Network


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

Hotspot


Dive into the research topics where Gundars Goldsteins is active.

Publication


Featured researches published by Gundars Goldsteins.


Proceedings of the National Academy of Sciences of the United States of America | 2002

β-Amyloid precursor protein transgenic mice that harbor diffuse Aβ deposits but do not form plaques show increased ischemic vulnerability: Role of inflammation

Milla Koistinaho; Mikko I. Kettunen; Gundars Goldsteins; Riitta Keinänen; Antero Salminen; Michael Ort; J. Bureš; David R. Liu; Risto A. Kauppinen; Linda S. Higgins; Jari Koistinaho

β-amyloid (Aβ), derived form the β-amyloid precursor protein (APP), is important for the pathogenesis of Alzheimers disease (AD), which is characterized by progressive decline of cognitive functions, formation of Aβ plaques and neurofibrillary tangles, and loss of neurons. However, introducing a human wild-type or mutant APP gene to rodent models of AD does not result in clear neurodegeneration, suggesting that contributory factors lowering the threshold of neuronal death may be present in AD. Because brain ischemia has recently been recognized to contribute to the pathogenesis of AD, we studied the effect of focal brain ischemia in 8- and 20-month-old mice overexpressing the 751-amino acid isoform of human APP. We found that APP751 mice have higher activity of p38 mitogen-activated protein kinase (p38 MAPK) in microglia, the main immune effector cells within the brain, and increased vulnerability to brain ischemia when compared with age-matched wild-type mice. These characteristics are associated with enhanced microglial activation and inflammation but not with altered regulation of cerebral blood flow, as assessed by MRI and laser Doppler flowmetry. Suppression of inflammation with aspirin or inhibition of p38 MAPK with a selective inhibitor, SD-282, abolishes the increased neuronal vulnerability in APP751 transgenic mice. SD-282 also suppresses the expression of inducible nitric-oxide synthase and the binding activity of activator protein 1. These findings elucidate molecular mechanisms of neuronal injury in AD and suggest that antiinflammatory compounds preventing activation of p38 MAPK in microglia may reduce neuronal vulnerability in AD.


Electrophoresis | 2002

Proteomic analysis of protein oxidation in Alzheimer's disease brain

Minna A. Korolainen; Gundars Goldsteins; Irina Alafuzoff; Jari Koistinaho; Tuula Pirttilä

There is a growing body of evidence that oxidative stress plays a major role in Alzheimers disease (AD) pathogenesis. Identification of oxidatively altered proteins in AD is important for understanding the relationship between protein oxidation, protein aggregation and neurodegeneration. In this communication, we report a method that can be applied to study oxidative changes of individual proteins in brain. In order to analyze protein oxidation by detection of protein‐bound carbonyls, cytosolic protein extracts were derivatized with 2,4‐dinitrophenylhydrazine (DNPH) and then separated by two‐dimensional (2‐D) gel electrophoresis. After electrotransfer to polyvinylidene difluoride (PVDF) membranes, proteins were first stained with Sypro Ruby protein stain, and then the oxidized proteins were detected with anti‐dinitrophenyl (DNP) antibody. About 150 proteins and more than 100 oxidized proteins were detected and quantified in both AD and control cases by 2‐D image analysis. The amount of protein‐bound carbonyls was decreased for six and increased for one protein in AD. The amount of protein was increased for three proteins in AD. Furthermore, the degree of oxidation was calculated as the ratio of protein‐bound carbonyls to the total amount of an individual protein. Two proteins showed a significant decrease in the degree of oxidation in AD. Our results suggest that the balance of protein oxidation and degradation is altered in AD.


Molecular and Cellular Neuroscience | 2008

Nuclear factor erythroid 2-related factor 2 protects against beta amyloid

Katja M. Kanninen; Tarja Malm; Henna-Kaisa Jyrkkänen; Gundars Goldsteins; Velta Keksa-Goldsteine; Heikki Tanila; Masayuki Yamamoto; Seppo Ylä-Herttuala; Anna-Liisa Levonen; Jari Koistinaho

Nuclear factor erythroid 2-related factor 2 (Nrf2) coordinates the up-regulation of cytoprotective genes via the antioxidant response element (ARE). In the pathogenesis of Alzheimers disease (AD) current evidence supports the role of oxidative stress. Considering the protective role of Nrf2 against oxidative injury, we studied Nrf2 and Nrf2-ARE target genes in transgenic AD mice and tested whether Nrf2 could confer neuroprotection against amyloid-beta peptides (Abeta). Nrf2-ARE pathway was attenuated in APP/PS1 transgenic mouse brain at the time of Abeta deposition. Boosting the activity of the Nrf2-ARE pathway by tert-butylhydroquinone treatment or adenoviral Nrf2 gene transfer protected against Abeta toxicity. This neuroprotection was associated with increased expression of Nrf2 target genes and reduced phosphorylation of p66Shc, a marker of increased susceptibility for oxidative stress. The findings suggest that the Nrf2-ARE pathway may be impaired in AD and that induction of the Nrf2-ARE defence mechanism may prevent or delay AD-like pathology.


The Journal of Neuroscience | 2007

Pyrrolidine Dithiocarbamate Activates Akt and Improves Spatial Learning in APP/PS1 Mice without Affecting β-Amyloid Burden

Tarja Malm; Henna Iivonen; Gundars Goldsteins; Velta Keksa-Goldsteine; Toni Ahtoniemi; Katja M. Kanninen; Antero Salminen; Seppo Auriola; Thomas van Groen; Heikki Tanila; Jari Koistinaho

Pyrrolidine dithiocarbamate (PDTC) is a clinically tolerated inhibitor of nuclear factor-κB (NF-κB), antioxidant and antiinflammatory agent, which provides protection in brain ischemia models. In neonatal hypoxia–ischemia model, PDTC activates Akt and reduces activation of glycogen synthase kinase 3β (GSK-3β). Because chronic inflammation, oxidative stress, and increased GSK-3β activity are features of Alzheimers disease (AD) pathology, we tested whether PDTC reduces brain pathology and improves cognitive function in a transgenic animal model of AD. A 7 month oral treatment with PDTC prevented the decline in cognition in AD mice without altering β-amyloid burden or gliosis. Moreover, marked oxidative stress and activation of NF-κB were not part of the brain pathology. Instead, the phosphorylated form of GSK-3β was decreased in the AD mouse brain, and PDTC treatment increased the phosphorylation of Akt and GSK-3β. Also, PDTC treatment increased the copper concentration in the brain. In addition, PDTC rescued cultured hippocampal neurons from the toxicity of oligomeric Aβ and reduced tau phosphorylation in the hippocampus of AD mice. Finally, astrocytic glutamate transporter GLT-1, known to be regulated by Akt pathway, was decreased in the transgenic AD mice but upregulated back to the wild-type levels by PDTC treatment. Thus, PDTC may improve spatial learning in AD by interfering with Akt–GSK pathway both in neurons and astrocytes. Because PDTC is capable of transferring external Cu2+ into a cell, and, in turn, Cu2+ is able to activate Akt, we hypothesize that PDTC provides the beneficial effect in transgenic AD mice through Cu2+-activated Akt pathway.


Journal of Neurochemistry | 2008

Age-related decrease in stimulated glutamate release and vesicular glutamate transporters in APP/PS1 transgenic and wild-type mice

Rimante Minkeviciene; Jouni Ihalainen; Tarja Malm; O. Matilainen; Velta Keksa-Goldsteine; Gundars Goldsteins; H. Iivonen; N. Leguit; J. Glennon; Jari Koistinaho; P. Banerjee; Heikki Tanila

We assessed baseline and KCl‐stimulated glutamate release by using microdialysis in freely moving young adult (7 months) and middle‐aged (17 months) transgenic mice carrying mutated human amyloid precursor protein and presenilin genes (APdE9 mice) and their wild‐type littermates. In addition, we assessed the age‐related development of amyloid pathology and spatial memory impaired in the water maze and changes in glutamate transporters. APdE9 mice showed gradual spatial memory impairment between 6 and 15 months of age. The stimulated glutamate release declined very robustly in 17‐month‐old APdE9 mice as compared to 7‐month‐old APdE9 mice. This age‐dependent decrease in stimulated glutamate release was also evident in wild‐type mice, although it was not as robust as in APdE9 mice. When compared to individual baselines, all aged wild‐type mice showed 25% or greater increase in glutamate release upon KCl stimulation, but none of the aged APdE9 mice. There was an age‐dependent decline in VGLUT1 levels, but not in the levels of VGLUT2, GLT‐1 or synaptophysin. Astrocyte activation as measured by glial acidic fibrillary protein was increased in middle‐aged APdE9 mice. Blunted pre‐synaptic glutamate response may contribute to memory deficit in middle‐aged APdE9 mice.


Journal of Cerebral Blood Flow and Metabolism | 2005

Minocycline protects against permanent cerebral ischemia in wild type but not in matrix metalloprotease-9-deficient mice

Milla Koistinaho; Tarja Malm; Mikko I. Kettunen; Gundars Goldsteins; Sofie Starckx; Risto A. Kauppinen; Ghislain Opdenakker; Jari Koistinaho

Minocycline is protective in models of transient middle cerebral artery occlusion (MCAO). We studied whether minocycline and doxycycline, another tetracycline derivative, provide protection in permanent MCAO. Because minocycline inhibits matrix metalloprotease-9 (MMP-9), we also compared minocyclines protective effect in wild type (wt) and MMP-9 knock-out (ko) mice. Wt FVB/N, Balb/C, and two lines of MMP-9 ko and their wt C57Bl/6 control mice were subjected to 24- or 72-hour permanent MCAO. Drug administration was started either 12 hours before or 2 hours after the onset of MCAO. Infarct size was determined by triphenyltetrazolium staining or T2-weighted MRI. Zymography was used to study the expression of MMPs. In wt strains, tetracycline treatments started before MCAO reduced the infarct size by 25% to 50%, whereas the treatment started after MCAO was not protective. Minocycline inhibited ischemia-provoked pro-MMP-9 induction in wt mice, but was not protective in MMP-9 ko mice. Pro-MMP-2 was induced by MCAO in wt and MMP-9 ko mice. MCAO-induced pro-MMP-2 was downregulated by minocycline treatment in wt mice but remained in MMP-9 ko mice at the same level as in saline-treated wt mice. Tetracyclines are protective in permanent MCAO when the treatment is started before the insult. Minocycline may provide protection by interfering with MMPs.


Circulation Research | 2008

Nrf2 Regulates Antioxidant Gene Expression Evoked by Oxidized Phospholipids in Endothelial Cells and Murine Arteries In Vivo

Henna-Kaisa Jyrkkänen; Emilia Kansanen; Matias Inkala; Annukka M. Kivelä; Hanna Hurttila; Suvi E. Heinonen; Gundars Goldsteins; Suvi Jauhiainen; Satu Tiainen; Harri Makkonen; Olga Oskolkova; Taras Afonyushkin; Jari Koistinaho; Masayuki Yamamoto; Valery N. Bochkov; Seppo Ylä-Herttuala; Anna-Liisa Levonen

Besides their well-characterized proinflammatory and proatherogenic effects, oxidized phospholipids, such as oxPAPC (oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-phosphocholine) have been shown to have beneficial responses in vascular cells via induction of antioxidant enzymes such as heme oxygenase-1. We therefore hypothesized that oxPAPC could evoke a general cytoprotective response via activation of antioxidative transcription factor Nrf2. Here, we show that oxPAPC increases nuclear accumulation of Nrf2. Using the small interfering RNA approach, we demonstrate that Nrf2 is critical in mediating the induction of glutamate-cysteine ligase modifier subunit (GCLM) and NAD(P)H quinone oxidoreductase-1 (NQO1) by oxPAPC in human endothelial cells, whereas the contribution to the induction of heme oxygenase-1 was less significant. The induction of GCLM and NQO1 was attenuated by reduction of electrophilic groups with sodium borohydrate, as well as treatment with thiol antioxidant N-acetylcysteine, suggesting that the thiol reactivity of oxPAPC is largely mediating its effect on Nrf2-responsive genes. Moreover, we show that oxidized phospholipid having a highly electrophilic isoprostane ring in its sn-2 position is a potent inducer of Nrf2 target genes. Finally, we demonstrate that the oxPAPC-inducible expression of heme oxygenase-1, GCLM, and NQO1 is lower in Nrf2-null than wild-type mouse carotid arteries in vivo. We suggest that the activation of Nrf2 by oxidized phospholipids provides a mechanism by which their deleterious effects are limited in the vasculature.


Neurobiology of Aging | 2006

Oxidative modification of proteins in the frontal cortex of Alzheimer's disease brain

Minna A. Korolainen; Gundars Goldsteins; Tuula A. Nyman; Irina Alafuzoff; Jari Koistinaho; Tuula Pirttilä

There is a large body of evidence highlighting the importance of oxidative stress in the pathogenesis of Alzheimers disease (AD). We have previously standardised a method that can be applied to study oxidative changes in individual brain proteins by using two-dimensional oxyblots (Korolainen MA, Goldsteins G, Alafuzoff I, Koistinaho J, Pirttilä T. Proteomic analysis of protein oxidation in Alzheimers disease brain. Electrophoresis 2002;23(19):3428-33). Here we have identified proteins that exhibited oxidative changes in AD when compared to age-matched controls and these protein changes have been further examined in relation to the neuropathological data. Indeed, several Tris-HCl soluble proteins tended to be less oxidised in AD when compared to controls. Two enzymes, mitochondrial glutamate dehydrogenase and cytosolic malate dehydrogenase, were increased in amount but showed significantly decreased degree of oxidation in AD brains when compared to controls. Furthermore, some changes related to the amounts or oxidation statuses of proteins were associated with the duration of the clinical impairment and also with the neuropathology. These results do not contradict the hypothesis of increased oxidative stress in AD but may represent co-existing compensatory changes in response to oxidative stress.


Journal of Neuroinflammation | 2010

Human intravenous immunoglobulin provides protection against Aβ toxicity by multiple mechanisms in a mouse model of Alzheimer's disease

Johanna Magga; Lakshman Puli; Rea Pihlaja; Katja M. Kanninen; Suvi Neulamaa; Tarja Malm; Wolfgang Härtig; Jens Grosche; Gundars Goldsteins; Heikki Tanila; Jari Koistinaho; Milla Koistinaho

BackgroundPurified intravenous immunoglobulin (IVIG) obtained from the plasma of healthy humans is indicated for the treatment of primary immunodeficiency disorders associated with defects in humoral immunity. IVIG contains naturally occurring auto-antibodies, including antibodies (Abs) against β-amyloid (Aβ) peptides accumulating in the brains of Alzheimers disease (AD) patients. IVIG has been shown to alleviate AD pathology when studied with mildly affected AD patients. Although its mechanisms-of-action have been broadly studied, it remains unresolved how IVIG affects the removal of natively formed brain Aβ deposits by primary astrocytes and microglia, two major cell types involved in the neuroinflammatory responses.MethodsWe first determined the effect of IVIG on Aβ toxicity in primary neuronal cell culture. The mechanisms-of-action of IVIG in reduction of Aβ burden was analyzed with ex vivo assay. We studied whether IVIG solubilizes natively formed Aβ deposits from brain sections of APP/PS1 mice or promotes Aβ removal by primary glial cells. We determined the role of lysosomal degradation pathway and Aβ Abs in the IVIG-promoted reduction of Aβ. Finally, we studied the penetration of IVIG into the brain parenchyma and interaction with brain deposits of human Aβ in a mouse model of AD in vivo.ResultsIVIG was protective against Aβ toxicity in a primary mouse hippocampal neuron culture. IVIG modestly inhibited the fibrillization of synthetic Aβ1-42 but did not solubilize natively formed brain Aβ deposits ex vivo. IVIG enhanced microglia-mediated Aβ clearance ex vivo, with a mechanism linked to Aβ Abs and lysosomal degradation. The IVIG-enhanced Aβ clearance appears specific for microglia since IVIG did not affect Aβ clearance by astrocytes. The cellular mechanisms of Aβ clearance we observed have potential relevance in vivo since after peripheral administration IVIG penetrated to mouse brain tissue reaching highest concentrations in the hippocampus and bound selectively to Aβ deposits in co-localization with microglia.ConclusionsOur results demonstrate that IVIG promotes recognition and removal of natively formed brain Aβ deposits by primary microglia involving natural Aβ Abs in IVIG. These findings may have therapeutic relevance in vivo as IVIG penetrates through the blood-brain barrier and specifically binds to Aβ deposits in brain parenchyma.


Neuroscience Letters | 2004

Glycosylation changes in Alzheimer’s disease as revealed by a proteomic approach

Katja M. Kanninen; Gundars Goldsteins; Seppo Auriola; Irina Alafuzoff; Jari Koistinaho

Glycosylation influences the biological activity of proteins and affects their folding and stability. Because aberrant glycosylation is associated with Alzheimers disease (AD), we applied proteome analysis together with Pro-Q Emerald 300 glycoprotein staining to investigate changes in glycosylated cytosolic proteins in AD and control brain. Frontal cortex proteins from 10 AD patients and 7 non-demented controls were subjected to separation by two-dimensional gel electrophoresis and subsequently stained with carbohydrate-specific Pro-Q Emerald 300 dye. Changes in glycosylation of separated proteins were quantified, and proteins of interest identified by mass spectrometry. Approximately 30% of all detectable proteins in the human frontal cortex appeared glycosylated, including heat shock cognate 71 stress protein and beta isoform of creatine kinase. The glycosylation of collapsin response mediator protein 2 (CRMP-2) and an unknown protein was reduced in AD, while the glycosylation of glial fibrillary acidic protein was increased. CRMP-2 regulates the assembly and polymerization of microtubules and is associated with neurofibrillary tangles in AD. Aberrant glycosylations in AD may help understand the mechanisms of neurodegenerative diseases.

Collaboration


Dive into the Gundars Goldsteins's collaboration.

Top Co-Authors

Avatar

Jari Koistinaho

University of Eastern Finland

View shared research outputs
Top Co-Authors

Avatar

Tarja Malm

University of Eastern Finland

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Toni Ahtoniemi

University of Eastern Finland

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Merja Jaronen

University of Eastern Finland

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Milla Koistinaho

University of Eastern Finland

View shared research outputs
Top Co-Authors

Avatar

Piia Valonen

University of Eastern Finland

View shared research outputs
Researchain Logo
Decentralizing Knowledge