Network


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

Hotspot


Dive into the research topics where Christian Hölscher is active.

Publication


Featured researches published by Christian Hölscher.


EMBO Reports | 2006

Aβ42‐driven cerebral amyloidosis in transgenic mice reveals early and robust pathology

Rebecca Radde; Tristan Bolmont; Stephan A. Kaeser; Janaky Coomaraswamy; Dennis Lindau; Lars Stoltze; Michael E. Calhoun; Fabienne Jäggi; Hartwig Wolburg; Simon Gengler; Christian Haass; Bernardino Ghetti; Christian Czech; Christian Hölscher; Paul M. Mathews; Mathias Jucker

We have generated a novel transgenic mouse model on a C57BL/6J genetic background that coexpresses KM670/671NL mutated amyloid precursor protein and L166P mutated presenilin 1 under the control of a neuron‐specific Thy1 promoter element (APPPS1 mice). Cerebral amyloidosis starts at 6–8 weeks and the ratio of human amyloid (A)β42 to Aβ40 is 1.5 and 5 in pre‐depositing and amyloid‐depositing mice, respectively. Consistent with this ratio, extensive congophilic parenchymal amyloid but minimal amyloid angiopathy is observed. Amyloid‐associated pathologies include dystrophic synaptic boutons, hyperphosphorylated tau‐positive neuritic structures and robust gliosis, with neocortical microglia number increasing threefold from 1 to 8 months of age. Global neocortical neuron loss is not apparent up to 8 months of age, but local neuron loss in the dentate gyrus is observed. Because of the early onset of amyloid lesions, the defined genetic background of the model and the facile breeding characteristics, APPPS1 mice are well suited for studying therapeutic strategies and the pathomechanism of amyloidosis by cross‐breeding to other genetically engineered mouse models.


Journal of Clinical Investigation | 2012

An anti-diabetes agent protects the mouse brain from defective insulin signaling caused by Alzheimer’s disease–associated Aβ oligomers

Theresa R. Bomfim; Leticia Forny-Germano; Luciana B. Sathler; Jordano Brito-Moreira; Jean-Christophe Houzel; Helena Decker; Michael A. Silverman; Hala Kazi; Helen M. Melo; Paula L. McClean; Christian Hölscher; Steven E. Arnold; Konrad Talbot; William L. Klein; Douglas P. Munoz; Sergio T. Ferreira; Fernanda G. De Felice

Defective brain insulin signaling has been suggested to contribute to the cognitive deficits in patients with Alzheimers disease (AD). Although a connection between AD and diabetes has been suggested, a major unknown is the mechanism(s) by which insulin resistance in the brain arises in individuals with AD. Here, we show that serine phosphorylation of IRS-1 (IRS-1pSer) is common to both diseases. Brain tissue from humans with AD had elevated levels of IRS-1pSer and activated JNK, analogous to what occurs in peripheral tissue in patients with diabetes. We found that amyloid-β peptide (Aβ) oligomers, synaptotoxins that accumulate in the brains of AD patients, activated the JNK/TNF-α pathway, induced IRS-1 phosphorylation at multiple serine residues, and inhibited physiological IRS-1pTyr in mature cultured hippocampal neurons. Impaired IRS-1 signaling was also present in the hippocampi of Tg mice with a brain condition that models AD. Importantly, intracerebroventricular injection of Aβ oligomers triggered hippocampal IRS-1pSer and JNK activation in cynomolgus monkeys. The oligomer-induced neuronal pathologies observed in vitro, including impaired axonal transport, were prevented by exposure to exendin-4 (exenatide), an anti-diabetes agent. In Tg mice, exendin-4 decreased levels of hippocampal IRS-1pSer and activated JNK and improved behavioral measures of cognition. By establishing molecular links between the dysregulated insulin signaling in AD and diabetes, our results open avenues for the investigation of new therapeutics in AD.


The Journal of Neuroscience | 2011

The Diabetes Drug Liraglutide Prevents Degenerative Processes in a Mouse Model of Alzheimer's Disease

Paula L. McClean; Vadivel Parthsarathy; Emilie Faivre; Christian Hölscher

Type 2 diabetes is a risk factor for Alzheimers disease, most likely linked to an impairment of insulin signaling in the brain. The incretin hormone glucagon-like peptide-1 (GLP-1) facilitates insulin signaling, and novel long-lasting GLP-1 analogs, such as liraglutide, are on the market as diabetes therapeutics. GLP-1 has been shown to have neuroprotective properties in vitro and in vivo. Here we tested the effects of peripherally injected liraglutide in an Alzheimer mouse model, APPswe/PS1ΔE9 (APP/PS1). Liraglutide was shown to cross the blood–brain barrier in an acute study. Liraglutide was injected for 8 weeks at 25 nmol/kg body weight i.p. once daily in 7-month-old APP/PS1 and wild-type littermate controls. In APP/PS1 mice, liraglutide prevented memory impairments in object recognition and water maze tasks, and prevented synapse loss and deterioration of synaptic plasticity in the hippocampus, commonly observed in this model. Overall β-amyloid plaque count in the cortex and dense-core plaque numbers were reduced by 40–50%, while levels of soluble amyloid oligomers were reduced by 25%. The inflammation response as measured by activated microglia numbers was halved in liraglutide-treated APP/PS1 mice. Numbers of young neurons in the dentate gyrus were increased in APP/PS1 mice with treatment. Liraglutide treatment had little effect on littermate control mice, whose behavior was comparable to wild-type saline controls; however, synaptic plasticity was enhanced in the drug group. Our results show that liraglutide prevents key neurodegenerative developments found in Alzheimers disease, suggesting that GLP-1 analogs represent a novel treatment strategy for Alzheimers disease.


CNS Drugs | 2013

Intranasal insulin as a treatment for Alzheimer's disease: a review of basic research and clinical evidence.

Jessica Freiherr; Manfred Hallschmid; William H. Frey; Yvonne F. Brünner; Colin D. Chapman; Christian Hölscher; Suzanne Craft; Fernanda G. De Felice; Christian Benedict

Research in animals and humans has associated Alzheimer’s disease (AD) with decreased cerebrospinal fluid levels of insulin in combination with decreased insulin sensitivity (insulin resistance) in the brain. This phenomenon is accompanied by attenuated receptor expression of insulin and insulin-like growth factor, enhanced serine phosphorylation of insulin receptor substrate-1, and impaired transport of insulin across the blood-brain barrier. Moreover, clinical trials have demonstrated that intranasal insulin improves both memory performance and metabolic integrity of the brain in patients suffering from AD or its prodrome, mild cognitive impairment. These results, in conjunction with the finding that insulin mitigates hippocampal synapse vulnerability to beta amyloid, a peptide thought to be causative in the development of AD, provide a strong rationale for hypothesizing that pharmacological strategies bolstering brain insulin signaling, such as intranasal administration of insulin, could have significant potential in the treatment and prevention of AD. With this view in mind, the review at hand will present molecular mechanisms potentially underlying the memory-enhancing and neuroprotective effects of intranasal insulin. Then, we will discuss the results of intranasal insulin studies that have demonstrated that enhancing brain insulin signaling improves memory and learning processes in both cognitively healthy and impaired humans. Finally, we will provide an overview of neuroimaging studies indicating that disturbances in insulin metabolism—such as insulin resistance in obesity, type 2 diabetes and AD—and altered brain responses to insulin are linked to decreased cerebral volume and especially to hippocampal atrophy.


BMC Neuroscience | 2012

Drugs developed to treat diabetes, liraglutide and lixisenatide, cross the blood brain barrier and enhance neurogenesis

