Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Brain Behav Immun ; 70: 118-130, 2018 05.
Article in English | MEDLINE | ID: mdl-29454023

ABSTRACT

Acute hypothermia treatment (HT) is the only clinically established intervention following neonatal hypoxic-ischemic brain injury. However, almost half of all cooled infants still die or suffer from long-lasting neurological impairments. Regenerative therapies, such as mesenchymal stem cells (MSC) appear promising as adjuvant therapy. In the present study, we hypothesized that HT combined with delayed MSC therapy results in augmented protection, improving long-term neurological outcome. Postnatal day 9 (P9) C57BL/6 mice were exposed to hypoxia-ischemia followed by 4 h HT. Murine bone marrow-derived MSC (1 × 106 cells/animal) were administered intranasally at P12. Cytokine and growth factor levels were assessed by ELISA and Luminex® multiplex assay 24 h following MSC delivery. One week after HI, tissue injury and neuroinflammatory responses were determined by immunohistochemistry and western blot. Long-term motor-cognitive outcome was assessed 5 weeks post injury. MSC responses to the brains' environment were evaluated by gene expression analysis in MSC, co-cultured with brain homogenates isolated at P12. Both, MSC and HT improved motor deficits, while cognitive function could only be restored by MSC. Compared to each single therapy, combined treatment led to increased long-lasting motor-cognitive deficits and exacerbated brain injury, accompanied by enhanced endothelial activation and peripheral immune cell infiltration. MSC co-cultured with brain extracts of HT-treated animals revealed increased pro-inflammatory cytokine and decreased growth factor expression. In vivo protein analysis showed higher pro-inflammatory cytokine levels after combined treatment compared to single therapy. Furthermore, HI-induced increase in growth factors was normalized to control levels by HT and MSC single therapy, while the combination induced a further decline below control levels. Our results suggest that alteration of the brains' microenvironment by acute HT modulates MSC function resulting in a pro-inflammatory environment combined with alteration of the homeostatic growth factor milieu in the neonatal hypoxic-ischemic brain. This study delineates potential unexpected side effects of cell-based therapies as add-on therapy for acute hypothermia treatment.


Subject(s)
Hypothermia/physiopathology , Hypoxia-Ischemia, Brain/physiopathology , Mesenchymal Stem Cells/physiology , Administration, Intranasal , Animals , Animals, Newborn/physiology , Behavior, Animal , Brain , Brain Injuries , Cell Proliferation , Disease Models, Animal , Humans , Hypothermia, Induced/methods , Mesenchymal Stem Cell Transplantation/methods , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins
2.
Exp Neurol ; 283(Pt A): 264-75, 2016 09.
Article in English | MEDLINE | ID: mdl-27349408

ABSTRACT

Hypothermia treatment (HT) is the only formally endorsed treatment recommended for hypoxic-ischemic encephalopathy (HIE). However, its success in protecting against brain injury is limited with a number to treat of 7-8. The identification of the target mechanisms of HIE in combination with HT will help to explain ineffective therapy outcomes but also requires stable experimental models in order to establish further neuroprotective therapies. Despite clinical and experimental indications for an endogenous thermoregulatory response to HIE, the potential effects on HIE-induced brain injury have largely been neglected in pre-clinical studies. In the present study we analyzed gray and white matter injury and neurobehavioral outcome in neonatal mice considering the endogenous thermoregulatory response during HIE combined with HT. HIE was induced in postnatal day (PND) 9 C57BL/6 mice through occlusion of the right common carotid artery followed by one hour of hypoxia. Hypoxia was performed at 8% or 10% oxygen (O2) at two different temperatures based on the nesting body core temperature. Using the model which mimics the clinical situation most closely, i.e. through maintenance of the nesting temperature during hypoxia we compared two mild HT protocols (rectal temperature difference 3°C for 4h), initiated either immediately after HIE or with delay of 2h. Injury was determined by histology, immunohistochemistry and western blot analyses at PND 16 and PND 51. Functional outcome was evaluated by Rota Rod, Elevated Plus Maze, Open Field and Novel Object Recognition testing at PND 30-PND 36 and PND 44-PND 50. We show that HIE modeling in neonatal mice is associated with a significant endogenous drop in body core temperature by 2°C resulting in profound neuroprotection, expressed by reduced neuropathological injury scores, reduced loss of neurons, axonal structures, myelin and decreased astrogliosis. Immediately applied post-hypoxic HT revealed slight advantages over a delayed onset of therapy on short- and long-term histological outcome demonstrated by reduced neuropathological injury scores and preservation of hippocampal structures. However, depending on the brain region analyzed neuroprotective effects were similar or even reduced compared to protection by endogenous cooling during HIE modeling. Moreover, long-term neurobehavioral outcome was only partially improved for motoric function (i.e. Rota Rod performance and rearing activity) while cognitive deficits (i.e. novel object recognition) remained unchanged. These findings emphasize the need to maintain the nesting temperature during the initiation of the pathological insult and highlight the urgency to develop and assess new adjuvant therapies for HT in well-defined experimental models.


Subject(s)
Body Temperature/physiology , Brain Injuries/etiology , Brain Injuries/therapy , Hypothermia, Induced , Hypoxia-Ischemia, Brain/complications , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Brain/pathology , Brain Injuries/pathology , Disease Models, Animal , Locomotion/physiology , Maze Learning , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Neurons/pathology , Oligodendroglia/metabolism , Oligodendroglia/pathology , Time Factors
3.
Brain Behav Immun ; 52: 106-119, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26456693

ABSTRACT

Cerebral white matter injury is a leading cause of adverse neurodevelopmental outcome in prematurely born infants involving cognitive deficits in later life. Despite increasing knowledge about the pathophysiology of perinatal brain injury, therapeutic options are limited. In the adult demyelinating disease multiple sclerosis the sphingosine-1-phosphate (S1P) receptor modulating substance fingolimod (FTY720) has beneficial effects. Herein, we evaluated the neuroprotective potential of FTY720 in a neonatal model of oxygen-toxicity, which is associated with hypomyelination and impaired neuro-cognitive outcome. A single dose of FTY720 (1mg/kg) at the onset of neonatal hyperoxia (24h 80% oxygen on postnatal day 6) resulted in improvement of neuro-cognitive development persisting into adulthood. This was associated with reduced microstructural white matter abnormalities 4 months after the insult. In search of the underlying mechanisms potential non-classical (i.e. lymphocyte-independent) pathways were analysed shortly after the insult, comprising modulation of oxidative stress and local inflammatory responses as well as myelination, oligodendrocyte degeneration and maturation. Treatment with FTY720 reduced hyperoxia-induced oxidative stress, microglia activation and associated pro-inflammatory cytokine expression. In vivo and in vitro analyses further revealed that oxygen-induced hypomyelination is restored to control levels, which was accompanied by reduced oligodendrocyte degeneration and enhanced maturation. Furthermore, hyperoxia-induced elevation of S1P receptor 1 (S1P1) protein expression on in vitro cultured oligodendrocyte precursor cells was reduced by activated FTY720 and protection from degeneration is abrogated after selective S1P1 blockade. Finally, FTY720s' classical mode of action (i.e. retention of immune cells within peripheral lymphoid organs) was analysed demonstrating that FTY720 diminished circulating lymphocyte counts independent from hyperoxia. Cerebral immune cell counts remained unchanged by hyperoxia and by FTY720 treatment. Taken together, these results suggest that beneficial effects of FTY720 in neonatal oxygen-induced brain injury may be rather attributed to its anti-oxidative and anti-inflammatory capacity acting in concert with a direct protection of developing oligodendrocytes than to a modulation of peripheral lymphocyte trafficking. Thus, FTY720 might be a potential new therapeutic option for the treatment of neonatal brain injury through reduction of white matter damage.


Subject(s)
Cognition Disorders/prevention & control , Fingolimod Hydrochloride/therapeutic use , Hyperoxia/drug therapy , White Matter/drug effects , Animals , Animals, Newborn , Brain/metabolism , Cognition Disorders/metabolism , Cognition Disorders/pathology , Diffusion Magnetic Resonance Imaging , Female , Hyperoxia/pathology , Lysophospholipids/metabolism , Male , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Nerve Fibers, Myelinated/drug effects , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Oligodendroglia/pathology , Oxygen/administration & dosage , Pregnancy , Random Allocation , Rats , Rats, Wistar , Receptors, Lysosphingolipid/antagonists & inhibitors , Receptors, Lysosphingolipid/metabolism , Sphingosine/analogs & derivatives , Sphingosine/metabolism , White Matter/metabolism , White Matter/pathology
4.
Acta Neuropathol ; 130(4): 603, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26341346

ABSTRACT

Erratum to: Acta Neuropathol (2012) 123:273­284. DOI 10.1007/s00401­011­0914­z. The authors would like to correct Fig. 3 of the original manuscript, since the image in Fig. 3b does not correspond to a VEGF treated animal. Corrected Fig. 3 is shown below. We apologize for this mistake.

5.
PLoS One ; 7(11): e49023, 2012.
Article in English | MEDLINE | ID: mdl-23155446

ABSTRACT

Intrauterine infection and inflammation are major reasons for preterm birth. The switch from placenta-mediated to lung-mediated oxygen supply during birth is associated with a sudden rise of tissue oxygen tension that amounts to relative hyperoxia in preterm infants. Both infection/inflammation and hyperoxia have been shown to be involved in brain injury of preterm infants. Hypothesizing that they might be additive or synergistic, we investigated the influence of a systemic lipopolysaccharide (LPS) application on hyperoxia-induced white matter damage (WMD) in newborn rats. Three-day-old Wistar rat pups received 0.25 mg/kg LPS i.p. and were subjected to 80% oxygen on P6 for 24 h. The extent of WMD was assessed by immunohistochemistry, western blots, and diffusion tensor (DT) magnetic resonance imaging (MRI). In addition, the effects of LPS and hyperoxia were studied in an in vitro co-culture system of primary rat oligodendrocytes and microglia cells. Both noxious stimuli, hyperoxia, and LPS caused hypomyelination as revealed by western blot, immunohistochemistry, and altered WM microstructure on DT-MRI. Even so, cellular changes resulting in hypomyelination seem to be different. While hyperoxia induces cell death, LPS induces oligodendrocyte maturity arrest without cell death as revealed by TUNEL-staining and immunohistological maturation analysis. In the two-hit scenario cell death is reduced compared with hyperoxia treated animals, nevertheless white matter alterations persist. Concordantly with these in vivo findings we demonstrate that LPS pre-incubation reduced premyelinating-oligodendrocyte susceptibility towards hyperoxia in vitro. This protective effect might be caused by upregulation of interleukin-10 and superoxide dismutase expression after LPS stimulation. Reduced expression of transcription factors controlling oligodendrocyte development and maturation further indicates oligodendrocyte maturity arrest. The knowledge about mechanisms that triggered hypomyelination contributes to a better understanding of WMD in premature born infants.


Subject(s)
Brain/pathology , Hyperoxia/pathology , Inflammation/pathology , Leukoencephalopathies/pathology , Nerve Fibers, Myelinated/pathology , Animals , Animals, Newborn , Apoptosis/drug effects , Apoptosis/physiology , Brain/drug effects , Brain/metabolism , Caspase 3/metabolism , Cells, Cultured , Hyperoxia/metabolism , Inflammation/chemically induced , Inflammation/metabolism , Leukoencephalopathies/metabolism , Lipopolysaccharides/pharmacology , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Nerve Fibers, Myelinated/drug effects , Nerve Fibers, Myelinated/metabolism , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Oligodendroglia/pathology , Rats , Rats, Sprague-Dawley , Rats, Wistar
6.
Neurobiol Dis ; 45(3): 1077-85, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22198574

ABSTRACT

Vascular endothelial growth factor (VEGF) has potent angiogenic and neuroprotective effects in the ischemic brain. Its effect on axonal plasticity and neurological recovery in the post-acute stroke phase was unknown. Using behavioral tests combined with anterograde tract tracing studies and with immunohistochemical and molecular biological experiments, we examined effects of a delayed i.c.v. delivery of recombinant human VEGF(165), starting 3 days after stroke, on functional neurological recovery, corticorubral plasticity and inflammatory brain responses in mice submitted to 30 min of middle cerebral artery occlusion. We herein show that the slowly progressive functional improvements of motor grip strength and coordination, which are induced by VEGF, are accompanied by enhanced sprouting of contralesional corticorubral fibres that branched off the pyramidal tract in order to cross the midline and innervate the ipsilesional parvocellular red nucleus. Infiltrates of CD45+ leukocytes were noticed in the ischemic striatum of vehicle-treated mice that closely corresponded to areas exhibiting Iba-1+ activated microglia. VEGF attenuated the CD45+ leukocyte infiltrates at 14 but not 30 days post ischemia and diminished the microglial activation. Notably, the VEGF-induced anti-inflammatory effect of VEGF was associated with a downregulation of a broad set of inflammatory cytokines and chemokines in both brain hemispheres. These data suggest a link between VEGF's immunosuppressive and plasticity-promoting actions that may be important for successful brain remodeling. Accordingly, growth factors with anti-inflammatory action may be promising therapeutics in the post-acute stroke phase.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Cerebral Cortex/drug effects , Functional Laterality/drug effects , Infarction, Middle Cerebral Artery/complications , Neuronal Plasticity/drug effects , Red Nucleus/drug effects , Vascular Endothelial Growth Factor A/administration & dosage , Animals , Biotin/analogs & derivatives , Brain Edema/etiology , Brain Edema/prevention & control , Cerebral Cortex/pathology , Cerebral Infarction/etiology , Cerebral Infarction/prevention & control , Cytokines/metabolism , Dextrans , Disease Models, Animal , Gene Expression Regulation/drug effects , Hand Strength/physiology , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/pathology , Injections, Intraventricular/methods , Male , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Nerve Degeneration/etiology , Nerve Degeneration/prevention & control , Neural Pathways/drug effects , Organometallic Compounds , Organophosphorus Compounds , Red Nucleus/pathology
7.
Acta Neuropathol ; 123(2): 273-84, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22109109

ABSTRACT

Vascular endothelial growth factor (VEGF) is a potent angiogenic factor, which also has neuroprotective activity. In view of these dual actions on vessels and neurons, we were interested whether VEGF promotes long distance axonal plasticity in the ischemic brain. Herein, we show that VEGF promotes neurological stroke recovery in mice when delivered in a delayed way starting 3 days after middle cerebral artery occlusion. Using anterograde tract-tracing experiments that we combined with histochemical and molecular biological studies, we demonstrate that although VEGF promoted angiogenesis predominantly in the ischemic hemisphere, pronounced axonal sprouting was induced by VEGF in the contralesional, but not the ipsilesional corticobulbar system. Corticobulbar plasticity was accompanied by the deactivation of the matrix metalloproteinase MMP9 in the lesioned hemisphere and the transient downregulation of the axonal growth inhibitors NG2 proteoglycan and brevican and the guidance molecules ephrin B1/2 in the contralesional hemisphere. The regulation of matrix proteinases, growth inhibitors, and guidance molecules offers insights how brain plasticity is controlled in the ischemic brain.


Subject(s)
Brain Ischemia/physiopathology , Infarction, Middle Cerebral Artery/physiopathology , Neuronal Plasticity/physiology , Recovery of Function/physiology , Vascular Endothelial Growth Factor A/physiology , Animals , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Disease Models, Animal , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/metabolism , Male , Mice , Mice, Inbred C57BL , Motor Cortex/drug effects , Motor Cortex/pathology , Motor Cortex/physiology , Neuronal Plasticity/drug effects , Recovery of Function/drug effects , Vascular Endothelial Growth Factor A/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...