Network


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

Hotspot


Dive into the research topics where Tuomas T. Rissanen is active.

Publication


Featured researches published by Tuomas T. Rissanen.


Circulation Research | 2003

VEGF-D Is the Strongest Angiogenic and Lymphangiogenic Effector Among VEGFs Delivered Into Skeletal Muscle via Adenoviruses

Tuomas T. Rissanen; Johanna E. Markkanen; Marcin Gruchała; Tommi Heikura; Antti Puranen; Mikko I. Kettunen; Ivana Kholová; Risto A. Kauppinen; Marc G. Achen; Steven A. Stacker; Kari Alitalo; Seppo Ylä-Herttuala

Abstract— Optimal angiogenic and lymphangiogenic gene therapy requires knowledge of the best growth factors for each purpose. We studied the therapeutic potential of human vascular endothelial growth factor (VEGF) family members VEGF-A, VEGF-B, VEGF-C, and VEGF-D as well as a VEGFR-3–specific mutant (VEGF-C156S) using adenoviral gene transfer in rabbit hindlimb skeletal muscle. The significance of proteolytic processing of VEGF-D was explored using adenoviruses encoding either full-length or mature (&Dgr;N&Dgr;C) VEGF-D. Adenoviruses expressing potent VEGFR-2 ligands, VEGF-A and VEGF-D&Dgr;N&Dgr;C, induced the strongest angiogenesis and vascular permeability effects as assessed by capillary vessel and perfusion measurements, modified Miles assay, and MRI. The most significant feature of angiogenesis induced by both VEGF-A and VEGF-D&Dgr;N&Dgr;C was a remarkable enlargement of microvessels with efficient recruitment of pericytes suggesting formation of arterioles or venules. VEGF-A also moderately increased capillary density and created glomeruloid bodies, clusters of tortuous vessels, whereas VEGF-D&Dgr;N&Dgr;C–induced angiogenesis was more diffuse. Vascular smooth muscle cell proliferation occurred in regions with increased plasma protein extravasation, indicating that arteriogenesis may be promoted by VEGF-A and VEGF-D&Dgr;N&Dgr;C. Full-length VEGF-C and VEGF-D induced predominantly and the selective VEGFR-3 ligand VEGF-C156S exclusively lymphangiogenesis. Unlike angiogenesis, lymphangiogenesis was not dependent on nitric oxide. The VEGFR-1 ligand VEGF-B did not promote either angiogenesis or lymphangiogenesis. Finally, we found a positive correlation between capillary size and vascular permeability. This study compares, for the first time, angiogenesis and lymphangiogenesis induced by gene transfer of different human VEGFs, and shows that VEGF-D is the most potent member when delivered via an adenoviral vector into skeletal muscle.


Proceedings of the National Academy of Sciences of the United States of America | 2009

VEGF-B is dispensable for blood vessel growth but critical for their survival, and VEGF-B targeting inhibits pathological angiogenesis

Fan Zhang; Zhongshu Tang; Xu Hou; Johan Lennartsson; Yang Li; Alexander W. Koch; Pierre Scotney; Chunsik Lee; Pachiappan Arjunan; Lijin Dong; Anil Kumar; Tuomas T. Rissanen; Bin Wang; Nobuo Nagai; Pierre Fons; Robert N. Fariss; Yongqing Zhang; Eric F. Wawrousek; Ginger Tansey; James Raber; Guo-Hua Fong; Hao Ding; David A. Greenberg; Kevin G. Becker; Jean-Marc Herbert; Andrew D. Nash; Seppo Ylä-Herttuala; Yihai Cao; Ryan J. Watts; Xuri Li

VEGF-B, a homolog of VEGF discovered a long time ago, has not been considered an important target in antiangiogenic therapy. Instead, it has received little attention from the field. In this study, using different animal models and multiple types of vascular cells, we revealed that although VEGF-B is dispensable for blood vessel growth, it is critical for their survival. Importantly, the survival effect of VEGF-B is not only on vascular endothelial cells, but also on pericytes, smooth muscle cells, and vascular stem/progenitor cells. In vivo, VEGF-B targeting inhibited both choroidal and retinal neovascularization. Mechanistically, we found that the vascular survival effect of VEGF-B is achieved by regulating the expression of many vascular prosurvival genes via both NP-1 and VEGFR-1. Our work thus indicates that the function of VEGF-B in the vascular system is to act as a “survival,” rather than an “angiogenic” factor and that VEGF-B inhibition may offer new therapeutic opportunities to treat neovascular diseases.


Circulation | 2004

Adenoviral Catheter-Mediated Intramyocardial Gene Transfer Using the Mature Form of Vascular Endothelial Growth Factor-D Induces Transmural Angiogenesis in Porcine Heart

Juha Rutanen; Tuomas T. Rissanen; Johanna E. Markkanen; Marcin Gruchała; Päivi Silvennoinen; Antti Kivelä; Antti Hedman; Marja Hedman; Tommi Heikura; Maija-Riitta Ordén; Steven A. Stacker; Marc G. Achen; Juha Hartikainen; Seppo Ylä-Herttuala

Background—It is unclear what is the most efficient vector and growth factor for induction of therapeutic vascular growth in the heart. Furthermore, the histological nature of angiogenesis and potential side effects caused by different vascular endothelial growth factors (VEGFs) in myocardium have not been documented. Methods and Results—Adenoviruses (Ad) at 2 doses (2×1011 and 2×1012 viral particles) or naked plasmids (1 mg) encoding Lac Z control, VEGF-A165, or the mature, soluble form of VEGF-D (VEGF-D&Dgr;N&Dgr;C) were injected intramyocardially with the NOGA catheter system into domestic pigs. AdVEGF-D&Dgr;N&Dgr;C gene transfer (GT) induced a dose-dependent myocardial protein production, as measured by ELISA, resulting in an efficient angiogenic effect 6 days after the injections. Also, AdVEGF-A165 produced high gene transfer efficacy, as demonstrated with immunohistochemistry, leading to prominent angiogenesis effects. Despite the catheter-mediated approach, angiogenesis induced by both AdVEGFs was transmural, with maximal effects in the epicardium. Histologically, strongly enlarged &agr;-smooth muscle actin–positive microvessels involving abundant cell proliferation were found in the transduced regions, whereas microvessel density did not change. Myocardial contrast echocardiography and microspheres showed marked increases in perfusion in the transduced areas. VEGF-D&Dgr;N&Dgr;C but not matrix-bound VEGF-A165 was detected in plasma after adenoviral GT. A modified Miles assay demonstrated myocardial edema resulting in pericardial effusion with the higher AdVEGF doses. All effects returned to baseline by 3 weeks. Naked plasmid–mediated GT did not induce detectable protein production or vascular effects. Conclusions—Like AdVEGF-A165, AdVEGF-D&Dgr;N&Dgr;C GT using the NOGA system produces efficient transmural angiogenesis and increases myocardial perfusion. AdVEGF-D&Dgr;N&Dgr;C could be useful for the induction of therapeutic vascular growth in the heart.


American Journal of Pathology | 2002

Expression of vascular endothelial growth factor and vascular endothelial growth factor receptor-2 (KDR/Flk-1) in ischemic skeletal muscle and its regeneration.

Tuomas T. Rissanen; Ismo Vajanto; Mikko Hiltunen; Juha Rutanen; Mikko I. Kettunen; Mari Niemi; Pia Leppänen; Mikko P. Turunen; Johanna E. Markkanen; Katja Arve; Esko Alhava; Risto A. Kauppinen; Seppo Ylä-Herttuala

Vascular endothelial growth factor (VEGF) is a hypoxia-inducible endothelial cell mitogen and survival factor. Its receptor VEGFR-2 (KDR/Flk-1) mediates these effects. We studied the expression of VEGF and VEGFR-2 in ischemic human and rabbit skeletal muscle by immunohistochemistry and in situ hybridization. Human samples were obtained from eight lower limb amputations because of acute or chronic critical ischemia. In chronically ischemic human skeletal muscle VEGF and VEGFR-2 expression was restricted to atrophic and regenerating skeletal myocytes, whereas in acutely ischemic limbs VEGF and VEGFR-2 were expressed diffusely in the affected muscle. Hypoxia-inducible factor-1alpha was associated with VEGF and VEGFR-2 expression both in acute and chronic ischemia but not in regeneration. Hindlimb ischemia was induced in 20 New Zealand White rabbits by excising the femoral artery. Magnetic resonance imaging and histological sections revealed extensive ischemic damage in the thigh and leg muscles of ischemic rabbit hindlimbs with VEGF expression similar to acute human lower limb ischemia. After 1 and 3 weeks of ischemia VEGF expression was restricted to regenerating myotubes and by 6 weeks regeneration and expression of VEGF was diminished. VEGFR-2 expression was co-localized with VEGF expression in regenerating myotubes. Macrophages and an increased number of capillaries were associated with areas of ischemic muscle expressing VEGF and VEGFR-2. In conclusion, two patterns of VEGF and VEGFR-2 expression in human and rabbit ischemic skeletal muscle are demonstrated. In acute skeletal muscle ischemia VEGF and VEGFR-2 are expressed diffusely in the affected muscle. In chronic skeletal muscle ischemia and in skeletal muscle recovering from ischemia VEGF and VEGFR-2 expression are restricted to atrophic and regenerating muscle cells suggesting the operation of an autocrine pathway that may promote survival and regeneration of myocytes.


Circulation | 2009

Vascular Endothelial Growth Factor-B Induces Myocardium-Specific Angiogenesis and Arteriogenesis via Vascular Endothelial Growth Factor Receptor-1– and Neuropilin Receptor-1–Dependent Mechanisms

Johanna Lähteenvuo; Markku Lähteenvuo; Antti Kivelä; Carolina Rosenlew; Annelie Falkevall; Joakim Klar; Tommi Heikura; Tuomas T. Rissanen; Elisa Vähäkangas; Petra Korpisalo; Berndt Enholm; Peter Carmeliet; Kari Alitalo; Ulf J. Eriksson; Seppo Ylä-Herttuala

Background— New revascularization therapies are urgently needed for patients with severe coronary heart disease who lack conventional treatment options. Methods and Results— We describe a new proangiogenic approach for these no-option patients using adenoviral (Ad) intramyocardial vascular endothelial growth factor (VEGF)-B186 gene transfer, which induces myocardium-specific angiogenesis and arteriogenesis in pigs and rabbits. After acute infarction, AdVEGF-B186 increased blood vessel area, perfusion, ejection fraction, and collateral artery formation and induced changes toward an ischemia-resistant myocardial phenotype. Soluble VEGF receptor-1 and soluble neuropilin receptor-1 reduced the effects of AdVEGF-B186, whereas neither soluble VEGF receptor-2 nor inhibition of nitric oxide production had this result. The effects of AdVEGF-B186 involved activation of neuropilin receptor-1, which is highly expressed in the myocardium, via recruitment of G-protein-&agr; interacting protein, terminus C (GIPC) and upregulation of G-protein-&agr; interacting protein. AdVEGF-B186 also induced an antiapoptotic gene expression profile in cardiomyocytes and had metabolic effects by inducing expression of fatty acid transport protein-4 and lipid and glycogen accumulation in the myocardium. Conclusions— VEGF-B186 displayed strikingly distinct effects compared with other VEGFs. These effects may be mediated at least in part via a G-protein signaling pathway. Tissue-specificity, high efficiency in ischemic myocardium, and induction of arteriogenesis and antiapoptotic and metabolic effects make AdVEGF-B186 a promising candidate for the treatment of myocardial ischemia.


The FASEB Journal | 2002

Fibroblast growth factor 4 induces vascular permeability, angiogenesis and arteriogenesis in a rabbit hindlimb ischemia model.

Tuomas T. Rissanen; Johanna E. Markkanen; Katja Arve; Juha Rutanen; Mikko I. Kettunen; Ismo Vajanto; Suvi Jauhiainen; Linda Cashion; Marcin Gruchała; Outi Närvänen; Pekka Taipale; Risto A. Kauppinen; Gabor M. Rubanyi; Seppo Ylä-Herttuala

Previous studies have shown that fibroblast growth factor (FGF)‐1, FGF‐2, and FGF‐5 induce therapeutic angiogenesis. Here, we investigated the potential of FGF‐4 for therapeutic neovascularization in comparison to vascular endothelial growth factor (VEGF), using adenoviral gene transfer in a novel rabbit hind limb ischemia model, with ischemia restricted to the calf. Magnetic resonance imaging and a modified Miles assay showed that both AdFGF‐4 and AdVEGF given intramuscularly (i.m.) resulted in increases in vascular permeability and edema in transduced muscles 6 days after the gene transfer. In contrast, recombinant FGF‐4 protein injected in the rabbit skin did not induce acute vascular permeability. Injections (i.m.) of AdFGF‐4 and AdVEGF, but not intra‐arterially administered AdVEGF, increased collateral growth, popliteal blood flow, and muscle perfusion compared with controls. The angiogenesis response consisted mainly of the enlargement of pre‐existing vessels rather than an increase in capillary density. Adenoviral FGF‐4 overexpression up‐regulated endogenous VEGF, which may explain many of the effects thought to be specific for VEGF such as the increase in vascular permeability. This study demonstrates for the first time that FGF‐4 induces vascular permeability, therapeutic angiogenesis, and arteriogenesis comparable to that of VEGF and could be useful for the treatment of peripheral vascular disease.


