Network


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

Hotspot


Dive into the research topics where Charlotte Israelsson is active.

Publication


Featured researches published by Charlotte Israelsson.


Journal of Neurotrauma | 2008

Distinct cellular patterns of upregulated chemokine expression supporting a prominent inflammatory role in traumatic brain injury

Charlotte Israelsson; Henrik Bengtsson; Annika Kylberg; Klas Kullander; Anders Lewén; Ted Ebendal

Cerebral gene expressions change in response to traumatic brain injury (TBI), and future trauma treatment may improve with increased knowledge about these regulations. We subjected C57BL/6J mice to injury by controlled cortical impact (CCI). At various time points post-injury, mRNA from neocortex and hippocampus was isolated, and transcriptional alterations studied using quantitative real-time polymerase chain reaction (PCR) and gene array analysis. Spatial distribution of enhanced expression was characterized by in situ hybridization. Products of the upregulated transcripts serve functions in a range of cellular mechanisms, including stress, inflammation and immune responses, and tissue remodeling. We also identified increased transcript levels characterizing reactive astrocytes, oligodendrocytes, and microglia, and furthermore, we demonstrated a novel pattern of scattered cell clusters expressing the chemokine Cxcl10. Notably, a sustained increase in integrin alpha X (Itgax), characterizing antigen-presenting dendritic cells, was found with the transcript located to similar cell clusters. In contrast, T-cell receptor alpha transcript showed only a modest increase. The induced P-selectin (Selp) expression level in endothelial cells, and chemokines from microglia, may guide perivascular accumulation of extravasating inflammatory monocytes differentiating into dendritic cells. In conclusion, our study shows that following TBI, secondary injury chiefly involves inflammatory processes and chemokine signaling, which comprise putative targets for pharmaceutical neuroprotection.


European Journal of Neuroscience | 2011

Neutralization of Interleukin-1β Reduces Cerebral Edema and Tissue loss and Improves Late Cognitive Outcome Following Traumatic Brain Injury in Mice

Fredrik Clausen; Anders Hånell; Charlotte Israelsson; Johanna Hedin; Ted Ebendal; Anis Khusro Mir; Hermann Gram; Niklas Marklund

Increasing evidence suggests that interleukin‐1β (IL‐1β) is a key mediator of the inflammatory response following traumatic brain injury (TBI). Recently, we showed that intracerebroventricular administration of an IL‐1β‐neutralizing antibody was neuroprotective following TBI in mice. In the present study, an anti‐IL‐1β antibody or control antibody was administered intraperitoneally following controlled cortical injury (CCI) TBI or sham injury in 105 mice and we extended our histological, immunological and behavioral analysis. First, we demonstrated that the treatment antibody reached target brain regions of brain‐injured animals in high concentrations (> 11 nm) remaining up to 8 days post‐TBI. At 48 h post‐injury, the anti‐IL‐1β treatment attenuated the TBI‐induced hemispheric edema (P < 0.05) but not the memory deficits evaluated using the Morris water maze (MWM). Neutralization of IL‐1β did not influence the TBI‐induced increases (P < 0.05) in the gene expression of the Ccl3 and Ccr2 chemokines, IL‐6 or Gfap. Up to 20 days post‐injury, neutralization of IL‐1β was associated with improved visuospatial learning in the MWM, reduced loss of hemispheric tissue and attenuation of the microglial activation caused by TBI (P < 0.05). Motor function using the rotarod and cylinder tests was not affected by the anti‐IL‐1β treatment. Our results suggest an important negative role for IL‐1β in TBI. The improved histological and behavioral outcome following anti‐IL‐1β treatment also implies that further exploration of IL‐1β‐neutralizing compounds as a treatment option for TBI patients is warranted.


Restorative Neurology and Neuroscience | 2010

Vimentin and GFAP responses in astrocytes after contusion trauma to the murine brain

Sara Ekmark-Lewén; Anders Lewén; Charlotte Israelsson; Gui Lin Li; Mohammad Farooque; Yngve Olsson; Ted Ebendal

PURPOSE Astroglial responses after traumatic brain injury are difficult to detect with routine morphological methods. The aims for this study were to compare the temporal and spatial expression pattern of vimentin- and glial fibrillary acidic protein (GFAP) in a weight drop model of mild cerebral contusion injury in the rat. We also wanted to study the vimentin response with immunohistochemistry and vimentin mRNA RT-PCR analysis in severe cortical contusion injury produced by the controlled cortical impact in the mouse. METHODS Vimentin and GFAP immunohistochemistry (1 day, 3 days and 7 days) combined with vimentin mRNA RT-PCR analysis (1 h, 4 h, 22 h, 3 days and 7 days) were used after experimental traumatic brain injury in the rat and mouse. RESULTS Increases in post-traumatic vimentin mRNA levels in the cortex and in the hippocampus appeared together with vimentin immunoreactivity in astrocytes in the perimeter of the cortical lesion, in the subcortical white matter and in the hippocampus starting at one day after severe trauma. GFAP immunostaining revealed hypertrophic astrocytes peaking at day 3 in the perifocal cortical region. There was no significant increase in GFAP immunoreactivity in the white matter in the rat. However, in the mouse there was a slight increase in the number of GFAP positive cells in this region, 3 days after trauma. Overall the pattern of vimentin immunoreactivity was very similar in the rat and mouse. CONCLUSIONS Vimentin immunoreactivity was more sensitive than the GFAP staining method to demonstrate the distribution and time course of astrocyte reactions after a contusion injury, especially in the white matter distant from the cortical lesion.


European Journal of Neuroscience | 2010

Appearance of Cxcl10-expressing cell clusters is common for traumatic brain injury and neurodegenerative disorders

Charlotte Israelsson; Henrik Bengtsson; Anna Lobell; Lars Nilsson; Annika Kylberg; Magnus Isaksson; Hanna Wootz; Lars Lannfelt; Klas Kullander; Ted Ebendal

Traumatic brain injury (TBI) in the mouse results in the rapid appearance of scattered clusters of cells expressing the chemokine Cxcl10 in cortical and subcortical areas. To extend the observation of this unique pattern, we used neuropathological mouse models using quantitative reverse transcriptase‐polymerase chain reaction, gene array analysis, in‐situ hybridization and flow cytometry. As for TBI, cell clusters of 150–200 μm expressing Cxcl10 characterize the cerebral cortex of mice carrying a transgene encoding the Swedish mutation of amyloid precursor protein, a model of amyloid Alzheimer pathology. The same pattern was found in experimental autoimmune encephalomyelitis in mice modelling multiple sclerosis. In contrast, mice carrying a SOD1G93A mutant mimicking amyotrophic lateral sclerosis pathology lacked such cell clusters in the cerebral cortex, whereas clusters appeared in the brainstem and spinal cord. Mice homozygous for a null mutation of the Cxcl10 gene did not show detectable levels of Cxcl10 transcript after TBI, confirming the quantitative reverse transcriptase‐polymerase chain reaction and in‐situ hybridization signals. Moreover, unbiased microarray expression analysis showed that Cxcl10 was among 112 transcripts in the neocortex upregulated at least threefold in both TBI and ageing TgSwe mice, many of them involved in inflammation. The identity of the Cxcl10+ cells remains unclear but flow cytometry showed increased numbers of activated microglia/macrophages as well as myeloid dendritic cells in the TBI and experimental autoimmune encephalomyelitis models. It is concluded that the Cxcl10+ cells appear in the inflamed central nervous system and may represent a novel population of cells that it may be possible to target pharmacologically in a broad range of neurodegenerative conditions.


Journal of Neuroscience Research | 2006

Genetically modified bone morphogenetic protein signalling alters traumatic brain injury-induced gene expression responses in the adult mouse

Charlotte Israelsson; Anders Lewén; Annika Kylberg; Dmitry Usoskin; Susanna Althini; Jonas Lindeberg; Chu-Xia Deng; Tomokazu Fukuda; Yun Wang; Vesa Kaartinen; Yuji Mishina; Ted Ebendal

Three genetic mouse models were examined to define effects of bone morphogenetic protein (BMP) signalling on gene expression in normal and injured adult brain. CaMKII‐Cre eliminated the BMP receptor Acvr1 (Alk2) and the common TGFβ superfamily signal mediator Smad4 or activated a constitutively active Acvr1 in postnatal forebrain neurons. All mutants followed mendelian ratios, with no overt phenotypic changes. In situ hybridization demonstrated normal patterns of the dendritic marker MAP2 (Mtap2) throughout cortex despite neuron‐specific losses of Acvr1 or Smad4. However, strong up‐regulation of Mtap2 transcript in these mice was found by quantitative RT‐PCR (qRT‐PCR), indicating that Mtap2 is normally suppressed by BMP. Traumatic brain injury (TBI) resulted in increases of histone‐associated DNA fragments in both control and Smad4‐deficient cortex. Several cell‐type‐specific transcripts known to be involved in injury‐related responses were measured by qRT‐PCR. Gfap mRNA was strongly up‐regulated in controls as well as in the loss‐of‐BMP‐signalling mutants. Notably, activated Acvr1 signalling gave significantly lower TBI‐induced up‐regulations of Gfap and Phox2a mRNA levels, indicating reductions in astroglial and neuronal reactions to injury. Strong impairment in injury‐induced Timp1 transcript up‐regulation was also seen in these mice. In contrast, osteopontin (Spp1) transcript levels in activated microglia were not reduced by Acvr1 signalling. Altogether, the data suggest that BMP signalling is dispensable in adult cortical neurons but that augmented BMP signalling affects molecular changes associated with neuronal lesions.


PLOS ONE | 2014

Interacting chemokine signals regulate dendritic cells in acute brain injury.

Charlotte Israelsson; Annika Kylberg; Henrik Bengtsson; Ted Ebendal

Brain trauma is known to activate inflammatory cells via various chemokine signals although their interactions remain to be characterized. Mice deficient in Ccl3, Ccr2 or Cxcl10 were compared with wildtype mice after controlled cortical impact injury. Expression of Ccl3 in wildtypes was rapidly upregulated in resident, regularly spaced reactive microglia. Ccl3-deficiency enhanced endothelial expression of platelet selectin and invasion of peripheral inflammatory cells. Appearance of Ccr2 transcripts, encoding the Ccl2 receptor, reflected invasion of lysozyme 2-expressing phagocytes and classical antigen-presenting dendritic cells expressing major histocompatibility complex class II. Ccr2 also directed clustered plasmacytoid dendritic cells positive for the T-cell attracting chemokine Cxcl10. A reduction in Ccr2 and dendritic cells was found in injured wildtype cortex after cyclophosphamide treatment resembling effects of Ccr2-deficiency. The findings demonstrate the feasibility to control inflammation in the injured brain by regulating chemokine-dependent pathways.


Journal of Neurotrauma | 2012

Functional and Histological Outcome after Focal Traumatic Brain Injury Is Not Improved in Conditional EphA4 Knockout Mice

Anders Hånell; Fredrik Clausen; Anders Djupsjö; Anna Vallstedt; Kalicharan Patra; Charlotte Israelsson; Martin Larhammar; Maria Björk; Sónia Paixão; Klas Kullander; Niklas Marklund

We investigated the role of the axon guidance molecule EphA4 following traumatic brain injury (TBI) in mice. Neutralization of EphA4 improved motor function and axonal regeneration following experimental spinal cord injury (SCI). We hypothesized that genetic absence of EphA4 could improve functional and histological outcome following TBI. Using qRT-PCR in wild-type (WT) mice, we evaluated the EphA4 mRNA levels following controlled cortical impact (CCI) TBI or sham injury and found it to be downregulated in the hippocampus (p<0.05) but not the cortex ipsilateral to the injury at 24 h post-injury. Next, we evaluated the behavioral and histological outcome following CCI using WT mice and Emx1-Cre-driven conditional knockout (cKO) mice. In cKO mice, EphA4 was completely absent in the hippocampus and markedly reduced in the cortical regions from embryonic day 16, which was confirmed using Western blot analysis. EphA4 cKO mice had similar learning and memory abilities at 3 weeks post-TBI compared to WT controls, although brain-injured animals performed worse than sham-injured controls (p<0.05). EphA4 cKO mice performed similarly to WT mice in the rotarod and cylinder tests of motor function up to 29 days post-injury. TBI increased cortical and hippocampal astrocytosis (GFAP immunohistochemistry, p<0.05) and hippocampal sprouting (Timm stain, p<0.05) and induced a marked loss of hemispheric tissue (p<0.05). EphA4 cKO did not alter the histological outcome. Although our results may argue against a beneficial role for EphA4 in the recovery process following TBI, further studies including post-injury pharmacological neutralization of EphA4 are needed to define the role for EphA4 following TBI.


Restorative Neurology and Neuroscience | 2014

Altered expression of myelin-associated inhibitors and their receptors after traumatic brain injury in the mouse

Charlotte Israelsson; Johanna Flygt; Elaine Astrand; Henrik Bengtsson; Niklas Marklund

PURPOSE When central nervous system axons are injured, regeneration is partly inhibited by myelin-associated inhibitors (MAIs). Following traumatic brain injury (TBI) in the rat, pharmacological neutralisation of the MAIs Nogo-A and myelin-associated glycoprotein (MAG) resulted in improved functional outcome. In contrast, genetic or pharmacological neutralization of the MAI receptors Nogo-66 receptor 1 (NgR1) or paired-immunoglobulin like receptor-B (PirB) showed an unaltered or impaired outcome following TBI in mice. The aim of the present study was thus to evaluate the MAI expression levels following TBI in mice. METHODS Quantitative reverse transcriptase PCR (qRT-PCR) was used to measure total RNA isolated from brains of young adult male C57BL/6 mice at one, three or seven days following controlled cortical impact TBI or sham injury. Hippocampal and neocortical tissue ipsi- and contralateral to the injury was analyzed for Nogo-A, oligodendrocyte-myelin glycoprotein (OMgp), MAG, and the MAI receptors PirB and NgR1, including its co-receptor Lingo1. RESULTS Compared to sham-injured controls, PirB neocortical expression was significantly upregulated at one day and NgR1 expression downregulated at seven days post-TBI. In the hippocampus, transcriptional upregulation was observed in Nogo-A (one day post-injury), MAG and PirB at seven days post-injury. In contrast, the hippocampal transcripts of NgR1 and Lingo1 were decreased at seven days post-injury. The expression of OMgp was unaltered at all time points post-injury. CONCLUSION These results suggest that early dynamic changes in MAI gene expression occur following TBI in the mouse, particularly in the hippocampus, which may play an inhibitory role for post-injury regeneration and plasticity.


Journal of Stroke & Cerebrovascular Diseases | 2018

Hyperglycemia Alters Expression of Cerebral Metabolic Genes after Cardiac Arrest

Rickard P.F. Lindblom; Maria Molnar; Charlotte Israelsson; Belinda Röjsäter; Lars Wiklund; Fredrik Lennmyr

BACKGROUND Survivors of cardiac arrest often experience neurologic deficits. To date, treatment options are limited. Associated hyperglycemia is believed to further worsen the neurologic outcome. The aim with this study was to characterize expression pathways induced by hyperglycemia in conjunction with global brain ischemia. METHODS Pigs were randomized to high or normal glucose levels, as regulated by glucose and insulin infusions with target levels of 8.5-10 mM and 4-5.5 mM, respectively. The animals were subjected to 5-minute cardiac arrest followed by 8 minutes of cardiopulmonary resuscitation and direct-current shock to restore spontaneous circulation. Global expression profiling of the cortex using microarrays was performed in both groups. RESULTS A total of 102 genes differed in expression at P < .001 between the hyperglycemic and the normoglycemic pigs. Several of the most strongly differentially regulated genes were involved in transport and metabolism of glucose. Functional clustering using bioinformatics tools revealed enrichment of multiple biological processes, including membrane processes, ion transport, and glycoproteins. CONCLUSIONS Hyperglycemia during cardiac arrest leads to differential early gene expression compared with normoglycemia. The functional relevance of these expressional changes cannot be deduced from the current study; however, the identified candidates have been linked to neuroprotective mechanisms and constitute interesting targets for further studies.


Journal of Neuroscience Research | 2015

Anti-inflammatory treatment of traumatic brain injury with Rabeximod reduces cerebral antigen presentation in mice

Charlotte Israelsson; Annika Kylberg; Ulf Björklund; Ted Ebendal

A major component of the damaging effect after traumatic brain injury (TBI) is activation of the inflammatory system. In particular, chemokines and chemokine‐regulated factors become activated in resident brain cells and signal to different invading immune cells. For evaluation of the effect on invading cells 3 days after injury, mice were treated with a single initial dose of the anti‐inflammatory agent Rabeximod in an experimental TBI model. For comparison, mice subjected to TBI were similarly injected with cyclophosphamide. TBI resulted in reduced body weight, an effect further enhanced by administration of Rabeximod, without obvious influence on motor performance. As revealed by quantitative RT‐PCR, microglial upregulation of chemokine Ccl3 in response to TBI was unaffected by Rabeximod. Also, injury‐induced expression of Cxcl10 in plasmacytoid dendritic cells (DCs) and endothelial expression of platelet selectin (Selp) were uninfluenced by Rabeximod. In contrast, Rabeximod robustly reduced the H2‐Aa transcript characteristic for classical DCs defined by CD11c/Itgax in the injured brain. In addition, the expression of lysozyme 2 in large phagocytic cells was impaired by Rabeximod. These results show that Rabeximod exerts a selective and potent inhibition of cells serving cortical antigen presentation after brain trauma.

Collaboration


Dive into the Charlotte Israelsson's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge