Network


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

Hotspot


Dive into the research topics where Casper Hempel is active.

Publication


Featured researches published by Casper Hempel.


Malaria Journal | 2008

Recombinant human erythropoietin increases survival and reduces neuronal apoptosis in a murine model of cerebral malaria.

Lothar Wiese; Casper Hempel; Milena Penkowa; Nikolai Kirkby; Jørgen A. L. Kurtzhals

BackgroundCerebral malaria (CM) is an acute encephalopathy with increased pro-inflammatory cytokines, sequestration of parasitized erythrocytes and localized ischaemia. In children CM induces cognitive impairment in about 10% of the survivors. Erythropoietin (Epo) has – besides of its well known haematopoietic properties – significant anti-inflammatory, antioxidant and anti-apoptotic effects in various brain disorders. The neurobiological responses to exogenously injected Epo during murine CM were examined.MethodsFemale C57BL/6j mice (4–6 weeks), infected with Plasmodium berghei ANKA, were treated with recombinant human Epo (rhEpo; 50–5000 U/kg/OD, i.p.) at different time points. The effect on survival was measured. Brain pathology was investigated by TUNEL (Terminal deoxynucleotidyl transferase (TdT)-mediated deoxyuridine triphosphate (dUTP)-digoxigenin nick end labelling), as a marker of apoptosis. Gene expression in brain tissue was measured by real time PCR.ResultsTreatment with rhEpo increased survival in mice with CM in a dose- and time-dependent manner and reduced apoptotic cell death of neurons as well as the expression of pro-inflammatory cytokines in the brain. This neuroprotective effect appeared to be independent of the haematopoietic effect.ConclusionThese results and its excellent safety profile in humans makes rhEpo a potential candidate for adjunct treatment of CM.


Biofactors | 2009

Metallothionein-I+II in neuroprotection

Mie Østergaard Pedersen; Rikke Beck Jensen; Dan Sonne Pedersen; Anders Daehli Skjolding; Casper Hempel; Lasse Maretty; Milena Penkowa

Metallothionein (MT)‐I+II synthesis is induced in the central nervous system (CNS) in response to practically any pathogen or disorder, where it is increased mainly in reactive glia. MT‐I+II are involved in host defence reactions and neuroprotection during neuropathological conditions, in which MT‐I+II decrease inflammation and secondary tissue damage (oxidative stress, neurodegeneration, and apoptosis) and promote post‐injury repair and regeneration (angiogenesis, neurogenesis, neuronal sprouting and tissue remodelling). Intracellularly the molecular MT‐I+II actions involve metal ion control and scavenging of reactive oxygen species (ROS) leading to cellular redox control. By regulating metal ions, MT‐I+II can control metal‐containing transcription factors, zinc‐finger proteins and p53. However, the neuroprotective functions of MT‐I+II also involve an extracellular component. MT‐I+II protects the neurons by signal transduction through the low‐density lipoprotein family of receptors on the cell surface involving lipoprotein receptor‐1 (LRP1) and megalin (LRP2). In this review we discuss the newest data on cerebral MT‐I+II functions following brain injury and experimental autoimmune encephalomyelitis.


Glia | 2010

In vivo expression of neuroglobin in reactive astrocytes during neuropathology in murine models of traumatic brain injury, cerebral malaria, and autoimmune encephalitis

Brian DellaValle; Casper Hempel; Jørgen A. L. Kurtzhals; Milena Penkowa

Neuroglobin (Ngb) is proposed to be a neuron‐specific, hypoxia‐responsive, neuroprotective protein. However, results are conflicting concerning both Ngbs physiological and pathological significance. This study was designed to investigate the in vivo localization and regulation of Ngb in different neuropathological models representing traumatic injury, infectious, autoimmune, and excitotoxic pathogeneses. We profiled Ngb immunohistochemistry in murine models of traumatic brain injury, cerebral malaria, experimental autoimmune encephalitis, and kainic acid (KA)‐mediated epileptic seizures that, to our knowledge, have not been studied in the context of Ngb. In control mice Ngb was expressed exclusively in neurons. In all pathological models except KA, in addition to neurons Ngb was present in reactive astrocytes. Ngb positive astrocytes were found within regions associated with most severe pathology and the astroglial scar. This is the first report of Ngb present in reactive astroglia and in scar‐forming astrocytes in response to different pathological conditions relevant to human disease. In light of previously reported cyto‐protective properties of Ngb, further insight may result in therapeutic ramifications.


Fems Immunology and Medical Microbiology | 2014

Formation of hydroxyl radicals contributes to the bactericidal activity of ciprofloxacin against Pseudomonas aeruginosa biofilms.

Peter Østrup Jensen; Alejandra Briales; Rikke Prejh Brochmann; Hengzhuang Wang; Kasper Nørskov Kragh; Mette Kolpen; Casper Hempel; Thomas Bjarnsholt; Niels Høiby; Oana Ciofu

Antibiotic-tolerant, biofilm-forming Pseudomonas aeruginosa has long been recognized as a major cause of chronic lung infections of cystic fibrosis patients. The mechanisms involved in the activity of antibiotics on biofilm are not completely clear. We have investigated whether the proposed induction of cytotoxic hydroxyl radicals (OH˙) during antibiotic treatment of planktonically grown cells may contribute to action of the commonly used antibiotic ciprofloxacin on P. aeruginosa biofilms. For this purpose, WT PAO1, a catalase deficient ΔkatA and a ciprofloxacin resistant mutant of PAO1 (gyrA), were grown as biofilms in microtiter plates and treated with ciprofloxacin. Formation of OH˙ and total amount of reactive oxygen species (ROS) was measured and viability was estimated. Formation of OH˙ and total ROS in PAO1 biofilms treated with ciprofloxacin was shown but higher levels were measured in ΔkatA biofilms, and no ROS production was seen in the gyrA biofilms. Treatment with ciprofloxacin decreased the viability of PAO1 and ΔkatA biofilms but not of gyrA biofilms. Addition of thiourea, a OH˙ scavenger, decreased the OH˙ levels and killing of PAO1 biofilm. Our study shows that OH˙ is produced by P. aeruginosa biofilms treated with ciprofloxacin, which may contribute to the killing of biofilm subpopulations.


Clinical and Experimental Immunology | 2014

Nitric oxide production by polymorphonuclear leucocytes in infected cystic fibrosis sputum consumes oxygen

Mette Kolpen; Thomas Bjarnsholt; C.R. Hansen; L. F. Rickelt; Michael Kühl; Casper Hempel; T. Pressler; Niels Høiby; Peter Østrup Jensen

Chronic Pseudomonas aeruginosa lung infection in cystic fibrosis (CF) patients is characterized by persisting mucoid biofilms in hypoxic endobronchial mucus. These biofilms are surrounded by numerous polymorphonuclear leucocytes (PMNs), which consume a major part of present molecular oxygen (O2) due to production of superoxide (O2−). In this study, we show that the PMNs also consume O2 for production of nitric oxide (NO) by the nitric oxide synthases (NOS) in the infected endobronchial mucus. Fresh expectorated sputum samples (n = 28) from chronically infected CF patients (n = 22) were analysed by quantifying and visualizing the NO production. NO production was detected by optode measurements combined with fluorescence microscopy, flow cytometry and spectrophotometry. Inhibition of nitric oxide synthases (NOS) with NG‐monomethyl‐L‐arginine (L‐NMMA) resulted in reduced O2 consumption (P < 0·0008, n = 8) and a lower fraction of cells with fluorescence from the NO‐indicator 4‐amino‐5‐methylamino‐2′,7′‐difluorofluorescein diacetate (DAF‐FM) (P < 0·002, n = 8). PMNs stained with DAF‐FM and the superoxide indicator hydroethidine (HE) and host cells with inducible NOS (iNOS) were identified in the sputum. In addition, the production of the stable end‐products of NO in CF sputum was correlated with the concentration of PMNs; NO3− (P < 0·04, r = 0·66, n = 10) and NO2− (P< 0·006, r = 0·78, n = 11). The present study suggests that besides consumption of O2 for production of reactive oxygen species, the PMNs in CF sputum also consume O2 for production of NO.


Annals of clinical and translational neurology | 2014

GLP-1 improves neuropathology after murine cold lesion brain trauma

Brian DellaValle; Casper Hempel; Flemming Fryd Johansen; Jørgen A. L. Kurtzhals

In this study, we address a gap in knowledge regarding the therapeutic potential of acute treatment with a glucagon‐like peptide‐1 (GLP‐1) receptor agonist after severe brain trauma. Moreover, it remains still unknown whether GLP‐1 treatment activates the protective, anti‐neurodegenerative cAMP response element binding protein (CREB) pathway in the brain in vivo, and whether activation leads to observable increases in protective, anti‐neurodegenerative proteins. Finally, we report the first use of a highly sensitive in vivo imaging agent to assess reactive species generation after brain trauma.


The International Journal of Biochemistry & Cell Biology | 2013

Brain mitochondrial function in a murine model of cerebral malaria and the therapeutic effects of rhEPO.

Michael Karlsson; Casper Hempel; Fredrik Sjövall; M. Hansson; Jørgen A. L. Kurtzhals; Eskil Elmér

Cerebral malaria (CM) is a life-threatening complication of Plasmodium falciparum infection. The pathogenesis of CM is complex. Cerebral metabolic dysfunction is implicated in CM, which may be caused by both an impaired cerebral microcirculation and a dysregulated inflammatory response affecting cellular respiration of mitochondria. Recombinant human erythropoietin (rhEPO) is a promising new therapy that has been shown to reduce mortality in a mouse model of CM. In order to further elucidate the metabolic dysfunction in CM the objective of the present study was to assess brain mitochondrial respiratory function in CM with and without rhEPO treatment. The P. berghei ANKA - C57BL/6 murine model of CM was used. Mitochondrial respiration was analyzed in brain homogenates using high-resolution respirometry and a multiple substrate and inhibitor protocol. The animals were divided into four groups; infected injected with saline or with rhEPO, non-infected injected with saline or with rhEPO. Infected mice developed CM and treatment with rhEPO attenuated clinical signs of disease. There were no differences in respiratory parameters of brain mitochondria between infected and non-infected mice and no connection between disease severity and mitochondrial respiratory function. Treatment with rhEPO similarly had no effect on respiratory function. Thus cerebral metabolic dysfunction in CM does not seem to be directly linked to altered mitochondrial respiratory capacity as analyzed in brain homogenates ex vivo. This article is part of a Directed Issue entitled: Bioenergetic dysfunction, adaptation and therapy.


Experimental Parasitology | 2011

Plasmodium berghei ANKA: erythropoietin activates neural stem cells in an experimental cerebral malaria model.

Andrew Core; Casper Hempel; Jørgen A. L. Kurtzhals; Milena Penkowa

Cerebral malaria (CM) causes substantial mortality and neurological sequelae in survivors, and no neuroprotective regimens are currently available for this condition. Erythropoietin (EPO) reduces neuropathology and improves survival in murine CM. Using the Plasmodium berghei model of CM, we investigated if EPOs neuroprotective effects include activation of endogenous neural stem cells (NSC). By using immunohistochemical markers of different NSC maturation stages, we show that EPO increased the number of nestin(+) cells in the dentate gyrus and in the sub-ventricular zone of the lateral ventricles, relative to control-treatment. 75% of the EPO-treated CM mice displayed migration as nestin(+) NSC. The NSC showed differentiation towards a neural cell lineage as shown by PSA-NCAM binding and NSC maturation and lineage commitment was significantly affected by exogenous EPO and by CM in the sub ventricular zone. These results indicate a rapid, EPO-dependent activation of NSC during CM pathology.


Frontiers in Immunology | 2014

Systemic and Cerebral Vascular Endothelial Growth Factor Levels Increase in Murine Cerebral Malaria along with Increased Calpain and Caspase Activity and Can be Reduced by Erythropoietin Treatment

Casper Hempel; Nils Hoyer; Anna Overgaard Kildemoes; Charlotte Bille Jendresen; Jørgen Anders Lindholm Kurtzhals

The pathogenesis of cerebral malaria (CM) includes compromised microvascular perfusion, increased inflammation, cytoadhesion, and endothelial activation. These events cause blood–brain barrier disruption and neuropathology and associations with the vascular endothelial growth factor (VEGF) signaling pathway have been shown. We studied this pathway in mice infected with Plasmodium berghei ANKA causing murine CM with or without the use of erythropoietin (EPO) as adjunct therapy. ELISA and western blotting was used for quantification of VEGF and relevant proteins in brain and plasma. CM increased levels of VEGF in brain and plasma and decreased plasma levels of soluble VEGF receptor 2. EPO treatment normalized VEGF receptor 2 levels and reduced brain VEGF levels. Hypoxia-inducible factor (HIF)-1α was significantly upregulated whereas cerebral HIF-2α and EPO levels remained unchanged. Furthermore, we noticed increased caspase-3 and calpain activity in terminally ill mice, as measured by protease-specific cleavage of α-spectrin and p35. In conclusion, we detected increased cerebral and systemic VEGF as well as HIF-1α, which in the brain were reduced to normal in EPO-treated mice. Also caspase and calpain activity was reduced markedly in EPO-treated mice.


PLOS ONE | 2013

Investigation of Hydrogen Sulfide Gas as a Treatment against P. falciparum, Murine Cerebral Malaria, and the Importance of Thiolation State in the Development of Cerebral Malaria

Brian DellaValle; Trine Staalsoe; Jørgen A. L. Kurtzhals; Casper Hempel

Introduction Cerebral malaria (CM) is a potentially fatal cerebrovascular disease of complex pathogenesis caused by Plasmodium falciparum. Hydrogen sulfide (HS) is a physiological gas, similar to nitric oxide and carbon monoxide, involved in cellular metabolism, vascular tension, inflammation, and cell death. HS treatment has shown promising results as a therapy for cardio- and neuro- pathology. This study investigates the effects of fast (NaHS) and slow (GYY4137) HS-releasing drugs on the growth and metabolism of P. falciparum and the development of P. berghei ANKA CM. Moreover, we investigate the role of free plasma thiols and cell surface thiols in the pathogenesis of CM. Methods P. falciparum was cultured in vitro with varying doses of HS releasing drugs compared with artesunate. Growth and metabolism were quantified. C57Bl/6 mice were infected with P. berghei ANKA and were treated with varying doses and regimes of HS-releasing drugs. Free plasma thiols and cell surface thiols were quantified in CM mice and age-matched healthy controls. Results HS-releasing drugs significantly and dose-dependently inhibited P. falciparum growth and metabolism. Treatment of CM did not affect P. berghei growth, or development of CM. Interestingly, CM was associated with lower free plasma thiols, reduced leukocyte+erythrocyte cell surface thiols (infection day 3), and markedly (5-fold) increased platelet cell surface thiols (infection day 7). Conclusions HS inhibits P. falciparum growth and metabolism in vitro. Reduction in free plasma thiols, cell surface thiols and a marked increase in platelet cell surface thiols are associated with development of CM. HS drugs were not effective in vivo against murine CM.

Collaboration


Dive into the Casper Hempel's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Brian DellaValle

Copenhagen University Hospital

View shared research outputs
Top Co-Authors

Avatar

Lasse Maretty

Copenhagen University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Milena Penkowa

University of Copenhagen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tom Vosch

University of Copenhagen

View shared research outputs
Top Co-Authors

Avatar

Mette Kolpen

University of Copenhagen

View shared research outputs
Top Co-Authors

Avatar

Niels Høiby

University of Copenhagen

View shared research outputs
Researchain Logo
Decentralizing Knowledge