Circulation | 2000

Intravascular Adenovirus-Mediated VEGF-C Gene Transfer Reduces Neointima Formation in Balloon-Denuded Rabbit Aorta

Mikko Hiltunen; Marja Laitinen; Mikko P. Turunen; Michael Jeltsch; Juha Hartikainen; Tuomas T. Rissanen; Johanna Laukkanen; Mari Niemi; Maija Kossila; Tomi Häkkinen; Antti Kivelä; Berndt Enholm; Hannu Mansukoski; Anna-Mari Turunen; Kari Alitalo; Seppo Yla-Herttuala

BackgroundGene transfer to the vessel wall may provide new possibilities for the treatment of vascular disorders, such as postangioplasty restenosis. In this study, we analyzed the effects of adenovirus-mediated vascular endothelial growth factor (VEGF)-C gene transfer on neointima formation after endothelial denudation in rabbits. For comparison, a second group was treated with VEGF-A adenovirus and a third group with lacZ adenovirus. Clinical-grade adenoviruses were used for the study. Methods and ResultsAortas of cholesterol-fed New Zealand White rabbits were balloon-denuded, and gene transfer was performed 3 days later. Animals were euthanized 2 and 4 weeks after the gene transfer, and intima/media ratio (I/M), histology, and cell proliferation were analyzed. Two weeks after the gene transfer, I/M in the lacZ-transfected control group was 0.57±0.04. VEGF-C gene transfer reduced I/M to 0.38±0.02 (P <0.05 versus lacZ group). I/M in VEGF-A–treated animals was 0.49±0.17 (P =NS). The tendency that both VEGF groups had smaller I/M persisted at the 4-week time point, when the lacZ group had an I/M of 0.73±0.16, the VEGF-C group 0.44±0.14, and the VEGF-A group 0.63±0.21 (P =NS). Expression of VEGF receptors 1, 2, and 3 was detected in the vessel wall by immunocytochemistry and in situ hybridization. As an additional control, the effect of adenovirus on cell proliferation was analyzed by performing gene transfer to intact aorta without endothelial denudation. No differences were seen in smooth muscle cell proliferation or I/M between lacZ adenovirus and 0.9% saline–treated animals. ConclusionsAdenovirus-mediated VEGF-C gene transfer may be useful for the treatment of postangioplasty restenosis and vessel wall thickening after vascular manipulations.


Journal of Gene Medicine | 2002

Evaluation of angiogenesis and side effects in ischemic rabbit hindlimbs after intramuscular injection of adenoviral vectors encoding VEGF and LacZ

Ismo Vajanto; Tuomas T. Rissanen; Juha Rutanen; Mikko Hiltunen; Tiina T. Tuomisto; Katja Arve; Outi Närvänen; Hannu Manninen; Heikki T. Räsänen; Mikko Hippeläinen; Esko Alhava; Seppo Ylä-Herttuala

Recent studies have suggested the therapeutic potential of vascular endothelial growth factor (VEGF) gene therapy in ischemic skeletal muscle. However, only limited information is available about the effects of VEGF gene therapy in different regions of ischemic limbs, effects of control adenoviruses, and biodistribution of the transgenes after intramuscular (i.m.) administration. Here we studied angiogenesis and side effects of adenovirus‐mediated VEGF and β‐galactosidase (LacZ) gene transfers in ischemic rabbit hindlimbs.


Circulation | 2005

Blood Flow Remodels Growing Vasculature During Vascular Endothelial Growth Factor Gene Therapy and Determines Between Capillary Arterialization and Sprouting Angiogenesis

Tuomas T. Rissanen; Petra Korpisalo; Johanna E. Markkanen; Timo Liimatainen; Maija-Riitta Ordén; Ivana Kholová; Anna de Goede; Tommi Heikura; Olli Gröhn; Seppo Ylä-Herttuala

Background— For clinically relevant proangiogenic therapy, it would be essential that the growth of the whole vascular tree is promoted. Vascular endothelial growth factor (VEGF) is well known to induce angiogenesis, but its capability to promote growth of larger vessels is controversial. We hypothesized that blood flow remodels vascular growth during VEGF gene therapy and may contribute to the growth of large vessels. Methods and Results— Adenoviral (Ad) VEGF or LacZ control gene transfer was performed in rabbit hindlimb semimembranous muscles with or without ligation of the profound femoral artery (PFA). Contrast-enhanced ultrasound and dynamic susceptibility contrast MRI demonstrated dramatic 23- to 27-fold increases in perfusion index and a strong decrease in peripheral resistance 6 days after AdVEGF gene transfer in normal muscles. Enlargement by 20-fold, increased pericyte coverage, and decreased alkaline phosphatase and dipeptidyl peptidase IV activities suggested the transformation of capillaries toward an arterial phenotype. Increase in muscle perfusion was attenuated, and blood vessel growth was more variable, showing more sprouting angiogenesis and formation of blood lacunae after AdVEGF gene transfer in muscles with ligated PFA than in normal muscles. Three-dimensional ultrasound reconstructions and histology showed that the whole vascular tree, including large arteries and veins, was enlarged manifold by AdVEGF. Blood flow was normalized and enlarged collaterals persisted in operated limbs 14 days after AdVEGF treatment. Conclusions— This study shows that (1) blood flow modulates vessel growth during VEGF gene therapy and (2) VEGF overexpression promotes growth of arteries and veins and induces capillary arterialization leading to supraphysiological blood flow in target muscles.


The FASEB Journal | 2000

Biodistribution of adenoviral vector to nontarget tissues after local in vivo gene transfer to arterial wall using intravascular and periadventitial gene delivery methods

Mikko Hiltunen; Mikko P. Turunen; Anna-Mari Turunen; Tuomas T. Rissanen; Marja Laitinen; Veli-Matti Kosma; Seppo Ylä-Herttuala

Expression of transgene other than in the target tissue may cause side effects and safety problems in gene therapy. We analyzed biodistribution of transgene expression after intravascular and periadventitial gene delivery methods using the first generation nuclear‐targeted lacZ adenovirus. RT‐PCR and X‐Gal stainings were used to study transgene expression 14 days after the gene transfer. After intravascular catheter‐mediated gene transfer to rabbit aorta mimicking angioplasty procedure, the target vessel showed 1.1% ± 0.5 gene transfer efficiency. Other tissues showed varying lacZ gene expression indicating a systemic leakage of the vector with the highest transfection efficiency in hepatocytes (0.7% ± 0.5). X‐Gal staining of blood cells 24 h after the intravascular gene transfer indicated that a significant portion (1.8% ± 0.8) of circulating monocytes was transfected. X‐Gal‐positive cells were also found in testis. After periadventitial gene transfer using a closed silicon capsule placed around the artery, 0.1% ± 0.1 lacZ‐positive cells were detected in the artery wall. Positive cells were also found in the liver and testis (<0.01%), indicating that the virus escapes even from the periadventitial space, although less extensively than during the intravascular application. We conclude that catheter‐mediated intravascular and, to a lesser extent, periadventitial gene transfer lead to leakage of adenovirus to systemic circulation, followed by expression of the transgene in several tissues. Possible consequences of the ectopic expression of the transgene should be evaluated in gene therapy trials even if local gene delivery methods are used.—Hiltunen, M. O., Turunen, M. P., Turunen, A.‐M., Rissanen, T. T., Laitinen, M., Kosma, V.‐M., Ylä‐Herttuala, S. Biodistribution of adenoviral vector to nontarget tissues after local in vivo gene transfer to arterial wall using intravascular and periadventitial gene delivery methods. FASEB J. 14, 2230–2236 (2000)

Collaboration


Dive into the Tuomas T. Rissanen's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Juha Rutanen

University of Eastern Finland

View shared research outputs
Top Co-Authors

Avatar

Petra Korpisalo

University of Eastern Finland

View shared research outputs
Top Co-Authors

Avatar

Johanna E. Markkanen

University of Eastern Finland

View shared research outputs
Top Co-Authors

Avatar

Mikko Hiltunen

University of Eastern Finland

View shared research outputs
Top Co-Authors

Avatar

Ismo Vajanto

University of Eastern Finland

View shared research outputs
Top Co-Authors

Avatar

Tommi Heikura

University of Eastern Finland

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Henna Karvinen

University of Eastern Finland

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge