Multiple Loci Modulate Opioid Therapy Response for Cancer Pain

Total Page:16

File Type:pdf, Size:1020Kb

Multiple Loci Modulate Opioid Therapy Response for Cancer Pain Author Manuscript Published OnlineFirst on May 27, 2011; DOI: 10.1158/1078-0432.CCR-10-3028 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 1 Multiple loci modulate opioid therapy response for 2 cancer pain 3 4 Antonella Galvan 1, Frank Skorpen 2, Pål Klepstad 2,3, Anne Kari Knudsen 2, Torill 5 Fladvad 2, Felicia S. Falvella 1, Alessandra Pigni 1, Cinzia Brunelli 1, Augusto 6 Caraceni 1,2, Stein Kaasa 2,4, Tommaso A. Dragani 1 7 8 9 10 1 Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy; 2 European Palliative 11 Care Research Center, Medical Faculty, Norwegian University of Science and 12 Technology, Trondheim, Norway; 3 Department of Anaesthesiology and Emergency 13 Medicine and 4 Department of Oncology, St. Olavs University Hospital, Trondheim, 14 Norway 15 16 17 Conflict of interest statement 18 None declared. 19 20 Correspondence to: 21 Tommaso A. Dragani, Department of Predictive and Preventive Medicine, 22 Fondazione IRCCS Istituto Nazionale Tumori, Via G.A. Amadeo 42, 20133 23 Milan, Italy. Tel.: +39-0223902642, Fax: +39-0223902764. E-Mail: 24 [email protected] 25 Running Title: Genetic control of cancer pain response 26 Keywords: opioids, pharmacogenomics, pain, polygenic diseases, single-nucleotide 27 polymorphisms. 28 1 Downloaded from clincancerres.aacrjournals.org on September 24, 2021. © 2011 American Association for Cancer Research. Author Manuscript Published OnlineFirst on May 27, 2011; DOI: 10.1158/1078-0432.CCR-10-3028 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 1 Abstract 2 Purpose: Patients treated with opioid drugs for cancer pain experience different 3 relief responses, raising the possibility that genetic factors play a role in opioid 4 therapy outcome. In this study, we tested the hypothesis that genetic variations may 5 control individual response to opioid drugs in cancer patients. 6 Experimental design: We tested one million SNPs in European cancer patients 7 selected in a first series for extremely poor (pain relief ≤40%; n=145) or good (pain 8 relief ≥90%; n=293) responses to opioid therapy using a DNA pooling approach. 9 Candidate SNPs identified by SNP-array were genotyped in individual samples 10 constituting DNA pools as well as in a second series of 570 patients. 11 Results: Association analysis in 1008 cancer patients identified 8 SNPs significantly 12 associated with pain relief at a statistical threshold of P<1.0 x 10-3, with rs12948783, 13 upstream of the RHBDF2 gene, showing the best statistical association (P=8.1 x 10- 14 9). Functional annotation analysis of SNP-tagged genes suggested the involvement 15 of genes acting on neurological system processes. 16 Conclusion: Our results indicate that the identified SNP panel can modulate the 17 response of cancer patients to opioid therapy and may provide a new tool for 18 personalized therapy of cancer pain. 19 2 Downloaded from clincancerres.aacrjournals.org on September 24, 2021. © 2011 American Association for Cancer Research. Author Manuscript Published OnlineFirst on May 27, 2011; DOI: 10.1158/1078-0432.CCR-10-3028 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 1 Translational relevance 2 Pain is a common complication of almost all types of advanced cancer, and 3 treatment with opioids produces variable responses of pain relief among individuals. 4 Indeed, a variable percentage of cancer patients experiences reduced or poor 5 benefit from opioid therapy, with impairment of their quality of life. Our association 6 study in 1008 European cancer patients identified 8 SNPs statistically associated 7 with pain relief, which together might modulate the response of cancer patients to 8 opioid therapy and might serve as targetable tools to design new drugs. Our 9 functional annotation analysis of SNP-tagged genes revealed for the first time the 10 involvement of genes acting on neurological system processes rather than on opioid 11 metabolism or pharmacodynamics. Thus, the identified SNP panel may provide a 12 new tool for personalized therapy of cancer pain. 13 3 Downloaded from clincancerres.aacrjournals.org on September 24, 2021. © 2011 American Association for Cancer Research. Author Manuscript Published OnlineFirst on May 27, 2011; DOI: 10.1158/1078-0432.CCR-10-3028 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 1 Introduction 2 Pain is the most dreaded symptom in patients with malignant disease. Indeed, 3 each year several million men and women of all ages suffer from some form of 4 invasive cancer associated with pain requiring chronic opioid treatment. At present, 5 opioids are virtually the only analgesics able to control moderate and severe cancer 6 pain. 7 While opioids are widely used to treat cancer pain, clinical studies indicate 8 great variability among individuals in the efficacy of these drugs and their side effects 9 (1, 2). Many studies have investigated the potential genetic basis of individual 10 variability in opioid drug response, but almost all have focused on a few candidate 11 genes involved in opioid pharmacokinetics or pharmacodynamics, and results are not 12 consistent among these often small studies (3, 4). For example, the catecholamines 13 dopamine, epinephrine and norepinephrine and their hydroxylated metabolites are 14 substrates for the enzyme catechol-O-methyltransferase encoded by the COMT 15 gene, in which genetic variations have been associated with variations in pain 16 perception and in doses of opioids required for pain treatment; however, 17 discrepancies in the results from different association studies on COMT 18 polymorphisms and pain perception have been reported, possibly due to the small 19 size of the studies (reviewed in (5)). 20 The human mu 1 opioid receptor (OPRM1) gene, which is the main 21 pharmacologic target of opioids, contains more than 100 polymorphisms, and these 22 polymorphisms might affect opioid binding affinity to the receptor and, consequently, 23 opioid-induced analgesia, tolerance, and dependence. However, a recent meta- 24 analysis casts doubt on the clinical relevance of the OPRM1 118A>G polymorphism 25 suggested by both preclinical and clinical studies as a candidate in personalized 26 cancer pain therapy (6). 27 A complex genetics underlying inter-individual differences in opioid response 28 is expected in light of the biochemical and biological complexity of the drug response 29 and of pain control. Indeed, opioid effects are governed by drug-related absorption, 30 distribution, metabolism, intrinsic efficacy of the receptors involved, and by signal 31 pathways downstream of the receptors. Since each of these steps in pain control is 32 modulated by several genes, each of which can exist in allelic forms in the general 4 Downloaded from clincancerres.aacrjournals.org on September 24, 2021. © 2011 American Association for Cancer Research. Author Manuscript Published OnlineFirst on May 27, 2011; DOI: 10.1158/1078-0432.CCR-10-3028 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 1 population, a genome-wide comprehensive analysis would be required to identify all 2 genetic factors involved. 3 Herein, we conducted the first genome-wide association study (GWAS) in 4 cancer patients treated with opioids using a 1-million single-nucleotide polymorphism 5 (SNP)-array to identify new genetic variations modulating individual pain relief. 6 Patients and methods 7 Study design 8 The European Pharmacogenetic Opioid Study (EPOS) represents a 9 multicenter effort to examine symptoms, pharmacology, and pharmacogenomics 10 related to the use of opioids. Recruitment criteria of patients included a regularly 11 scheduled opioid treatment stable for at least 3 days. Opioids administered to >5% of 12 patients included morphine, oxycodone, and fentanyl. The study includes 2294 13 cancer patients treated with opioids for moderate or severe pain and recruited from 14 17 centers of 11 European countries. The participation of the Research Steering 15 Committee from the European Association for Palliative Care (EAPC) facilitated 16 international cooperation in the study. 17 The study collected demographic and personal data, as well as malignant 18 diagnosis and opioid treatment from cancer patients. Opioid doses were converted to 19 equivalent total daily oral morphine doses (7). Pain was assessed by completing the 20 Brief Pain Inventory (BPI) (8). Functional status was measured by the Karnofsky 21 performance status score. Further details about the EPOS series, including opioid 22 doses and recruitments, have been reported (7). The study collected whole blood for 23 genetic analysis and has established a biobank. 24 Pain relief phenotype and DNA pool construction 25 The pain relief phenotype under study is semi-quantitative and determined 26 based on the BPI, a robust and psychometric validated method to assess the 27 subjective severity of pain. Pain relief is measured using an 11-point numerical rating 28 scale, from 0%, representing “no pain relief,” to 100% or “complete pain relief", i.e., 29 0%, 10%, 20%, etc. (9). 30 For DNA pool preparation, we first defined the cancer patients as “good” or 5 Downloaded from clincancerres.aacrjournals.org on September 24, 2021. © 2011 American Association for Cancer Research. Author Manuscript Published OnlineFirst on May 27, 2011; DOI: 10.1158/1078-0432.CCR-10-3028 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 1 “poor” responders to opioid therapy, based on their pain relief phenotype score 2 ≥90% or ≤40%, respectively (Fig. 1). Selection was stratified by gender and country 3 of origin, with a 1:2 matching of “poor” and “good” responders, to obtain a balanced 4 representation of patients in the two groups. DNA pools of “good” (n=293, mean pain 5 relief, 94.1%) and “poor” (n=145, mean pain relief, 27.0%) responders were created 6 using equal amounts of fluorimetrically measured DNA from each sample.
Recommended publications
  • A Gene-Level Methylome-Wide Association Analysis Identifies Novel
    bioRxiv preprint doi: https://doi.org/10.1101/2020.07.13.201376; this version posted July 14, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. 1 A gene-level methylome-wide association analysis identifies novel 2 Alzheimer’s disease genes 1 1 2 3 4 3 Chong Wu , Jonathan Bradley , Yanming Li , Lang Wu , and Hong-Wen Deng 1 4 Department of Statistics, Florida State University; 2 5 Department of Biostatistics & Data Science, University of Kansas Medical Center; 3 6 Population Sciences in the Pacific Program, University of Hawaii Cancer center; 4 7 Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, 8 Tulane University School of Medicine 9 Corresponding to: Chong Wu, Assistant Professor, Department of Statistics, Florida State 10 University, email: [email protected] 1 bioRxiv preprint doi: https://doi.org/10.1101/2020.07.13.201376; this version posted July 14, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. 11 Abstract 12 Motivation: Transcriptome-wide association studies (TWAS) have successfully facilitated the dis- 13 covery of novel genetic risk loci for many complex traits, including late-onset Alzheimer’s disease 14 (AD). However, most existing TWAS methods rely only on gene expression and ignore epige- 15 netic modification (i.e., DNA methylation) and functional regulatory information (i.e., enhancer- 16 promoter interactions), both of which contribute significantly to the genetic basis ofAD.
    [Show full text]
  • Genome-Wide Association Study in Collaborative Cross Mice Revealed a Skeletal
    bioRxiv preprint doi: https://doi.org/10.1101/094698; this version posted December 16, 2016. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC 4.0 International license. Title Genome-wide association study in Collaborative Cross mice revealed a skeletal role for Rhbdf2 Authors Roei Levy1,2, Clemence Levet3, Keren Cohen1, Matthew Freeman3, Richard Mott4, Fuad Iraqi5, Yankel Gabet1 Affiliations 1 Department of Anatomy and Anthropology, 2 Department of Human Molecular Genetics and Biochemistry, and 5 Department of Clinical Microbiology and Immunology, Sackler Medical School, Tel Aviv University. 3 Dunn School of Pathology, South Parks Road, Oxford OX1 3RE. 4 UCL Genetics Institute, University College London, Gower St., London, WC1E 6BT, UK. Abstract A growing concern that overshadows the increased life expectancy developed countries have been witnessing during the last decades is an accompanying bone loss, which often manifests as osteoporosis. Despite ongoing efforts in utilizing genomic data to fully map the genes responsible for bone remodeling, a detailed picture remains to be desired. Here we took advantage of the phenotypic and genetic diversity innate in Collaborative Cross (CC) mice to identify genetic variants associated with microstructural bone characteristics. 1 bioRxiv preprint doi: https://doi.org/10.1101/094698; this version posted December 16, 2016. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity.
    [Show full text]
  • Genetic Deletion of Amphiregulin Restores the Normal Skin Phenotype in a Mouse Model of the Human Skin Disease Tylosis Vishnu Hosur*, Benjamin E
    © 2017. Published by The Company of Biologists Ltd | Biology Open (2017) 6, 1174-1179 doi:10.1242/bio.026260 RESEARCH ARTICLE Genetic deletion of amphiregulin restores the normal skin phenotype in a mouse model of the human skin disease tylosis Vishnu Hosur*, Benjamin E. Low, Leonard D. Shultz and Michael V. Wiles ABSTRACT mutations in human RHBDF2 lead to enhanced AREG secretion In humans, gain-of-function (GOF) mutations in RHBDF2 cause the and tylosis. We provide evidence that Rhbdf2 GOF mutations skin disease tylosis. We generated a mouse model of human tylosis enhance AREG secretion to cause hyperplasia and hyperkeratosis. and show that GOF mutations in RHBDF2 cause tylosis by enhancing In addition, we show that genetic ablation of AREG attenuates skin P159L/P159L the amount of amphiregulin (AREG) secretion. Furthermore, we show disease in the Rhbdf2 mouse model of human tylosis. that genetic disruption of AREG ameliorates skin pathology in mice Together, our data strongly suggest that inhibition of AREG could carrying the human tylosis disease mutation. Collectively, our data have potential therapeutic value in the treatment of tylosis. suggest that RHBDF2 plays a critical role in regulating EGFR signaling and its downstream events, including development of RESULTS AND DISCUSSION tylosis, by facilitating enhanced secretion of AREG. Thus, targeting The human tylosis disease mutation enhances AREG AREG could have therapeutic benefit in the treatment of tylosis. secretion Autosomal dominant mutations in the human RHBDF2 gene cause KEY WORDS: CRISPR/Cas9, EGFR, RHBDF2, Amphiregulin, Tylosis tylosis (Blaydon et al., 2012; Saarinen et al., 2012). Substantial evidence implicates the involvement of AREG-induced constitutive INTRODUCTION activation of the EGFR pathway (Blaydon et al., 2012; Brooke et al., The role of RHBDF2 in enhancing amphiregulin (AREG) secretion, 2014; Hosur et al., 2014) in human tylosis.
    [Show full text]
  • Multiple Loci Modulate Opioid Therapy Response for Cancer Pain
    Published OnlineFirst May 27, 2011; DOI: 10.1158/1078-0432.CCR-10-3028 Clinical Cancer Predictive Biomarkers and Personalized Medicine Research Multiple Loci Modulate Opioid Therapy Response for Cancer Pain Antonella Galvan1, Frank Skorpen2,Pal Klepstad2,3, Anne Kari Knudsen2, Torill Fladvad2, Felicia S. Falvella1, Alessandra Pigni1, Cinzia Brunelli1, Augusto Caraceni1,2, Stein Kaasa2,4, and Tommaso A. Dragani1 Abstract Purpose: Patients treated with opioid drugs for cancer pain experience different relief responses, raising the possibility that genetic factors play a role in opioid therapy outcome. In this study, we tested the hypothesis that genetic variations may control individual response to opioid drugs in cancer patients. Experimental Design: We tested 1 million single-nucleotide polymorphisms (SNP) in European cancer patients, selected in a first series, for extremely poor (pain relief 40%; n ¼ 145) or good (pain relief 90%; n ¼ 293) responses to opioid therapy using a DNA-pooling approach. Candidate SNPs identified by SNP- array were genotyped in individual samples constituting DNA pools as well as in a second series of 570 patients. Results: Association analysis in 1,008 cancer patients identified eight SNPs significantly associated with À pain relief at a statistical threshold of P < 1.0 Â 10 3, with rs12948783, upstream of the RHBDF2 gene, À showing the best statistical association (P ¼ 8.1 Â 10 9). Functional annotation analysis of SNP-tagged genes suggested the involvement of genes acting on processes of the neurologic system. Conclusion: Our results indicate that the identified SNP panel can modulate the response of cancer patients to opioid therapy and may provide a new tool for personalized therapy of cancer pain.
    [Show full text]
  • Collagen-Induced Arthritis Analysis in Rhbdf2 Knockout Mouse
    Original Article Biomol Ther 26(3), 298-305 (2018) Collagen-Induced Arthritis Analysis in Rhbdf2 Knockout Mouse Min­Young Lee1,2, Ju­Seong Kang1, Ryeo­Eun Go2, Yong­Sub Byun1, Young Jin Wi3, Kyung­A Hwang2, Jae­Hoon Choi3, Hyoung­Chin Kim1, Kyung­Chul Choi2,* and Ki­Hoan Nam1,* 1Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Cheongwon 28116, 2Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, 3Department of Life Science, College of Natureal Sciences, Research Institute of Natural Sciences, Hanyang University, Seoul 04763, Republic of Korea Abstract Rhomboid family member 2 gene (Rhbdf2) is an inactive homologue lacking essential catalytic residues of rhomboid intramem- brane serine proteases. The protein is necessary for maturation of tumor necrosis factor-alpha (TNF-α) converting enzyme, which is the molecule responsible for the release of TNF-α. In this study, Rhbdf2 knockout (KO) mice were produced by CRISPR/CAS9. To see the effects of the failure of TNF-α release induced by Rhbdf2 gene KO, collagen-induced arthritis (CIA), which is the rep- resentative TNF-α related disease, was induced in the Rhbdf2 mutant mouse using chicken collagen type II. The severity of the CIA was measured by traditional clinical scores and histopathological analysis of hind limb joints. A rota-rod test and grip strength test were employed to evaluate the severity of CIA based on losses of physical functions. The results indicated that Rhbdf2 mutant mice showed clear alleviation of the clinical severity of CIA as demonstrated by the significantly lower severity indexes.
    [Show full text]
  • Sex-Specific Transcriptome Differences in Human Adipose
    G C A T T A C G G C A T genes Article Sex-Specific Transcriptome Differences in Human Adipose Mesenchymal Stem Cells 1, 2, 3 1,3 Eva Bianconi y, Raffaella Casadei y , Flavia Frabetti , Carlo Ventura , Federica Facchin 1,3,* and Silvia Canaider 1,3 1 National Laboratory of Molecular Biology and Stem Cell Bioengineering of the National Institute of Biostructures and Biosystems (NIBB)—Eldor Lab, at the Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy; [email protected] (E.B.); [email protected] (C.V.); [email protected] (S.C.) 2 Department for Life Quality Studies (QuVi), University of Bologna, Corso D’Augusto 237, 47921 Rimini, Italy; [email protected] 3 Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy; fl[email protected] * Correspondence: [email protected]; Tel.: +39-051-2094114 These authors contributed equally to this work. y Received: 1 July 2020; Accepted: 6 August 2020; Published: 8 August 2020 Abstract: In humans, sexual dimorphism can manifest in many ways and it is widely studied in several knowledge fields. It is increasing the evidence that also cells differ according to sex, a correlation still little studied and poorly considered when cells are used in scientific research. Specifically, our interest is on the sex-related dimorphism on the human mesenchymal stem cells (hMSCs) transcriptome. A systematic meta-analysis of hMSC microarrays was performed by using the Transcriptome Mapper (TRAM) software. This bioinformatic tool was used to integrate and normalize datasets from multiple sources and allowed us to highlight chromosomal segments and genes differently expressed in hMSCs derived from adipose tissue (hADSCs) of male and female donors.
    [Show full text]
  • The Mutational Landscape of Human Olfactory G Protein-Coupled Receptors
    Jimenez et al. BMC Biology (2021) 19:21 https://doi.org/10.1186/s12915-021-00962-0 RESEARCH ARTICLE Open Access The mutational landscape of human olfactory G protein-coupled receptors Ramón Cierco Jimenez1,2, Nil Casajuana-Martin1, Adrián García-Recio1, Lidia Alcántara1, Leonardo Pardo1, Mercedes Campillo1 and Angel Gonzalez1* Abstract Background: Olfactory receptors (ORs) constitute a large family of sensory proteins that enable us to recognize a wide range of chemical volatiles in the environment. By contrast to the extensive information about human olfactory thresholds for thousands of odorants, studies of the genetic influence on olfaction are limited to a few examples. To annotate on a broad scale the impact of mutations at the structural level, here we analyzed a compendium of 119,069 natural variants in human ORs collected from the public domain. Results: OR mutations were categorized depending on their genomic and protein contexts, as well as their frequency of occurrence in several human populations. Functional interpretation of the natural changes was estimated from the increasing knowledge of the structure and function of the G protein-coupled receptor (GPCR) family, to which ORs belong. Our analysis reveals an extraordinary diversity of natural variations in the olfactory gene repertoire between individuals and populations, with a significant number of changes occurring at the structurally conserved regions. A particular attention is paid to mutations in positions linked to the conserved GPCR activation mechanism that could imply phenotypic variation in the olfactory perception. An interactive web application (hORMdb, Human Olfactory Receptor Mutation Database) was developed for the management and visualization of this mutational dataset.
    [Show full text]
  • Science Advances | Research Article
    SCIENCE ADVANCES | RESEARCH ARTICLE NEUROSCIENCE Copyright © 2019 The Authors, some rights reserved; Sleep fragmentation, microglial aging, and exclusive licensee American Association cognitive impairment in adults with and without for the Advancement of Science. No claim to Alzheimer’s dementia original U.S. Government Kirusanthy Kaneshwaran1, Marta Olah2, Shinya Tasaki3,4, Lei Yu3,4, Works. Distributed 2 3,4 3,4 under a Creative Elizabeth M. Bradshaw , Julie A. Schneider , Aron S. Buchman , Commons Attribution 3,4 2 1 David A. Bennett , Philip L. De Jager , Andrew S. P. Lim * NonCommercial License 4.0 (CC BY-NC). Sleep disruption is associated with cognitive decline and dementia in older adults; however, the underlying mechanisms are unclear. In rodents, sleep disruption causes microglial activation, inhibition of which improves cognition. However, data from humans are lacking. We studied participants in two cohort studies of older persons—the Rush Memory and Aging Project and the Religious Orders Study. We assessed sleep fragmentation by actigraphy and related this to cognitive function, to neocortical microglial marker gene expression measured by RNA sequencing, and to the neocortical density of microglia assessed by immunohistochemistry. Greater Downloaded from sleep fragmentation was associated with higher neocortical expression of genes characteristic of aged microglia, and a higher proportion of morphologically activated microglia, independent of chronological age- and dementia-related neuropathologies. Furthermore, these were, in turn, associated with worse cognition. This suggests that sleep fragmentation is accompanied by accelerated microglial aging and activation, which may partially underlie its association with cognitive impairment. http://advances.sciencemag.org/ INTRODUCTION development of therapies to mitigate the deleterious impact of sleep Cognitive impairment and dementia constitute a growing public disruption on cognition and dementia.
    [Show full text]
  • Biological Pathways, Candidate Genes, and Molecular Markers Associated with Quality-Of-Life Domains: an Update
    Qual Life Res (2014) 23:1997–2013 DOI 10.1007/s11136-014-0656-1 REVIEW Biological pathways, candidate genes, and molecular markers associated with quality-of-life domains: an update Mirjam A. G. Sprangers • Melissa S. Y. Thong • Meike Bartels • Andrea Barsevick • Juan Ordon˜ana • Qiuling Shi • Xin Shelley Wang • Pa˚l Klepstad • Eddy A. Wierenga • Jasvinder A. Singh • Jeff A. Sloan Accepted: 19 February 2014 / Published online: 7 March 2014 Ó Springer International Publishing Switzerland 2014 Abstract (depressed mood) and positive (well-being/happiness) Background There is compelling evidence of a genetic emotional functioning, social functioning, and overall foundation of patient-reported quality of life (QOL). Given QOL. the rapid development of substantial scientific advances in Methods We followed a purposeful search algorithm of this area of research, the current paper updates and extends existing literature to capture empirical papers investigating reviews published in 2010. the relationship between biological pathways and molecu- Objectives The objective was to provide an updated lar markers and the identified QOL domains. overview of the biological pathways, candidate genes, and Results Multiple major pathways are involved in each molecular markers involved in fatigue, pain, negative QOL domain. The inflammatory pathway has the strongest evidence as a controlling mechanism underlying fatigue. Inflammation and neurotransmission are key processes On behalf of the GeneQol Consortium. involved in pain perception, and the catechol-O-methyl- transferase (COMT) gene is associated with multiple sorts Electronic supplementary material The online version of this article (doi:10.1007/s11136-014-0656-1) contains supplementary of pain. The neurotransmitter and neuroplasticity theories material, which is available to authorized users.
    [Show full text]
  • A Concise Review of Human Brain Methylome During Aging and Neurodegenerative Diseases
    BMB Rep. 2019; 52(10): 577-588 BMB www.bmbreports.org Reports Invited Mini Review A concise review of human brain methylome during aging and neurodegenerative diseases Renuka Prasad G & Eek-hoon Jho* Department of Life Science, University of Seoul, Seoul 02504, Korea DNA methylation at CpG sites is an essential epigenetic mark position of carbon in the cytosine within CG dinucleotides that regulates gene expression during mammalian development with resultant formation of 5mC. The symmetrical CG and diseases. Methylome refers to the entire set of methylation dinucleotides are also called as CpG, due to the presence of modifications present in the whole genome. Over the last phosphodiester bond between cytosine and guanine. The several years, an increasing number of reports on brain DNA human genome contains short lengths of DNA (∼1,000 bp) in methylome reported the association between aberrant which CpG is commonly located (∼1 per 10 bp) in methylation and the abnormalities in the expression of critical unmethylated form and referred as CpG islands; they genes known to have critical roles during aging and neuro- commonly overlap with the transcription start sites (TSSs) of degenerative diseases. Consequently, the role of methylation genes. In human DNA, 5mC is present in approximately 1.5% in understanding neurodegenerative diseases has been under of the whole genome and CpG base pairs are 5-fold enriched focus. This review outlines the current knowledge of the human in CpG islands than other regions of the genome (3, 4). CpG brain DNA methylomes during aging and neurodegenerative islands have the following salient features. In the human diseases.
    [Show full text]
  • IPP17 Carcinoma IPMN Gene Symbol Transcript Accession Nucleotide
    IPP17 Carcinoma IPMN Gene Transcript Amino Gene Transcript Amino Allele Nucleotide change Consequence Allele Frequency Nucleotide change Consequence Symbol Accession Acid Symbol Accession Acid Frequency ACAP2 CCDS33924.1 chr3: 195063234 C>T C165Y Missense 19 ADCY8 CCDS33924.1 chr8: 131964235 C>T V374M Missense 29 ADAMTS1 CCDS33524.1 chr21: 28212054 T>G E627A Missense 11 ASH1L CCDS6363.1 chr1: 155307924 T>C N2920S Missense 16 ADCY8 CCDS6363.1 chr8: 131964235 C>T V374M Missense 36 CLNK CCDS1113.2 chr4: 10542196 G>T P175H Missense 14 AR CCDS14387.1 chrX: 66765158 T>A L57Q Missense 15 CLU CCDS47024.1 chr1: 155307924 T>C R127H Missense 23 ARL11 CCDS9419.1 chr13: 50204931 delCTT F117del In-frame deletion 10 CPN1 CCDS47832.1 chr10: 101816891 T>A Y297F In-frame deletion 13 ASH1L CCDS1113.2 chr1: 155307924 T>C N2920S Missense 16 CSRNP2 CCDS7486.1 chr12: 51458139 delTCT E340del In-frame deletion 13 C7orf26 CCDS5353.1 chr7: 6647641 C>T A400V Missense 13 CTNNB1 CCDS8807.1 chr3: 41266136 T>C S45P Missense 11 CLNK CCDS47024.1 chr4: 10542196 G>T P175H Missense 14 ERF CCDS2694.1 chr19: 42752892 G>A R458C Missense 15 COMT CCDS13770.1 chr22: 19951169 G>A V124M Missense 11 FBP2 CCDS12600.1 chr9: 97325736 C>A S238I Missense 13 CORIN CCDS3477.1 chr4: 47645243 G>C A663G Missense 14 HOXB3 CCDS6711.1 chr17: 46629769 C>T G23D Missense 13 CPN1 CCDS8807.1 chr10: 101816891 T>A Y297F In-frame deletion 9 KRTAP13 CCDS11528.1 chr21: 31797951 T>C T94A Missense 14 CSRNP2 CCDS2694.1 chr12: 51458139 delTCT E340del In-frame deletion 17 MAGEB4 CCDS13591.1 chrX: 30260590 C>T
    [Show full text]
  • The Hypothalamus As a Hub for SARS-Cov-2 Brain Infection and Pathogenesis
    bioRxiv preprint doi: https://doi.org/10.1101/2020.06.08.139329; this version posted June 19, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. The hypothalamus as a hub for SARS-CoV-2 brain infection and pathogenesis Sreekala Nampoothiri1,2#, Florent Sauve1,2#, Gaëtan Ternier1,2ƒ, Daniela Fernandois1,2 ƒ, Caio Coelho1,2, Monica ImBernon1,2, Eleonora Deligia1,2, Romain PerBet1, Vincent Florent1,2,3, Marc Baroncini1,2, Florence Pasquier1,4, François Trottein5, Claude-Alain Maurage1,2, Virginie Mattot1,2‡, Paolo GiacoBini1,2‡, S. Rasika1,2‡*, Vincent Prevot1,2‡* 1 Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, DistAlz, UMR-S 1172, Lille, France 2 LaBoratorY of Development and PlasticitY of the Neuroendocrine Brain, FHU 1000 daYs for health, EGID, School of Medicine, Lille, France 3 Nutrition, Arras General Hospital, Arras, France 4 Centre mémoire ressources et recherche, CHU Lille, LiCEND, Lille, France 5 Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Center for Infection and ImmunitY of Lille (CIIL), Lille, France. # and ƒ These authors contriButed equallY to this work. ‡ These authors directed this work *Correspondence to: [email protected] and [email protected] Short title: Covid-19: the hypothalamic hypothesis 1 bioRxiv preprint doi: https://doi.org/10.1101/2020.06.08.139329; this version posted June 19, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity.
    [Show full text]