Kerry Hunter; Christian Hölscher

BackgroundType 2 diabetes is a risk factor for Alzheimers disease (AD), most likely linked to an impairment of insulin signalling in the brain. Therefore, drugs that enhance insulin signalling may have therapeutic potential for AD. Liraglutide (Victoza) and exenatide (Byetta) are novel long-lasting analogues of the GLP-1 incretin hormone and are currently available to treat diabetes. They facilitate insulin signalling via the GLP-1 receptor (GLP-1R). Numerous in vitro and in vivo studies have shown that GLP-1 analogues have a range of neuroprotective properties. GLP-1Rs are expressed in the hippocampal area of the brain an important site of adult neurogenesis and maintenance of cognition and memory formation. Therefore, if GLP-1 analogues can cross the blood brain barrier, diffuse through the brain to reach the receptors and most importantly activate them, their neuroprotective effects may be realized.ResultsIn the present study we profiled the GLP-1 receptor agonists liraglutide (Victoza) and lixisenatide (Lyxumia). We measured the kinetics of crossing the blood brain barrier (BBB), activation of the GLP-1R by measuring cAMP levels, and physiological effects in the brain on neuronal stem cell proliferation and neurogenesis. Both drugs were able to cross the BBB. Lixisenatide crossed the BBB at all doses tested (2.5, 25, or 250 nmol/kg bw ip.) when measured 30 min post-injection and at 2.5-25 nmol/kg bw ip. 3 h post-injection. Lixisenatide also enhanced neurogenesis in the brain. Liraglutide crossed the BBB at 25 and 250 nmol/kg ip. but no increase was detectable at 2.5 nmol/kg ip. 30 min post-injection, and at 250 nmol/kg ip. at 3 h post-injection. Liraglutide and lixisenatide enhanced cAMP levels in the brain, with lixisenatide being more effective.ConclusionsOur results suggest that these novel incretin analogues cross the BBB and show physiological activity and neurogenesis in the brain, which may be of use as a treatment of neurodegenerative diseases.


Behavioural Brain Research | 1999

Stress impairs performance in spatial water maze learning tasks.

Christian Hölscher

The water maze task has been developed to test spatial learning abilities in rats or mice, and is widely used. Though it has been reported before that numerous cognitive abilities are of importance for learning this task, poor performance is usually interpreted as an impairment of spatial memory formation. Previous investigations that tried to correlate long-term potentiation (LTP) of synaptic transmission with spatial learning abilities in rats reported that injection of drugs or specific gene deletions which blocked the expression of LTP correlated with learning impairments of spatial tasks in a water maze. Recent studies, however, have shown that pretraining enables these animals to learn such spatial tasks even though LTP was still found to be blocked. I investigated to what degree altered fear condition and stress perception could account for the impaired spatial learning when no pretraining is given. In a fear habituation task, unhandled rats preferred a dark over a well lit chamber more than handled animals did, but unhandled rats favoured the lit chamber more in an active avoidance task. They also performed poorly in a spatial water maze task compared with handled rats. Rats pretrained in a radial arm maze performed better in a water maze than non-pretrained rats. No difference between groups was found in a non-spatial water maze task. On the other hand, when pretrained in a water maze, rats performed only marginally better in a radial arm maze compared to non-pretrained animals. Since animals have to be handled to learn a radial arm maze, the difference in this task was not due to stress but most probably due to getting accustomed to the room dimensions prior to learning the spatial task. The results suggest that impaired learning of spatial tasks in the water maze can be due to increased stress and decreased fear conditioning without actually affecting spatial learning abilities. These results question the interpretations of the results of some previously published results of spatial water maze tasks.


Behavioural Brain Research | 2009

Impairment of synaptic plasticity and memory formation in GLP-1 receptor KO mice: Interaction between type 2 diabetes and Alzheimer's disease

Talib Abbas; Emilie Faivre; Christian Hölscher

Type 2 diabetes has been identified as a risk factor for Alzheimer disease (AD). Insulin signalling is often impaired in AD, contributing to the neurodegeneration seen in AD. Previous studies have shown that the incretin glucagon-like peptide 1 (GLP-1) helps to normalise insulin signalling in type 2 diabetes. GLP-1 also plays important roles in neuronal activity and brain functions. We tested the specific role of GLP-1 receptors in synaptic plasticity and cognitive processes in a GLP-1 receptor knockout (Glp1r(-/-)) mouse model. In an open field assessment, no general difference in exploratory and anxiety was found except for a small decrease in running speed was found (p<0.05). In an object recognition task, Glp1r(-/-) mice explored objects in a similar way to WT controls but did not learn to differentiate between novel and familiar objects (p<0.05) while in an object relocation task, no impairment was observed. In a water maze task, Glp1r(-/-) mice were impaired in the acquisition phase (p<0.001), and also in the probe recall task (p<0.05). LTP in area CA1 of the hippocampus was severely impaired in Glp1r(-/-) mice (p<0.0001). Paired-pulse facilitation was also impaired at 25ms interstimulus interval (p<0.05) but not at longer intervals. The results demonstrate that the murine GLP-1R plays an important role in the control of synaptic plasticity and in some forms of memory formation. The results shed light on the molecular processes that underlie the neuroprotective properties of GLP-1 analogues in animal models of Alzheimers disease.


Neuroreport | 2009

Receptors for the incretin glucagon-like peptide-1 are expressed on neurons in the central nervous system

Alison Hamilton; Christian Hölscher

Glucagon-like-peptide-1 is an incretin hormone that also has neuroprotective properties. Here we analyse where glucagon-like-peptide-1receptors are expressed in the brain. The receptor is found only on neurons, not on glia cells. The pyramidal cell layer of the CA region and the granule cell layer of the dentate gyrus in the hippocampus show intense staining. In the neocortex, larger pyramidal neurons express the receptor. In the cerebellum, only Purkinje neurons express the receptor. Dendrites of larger neurons were stained; in particular, pyramidal cells in area CA and dendrites of Purkinje cells. The fact that the receptor is located on neurons and dendrites suggests that the neuroprotective action is caused by the modulation of neuronal excitation.


Neurobiology of Aging | 2012

Val(8)GLP-1 rescues synaptic plasticity and reduces dense core plaques in APP/PS1 mice

Simon Gengler; Paula L. McClean; Ruth McCurtin; Victor Gault; Christian Hölscher

Diabetes is a risk factor for Alzheimers disease. We tested the effects of Val(8)GLP-1, an enzyme-resistant analogue of the incretin hormone glucagon-like peptide 1 originally developed to treat diabetes in a mouse model of Alzheimers disease that expresses mutated amyloid precursor protein (APP) and presenilin-1. We tested long term potentiation (LTP) of synaptic plasticity, inflammation response, and plaque formation. Val(8)GLP-1 crosses the blood-brain barrier when administered via intraperitoneal injection. Val(8)GLP-1 protected LTP in 9- and 18-month-old Alzheimers disease mice when given for 3 weeks at 25 nmol/kg intraperitoneally. LTP was also enhanced in 18-month-old wild type mice, indicating that Val(8)GLP-1 also ameliorates age-related synaptic degenerative processes. Paired-pulse facilitation was also enhanced. The number of beta-amyloid plaques and microglia activation in the cortex increased with age but was not reduced by Val(8)GLP-1. In 18-month-old mice, however, the number of Congo red positive dense-core amyloid plaques was reduced. Treatment with Val(8)GLP-1 might prevent or delay neurodegenerative processes.


Journal of Neuroscience Research | 2011

Novel GLP-1 Mimetics Developed To Treat Type 2 Diabetes Promote Progenitor Cell Proliferation in the Brain

Alison Hamilton; Steven Patterson; David W. Porter; Victor Gault; Christian Hölscher

One of the symptoms of diabetes is the progressive development of neuropathies. One mechanism to replace neurons in the CNS is through the activation of stem cells and neuronal progenitor cells. We have tested the effects of the novel GLP‐1 mimetics exenatide (exendin‐4; Byetta) and liraglutide (NN2211; Victoza), which are already on the market as treatments for type 2 diabetes, on the proliferation rate of progenitor cells and differentiation into neurons in the dentate gyrus of brains of mouse models of diabetes. GLP‐1 analogues were injected subcutaneously for 4, 6, or 10 weeks once daily in three mouse models of diabetes: ob/ob mice, db/db mice, or high‐fat‐diet‐fed mice. Twenty‐four hours before perfusion, animals were injected with 5′‐bromo‐2′‐deoxyuridine (BrdU) to mark dividing progenitor cells. By using immunohistochemistry and stereological methods, the number of progenitor cells or doublecortin‐positive young neurons in the dentate gyrus was estimated. We found that, in all three mouse models, progenitor cell division was enhanced compared with nondiabetic controls after chronic i.p. injection of either liraglutide or exendin‐4 by 100–150% (P < 0.001). We also found an increase in young neurons in the DG of high‐fat‐diet‐fed mice after drug treatment (P < 0.001). The GLP‐1 receptor antagonist exendin(9–36) reduced progenitor cell proliferation in these mice. The results demonstrate that GLP‐1 mimetics show promise as a treatment for neurodegenerative diseases such as Alzheimers disease, because these novel drugs cross the blood–brain barrier and increase neuroneogenesis.

Collaboration


Dive into the Christian Hölscher's collaboration.

Top Co-Authors

Avatar

Lin Li

Shanxi Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Brian D. Green

Queen's University Belfast

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jin-Shun Qi

Shanxi Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Guanglai Li

Shanxi Medical University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge