Network


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

Hotspot


Dive into the research topics where Pernilla Svedin is active.

Publication


Featured researches published by Pernilla Svedin.


The Journal of Neuroscience | 2007

Matrix Metalloproteinase-9 Gene Knock-out Protects the Immature Brain after Cerebral Hypoxia–Ischemia

Pernilla Svedin; Henrik Hagberg; Karin Sävman; Changlian Zhu; Carina Mallard

Inhibition of matrix metalloproteinase-9 (MMP-9) protects the adult brain after cerebral ischemia. However, the role of MMP-9 in the immature brain after hypoxia–ischemia (HI) is unknown. We exposed MMP-9(−/−) [MMP-9 knock-out (KO)] and wild-type (WT) mice to HI on postnatal day 9. HI was induced by unilateral ligation of the left carotid artery followed by hypoxia (10% O2; 36°C). Gelatin zymography showed that MMP-9 activity was transiently increased at 24 h after HI in the ipsilateral hemisphere and MMP-9-positive cells were colocalized with activated microglia. Seven days after 50 min of HI, cerebral tissue volume loss was reduced in MMP-9 KO (21.8 ± 1.7 mm3; n = 22) compared with WT (32.3 ± 2.1 mm3; n = 22; p < 0.001) pups, and loss of white-matter components was reduced in MMP-9 KO compared with WT pups (neurofilament: WT, 50.9 ± 5.4%; KO, 18.4 ± 3.1%; p < 0.0001; myelin basic protein: WT, 57.5 ± 5.8%; KO, 23.2 ± 3.5%; p = 0.0001). The neuropathological changes were associated with a delayed and diminished leakage of the blood–brain barrier (BBB) and a decrease in inflammation in MMP-9-deficient animals. In contrast, the neuroprotective effects after HI in MMP-9-deficient animals were not linked to either caspase-dependent (caspase-3 and cytochrome c) or caspase-independent (apoptosis-inducing factor) processes. This study demonstrates that excessive activation of MMP-9 is deleterious to the immature brain, which is associated with the degree of BBB leakage and inflammation. In contrast, apoptosis does not appear to be a major contributing factor.


Pediatric Research | 2007

Melatonin reduces inflammation and cell death in white matter in the mid-gestation fetal sheep following umbilical cord occlusion

Anne-Karin Welin; Pernilla Svedin; Risto Lapatto; Bo Sultan; Henrik Hagberg; Pierre Gressens; Ingemar Kjellmer; Carina Mallard

The premature infant is at increased risk of cerebral white matter injury. Melatonin is neuroprotective in adult models of focal cerebral ischemia and attenuates ibotenate-induced white matter cysts in neonatal mice. Clinically, melatonin has been used to treat sleep disorders in children without major side effects. The aim of this study was to investigate the protective and anti-inflammatory effects of melatonin in the immature brain following intrauterine asphyxia. Fetal sheep at 90 d of gestation were subjected to umbilical cord occlusion. Melatonin (20 mg/kg, n = 9) or vehicle (n = 10) was administered IV to the fetus, starting 10 min after the start of reperfusion and continued for 6 h. Melatonin treatment resulted in a slower recovery of fetal blood pressure following umbilical cord occlusion, but without changes in fetal heart rate, acid base status or mortality. The production of 8-isoprostanes following umbilical cord occlusion was attenuated and there was a reduction in the number of activated microglia cells and TUNEL-positive cells in melatonin treated fetuses, suggesting a protective effect of melatonin. In conclusion, this study shows that melatonin attenuates cell death in the fetal brain in association with a reduced inflammatory response in the blood and the brain following intrauterine asphyxia in mid-gestation fetal sheep.


Annals of Neurology | 2007

N-acetylcysteine reduces lipopolysaccharide-sensitized hypoxic-ischemic brain injury.

Xiaoyang Wang; Pernilla Svedin; Chunxia Nie; Risto Lapatto; Changlian Zhu; Malin Gustavsson; Mats Sandberg; Jan-Olof Karlsson; Roberto Romero; Henrik Hagberg; Carina Mallard

Maternal inflammation/infection alone or in combination with birth asphyxia increases the risk for perinatal brain injury. Free radicals are implicated as major mediators of inflammation and hypoxia‐ischemia (HI)–induced perinatal brain injury. This study evaluated the neuroprotective efficacy of a scavenging agent, N‐acetylcysteine (NAC), in a clinically relevant model.


Journal of Cerebral Blood Flow and Metabolism | 2015

Brain Barrier Properties and Cerebral Blood Flow in Neonatal Mice Exposed to Cerebral Hypoxia-Ischemia

C. Joakim Ek; Barbara D'Angelo; Ana A. Baburamani; Christine Lehner; Anna-Lena Leverin; Peter L.P. Smith; Holger Nilsson; Pernilla Svedin; Henrik Hagberg; Carina Mallard

Insults to the developing brain often result in irreparable damage resulting in long-term deficits in motor and cognitive functions. The only treatment today for hypoxic-ischemic encephalopathy (HIE) in newborns is hypothermia, which has limited clinical benefit. We have studied changes to the blood–brain barriers (BBB) as well as regional cerebral blood flow (rCBF) in a neonatal model of HIE to further understand the underlying pathologic mechanisms. Nine-day old mice pups, brain roughly equivalent to the near-term human fetus, were subjected to hypoxia-ischemia. Hypoxia-ischemia increased BBB permeability to small and large molecules within hours after the insult, which normalized in the following days. The opening of the BBB was associated with changes to BBB protein expression whereas gene transcript levels were increased showing direct molecular damage to the BBB but also suggesting compensatory mechanisms. Brain pathology was closely related to reductions in rCBF during the hypoxia as well as the areas with compromised BBB showing that these are intimately linked. The transient opening of the BBB after the insult is likely to contribute to the pathology but at the same time provides an opportunity for therapeutics to better reach the infarcted areas in the brain.


Pediatric Research | 2012

Increased MMP-9 and TIMP-1 in mouse neonatal brain and plasma and in human neonatal plasma after hypoxia-ischemia: a potential marker of neonatal encephalopathy.

Nathalie Bednarek; Pernilla Svedin; Roselyne Garnotel; Géraldine Favrais; Gauthier Loron; Leslie Schwendiman; Henrik Hagberg; Patrice Morville; Carina Mallard; Pierre Gressens

Introduction:To implement neuroprotective strategies in newborns, sensitive and specific biomarkers are needed for identifying those who are at risk for brain damage. We evaluated the effectiveness of matrix metalloproteinases (MMPs) and their naturally occurring tissue inhibitors of metalloproteinases (TIMPs) in predicting neonatal encephalopathy (NE) damage in newborns.Results:Plasma MMP-9 and TIMP-1 levels were upregulated as early as 1 h after the HI insult but not did not show such elevations after other types of injury (ibotenate-induced excitotoxicity, hypoxia, lipopolysaccharide-induced inflammation), and brain levels reflected this increase soon thereafter. We confirmed these results by carrying out plasma MMP-9 and TIMP-1 measurements in human newborns with NE. In these infants, protein levels of MMP-9 and TIMP-1 were found to be elevated during a short window up to 6 h after birth.Discussion:This feature is particularly useful in identifying newborns in need of neuroprotection. A second peak observed 72 h after birth is possibly related to the second phase of energy failure after a HI insult. Our data, although preliminary, support the use of MMP-9 and TIMP-1 as early biomarkers for the presence and extent of perinatal brain injury in human term newborns.Methods:We first used a mouse model of neonatal HI injury to explore mechanistic aspects such as the time course of these markers after the hypoxia–ischemia event, and the correlation between the levels of these candidate markers in brain and plasma.


Developmental Neuroscience | 2007

Delayed Peripheral Administration of a GPE Analogue Induces Astrogliosis and Angiogenesis and Reduces Inflammation and Brain Injury following Hypoxia-Ischemia in the Neonatal Rat

Pernilla Svedin; Jian Guan; Sam Mathai; Rong Zhang; Xiaoyang Wang; Malin Gustavsson; Henrik Hagberg; Carina Mallard

Glycine 2-methyl proline glutamate (G-2mPE) is a proline-modified analogue to the naturally existing N-terminal tripeptide glycine-proline-glutamate that is a cleaved product from insulin-like growth factor-1. G-2mPE is designed to be more enzymatically resistant than glycine-proline-glutamate and to increase its bioavailability. The current study has investigated the protective effects of G-2mPE following hypoxic-ischemic brain injury in the neonatal brain. On postnatal day 7, Wistar rats were exposed to hypoxia-ischemia (HI). HI was induced by unilateral ligation of the left carotid artery followed by hypoxia (7.7% O2, 36°C) for 60 min. The drug treatment started 2 h after the insult, and the pups were given either 1.2 mg/kg (bolus), 1.2 mg/ml once a day for 7 days, or vehicle. The degree of brain damage was determined histochemically by thionin/acid fuchsin staining. G-2mPE’s anti-inflammatory properties were investigated by IL-1β, IL-6, and IL-18 ELISA, and effects on apoptosis by caspase 3 activity. Vascularization was determined immunohistochemically by the total length of isolectin-positive blood vessels. Effect on astrocytosis was also determined in the hippocampus. Animals treated with multiple doses of G-2mPE demonstrated reduced overall brain injury 7 days after HI, particularly in the hippocampus and thalamus compared to vehicle-treated rats. The expression of IL-6 was decreased in G-2mPE-treated animals compared to vehicle-treated pups, and both the capillary length and astrogliosis were increased in the drug-treated animals. There was no effect on caspase 3 activity. This study indicates that peripheral administration of G-2mPE, starting 2 h after a hypoxic-ischemic insult, reduces the degree of brain injury in the immature rat brain. The normalization of IL-6 levels and the promotion of both neovascularization and reactive astrocytosis may be potential mechanisms that underlie its protective effects.


Translational Stroke Research | 2013

Microglia/Macrophage-Derived Inflammatory Mediators Galectin-3 and Quinolinic Acid are Elevated in Cerebrospinal Fluid from Newborn Infants After Birth Asphyxia

Karin Sävman; Melvyn P. Heyes; Pernilla Svedin; Anna Karlsson

Activation of microglia/macrophages is important in neonatal hypoxic–ischemic (HI) brain injury. Based on experimental studies, we identified macrophage/microglia-derived mediators with potential neurotoxic effects after neonatal HI and examined them in cerebrospinal fluid (CSF) from newborn infants after birth asphyxia. Galectin-3 is a novel inflammatory mediator produced by microglia/macrophages. Galectin-3 is chemotactic for inflammatory cells and activates nicotinamide adenine dinucleotide phosphate (NADPH) oxidase resulting in production and release of reactive oxygen species (ROS). Matrix metalloproteinase-9 (MMP-9) is a tissue-degrading protease expressed by activated microglia in the immature brain after HI. Both galectin-3 and MMP-9 contribute to brain injury in animal models for neonatal HI. Quinolinic acid (QUIN) is a neurotoxic N-methyl-d-aspartate (NMDA) receptor agonist also produced by activated microglia/macrophages. Galectin-3 and MMP-9 were measured by ELISA and QUIN by mass spectrometry. Asphyxiated infants (n = 20) had higher levels of galectin-3 (mean (SEM) 2.64 (0.43) ng/mL) and QUIN (335.42 (58.9) nM) than controls (n = 15) (1.36 (0.46) ng/mL and 116.56 (16.46) nM, respectively), p < 0.05 and p < 0.01. Infants with septic infections (n = 10) did not differ from controls. Asphyxiated infants with abnormal outcome had higher levels of galectin-3 (3.96 (0.67) ng/mL) than those with normal outcome (1.76 (0.32) ng/mL), p = 0.02, and the difference remained significant in the clinically relevant group of infants with moderate encephalopathy. MMP-9 was detected in few infants with no difference between groups. The potentially neurotoxic macrophage/microglia-derived mediators galectin-3 and QUIN are increased in CSF after birth asphyxia and could serve as markers and may contribute to injury.


Developmental Neuroscience | 2009

Expression of MMP-12 after Neonatal Hypoxic-Ischemic Brain Injury in Mice

Pernilla Svedin; Henrik Hagberg; Carina Mallard

Matrix metalloproteinase-12 (MMP-12) is expressed in the brain and is important for myelin formation in the developing brain, while MMP-12 deficiency protects against spinal cord injury in the adult, suggesting a role for MMP-12 after brain injury. However, the role of MMP-12 in neonatal hypoxic-ischemic brain injury is not known. The purpose of this study was to investigate the expression of MMP-12 in the brain after neonatal hypoxia-ischemia (HI). HI was induced by unilateral ligation of the left carotid artery followed by hypoxia (10% O2, 36°C) for 50 min in postnatal day 9 C57/Bl6J mice. At 24 and 72 h after HI, the mRNA and protein expression of MMP-12 were measured by real-time PCR and Western blot, respectively. Distribution and cellular expression of MMP-12 were examined by immunohistochemistry. At 72 h after HI, both MMP-12 mRNA and protein expression were significantly increased in the ipsilateral hemisphere compared to the contralateral hemisphere and sham-operated animals (p < 0.05). The extent of tissue loss in the ipsilateral hemisphere at 72 h after HI was positively correlated with the degree of MMP-12 protein expression (r = 0.900, p < 0.05). In nonischemic animals, immunohistochemical analysis demonstrated weak cytoplasmic MMP-12 immunoreactivity throughout the brain in cells which resembled neurons and stronger immunoreactivity was observed in blood vessels. After HI, the MMP-12 staining became more prominent in the ipsilateral hemisphere and immunohistochemical double-labeling experiments identified the MMP-12-positive cells as neurons, isolectin and Olig2-positive cells. This study demonstrates that MMP-12 is upregulated after HI, suggesting that MMP-12 may contribute to injury in the immature brain, similar to that seen in the adult.


Journal of Cerebral Blood Flow and Metabolism | 2017

Systemic activation of Toll-like receptor 2 suppresses mitochondrial respiration and exacerbates hypoxic-ischemic injury in the developing brain

Amin Mottahedin; Pernilla Svedin; Syam Nair; Carl-Johan Mohn; Xiaoyang Wang; Henrik Hagberg; Joakim Ek; Carina Mallard

Infection and inflammation are known risk factors for neonatal brain injury. Mycoplasma and Gram-positive bacteria, for which Toll-like receptor 2 (TLR2) plays a key role in recognition and inflammatory response, are among the most common pathogens in the perinatal period. Here, we report that systemic activation of TLR2 by Pam3CSK4 (P3C) increases neural tissue loss and demyelination induced by subsequent hypoxia–ischemia (HI) in neonatal mice. High-resolution respirometry of brain isolated mitochondria revealed that P3C suppresses ADP-induced oxidative phosphorylation, the main pathway of cellular energy production. The results suggest that infection and inflammation might contribute to HI-induced energy failure.


Frontiers in Cellular Neuroscience | 2016

Temporal Characterization of Microglia/Macrophage Phenotypes in a Mouse Model of Neonatal Hypoxic-Ischemic Brain Injury

Nina Hellström Erkenstam; Peter L.P. Smith; Bobbi Fleiss; Syam Nair; Pernilla Svedin; Wei Wang; Martina Boström; Pierre Gressens; Henrik Hagberg; Kelly L. Brown; Karin Sävman; Carina Mallard

Immune cells display a high degree of phenotypic plasticity, which may facilitate their participation in both the progression and resolution of injury-induced inflammation. The purpose of this study was to investigate the temporal expression of genes associated with classical and alternative polarization phenotypes described for macrophages and to identify related cell populations in the brain following neonatal hypoxia-ischemia (HI). HI was induced in 9-day old mice and brain tissue was collected up to 7 days post-insult to investigate expression of genes associated with macrophage activation. Using cell-markers, CD86 (classic activation) and CD206 (alternative activation), we assessed temporal changes of CD11b+ cell populations in the brain and studied the protein expression of the immunomodulatory factor galectin-3 in these cells. HI induced a rapid regulation (6 h) of genes associated with both classical and alternative polarization phenotypes in the injured hemisphere. FACS analysis showed a marked increase in the number of CD11b+CD86+ cells at 24 h after HI (+3667%), which was coupled with a relative suppression of CD11b+CD206+ cells and cells that did not express neither CD86 nor CD206. The CD11b+CD206+ population was mixed with some cells also expressing CD86. Confocal microscopy confirmed that a subset of cells expressed both CD86 and CD206, particularly in injured gray and white matter. Protein concentration of galectin-3 was markedly increased mainly in the cell population lacking CD86 or CD206 in the injured hemisphere. These cells were predominantly resident microglia as very few galectin-3 positive cells co-localized with infiltrating myeloid cells in Lys-EGFP-ki mice after HI. In summary, HI was characterized by an early mixed gene response, but with a large expansion of mainly the CD86 positive population during the first day. However, the injured hemisphere also contained a subset of cells expressing both CD86 and CD206 and a large population that expressed neither activation marker CD86 nor CD206. Interestingly, these cells expressed the highest levels of galectin-3 and were found to be predominantly resident microglia. Galectin-3 is a protein involved in chemotaxis and macrophage polarization suggesting a novel role in cell infiltration and immunomodulation for this cell population after neonatal injury.

Collaboration


Dive into the Pernilla Svedin's collaboration.

Top Co-Authors

Avatar

Carina Mallard

University of Gothenburg

View shared research outputs
Top Co-Authors

Avatar

Henrik Hagberg

University of Gothenburg

View shared research outputs
Top Co-Authors

Avatar

Xiaoyang Wang

University of Gothenburg

View shared research outputs
Top Co-Authors

Avatar

Karin Sävman

University of Gothenburg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Aurimantas Pelanis

Sahlgrenska University Hospital

View shared research outputs
Top Co-Authors

Avatar

Changlian Zhu

University of Gothenburg

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge