGF-B ligand—was found in the intima of arteries, and the expression of PDGF-A and PDGF-C was seen in cardiomyocytes. Intracoronary adeno-associated virus-2 (AAV2)-mediated PDGF-A and -D gene transfer enhanced cardiac allograft inflammation. AAV2-PDGF-A, AAV2-PDGF-C, and AAV2-PDGF-D significantly upregulated profibrotic TGF-&bgr;1 mRNA and accelerated cardiac fibrosis and arteriosclerosis. In contrast, AAV2-PDGF-B did not aggravate chronic rejection. Conclusions—We found that alloimmune response induces PDGF-A, PDGF-C, and PDGF-D expression in the graft vasculature. PDGF-A, PDGF-C, and PDGF-D mediated profibrotic and proarteriosclerotic effects in transplanted hearts involving the TGF-&bgr;1 pathway. Inhibition of signaling of all PDGF-ligands except that of PDGF-B may thus be needed to inhibit chronic rejection in cardiac allografts.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2013

Genomic, Transcriptomic, and Lipidomic Profiling Highlights the Role of Inflammation in Individuals With Low High-density Lipoprotein Cholesterol

Pirkka-Pekka Laurila; Ida Surakka; Antti-Pekka Sarin; Laxman Yetukuri; Tuulia Hyötyläinen; Sanni Söderlund; Jussi Naukkarinen; Jing Tang; Johannes Kettunen; Daniel B. Mirel; Jarkko Soronen; Terho Lehtimäki; Aimo Ruokonen; Christian Ehnholm; Johan G. Eriksson; Veikko Salomaa; Antti Jula; Olli T. Raitakari; Marjo-Riitta Järvelin; Aarno Palotie; Leena Peltonen; Matej Orešič; Matti Jauhiainen; Marja-Riitta Taskinen; Samuli Ripatti

Objective—Low high-density lipoprotein cholesterol (HDL-C) is associated with cardiometabolic pathologies. In this study, we investigate the biological pathways and individual genes behind low HDL-C by integrating results from 3 high-throughput data sources: adipose tissue transcriptomics, HDL lipidomics, and dense marker genotypes from Finnish individuals with low or high HDL-C (n=450). Approach and Results—In the pathway analysis of genetic data, we demonstrate that genetic variants within inflammatory pathways were enriched among low HDL-C associated single-nucleotide polymorphisms, and the expression of these pathways upregulated in the adipose tissue of low HDL-C subjects. The lipidomic analysis highlighted the change in HDL particle quality toward putatively more inflammatory and less vasoprotective state in subjects with low HDL-C, as evidenced by their decreased antioxidative plasmalogen contents. We show that the focal point of these inflammatory pathways seems to be the HLA region with its low HDL-associated alleles also associating with more abundant local transcript levels in adipose tissue, increased plasma vascular cell adhesion molecule 1 (VCAM1) levels, and decreased HDL particle plasmalogen contents, markers of adipose tissue inflammation, vascular inflammation, and HDL antioxidative potential, respectively. In a population-based look-up of the inflammatory pathway single-nucleotide polymorphisms in a large Finnish cohorts (n=11 211), no association of the HLA region was detected for HDL-C as quantitative trait, but with extreme HDL-C phenotypes, implying the presence of low or high HDL genes in addition to the population-genomewide association studies–identified HDL genes. Conclusions—Our study highlights the role of inflammation with a genetic component in subjects with low HDL-C and identifies novel cis-expression quantitative trait loci (cis-eQTL) variants in HLA region to be associated with low HDL-C.


The Journal of Clinical Endocrinology and Metabolism | 2015

Regulation of Angiopoietin-Like Proteins (ANGPTLs) 3 and 8 by Insulin.

P.A. Nidhina Haridas; Jarkko Soronen; Sanja Sädevirta; Raghavendra Mysore; Fabiana Quagliarini; Arja Pasternack; Jari Metso; Julia Perttilä; Marja Leivonen; Cynthia M. Smas; Pamela Fischer-Posovszky; Martin Wabitsch; Christian Ehnholm; Olli Ritvos; Matti Jauhiainen; Vesa M. Olkkonen; Hannele Yki-Järvinen

OBJECTIVE Circulating ANGPTL8 has recently been used as a marker of insulin action. We studied expression and insulin regulation of ANGPTL8 and ANGPTL3 in vivo and in vitro. DESIGN AND METHODS Expression of ANGPTL8 and ANGPTL3 was studied in 34 paired samples of human liver and adipose tissue. Effects of insulin on 1) plasma concentrations and adipose tissue expression of ANGPTL8 and ANGPTL3 (in vivo 6-h euglycemic hyperinsulinemia; n = 18), and 2) ANGPTL8 and ANGPTL3 gene and protein expression in immortalized human hepatocytes (IHH) and adipocytes were measured. Effect of ANGPTL3 on secretion of ANGPTL8 in cells stably overexpressing ANGPTL3, -8, or both was determined. RESULTS ANGPTL3 was only expressed in the liver, whereas ANGPTL8 was expressed in both tissues. In vivo hyperinsulinemia significantly decreased both plasma ANGPTL8 and ANGPTL3 at 3 and 6 hours. Insulin increased ANGPTL8 expression in human adipose tissue 14- and 18-fold at 3 and 6 hours and ANGPTL8 was the most insulin-responsive transcript on microarray. Insulin also increased ANPGTL8 in cultured adipocytes and IHH but the protein mainly remained intracellular. In vitro in IHH, insulin decreased ANGPTL3 gene expression and secretion of ANGPTL3 into growth medium. Overexpression of ANGPTL8 in CHO cells did not result in its release into culture medium while abundant secretion occurred in cells co-expressing ANGPTL3 and -8. CONCLUSIONS Insulin decreases plasma ANGPTL3 by decreasing ANGPTL3 expression in the liver. Insulin markedly increases ANGPTL8 in adipose tissue and the liver but not in plasma. These data show that measurement of plasma ANGPTL3 but not -8 reflects insulin action in target tissues.


The Journal of Clinical Endocrinology and Metabolism | 2013

17β-Estradiol and estradiol fatty acyl esters and estrogen-converting enzyme expression in adipose tissue in obese men and women.

Feng Wang; Veera Vihma; Jarkko Soronen; Ursula Turpeinen; Esa Hämäläinen; Hanna Savolainen-Peltonen; Tomi S. Mikkola; Jussi Naukkarinen; Kirsi H. Pietiläinen; Matti Jauhiainen; Hannele Yki-Järvinen; Matti J. Tikkanen

CONTEXT Obesity is associated with increased circulating 17β-estradiol (E₂), but less is known about E₂ concentrations in adipose tissue. In addition to E₂, adipose tissue synthesizes E₂ fatty acyl esters (E₂-FAE). OBJECTIVE The aim was to compare estrogen concentrations and expression of estrogen-converting enzymes in adipose tissue between severely obese men and women. DESIGN AND SETTING Tissue samples were obtained during elective surgery in University Central Hospital in the years 2008 through 2011. PATIENTS We studied 14 men and 22 premenopausal women undergoing bariatric surgery and 10 control women operated for nonmalignant reasons. INTERVENTIONS Paired samples were taken from abdominal sc and visceral adipose tissue and serum and analyzed for E₂ and E₂-FAE by fluoroimmunoassay and liquid chromatography-tandem mass spectrometry. mRNA expression of genes was analyzed by quantitative PCR. RESULTS Compared with men, E₂ levels in sc adipose tissue in obese women were higher, along with higher relative mRNA expression of steroid sulfatase and 17β-hydroxysteroid dehydrogenases 1, 7, and 12. In men, E₂-FAE concentrations in adipose tissue were similar to E₂ but in women significantly lower compared with E₂. Adipose tissue E₂-FAE and serum E₂-FAE levels correlated positively in obese subjects. Serum E₂ did not significantly correlate with E₂ concentration or mRNA expression of genes in adipose tissue in obese men or women. CONCLUSIONS The production of E₂ by the large adipose mass was not reflected by increased circulating E₂ concentrations in severely obese men or women. However, adipose tissue may contribute to concentrations of serum E₂-FAE.


Science Translational Medicine | 2016

USF1 deficiency activates brown adipose tissue and improves cardiometabolic health

Pirkka-Pekka Laurila; Jarkko Soronen; Sander Kooijman; Saara Forsström; Mariëtte R. Boon; Ida Surakka; Essi Kaiharju; Claudia P. Coomans; Sjoerd A. A. van den Berg; Anu Autio; Antti-Pekka Sarin; Johannes Kettunen; Emmi Tikkanen; Tuula Manninen; Jari Metso; Reija Silvennoinen; Krista Merikanto; Maija Ruuth; Julia Perttilä; Anne Mäkelä; Ayaka Isomi; Anita M. Tuomainen; Anna Tikka; Usama Abo Ramadan; Ilkka Seppälä; Terho Lehtimäki; Johan G. Eriksson; Aki S. Havulinna; Antti Jula; Pekka J. Karhunen

Deficiency of USF1 protects against obesity, insulin resistance, and cardiovascular disease in mice and humans, and induces brown adipose tissue to burn triglycerides and glucose. Boosting metabolism with low USF1 Laurila et al. show that a deficiency of the transcription factor USF1 protects against obesity, insulin resistance, and cardiovascular disease. Even when fed a high-fat diet, USF1-deficient mice stayed lean and maintained a beneficial lipid profile with low triglycerides and high high-density lipoprotein (HDL) cholesterol. The mice had elevated energy expenditure because their brown adipose tissue was more active. In USF1-deficient mice, glucose and lipids were rapidly cleared from the circulation to be burned by brown fat. In humans, individuals with reduced USF1 expression also had higher plasma HDL cholesterol and lower triglycerides, and were more insulin-sensitive and less prone to hardening of the arteries. These findings identify USF1 as a potential therapeutic target for treating metabolic and cardiac diseases. USF1 (upstream stimulatory factor 1) is a transcription factor associated with familial combined hyperlipidemia and coronary artery disease in humans. However, whether USF1 is beneficial or detrimental to cardiometabolic health has not been addressed. By inactivating USF1 in mice, we demonstrate protection against diet-induced dyslipidemia, obesity, insulin resistance, hepatic steatosis, and atherosclerosis. The favorable plasma lipid profile, including increased high-density lipoprotein cholesterol and decreased triglycerides, was coupled with increased energy expenditure due to activation of brown adipose tissue (BAT). Usf1 inactivation directs triglycerides from the circulation to BAT for combustion via a lipoprotein lipase–dependent mechanism, thus enhancing plasma triglyceride clearance. Mice lacking Usf1 displayed increased BAT-facilitated, diet-induced thermogenesis with up-regulation of mitochondrial respiratory chain complexes, as well as increased BAT activity even at thermoneutrality and after BAT sympathectomy. A direct effect of USF1 on BAT activation was demonstrated by an amplified adrenergic response in brown adipocytes after Usf1 silencing, and by augmented norepinephrine-induced thermogenesis in mice lacking Usf1. In humans, individuals carrying SNP (single-nucleotide polymorphism) alleles that reduced USF1 mRNA expression also displayed a beneficial cardiometabolic profile, featuring improved insulin sensitivity, a favorable lipid profile, and reduced atherosclerosis. Our findings identify a new molecular link between lipid metabolism and energy expenditure, and point to the potential of USF1 as a therapeutic target for cardiometabolic disease.


Physiological Reports | 2016

Novel hepatic microRNAs upregulated in human nonalcoholic fatty liver disease

Jarkko Soronen; Hannele Yki-Järvinen; You Zhou; Sanja Sädevirta; Antti-Pekka Sarin; Marja Leivonen; Ksenia Sevastianova; Julia Perttilä; Pirkka-Pekka Laurila; Alexander Sigruener; Gerd Schmitz; Vesa M. Olkkonen

MicroRNAs (miRNAs) control gene expression by reducing mRNA stability and translation. We aimed to identify alterations in human liver miRNA expression/function in nonalcoholic fatty liver disease (NAFLD). Subjects with the highest (median liver fat 30%, n = 15) and lowest (0%, n = 15) liver fat content were selected from >100 obese patients for miRNA profiling of liver biopsies on microarrays carrying probes for 1438 human miRNAs (a cross‐sectional study). Target mRNAs and pathways were predicted for the miRNAs most significantly upregulated in NAFLD, their cell‐type‐specific expression was investigated by quantitative PCR (qPCR), and the transcriptome of immortalized human hepatocytes (IHH) transfected with the miRNA with the highest number of predicted targets, miR‐576‐5p, was studied. The screen revealed 42 miRNAs up‐ and two downregulated in the NAFLD as compared to non‐NAFLD liver. The miRNAs differing most significantly between the groups, miR‐103a‐2*, miR‐106b, miR‐576‐5p, miRPlus‐I137*, miR‐892a, miR‐1282, miR‐3663‐5p, and miR‐3924, were all upregulated in NAFLD liver. Target pathways predicted for these miRNAs included ones involved in cancer, metabolic regulation, insulin signaling, and inflammation. Consistent transcriptome changes were observed in IHH transfected with miR‐576‐5p, and western analysis revealed a marked reduction of the RAC1 protein belonging to several miR‐576‐5p target pathways. To conclude, we identified 44 miRNAs differentially expressed in NAFLD versus non‐NAFLD liver, 42 of these being novel in the context of NAFLD. The study demonstrates that by applying a novel study set‐up and a broad‐coverage array platform one can reveal a wealth of previously undiscovered miRNA dysregulation in metabolic disease.

Collaboration


Dive into the Jarkko Soronen's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Christian Ehnholm

National Institute for Health and Welfare

View shared research outputs
Top Co-Authors

Avatar

Essi Kaiharju

National Institute for Health and Welfare

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jari Metso

National Institute for Health and Welfare

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anna Tikka

National Institute for Health and Welfare

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge