Network


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

Hotspot


Dive into the research topics where Raimo K. Tuominen is active.

Publication


Featured researches published by Raimo K. Tuominen.


Nature | 2007

Novel neurotrophic factor CDNF protects and rescues midbrain dopamine neurons in vivo

Päivi Lindholm; Merja H. Voutilainen; Juha Laurén; Johan Peränen; Veli-Matti Leppänen; Jaan-Olle Andressoo; Maria Lindahl; Sanna Janhunen; Nisse Kalkkinen; Tõnis Timmusk; Raimo K. Tuominen; Mart Saarma

In Parkinson’s disease, brain dopamine neurons degenerate most prominently in the substantia nigra. Neurotrophic factors promote survival, differentiation and maintenance of neurons in developing and adult vertebrate nervous system. The most potent neurotrophic factor for dopamine neurons described so far is the glial-cell-line-derived neurotrophic factor (GDNF). Here we have identified a conserved dopamine neurotrophic factor (CDNF) as a trophic factor for dopamine neurons. CDNF, together with its previously described vertebrate and invertebrate homologue the mesencephalic-astrocyte-derived neurotrophic factor, is a secreted protein with eight conserved cysteine residues, predicting a unique protein fold and defining a new, evolutionarily conserved protein family. CDNF (Armetl1) is expressed in several tissues of mouse and human, including the mouse embryonic and postnatal brain. In vivo, CDNF prevented the 6-hydroxydopamine (6-OHDA)-induced degeneration of dopaminergic neurons in a rat experimental model of Parkinson’s disease. A single injection of CDNF before 6-OHDA delivery into the striatum significantly reduced amphetamine-induced ipsilateral turning behaviour and almost completely rescued dopaminergic tyrosine-hydroxylase-positive cells in the substantia nigra. When administered four weeks after 6-OHDA, intrastriatal injection of CDNF was able to restore the dopaminergic function and prevent the degeneration of dopaminergic neurons in substantia nigra. Thus, CDNF was at least as efficient as GDNF in both experimental settings. Our results suggest that CDNF might be beneficial for the treatment of Parkinson’s disease.


Neuron | 1999

Retarded Growth and Deficits in the Enteric and Parasympathetic Nervous System in Mice Lacking GFRα2, a Functional Neurturin Receptor

Jari Rossi; Keijo Luukko; Dmitry Poteryaev; Antti Laurikainen; Yun Fu Sun; Tiina Laakso; Simo Eerikäinen; Raimo K. Tuominen; Merja Lakso; Heikki Rauvala; Urmas Arumäe; Michael Pasternack; Mart Saarma; Matti S. Airaksinen

Glial cell line-derived neurotrophic factor (GDNF) and a related protein, neurturin (NTN), require a GPI-linked coreceptor, either GFR alpha1 or GFR alpha2, for signaling via the transmembrane Ret tyrosine kinase. We show that mice lacking functional GFR alpha2 coreceptor (Gfra2-/-) are viable and fertile but have dry eyes and grow poorly after weaning, presumably due to malnutrition. While the sympathetic innervation appeared normal, the parasympathetic cholinergic innervation was almost absent in the lacrimal and salivary glands and severely reduced in the small bowel. Neurite outgrowth and trophic effects of NTN at low concentrations were lacking in Gfra2-/- trigeminal neurons in vitro, whereas responses to GDNF were similar between the genotypes. Thus, GFR alpha2 is a physiological NTN receptor, essential for the development of specific postganglionic parasympathetic neurons.


The Journal of Neuroscience | 2009

Mesencephalic Astrocyte-Derived Neurotrophic Factor Is Neurorestorative in Rat Model of Parkinson's Disease

Merja H. Voutilainen; Susanne Bäck; Eeva Pörsti; Liisa Toppinen; Lauri Lindgren; Päivi Lindholm; Johan Peränen; Mart Saarma; Raimo K. Tuominen

Neurotrophic factors are promising candidates for the treatment of Parkinsons disease (PD). Mesencephalic astrocyte-derived neurotrophic factor (MANF) belongs to a novel evolutionarily conserved family of neurotrophic factors. We examined whether MANF has neuroprotective and neurorestorative effect in an experimental model of PD in rats. We also studied the distribution and transportation of intrastriatally injected MANF in the brain and compared it with glial cell line-derived neurotrophic factor (GDNF). Unilateral lesion of nigrostriatal dopaminergic system was induced by intrastriatal injection of 6-hydroxydopamine (6-OHDA). Amphetamine-induced turning behavior was monitored up to 12 weeks after the unilateral lesion. The local diffusion at the injection site and transportation profiles of intrastriatally injected MANF and GDNF were studied by immunohistochemical detection of the unlabeled growth factors as well as by autoradiographic and gamma counting detection of 125I-labeled trophic factors. Intrastriatally injected MANF protected nigrostriatal dopaminergic nerves from 6-OHDA-induced degeneration as evaluated by counting tyrosine hydroxylase (TH)-positive cell bodies in the substantia nigra (SN) and TH-positive fibers in the striatum. More importantly, MANF also restored the function of the nigrostriatal dopaminergic system when administered either 6 h before or 4 weeks after 6-OHDA administration in the striatum. MANF was distributed throughout the striatum more readily than GDNF. The mechanism of MANF action differs from that of GDNF because intrastriatally injected 125I-MANF was transported to the frontal cortex, whereas 125I-GDNF was transported to the SN. Our results suggest that MANF is readily distributed throughout the striatum and has significant therapeutic potential for the treatment of PD.


Toxicological Sciences | 1989

Target tissue morphology and serum biochemistry following 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) exposure in a TCDD-susceptible and a TCDD-resistant rat strain.

Raimo Pohjanvirta; Tauno Kulju; Antonius F. W. Morselt; Raimo K. Tuominen; Risto O. Juvonen; Pekka T. Männistö; Yrjö Collan; Eeva-Liisa Sainio; Jouko Tuomisto

The mode of action of the highly toxic environmental contaminant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is unknown. It was recently discovered that two strains of rat, Long-Evans (L-E) and Han/Wistar (H/W), differ widely in susceptibility to TCDD. Employing this strain divergence as a probe, the present study set out to assess the role of various biochemical and morphological effects in TCDD lethality. In the main experiment, the rats were treated once ip with 0,5,50, or (H/W) 500 micrograms/kg TCDD and killed 1 to 16 days postexposure. Several target organs were evaluated by light microscopy and a number of serum lipid and carbohydrate parameters as well as a few major regulatory hormones were analyzed. The results demonstrated that most alterations caused by TCDD were essentially similar in both strains. TCDD reduced circulating thyroxine to a slightly greater extent and more permanently in the sensitive L-E strain. Moreover, a highly significant interaction on thyroid-stimulating hormone was found among strain, dose, and time. Serum concentrations of corticosterone and free fatty acids were increased only in the L-E rats given 50 micrograms/kg TCDD, i.e., at an apparent LD100 dose level for this strain. Yet, the most striking interstrain difference was seen in the liver which was distinctly affected after Day 4 in L-E rats given 50 micrograms/kg TCDD but only marginally affected in rats from any H/W group. The lesion, while showing no necrotic cell changes, was suggestive of plasma membrane damage, possibly reflecting the production of free radicals. The relation of the findings to possible mechanisms of TCDD action is discussed.


British Journal of Pharmacology | 1992

Different in vivo properties of three new inhibitors of catechol O‐methyltransferase in the rat

Pekka T. Männistö; Päivi Tuomainen; Raimo K. Tuominen

1 We compared three new catechol O‐methyltransferase (COMT) inhibitors (OR‐611, Ro 40–7592 and CGP 28014; 10 and 30 mg kg−1, i.p.) in male rats given levodopa (l‐DOPA, 50 mg kg−1, i.p.) and carbidopa ((−)‐l‐α‐methyl dopa, 50 mg kg−1, i.p.). In some studies pretreatment with pargyline (80 mg kg−1, i.p.) was used to block the function of monoamine oxidase (MAO). 2 Decreases of hypothalamic and striatal 3‐O‐methyl‐dopa (3‐OMD) levels were used as measures of the inhibition of peripheral COMT. The inhibition of brain COMT activity was estimated by decreases of hypothalamic and striatal homovanillic acid (HVA) and 3‐methoxytyramine (3‐MT; after pargyline) levels. 3 The three COMT inhibitors studied had different individual characteristics. OR‐611 was primarily a peripherally acting COMT inhibitor, decreasing 3‐OMD levels in the striatum (to 31–52%) and in the hypothalamus (to 16–27%) both in the control and pargyline‐treated animals at 1 and 3 h. It did not have any effect on brain HVA and 3‐MT. 3 Ro 40–7592 was a broad spectrum COMT inhibitor decreasing striatal and hypothalamic 3‐OMD (always to < 30%), HVA (to < 50%) and 3‐MT levels (to < 23%) significantly both at 1 and 3 h. It was more potent than OR‐611. 4 CGP 28014 functioned as a weak COMT inhibitor in the periphery inhibiting 3‐OMD formation only at 3 h. In contrast, it was fairly potent in decreasing the brain HVA and 3‐MT levels at 1 h (to 37–22% and 42–35% in the striatum, and to 57–33% and 64–35% in the hypothalamus, respectively) but not at 3 h. Since CGP 28014, unlike OR‐611 and Ro 40–7592, did not generally increase the brain DOPA, dopamine or DOPAC levels, it was not a typical COMT inhibitor.


Experimental Neurology | 2011

Chronic infusion of CDNF prevents 6-OHDA-induced deficits in a rat model of Parkinson's disease.

Merja H. Voutilainen; Susanne Bäck; Johan Peränen; Päivi Lindholm; Atso Raasmaja; Pekka T. Männistö; Mart Saarma; Raimo K. Tuominen

Cerebral dopamine neurotrophic factor (CDNF) and mesencephalic astrocyte-derived neurotrophic factor (MANF) constitute a novel, evolutionarily conserved family of neurotrophic factors (NTF) expressed in vertebrates and invertebrates. The effects of two-week infusions of CDNF, MANF and glial cell line-derived neurotrophic factor (GDNF) were studied in a rat 6-hydroxydopamine (6-OHDA) hemiparkinsonian model. Degeneration of nigrostriatal dopamine nerve tract after toxin injection was assessed by measuring amphetamine-induced rotational behavior, and at the end of the experiment by quantifying tyrosine hydroxylase (TH)-positive neurons in the substantia nigra pars compacta (SNpc) and TH-positive fibers in the striatum. The diffusion of the NTFs into the brain tissue following chronic infusion was also studied. Finally, we examined the transportation of intrastriatally injected (125)I-CDNF within the brain. The amphetamine-induced rotational behavior was gradually normalized in rats treated with CDNF for two weeks following the intrastriatal 6-OHDA injection. CDNF was also able to inhibit 6-OHDA-induced loss of TH-immunoreactive cells of the SNpc and TH-positive fibers in the striatum. MANF and GDNF had no statistically significant effect in any of the above measures. The volume of distribution for MANF in the striatum was significantly larger than that of GDNF after 3-day infusions. Both (125)I-CDNF and (125)I-GDNF were retrogradely transported from the striatum to the SN. No behavioral signs of toxicity were observed during treatment with the three NTFs. These results imply that CDNF may have potential as a neuroprotective or even neurorestorative therapy of PD.


Cell Transplantation | 2012

CDNF Protects the Nigrostriatal Dopamine System and Promotes Recovery after MPTP Treatment in Mice

Mikko Airavaara; Brandon K. Harvey; Merja H. Voutilainen; Hui Shen; Jenny Chou; Päivi Lindholm; Maria Lindahl; Raimo K. Tuominen; Mart Saarma; Barry J. Hoffer; Yun Wang

Cerebral dopamine neurotrophic factor (CDNF) is a recently discovered protein, which belongs to the evolutionarily conserved CDNF/MANF family of neurotrophic factors. The degeneration of dopamine neurons following 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) treatment is well characterized, and efficacy in this model is considered a standard criterion for development of parkinsonian therapies. MPTP is a neurotoxin, which produces parkinsonian symptoms in humans and in C57/Bl6 mice. To date, there are no reports about the effects of CDNF on dopamine neuron survival or function in the MPTP rodent model, a critical gap. Therefore, we studied whether CDNF has neuroprotective and neurorestorative properties for the nigrostriatal dopamine system after MPTP injections in C57/Bl6 mice. We found that bilateral striatal CDNF injections, given 20 h before MPTP, improved horizontal and vertical motor behavior. CDNF pretreatment increased tyrosine hydroxylase (TH) immunoreactivity in the striatum and in the substantia nigra pars reticulata (SNpr), as well as the number of TH-positive cells in substantia nigra pars compacta (SNpc). Posttreatment with CDNF, given 1 week after MPTP injections, increased horizontal and vertical motor behavior of mice, as well as dopamine fiber densities in the striatum and the number of TH-positive cells in SNpc. CDNF did not alter any of the analyzed dopaminergic biomarkers or locomotor behavior in MPTP-untreated animals. We conclude that striatal CDNF administration is both neuroprotective and neurorestorative for the TH-positive cells in the nigrostriatal dopamine system in the MPTP model, which supports the development of CDNF-based treatment for Parkinsons disease.


British Journal of Pharmacology | 2006

Inhibition of classical PKC isoenzymes downregulates STAT1 activation and iNOS expression in LPS-treated murine J774 macrophages

Tiina Salonen; Outi Sareila; Ulla Jalonen; Hannu Kankaanranta; Raimo K. Tuominen; Eeva Moilanen

Proinflammatory cytokines and bacterial products trigger inducible nitric oxide synthase (iNOS) expression and nitric oxide (NO) production in inflammatory and tissue cells. In inflammation, NO acts as an important mediator having both proinflammatory and destructive effects. Protein kinase C (PKC) is a family of serine–threonine protein kinase isoenzymes involved in signal transduction pathways related to inflammatory responses. The aim of the present study was to investigate the role of classical PKC (cPKC) isoenzymes in the regulation of iNOS expression and NO production in murine J774 macrophages and the mechanisms involved. RO318220 (inhibits PKCβ, PKCγ and PKCɛ), GÖ6976 (inhibits cPKC isoenzymes PKCα and PKCβ) and LY333531 (inhibits PKCβ) reduced lipopolysaccharide (LPS)‐induced NO production and iNOS expression in a dose‐dependent manner as did 6 h pretreatment with 1 μM phorbol 12‐myristate 13‐acetate (PMA) (which was shown to downregulate PKC expression). PKC inhibitors also reduced LPS‐induced iNOS mRNA levels, but they did not affect the half‐life of iNOS mRNA. PKC inhibitors did not alter LPS‐induced activation of NF‐κB as measured by electrophoretic mobility shift assay. All PKC inhibitors used and pretreatment with 1 μM PMA inhibited signal transducer and activator of transcription 1 (STAT1) activation as measured by the translocation of STAT1α from the cytosol to the nucleus by Western blot. In addition, inhibition of STAT1 activation by AG‐490, an inhibitor of JAK‐2, also reduced NO production. These results suggest that cPKC isoenzymes, especially PKCβ, mediate the upregulation of iNOS expression and NO production in activated macrophages in an NF‐κB‐independent manner, possibly through the activation of transcription factor STAT1.


Life Sciences | 1988

Catechol-O-methyltransferase activity in human and rat small intestine

Erkki Nissinen; Raimo K. Tuominen; Vesa Perhoniemi; Seppo Kaakkola

The activities of soluble (S-COMT) and membrane-bound (MB-COMT) forms of catechol-O-methyltransferase in different regions of human and rat small intestine were determined. S-COMT was the predominant form both in the mucous membrane and in the muscle layers. The activity of S-COMT was more than twice as high in the mucous membrane as in the muscle layer. The activity of MB-COMT was almost equal in the different regions of the gut as well as in the mucous membrane and muscle layers. However, the ratio of the soluble form to the membrane-bound form was close to one in the human muscle layers of the jejunum and ileum.


Brain and behavior | 2013

Gene therapy with AAV2-CDNF provides functional benefits in a rat model of Parkinson's disease

Susanne Bäck; Johan Peränen; Emilia Galli; Päivi Pulkkila; Liina Lonka-Nevalaita; Tuulia Tamminen; Merja H. Voutilainen; Atso Raasmaja; Mart Saarma; Pekka T. Männistö; Raimo K. Tuominen

Cerebral dopamine neurotrophic factor (CDNF) protein has been shown to protect the nigrostriatal dopaminergic pathway when given as intrastriatal infusions in rat and mouse models of Parkinsons disease (PD). In this study, we assessed the neuroprotective effect of CDNF delivered with a recombinant adeno‐associated viral (AAV) serotype 2 vector in a rat 6‐hydroxydopamine (6‐OHDA) model of PD. AAV2 vectors encoding CDNF, glial cell line–derived neurotrophic factor (GDNF), or green fluorescent protein were injected into the rat striatum. Protein expression analysis showed that our AAV2 vector efficiently delivered the neurotrophic factor genes into the brain and gave rise to a long‐lasting expression of the proteins. Two weeks after AAV2 vector injection, 6‐OHDA was injected into the rat striatum, creating a progressive degeneration of the nigrostriatal dopaminergic system. Treatment with AAV2‐CDNF resulted in a marked decrease in amphetamine‐induced ipsilateral rotations while it provided only partial protection of tyrosine hydroxylase (TH)‐immunoreactive cells in the rat substantia nigra pars compacta and TH‐reactive fibers in the striatum. Results from this study provide additional evidence that CDNF can be considered a potential treatment of Parkinsons disease.

Collaboration


Dive into the Raimo K. Tuominen's collaboration.

Researchain Logo
Decentralizing Knowledge