Network


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

Hotspot


Dive into the research topics where Charles Pyke is active.

Publication


Featured researches published by Charles Pyke.


Apmis | 1999

Cancer invasion and tissue remodeling : cooperation of protease systems and cell types

Keld Danø; John Rømer; Boye Schnack Nielsen; Signe F.Bjørn; Charles Pyke; Jørgen Rygaard; Leif R. Lund

Proteolytic degradation of the extracellular matrix plays a crucial role in both cancer invasion and non‐neoplastic tissue remodeling processes. In human cancers the components of matrix degrading protease systems (uPA, uPAR, PAI‐1 and MMPs) can be expressed by either the non‐neoplastic stromal cells, the cancer cells or both. Studies of the prognostic impact of these components in human cancer and the effect of targeted gene inactivation on cancer metastasis in mice support the assumption that proteases promote cancer progression, independent of whether they are expressed by cancer cells or stromal cells. The pattern of expression of components of protease systems is usually very similar in different cases of the same type of cancer, while it varies between different types of cancer. There are intriguing similarities between the cellular expression pattern of components of protease systems seen in cancer invasion and in certain types of non‐neoplastic tissue remodeling. We propose that cancer invasion can be viewed as tissue remodeling gone out of control. The stromal cell involvement in cancer invasion represents a new paradigm with important implications for cancer pathophysiology and cancer therapy.


Endocrinology | 2014

GLP-1 Receptor Localization in Monkey and Human Tissue: Novel Distribution Revealed With Extensively Validated Monoclonal Antibody

Charles Pyke; R. Scott Heller; Rikke Kaae Kirk; Cathrine Ørskov; Steffen Reedtz-Runge; Peter Kaastrup; Anders Hvelplund; Linda Bardram; Dan Calatayud; Lotte Bjerre Knudsen

Glucagon-like peptide 1 (GLP-1) analogs are increasingly being used in the treatment of type 2 diabetes. It is clear that these drugs lower blood glucose through an increase in insulin secretion and a lowering of glucagon secretion; in addition, they lower body weight and systolic blood pressure and increase heart rate. Using a new monoclonal antibody for immunohistochemistry, we detected GLP-1 receptor (GLP-1R) in important target organs in humans and monkeys. In the pancreas, GLP-1R was predominantly localized in β-cells with a markedly weaker expression in acinar cells. Pancreatic ductal epithelial cells did not express GLP-1R. In the kidney and lung, GLP-1R was exclusively expressed in smooth muscle cells in the walls of arteries and arterioles. In the heart, GLP-1R was localized in myocytes of the sinoatrial node. In the gastrointestinal tract, the highest GLP-1R expression was seen in the Brunners gland in the duodenum, with lower level expression in parietal cells and smooth muscle cells in the muscularis externa in the stomach and in myenteric plexus neurons throughout the gut. No GLP-1R was seen in primate liver and thyroid. GLP-1R expression seen with immunohistochemistry was confirmed by functional expression using in situ ligand binding with (125)I-GLP-1. In conclusion, these results give important new insight into the molecular mode of action of GLP-1 analogs by identifying the exact cellular localization of GLP-1R.


Journal of Clinical Investigation | 2014

The arcuate nucleus mediates GLP-1 receptor agonist liraglutide-dependent weight loss

Anna Secher; Jacob Jelsing; Arian F. Baquero; Jacob Hecksher-Sørensen; Michael A. Cowley; Louise S. Dalbøge; Gitte Hansen; Kevin L. Grove; Charles Pyke; Kirsten Raun; Lauge Schäffer; Mads Tang-Christensen; Saurabh Verma; Brent M. Witgen; Niels Vrang; Lotte Bjerre Knudsen

Liraglutide is a glucagon-like peptide-1 (GLP-1) analog marketed for the treatment of type 2 diabetes. Besides lowering blood glucose, liraglutide also reduces body weight. It is not fully understood how liraglutide induces weight loss or to what degree liraglutide acts directly in the brain. Here, we determined that liraglutide does not activate GLP-1-producing neurons in the hindbrain, and liraglutide-dependent body weight reduction in rats was independent of GLP-1 receptors (GLP-1Rs) in the vagus nerve, area postrema, and paraventricular nucleus. Peripheral injection of fluorescently labeled liraglutide in mice revealed the presence of the drug in the circumventricular organs. Moreover, labeled liraglutide bound neurons within the arcuate nucleus (ARC) and other discrete sites in the hypothalamus. GLP-1R was necessary for liraglutide uptake in the brain, as liraglutide binding was not seen in Glp1r(-/-) mice. In the ARC, liraglutide was internalized in neurons expressing proopiomelanocortin (POMC) and cocaine- and amphetamine-regulated transcript (CART). Electrophysiological measurements of murine brain slices revealed that GLP-1 directly stimulates POMC/CART neurons and indirectly inhibits neurotransmission in neurons expressing neuropeptide Y (NPY) and agouti-related peptide (AgRP) via GABA-dependent signaling. Collectively, our findings indicate that the GLP-1R on POMC/CART-expressing ARC neurons likely mediates liraglutide-induced weight loss.


American Journal of Pathology | 2000

Hypoxia-Inducible Angiopoietin-2 Expression Is Mimicked by Iodonium Compounds and Occurs in the Rat Brain and Skin in Response to Systemic Hypoxia and Tissue Ischemia

Stefano J. Mandriota; Charles Pyke; Corinne Di Sanza; Pierre Dominique Quinodoz; Brigitte Pittet; Michael S. Pepper

Angiopoietins are ligands for the endothelial cell tyrosine kinase receptor Tie-2. Ang-1, the major physiological activator of Tie-2, promotes blood vessel maturation and stability. Ang-2 counteracts this effect by competitively inhibiting the binding of Ang-1 to Tie-2. Using a combined RNase protection/semiquantitative reverse transcriptase-polymerase chain reaction approach, we demonstrate that hypoxia up-regulates Ang-2 mRNA levels by up to 3.3-fold in two human endothelial cell lines. In bovine microvascular endothelial (BME) cells, the flavoprotein oxidoreductase inhibitor diphenylene iodonium (DPI) and the related compound iodonium diphenyl mimic induction of Ang-2 but not vascular endothelial growth factor (VEGF) by hypoxia; in combination with hypoxia, DPI further increases Ang-2 expression but has no effect on the induction of VEGF by hypoxia. Neither Ang-2 or VEGF was increased by cyanide or rotenone, suggesting that failure in mitochondrial electron transport is not involved in the oxygen-sensing system that controls their expression. In ischemic rat dorsal skin flaps or in the brain of rats maintained for 12 hours under conditions of hypoxia, Ang-2 mRNA was up-regulated 7.5- or 17.6- fold, respectively. VEGF was concomitantly increased, whereas expression of Ang-1, Tie-2, and the related receptor Tie-1 was unaltered. In situ hybridization localized Ang-2 mRNA to endothelial cells in hypoxic skin. These findings 1) show that up-regulation of Ang-2 by hypoxia occurs widely in endothelial cells in vitro and in vivo; 2) suggest that induction of Ang-2, but not VEGF, by hypoxia in BME cells is controlled by a flavoprotein oxidoreductase that is sensitive to iodonium compounds; and 3) point to Ang-2 and VEGF as independently regulated and selective effectors of hypoxia-induced vascular sprouting.


Endocrinology | 2013

The Glucagon-Like Peptide-1 Receptor—or Not?

Charles Pyke; Lotte Bjerre Knudsen

Glucagon-like peptide-1 (GLP-1)-based diabetes therapy is increasingly used in the treatment of type 2 diabetes. Two GLP-1 analogs, liraglutide and exenatide, have been approved for clinical use and act as agonists of the GLP-1 receptor (GLP-1R), a member of the glucagon receptor family of G protein-coupled receptors (GPCRs) (1). Also, the clinical effect of DiPeptidyl Peptidase-4 (DPP-4) inhibitors is at least partially resulting from an increased level of active GLP-1 (2). The unambiguous identification of GLP-1R-expressing cells in humans and animal species constitutes critically important knowledge for fully understanding the pharmacological effects of GLP-1R agonists, both those associated with the documented clinical benefits as well as any potential undesired effects of treatment. The GLP-1R was cloned in 1992 (3). Preceding that, the receptor was identified in numerous cell types and organs, most obviously the pancreatic -cell but also in rat lung and brain by studies using either ligand binding or functional assays (4, 5). After the receptor was cloned, RT-PCR and in situ hybridization were used to report tissue distribution, and the receptor was located in the intestine, stomach, kidney, lungs, heart, and brain (6, 7). Later studies have used Western blotting and immunohistochemistry to identify the exact cellular localization of the receptor. Using such techniques, the receptor has been identified in mouse cardiomyocytes and smooth muscle cells in the heart, and in porcine tubular cells in the kidney (8, 9). Human GLP-1R expression has been mapped by in situ ligand binding across a wide range of both normal and tumor tissues (10). In this issue of Endocrinology, Panjwani et al. (11) reports new knowledge about the mechanism of action of GLP-1 analogs in atherosclerosis and hepatic steatosis and challenges some GLP-1R expression data in macrophages and hepatocytes that have previously been published. When Panjwani et al. (11) could not identify GLP-1R mRNA on macrophages or isolated hepatocytes despite previous reports of such localization, they did a careful Western blotting characterization of three commercially available polyclonal antibodies (PAbs) against the GLP-1R that are commonly used for Western blotting. They used mouse wild-type lung tissue that has a very high expression of GLP-1R as a positive control and lung tissue from GLP-1R / mice as a negative control. They also employed a very thorough procedure with immunoprecipitation in an attempt to increase the sensitivity of the analysis but found no GLP-1R-specific expression. The authors conclude that multiple commercially available GLP-1R antibodies do not detect authentic GLP-1R protein, even using optimally enhanced methods. This technical difficulty at first glance seems to have little bearing on aspects of GLP-1 biology unrelated to the scope of the article; however, on further consideration, it brings into focus a potentially serious problem with the validity of any GLP-1R expression data generated through the use of antibodies. Adding to the complexity, for some GLP-1R-containing organs, markedly different GLP-1R expression patterns have been reported between human and rodent species (10, 12). Rodent thyroid C-cells express much higher levels of GLP-1R than primates (10, 12), and there may be an important species difference, where the GLP-1R in the rodent thyroid is important for the high bone turnover in those species but less important in humans (13, 14). In the lung, the number of GLP1R-expressing cells and the receptor density is much


Endocrinology | 2015

Expression and distribution of glucagon-like peptide-1 receptor mRNA, protein and binding in the male nonhuman primate (Macaca mulatta) brain.

Kristy M. Heppner; Melissa A. Kirigiti; Anna Secher; Sarah Juel Paulsen; Rikley Buckingham; Charles Pyke; Lotte Bjerre Knudsen; Niels Vrang; Kevin L. Grove

Glucagon-like peptide-1 (GLP-1) is released from endocrine L-cells lining the gut in response to food ingestion. However, GLP-1 is also produced in the nucleus of the solitary tract, where it acts as an anorectic neurotransmitter and key regulator of many autonomic and neuroendocrine functions. The expression and projections of GLP-1-producing neurons is highly conserved between rodent and primate brain, although a few key differences have been identified. The GLP-1 receptor (GLP-1R) has been mapped in the rodent brain, but no studies have described the distribution of GLP-1Rs in the nonhuman primate central nervous system. Here, we characterized the distribution of GLP-1R mRNA and protein in the adult macaque brain using in situ hybridization, radioligand receptor autoradiography, and immunohistochemistry with a primate specific GLP-1R antibody. Immunohistochemistry demonstrated that the GLP-1R is localized to cell bodies and fiber terminals in a very selective distribution throughout the brain. Consistent with the functional role of the GLP-1R system, we find the highest concentration of GLP-1R-immunoreactivity present in select hypothalamic and brainstem regions that regulate feeding, including the paraventricular and arcuate hypothalamic nuclei, as well as the area postrema, nucleus of the solitary tract, and dorsal motor nucleus of the vagus. Together, our data demonstrate that GLP-1R distribution is highly conserved between rodent and primate, although a few key species differences were identified, including the amygdala, where GLP-1R expression is much higher in primate than in rodent.


Journal of Alzheimer's Disease | 2015

The GLP-1 Receptor Agonist Liraglutide Improves Memory Function and Increases Hippocampal CA1 Neuronal Numbers in a Senescence-Accelerated Mouse Model of Alzheimer’s Disease

Henrik H. Hansen; Katrine Fabricius; Pernille Barkholt; Michael L. Niehoff; John E. Morley; Jacob Jelsing; Charles Pyke; Lotte Bjerre Knudsen; Susan A. Farr; Niels Vrang

Abstract Recent studies indicate that glucagon-like peptide 1 (GLP-1) receptor agonists, currently used in the management of type 2 diabetes, exhibit neurotrophic and neuroprotective effects in amyloid-β (Aβ) toxicity models of Alzheimer’s disease (AD). We investigated the potential pro-cognitive and neuroprotective effects of the once-daily GLP-1 receptor agonist liraglutide in senescence-accelerated mouse prone 8 (SAMP8) mice, a model of age-related sporadic AD not dominated by amyloid plaques. Six-month-old SAMP8 mice received liraglutide (100 or 500 μg/kg/day, s.c.) or vehicle once daily for 4 months. Vehicle-dosed age-matched 50% back-crossed as well as untreated young (4-month-old) SAMP8 mice were used as control groups for normal memory function. Vehicle-dosed 10-month-old SAMP8 mice showed significant learning and memory retention deficits in an active-avoidance T-maze, as compared to both control groups. Also, 10-month-old SAMP8 mice displayed no immunohistological signatures of amyloid-β plaques or hyperphosphorylated tau, indicating the onset of cognitive deficits prior to deposition of amyloid plaques and neurofibrillary tangles in this AD model. Liraglutide significantly increased memory retention and total hippocampal CA1 pyramidal neuron numbers in SAMP8 mice, as compared to age-matched vehicle-dosed SAMP8 mice. In conclusion, liraglutide delayed or partially halted the progressive decline in memory function associated with hippocampal neuronal loss in a mouse model of pathological aging with characteristics of neurobehavioral and neuropathological impairments observed in early-stage sporadic AD.


Thrombosis Research | 2003

Recombinant factor VIIa reduces bleeding in severely thrombocytopenic rabbits.

Mikael Tranholm; Rasmus Rojkjaer; Charles Pyke; Annemarie T. Kristensen; Birgitte Klitgaard; Karsten Lollike; Morris A. Blajchman

Severe thrombocytopenia is a common complication to intensive chemotherapeutic regimens. For bleeding episodes associated with severe thrombocytopenia, the current standard treatment is platelet transfusion. However, due to several transfusion complications such as transfusion-transmitted diseases, platelet refractoriness and immunomodulation, as well as increasing problems with sufficient supply of platelet products, it is imperative to search for alternatives to platelet transfusion. To test the efficacy of recombinant activated human coagulation factor VII (rFVIIa, NovoSeven) in thrombocytopenia, a preclinical study was conducted in thrombocytopenic rabbits. Thrombocytopenia was induced by a combination of gamma-irradiation and the use of platelet antibodies, and the effect of rFVIIa on nail cuticle bleeding was determined. Administration of rFVIIa at 2 mg/kg significantly shortened the prolonged bleeding time in thrombocytopenic animals (rFVIIa vs. control, median 23 min 41 s vs. 60 min, p=0.016) as well as significantly reducing the blood loss (rFVIIa vs. control, median: 8.8 vs. 12.2 nmol hemoglobin/ml, p=0.016). This effect was also reflected by a significant reduction of the prothrombin time, activated partial thromboplastin time, as well as improvement in clotting parameters in an in vitro thromboelastography thrombocytopenia model. Histopathological evaluation of kidney biopsies for the presence of micro thrombi did not reveal evidence of prothrombotic effects of rFVIIa in this model. These data demonstrate the haemostatic efficacy of rFVIIa in a rabbit model of severe thrombocytopenia. Clinical trials will be needed to further explore the potential of NovoSeven as a haemostatic agent in thrombocytopenic patients.


Nature Communications | 2016

Analytic framework for peptidomics applied to large-scale neuropeptide identification

Anna Secher; Christian D. Kelstrup; Kilian W. Conde-Frieboes; Charles Pyke; Kirsten Raun; Birgitte S. Wulff; J. Olsen

Large-scale mass spectrometry-based peptidomics for drug discovery is relatively unexplored because of challenges in peptide degradation and identification following tissue extraction. Here we present a streamlined analytical pipeline for large-scale peptidomics. We developed an optimized sample preparation protocol to achieve fast, reproducible and effective extraction of endogenous peptides from sub-dissected organs such as the brain, while diminishing unspecific protease activity. Each peptidome sample was analysed by high-resolution tandem mass spectrometry and the resulting data set was integrated with publically available databases. We developed and applied an algorithm that reduces the peptide complexity for identification of biologically relevant peptides. The developed pipeline was applied to rat hypothalamus and identifies thousands of neuropeptides and their post-translational modifications, which is combined in a resource format for visualization, qualitative and quantitative analyses.


Brain Research | 2016

The GLP-1 receptor agonist liraglutide reduces pathology-specific tau phosphorylation and improves motor function in a transgenic hTauP301L mouse model of tauopathy

Henrik H. Hansen; Pernille Barkholt; Katrine Fabricius; Jacob Jelsing; Dick Terwel; Charles Pyke; Lotte Bjerre Knudsen; Niels Vrang

In addition to a prominent role in glycemic control, glucagon-like peptide 1 (GLP-1) receptor agonists exhibit neuroprotective properties. There is mounting experimental evidence that GLP-1 receptor agonists, including liraglutide, may enhance synaptic plasticity, counteract cognitive deficits and ameliorate neurodegenerative features in preclinical models of Alzheimers disease (AD), predominantly in the context of β-amyloid toxicity. Here we characterized the effects of liraglutide in a transgenic mutant tau (hTauP301L) mouse tauopathy model, which develops age-dependent pathology-specific neuronal tau phosphorylation and neurofibrillary tangle formation with progressively compromised motor function (limb clasping). Liraglutide (500 µg/kg/day, s.c., q.d., n=18) or vehicle (n=18) was administered to hTauP301L mice for 6 months from the age of three months. Vehicle-dosed wild-type FVB/N mice served as normal control (n=17). The onset and severity of hind limb clasping was markedly different in liraglutide and vehicle-dosed transgenic mice. Clasping behavior was observed in 61% of vehicle-dosed hTauP301L mice with a 55% survival rate in 9-month old transgenic mice. In contrast, liraglutide treatment reduced the clasping rate to 39% of hTauP301L mice, and fully prevented clasping-associated lethality resulting in a survival rate of 89%. Stereological analyses demonstrated that hTauP301L mice exhibited hindbrain-dominant neuronal accumulation of phosphorylated tau closely correlated to the severity of clasping behavior. In correspondence, liraglutide treatment significantly reduced neuronal phospho-tau load by 61.9±10.2% (p<0.001) in hTauP301L mice, as compared to vehicle-dosed controls. In conclusion, liraglutide significantly reduced tau pathology in a transgenic mouse tauopathy model.

Collaboration


Dive into the Charles Pyke's collaboration.

Top Co-Authors

Avatar

Niels Vrang

University of Copenhagen

View shared research outputs
Top Co-Authors

Avatar

Anna Secher

University of Copenhagen

View shared research outputs
Top Co-Authors

Avatar

Jacob Jelsing

Copenhagen University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Leif R. Lund

University of Copenhagen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge