Network


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

Hotspot


Dive into the research topics where Peter J. Crick is active.

Publication


Featured researches published by Peter J. Crick.


Biochemical Pharmacology | 2013

Methods for oxysterol analysis: Past, present and future

William J. Griffiths; Peter J. Crick; Yuqin Wang

Oxysterols are oxidised forms of cholesterol or its precursors. In this article we will concentrate specifically on those formed in mammalian systems. Oxidation may be catalysed by endogenous enzymes or through reactive oxygen species forming a myriad of potential products. A number of these products are biologically active, and oxysterols may have roles in cholesterol homeostasis, neurogenesis, protein prenylation and in the immune system. Oxysterols are also implicated in aetiology of disease states including atherosclerosis, neurodegenerative and inflammatory diseases. Reports indicating the levels of oxysterols in plasma, cerebrospinal fluid and various tissues are in many cases unrealistic owing to a lack of attention to the possibility of autoxidation, a process by which oxysterols are formed from cholesterol by oxygen in air. This article comprises a critical assessment of the technical difficulties of oxysterol analysis, highlights methodologies utilising best practise and discusses newer procedures.


Journal of Clinical Investigation | 2014

Cholestenoic acids regulate motor neuron survival via liver X receptors

Spyridon Theofilopoulos; William J. Griffiths; Peter J. Crick; Shanzheng Yang; Anna Meljon; Michael Ogundare; Satish Srinivas Kitambi; Andrew Lockhart; Karin Tuschl; Peter Clayton; Andrew Morris; Adelaida Martinez; M. Ashwin Reddy; Andrea Martinuzzi; Maria Teresa Bassi; Akira Honda; Tatsuki Mizuochi; Akihiko Kimura; Hiroshi Nittono; Giuseppe De Michele; Rosa Carbone; Chiara Criscuolo; Joyce L.W. Yau; Jonathan R. Seckl; Rebecca Schüle; Ludger Schöls; Andreas W. Sailer; Jens Kuhle; Matthew J. Fraidakis; Jan Åke Gustafsson

Cholestenoic acids are formed as intermediates in metabolism of cholesterol to bile acids, and the biosynthetic enzymes that generate cholestenoic acids are expressed in the mammalian CNS. Here, we evaluated the cholestenoic acid profile of mammalian cerebrospinal fluid (CSF) and determined that specific cholestenoic acids activate the liver X receptors (LXRs), enhance islet-1 expression in zebrafish, and increase the number of oculomotor neurons in the developing mouse in vitro and in vivo. While 3β,7α-dihydroxycholest-5-en-26-oic acid (3β,7α-diHCA) promoted motor neuron survival in an LXR-dependent manner, 3β-hydroxy-7-oxocholest-5-en-26-oic acid (3βH,7O-CA) promoted maturation of precursors into islet-1+ cells. Unlike 3β,7α-diHCA and 3βH,7O-CA, 3β-hydroxycholest-5-en-26-oic acid (3β-HCA) caused motor neuron cell loss in mice. Mutations in CYP7B1 or CYP27A1, which encode enzymes involved in cholestenoic acid metabolism, result in different neurological diseases, hereditary spastic paresis type 5 (SPG5) and cerebrotendinous xanthomatosis (CTX), respectively. SPG5 is characterized by spastic paresis, and similar symptoms may occur in CTX. Analysis of CSF and plasma from patients with SPG5 revealed an excess of the toxic LXR ligand, 3β-HCA, while patients with CTX and SPG5 exhibited low levels of the survival-promoting LXR ligand 3β,7α-diHCA. Moreover, 3β,7α-diHCA prevented the loss of motor neurons induced by 3β-HCA in the developing mouse midbrain in vivo.Our results indicate that specific cholestenoic acids selectively work on motor neurons, via LXR, to regulate the balance between survival and death.


Free Radical Biology and Medicine | 2013

Analytical strategies for characterization of oxysterol lipidomes: Liver X receptor ligands in plasma.

William J. Griffiths; Peter J. Crick; Yuchen Wang; Michael Ogundare; Karin Tuschl; Andrew Morris; Brian Bigger; Peter Clayton; Yuqin Wang

Bile acids, bile alcohols, and hormonal steroids represent the ultimate biologically active products of cholesterol metabolism in vertebrates. However, intermediates in their formation, including oxysterols and cholestenoic acids, also possess known, e.g., as ligands to nuclear and G-protein-coupled receptors, and unknown regulatory activities. The potential diversity of molecules originating from the cholesterol structure is very broad and their abundance in biological materials ranges over several orders of magnitude. Here we describe the application of enzyme-assisted derivatization for sterol analysis (EADSA) in combination with liquid chromatography-electrospray ionization-mass spectrometry to define the oxysterol and cholestenoic acid metabolomes of human plasma. Quantitative profiling of adult plasma using EADSA leads to the detection of over 30 metabolites derived from cholesterol, some of which are ligands to the nuclear receptors LXR, FXR, and pregnane X receptor or the G-protein-coupled receptor Epstein-Barr virus-induced gene 2. The potential of the EADSA technique in screening for inborn errors of cholesterol metabolism and biosynthesis is demonstrated by the unique plasma profile of patients suffering from cerebrotendinous xanthomatosis. The analytical methods described are easily adapted to the analysis of other biological fluids, including cerebrospinal fluid, and also tissues, e.g., brain, in which nuclear and G-protein-coupled receptors may have important regulatory roles.


Steroids | 2015

Cholesterol metabolites exported from human brain

Luigi Iuliano; Peter J. Crick; Chiara Zerbinati; Luigi Tritapepe; Jonas Abdel-Khalik; Marc Poirot; Yuqin Wang; William J. Griffiths

Graphical abstract


Clinical Chemistry | 2015

Quantitative Charge-Tags for Sterol and Oxysterol Analysis

Peter J. Crick; T. William Bentley; Jonas Abdel-Khalik; Ian Matthews; Peter Clayton; Andrew Morris; Brian Bigger; Chiara Zerbinati; Luigi Tritapepe; Luigi Iuliano; Yuqin Wang; William J. Griffiths

BACKGROUND Global sterol analysis is challenging owing to the extreme diversity of sterol natural products, the tendency of cholesterol to dominate in abundance over all other sterols, and the structural lack of a strong chromophore or readily ionized functional group. We developed a method to overcome these challenges by using different isotope-labeled versions of the Girard P reagent (GP) as quantitative charge-tags for the LC-MS analysis of sterols including oxysterols. METHODS Sterols/oxysterols in plasma were extracted in ethanol containing deuterated internal standards, separated by C18 solid-phase extraction, and derivatized with GP, with or without prior oxidation of 3β-hydroxy to 3-oxo groups. RESULTS By use of different isotope-labeled GPs, it was possible to analyze in a single LC-MS analysis both sterols/oxysterols that naturally possess a 3-oxo group and those with a 3β-hydroxy group. Intra- and interassay CVs were <15%, and recoveries for representative oxysterols and cholestenoic acids were 85%-108%. By adopting a multiplex approach to isotope labeling, we analyzed up to 4 different samples in a single run. Using plasma samples, we could demonstrate the diagnosis of inborn errors of metabolism and also the export of oxysterols from brain via the jugular vein. CONCLUSIONS This method allows the profiling of the widest range of sterols/oxysterols in a single analytical run and can be used to identify inborn errors of cholesterol synthesis and metabolism.


Journal of Biological Chemistry | 2014

Effects of a Disrupted Blood-Brain Barrier on Cholesterol Homeostasis in the Brain

Ahmed Saeed; Guillem Genové; Tian Li; Dieter Lütjohann; Maria Olin; Natalia Mast; Irina A. Pikuleva; Peter J. Crick; Yuqin Wang; William J. Griffiths; Christer Betsholtz; Ingemar Björkhem

Background: The role of the blood-brain barrier for cholesterol homeostasis in the brain is not known. Results: Significant influx of cholesterol into the brain and increased efflux of 24(S)-hydroxycholesterol were observed in mice with a defect blood-brain barrier. Conclusion: A defect blood-brain barrier increases permeability for steroid flux in both directions. Significance: Elucidation of the role of the blood-brain barrier for brain cholesterol turnover. The presence of the blood-brain barrier (BBB) is critical for cholesterol metabolism in the brain, preventing uptake of lipoprotein-bound cholesterol from the circulation. The metabolic consequences of a leaking BBB for cholesterol metabolism have not been studied previously. Here we used a pericyte-deficient mouse model, Pdgfbret/ret, shown to have increased permeability of the BBB to a range of low-molecular mass and high-molecular mass tracers. There was a significant accumulation of plant sterols in the brains of the Pdgfbret/ret mice. By dietary treatment with 0.3% deuterium-labeled cholesterol, we could demonstrate a significant flux of cholesterol from the circulation into the brains of the mutant mice roughly corresponding to about half of the measured turnover of cholesterol in the brain. We expected the cholesterol flux into the brain to cause a down-regulation of cholesterol synthesis. Instead, cholesterol synthesis was increased by about 60%. The levels of 24(S)-hydroxycholesterol (24S-OHC) were significantly reduced in the brains of the pericyte-deficient mice but increased in the circulation. After treatment with 1% cholesterol in diet, the difference in cholesterol synthesis between mutants and controls disappeared. The findings are consistent with increased leakage of 24S-OHC from the brain into the circulation in the pericyte-deficient mice. This oxysterol is an efficient suppressor of cholesterol synthesis, and the results are consistent with a regulatory role of 24S-OHC in the brain. To our knowledge, this is the first demonstration that a defective BBB may lead to increased flux of a lipophilic compound out from the brain. The relevance of the findings for the human situation is discussed.


Journal of Lipid Research | 2017

Defective cholesterol metabolism in amyotrophic lateral sclerosis

Jonas Abdel-Khalik; Eylan Yutuc; Peter J. Crick; Jan Åke Gustafsson; Margaret Warner; Gustavo Roman; Kevin Talbot; Elizabeth Gray; William J. Griffiths; Martin Turner; Yuqin Wang

As neurons die, cholesterol is released in the central nervous system (CNS); hence, this sterol and its metabolites may represent a biomarker of neurodegeneration, including in amyotrophic lateral sclerosis (ALS), in which altered cholesterol levels have been linked to prognosis. More than 40 different sterols were quantified in serum and cerebrospinal fluid (CSF) from ALS patients and healthy controls. In CSF, the concentration of cholesterol was found to be elevated in ALS samples. When CSF metabolite levels were normalized to cholesterol, the cholesterol metabolite 3β,7α-dihydroxycholest-5-en-26-oic acid, along with its precursor 3β-hydroxycholest-5-en-26-oic acid and product 7α-hydroxy-3-oxocholest-4-en-26-oic acid, were reduced in concentration, whereas metabolites known to be imported from the circulation into the CNS were not found to differ in concentration between groups. Analysis of serum revealed that (25R)26-hydroxycholesterol, the immediate precursor of 3β-hydroxycholest-5-en-26-oic acid, was reduced in concentration in ALS patients compared with controls. We conclude that the acidic branch of bile acid biosynthesis, known to be operative in-part in the brain, is defective in ALS, leading to a failure of the CNS to remove excess cholesterol, which may be toxic to neuronal cells, compounded by a reduction in neuroprotective 3β,7α-dihydroxycholest-5-en-26-oic acid.


PLOS Biology | 2016

An Interferon Regulated MicroRNA Provides Broad Cell-Intrinsic Antiviral Immunity through Multihit Host-Directed Targeting of the Sterol Pathway

Kevin Robertson; Wei Yuan Hsieh; Thorsten Forster; Mathieu Blanc; Hongjin Lu; Peter J. Crick; Eylan Yutuc; Steven Watterson; Kimberly Martin; Samantha J. Griffiths; Anton J. Enright; Mami Yamamoto; Madapura M. Pradeepa; Kimberly Ann Lennox; Mark A. Behlke; Simon G. Talbot; Jürgen Haas; Lars Dölken; William J. Griffiths; Yuqin Wang; Ana Angulo; Peter Ghazal

In invertebrates, small interfering RNAs are at the vanguard of cell-autonomous antiviral immunity. In contrast, antiviral mechanisms initiated by interferon (IFN) signaling predominate in mammals. Whilst mammalian IFN-induced miRNA are known to inhibit specific viruses, it is not known whether host-directed microRNAs, downstream of IFN-signaling, have a role in mediating broad antiviral resistance. By performing an integrative, systematic, global analysis of RNA turnover utilizing 4-thiouridine labeling of newly transcribed RNA and pri/pre-miRNA in IFN-activated macrophages, we identify a new post-transcriptional viral defense mechanism mediated by miR-342-5p. On the basis of ChIP and site-directed promoter mutagenesis experiments, we find the synthesis of miR-342-5p is coupled to the antiviral IFN response via the IFN-induced transcription factor, IRF1. Strikingly, we find miR-342-5p targets mevalonate-sterol biosynthesis using a multihit mechanism suppressing the pathway at different functional levels: transcriptionally via SREBF2, post-transcriptionally via miR-33, and enzymatically via IDI1 and SC4MOL. Mass spectrometry-based lipidomics and enzymatic assays demonstrate the targeting mechanisms reduce intermediate sterol pathway metabolites and total cholesterol in macrophages. These results reveal a previously unrecognized mechanism by which IFN regulates the sterol pathway. The sterol pathway is known to be an integral part of the macrophage IFN antiviral response, and we show that miR-342-5p exerts broad antiviral effects against multiple, unrelated pathogenic viruses such Cytomegalovirus and Influenza A (H1N1). Metabolic rescue experiments confirm the specificity of these effects and demonstrate that unrelated viruses have differential mevalonate and sterol pathway requirements for their replication. This study, therefore, advances the general concept of broad antiviral defense through multihit targeting of a single host pathway.


The Journal of Steroid Biochemistry and Molecular Biology | 2016

New methods for analysis of oxysterols and related compounds by LC-MS.

William J. Griffiths; Jonas Abdel-Khalik; Peter J. Crick; Eylan Yutuc; Yuqin Wang

Oxysterols are oxygenated forms of cholesterol or its precursors. They are formed enzymatically and via reactive oxygen species. Oxysterols are intermediates in bile acid and steroid hormone biosynthetic pathways and are also bioactive molecules in their own right, being ligands to nuclear receptors and also regulators of the processing of steroid regulatory element-binding proteins (SREBPs) to their active forms as transcription factors regulating cholesterol and fatty acid biosynthesis. Oxysterols are implicated in the pathogenesis of multiple disease states ranging from atherosclerosis and cancer to multiple sclerosis and other neurodegenerative diseases including Alzheimers and Parkinsons disease. Analysis of oxysterols is challenging on account of their low abundance in biological systems in comparison to cholesterol, and due to the propensity of cholesterol to undergo oxidation in air to generate oxysterols with the same structures as those present endogenously. In this article we review the mass spectrometry-based methods for oxysterol analysis paying particular attention to analysis by liquid chromatography-mass spectrometry (LC-MS).


The Journal of Steroid Biochemistry and Molecular Biology | 2017

Sterols and oxysterols in plasma from Smith-Lemli-Opitz syndrome patients.

William J. Griffiths; Jonas Abdel-Khalik; Peter J. Crick; Michael Ogundare; Cedric Shackleton; Karin Tuschl; Mei Kwun Kwok; Brian Bigger; Andrew Morris; Akira Honda; Libin Xu; Ned A. Porter; Ingemar Björkhem; Peter Clayton; Yuqin Wang

Smith-Lemli-Opitz syndrome (SLOS) is a severe autosomal recessive disorder resulting from defects in the cholesterol synthesising enzyme 7-dehydrocholesterol reductase (Δ7-sterol reductase, DHCR7, EC 1.3.1.21) leading to a build-up of the cholesterol precursor 7-dehydrocholesterol (7-DHC) in tissues and blood plasma. Although the underling enzyme deficiency associated with SLOS is clear there are likely to be multiple mechanisms responsible for SLOS pathology. In an effort to learn more of the aetiology of SLOS we have analysed plasma from SLOS patients to search for metabolites derived from 7-DHC which may be responsible for some of the pathology. We have identified a novel hydroxy-8-dehydrocholesterol, which is either 24- or 25-hydroxy-8-dehydrocholesterol and also the known metabolites 26-hydroxy-8-dehydrocholesterol, 4-hydroxy-7-dehydrocholesterol, 3β,5α-dihydroxycholest-7-en-6-one and 7α,8α-epoxycholesterol. None of these metabolites are detected in control plasma at quantifiable levels (0.5ng/mL).

Collaboration


Dive into the Peter J. Crick's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Peter Clayton

University of Manchester

View shared research outputs
Top Co-Authors

Avatar

Brian Bigger

University of Manchester

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ingemar Björkhem

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge