Maternal Folic Acid Impacts DNA Methylation Profile in Male Rat Offspring Implicated in Neurodevelopment and Learning/Memory

Total Page:16

File Type:pdf, Size:1020Kb

Maternal Folic Acid Impacts DNA Methylation Profile in Male Rat Offspring Implicated in Neurodevelopment and Learning/Memory Wang et al. Genes & Nutrition (2021) 16:1 https://doi.org/10.1186/s12263-020-00681-1 RESEARCH Open Access Maternal folic acid impacts DNA methylation profile in male rat offspring implicated in neurodevelopment and learning/memory abilities Xinyan Wang1, Zhenshu Li1, Yun Zhu2,3, Jing Yan3,4, Huan Liu1,3, Guowei Huang1,3 and Wen Li1,3* Abstract Background: Periconceptional folic acid (FA) supplementation not only reduces the incidence of neural tube defects, but also improves cognitive performances in offspring. However, the genes or pathways that are epigenetically regulated by FA in neurodevelopment were rarely reported. Methods: To elucidate the underlying mechanism, the effect of FA on the methylation profiles in brain tissue of male rat offspring was assessed by methylated DNA immunoprecipitation chip. Differentially methylated genes (DMGs) and gene network analysis were identified using DAVID and KEGG pathway analysis. Results: Compared with the folate-normal diet group, 1939 DMGs were identified in the folate-deficient diet group, and 1498 DMGs were identified in the folate-supplemented diet group, among which 298 DMGs were overlapped. The pathways associated with neurodevelopment and learning/memory abilities were differentially methylated in response to maternal FA intake during pregnancy, and there were some identical and distinctive potential mechanisms under FA deficiency or FA-supplemented conditions. Conclusions: In conclusion, genes and pathways associated with neurodevelopment and learning/memory abilities were differentially methylated in male rat offspring in response to maternal FA deficiency or supplementation during pregnancy. Keywords: Folic acid, Pregnancy, DNA methylation, Learning/memory, Synaptic plasticity Introduction demonstrated that maternal FA supplementation during Emerging evidence has indicated that early life nutrition pregnancy promoted offspring’s neurogenesis and synap- influences brain development and exerts long-term conse- togenesis at birth [5], stimulated sensory-motor reflex de- quences for memory abilities profoundly [1, 2]. Pericon- velopment in infancy [6, 7], and exerted long-term ceptional folic acid (FA) supplementation reduces the beneficial effects on learning and memory abilities in ado- incidence of neural tube defects and improves cognitive lescence and adult in rat offspring [6]. Our previous study performances in offspring [3, 4]. Our previous studies also found maternal folic acid supplementation increased in the offspring’s brains the methylation potential, global DNA methylation level, and DNMT expression and activ- * Correspondence: [email protected] 1Department of Nutrition and Food Science, School of Public Health, Tianjin ity [7]. However, the genes or pathways that are epigeneti- Medical University, Tianjin 300070, China cally regulated by FA in neurodevelopment were rarely 3 Tianjin Key Laboratory of Environment, Nutrition and Public Health, Center reported. for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China Through one-carbon metabolism, folate provides a la- Full list of author information is available at the end of the article bile source of methyl groups for numerous methylation © The Author(s). 2021 Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. Wang et al. Genes & Nutrition (2021) 16:1 Page 2 of 11 reactions, including DNA methylation [8]. DNA methy- diet added a 1.4 mg folic acid/kg diet compared to the lation and histone modification are the most studied epi- folate-normal diet, which in rats is equivalent to con- genetic modifications reprogrammed considerably in suming a 400 μg folic acid supplement daily on consum- early embryonic development [9]. Epigenetic alterations ing a healthy diet in human. The FA-S group had the are thought to module gene expression and silencing highest body weight and brain weight of neonatal off- [10], and their dysregulation may contribute to numer- spring; however, there were no differences in brain ous neurodevelopment disorders [11, 12]. Maternal FA weight-to-body weight ratio of neonatal offspring within supplementation during pregnancy increased offspring’s groups. For running methylation analysis, three dams global DNA methylation level in rats [7] and influenced were chosen from each group using random numbers, the offspring’s repeat element and imprinted gene and then three neonatal male offspring derived from methylation in humans [13]. A meta-analysis that in- each dam also used random numbers. There were no cluded 1988 newborns from two European birth cohorts between-group differences in conception rate, live birth revealed that maternal plasma folate during pregnancy rate, stillbirth rate, absorbed embryo rate, or sex impacted epigenome-wide DNA methylation in the cord distribution. blood. And some of the implicated genes had functional The neonatal male offspring were sacrificed within 24 h relevance to neurodevelopment [4]. Lambrot et al. [10] after birth, and cerebral hemispheres were collected and provided evidence in mice that lifetime exposure to fol- stored at − 80 °C after liquid nitrogen flash-freezing for ate deficiency may cause an altered sperm epigenome further methylated DNA immunoprecipitation (MeDIP) and was associated with adverse reproductive outcomes. chip assay. Transient inhibition of DNA methylation by 5- Azacytidine treatment during development could induce MeDIP-Chip assay neurodegeneration in neonatal mice and long-lasting Genomic DNA was extracted from brain tissue using a deficits in synaptic plasticity and memory abilities in DNeasy Blood & Tissue Kit (Qiagen, USA). The purified adult mice [14]. Thereby DNA methylation modification DNA was quantified using nanodrop ND-1000. Then may play critical roles in neurodevelopment and learn- the genomic DNA was sonicated (Bioruptor sonicator, ing/memory ability in offspring. Diagenode, Belgian) to fragments between 200 and 1000 In the present study, male offspring derived from dams bp. The MeDIP was prepared as follows: 1 μg of soni- fed with FA-deficient, FA-normal, and FA-supplemented cated genomic DNA fragments was denatured for 10 diets throughout pregnancy were sacrificed within 24 h min at 94 °C, immunoprecipitated with mouse anti-5- after birth to detect DNA methylation profiles. We hy- methylcytosine antibody (1:400, Diagenode, Belgian) pothesized that maternal FA intake during pregnancy overnight at 4 °C, and incubated with anti-mouse IgG might alter DNA methylation profiles established in magnetic beads for 2 h at 4 °C. After washing, the beads- utero that subsequently regulated neurodevelopment DNA complex was resuspended in TE buffer with 0.25% and long-term learning/memory abilities in male SDS and 0.25 mg/mL proteinase K for 2 h at 65 °C. offspring. Lastly, the MeDIP DNA was purified using Qiagen MinElute columns (Qiagen, USA). Methods MeDIP DNA samples and Input samples were labeled Rats and dietary treatment with Cy5-9mer and Cy3-9mer primer, respectively, using As our previous study [6], 3-month-old female Sprague- a NimbleGen Dual-Color DNA Labeling Kit according Dawley rats (Charles River Laboratories, China) were to the manufacturer’s instruction (Nimblegen Systems, assigned randomly into three dietary groups (12 rats/ Inc., USA). Then the samples were hybridized to the group): (1) FA-deficient diet (FA-D), (2) FA-normal diet Arraystar Rat RefSeq Promoter Array, which is designed (FA-N), and (3) FA-supplemented diet (FA-S). Dietary to investigate the epigenetic modifications and transcrip- treatment began from 1 week before mating and tion factor binding sites within 15,987 RefSeq Gene pro- throughout the end of pregnancy. Rats were housed in a moter regions covered by approximately 180,000 probes. specific pathogen-free facility under a controlled 12-h Scanning was performed with the Agilent Scanner light/dark cycle and were provided with food and water G2505C microarray scanner. ad libitum. All animal procedures were approved by the Tianjin Medical University Animal Ethics Committee Identification of differentially methylated regions (TMUaEC2015001). The log2 (MeDIP/Input) getting from raw data was normal- Diet compositions were modified from the AIN-93G ized using the Median-centering, quantile normalization, and diet (TestDiet, USA), which contains 2.1 mg FA/kg diet. linear smoothing analysis by Bioconductor packages Ringo, The FA-D and FA-S diets contain 0.1 mg FA/kg diet and limma, and MEDME. Then from the normalized log2 3.5 mg FA/kg diet, respectively. The folate-supplemented (MeDIP/Input) data, a sliding-window peak-finding Wang et al. Genes & Nutrition (2021) 16:1 Page 3 of 11 algorithm provided by NimbleScan v2.5 (Roche-NimbleGen) Since one single peak can
Recommended publications
  • Original Article Upregulation of HOXA13 As a Potential Tumorigenesis and Progression Promoter of LUSC Based on Qrt-PCR and Bioinformatics
    Int J Clin Exp Pathol 2017;10(10):10650-10665 www.ijcep.com /ISSN:1936-2625/IJCEP0065149 Original Article Upregulation of HOXA13 as a potential tumorigenesis and progression promoter of LUSC based on qRT-PCR and bioinformatics Rui Zhang1*, Yun Deng1*, Yu Zhang1, Gao-Qiang Zhai1, Rong-Quan He2, Xiao-Hua Hu2, Dan-Ming Wei1, Zhen-Bo Feng1, Gang Chen1 Departments of 1Pathology, 2Medical Oncology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China. *Equal contributors. Received September 7, 2017; Accepted September 29, 2017; Epub October 1, 2017; Published October 15, 2017 Abstract: In this study, we investigated the levels of homeobox A13 (HOXA13) and the mechanisms underlying the co-expressed genes of HOXA13 in lung squamous cancer (LUSC), the signaling pathways in which the co-ex- pressed genes of HOXA13 are involved and their functional roles in LUSC. The clinical significance of 23 paired LUSC tissues and adjacent non-tumor tissues were gathered. HOXA13 levels in LUSC were detected by quantita- tive real-time polymerase chain reaction (qRT-PCR). HOXA13 levels in LUSC from The Cancer Genome Atlas (TCGA) and Oncomine were analyzed. We performed receiver operator characteristic (ROC) curves of various clinicopath- ological features of LUSC. Co-expressed of HOXA13 were collected from MEM, cBioPortal and GEPIA. The func- tions and pathways of the most reliable overlapped genes were achieved from the Gene Otology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) databases, respectively. The protein-protein interaction (PPI) net- works were mapped using STRING. HOXA13 in LUSC were markedly upregulated compared with those in the non- cancerous controls as demonstrated by qRT-PCR (LUSC: 0.330±0.360; CONTROLS: 0.155±0.142; P=0.021).
    [Show full text]
  • Dynamin Functions and Ligands: Classical Mechanisms Behind
    1521-0111/91/2/123–134$25.00 http://dx.doi.org/10.1124/mol.116.105064 MOLECULAR PHARMACOLOGY Mol Pharmacol 91:123–134, February 2017 Copyright ª 2017 by The American Society for Pharmacology and Experimental Therapeutics MINIREVIEW Dynamin Functions and Ligands: Classical Mechanisms Behind Mahaveer Singh, Hemant R. Jadhav, and Tanya Bhatt Department of Pharmacy, Birla Institute of Technology and Sciences Pilani, Pilani Campus, Rajasthan, India Received May 5, 2016; accepted November 17, 2016 Downloaded from ABSTRACT Dynamin is a GTPase that plays a vital role in clathrin-dependent pathophysiology of various disorders, such as Alzheimer’s disease, endocytosis and other vesicular trafficking processes by acting Parkinson’s disease, Huntington’s disease, Charcot-Marie-Tooth as a pair of molecular scissors for newly formed vesicles originating disease, heart failure, schizophrenia, epilepsy, cancer, dominant ’ from the plasma membrane. Dynamins and related proteins are optic atrophy, osteoporosis, and Down s syndrome. This review is molpharm.aspetjournals.org important components for the cleavage of clathrin-coated vesicles, an attempt to illustrate the dynamin-related mechanisms involved phagosomes, and mitochondria. These proteins help in organelle in the above-mentioned disorders and to help medicinal chemists division, viral resistance, and mitochondrial fusion/fission. Dys- to design novel dynamin ligands, which could be useful in the function and mutations in dynamin have been implicated in the treatment of dynamin-related disorders. Introduction GTP hydrolysis–dependent conformational change of GTPase dynamin assists in membrane fission, leading to the generation Dynamins were originally discovered in the brain and identi- of endocytic vesicles (Praefcke and McMahon, 2004; Ferguson at ASPET Journals on September 23, 2021 fied as microtubule binding partners.
    [Show full text]
  • PERK Antibody / EIF2AK3 (RQ4206)
    PERK Antibody / EIF2AK3 (RQ4206) Catalog No. Formulation Size RQ4206 0.5mg/ml if reconstituted with 0.2ml sterile DI water 100 ug Bulk quote request Availability 1-3 business days Species Reactivity Human, Mouse, Rat Format Antigen affinity purified Clonality Polyclonal (rabbit origin) Isotype Rabbit IgG Purity Antigen affinity purified Buffer Lyophilized from 1X PBS with 2% Trehalose and 0.025% sodium azide UniProt Q9NZJ5 Applications Western Blot : 0.5-1ug/ml Flow cytometry : 1-3ug/10^6 cells Direct ELISA : 0.1-0.5ug/ml Limitations This PERK antibody is available for research use only. Western blot testing of human 1) HeLa, 2) COLO320, 3) A549, 4) SK-OV-3, 5) A431, 6) rat brain and 7) mouse brain lysate with PERK antibody at 0.5ug/ml. Predicted molecular weight ~125 kDa, observed here at ~140 kDa. Flow cytometry testing of human HepG2 cells with PERK antibody at 1ug/10^6 cells (blocked with goat sera); Red=cells alone, Green=isotype control, Blue= PERK antibody. Description Eukaryotic translation initiation factor 2-alpha kinase 3, also known as protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK), is an enzyme that in humans is encoded by the EIF2AK3 gene. The protein encoded by this gene phosphorylates the alpha subunit of eukaryotic translation-initiation factor 2, leading to its inactivation, and thus to a rapid reduction of translational initiation and repression of global protein synthesis. This protein is thought to modulate mitochondrial function. It is a type I membrane protein located in the endoplasmic reticulum (ER), where it is induced by ER stress caused by malfolded proteins.
    [Show full text]
  • The Rise and Fall of the Bovine Corpus Luteum
    University of Nebraska Medical Center DigitalCommons@UNMC Theses & Dissertations Graduate Studies Spring 5-6-2017 The Rise and Fall of the Bovine Corpus Luteum Heather Talbott University of Nebraska Medical Center Follow this and additional works at: https://digitalcommons.unmc.edu/etd Part of the Biochemistry Commons, Molecular Biology Commons, and the Obstetrics and Gynecology Commons Recommended Citation Talbott, Heather, "The Rise and Fall of the Bovine Corpus Luteum" (2017). Theses & Dissertations. 207. https://digitalcommons.unmc.edu/etd/207 This Dissertation is brought to you for free and open access by the Graduate Studies at DigitalCommons@UNMC. It has been accepted for inclusion in Theses & Dissertations by an authorized administrator of DigitalCommons@UNMC. For more information, please contact [email protected]. THE RISE AND FALL OF THE BOVINE CORPUS LUTEUM by Heather Talbott A DISSERTATION Presented to the Faculty of the University of Nebraska Graduate College in Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy Biochemistry and Molecular Biology Graduate Program Under the Supervision of Professor John S. Davis University of Nebraska Medical Center Omaha, Nebraska May, 2017 Supervisory Committee: Carol A. Casey, Ph.D. Andrea S. Cupp, Ph.D. Parmender P. Mehta, Ph.D. Justin L. Mott, Ph.D. i ACKNOWLEDGEMENTS This dissertation was supported by the Agriculture and Food Research Initiative from the USDA National Institute of Food and Agriculture (NIFA) Pre-doctoral award; University of Nebraska Medical Center Graduate Student Assistantship; University of Nebraska Medical Center Exceptional Incoming Graduate Student Award; the VA Nebraska-Western Iowa Health Care System Department of Veterans Affairs; and The Olson Center for Women’s Health, Department of Obstetrics and Gynecology, Nebraska Medical Center.
    [Show full text]
  • Adenylate Cyclase 3: a New Target for Anti‐Obesity Drug Development
    obesity reviews doi: 10.1111/obr.12430 Obesity Pharmacotherapy Adenylate cyclase 3: a new target for anti-obesity drug development L. Wu,1 C. Shen,2 M. Seed Ahmed,3 C.-G. Östenson4 and H. F. Gu5,6 1Jiangsu Key Laboratory of Drug Screening, Summary China Pharmaceutical University, Nanjing Obesity has become epidemic worldwide, and abdominal obesity has a negative im- 210009, China, 2Department of Epidemiology, pact on health. Current treatment options on obesity, however, still remain limited. School of Public Health, Nanjing Medical It is then of importance to find a new target for anti-obesity drug development University, Nanjing 211166, China, 3Unit for based upon recent molecular studies in obesity. Adenylate cyclase 3 (ADCY3) is Medical Education, Centre for Learning and the third member of adenylyl cyclase family and catalyses the synthesis of cAMP Knowledge, Department of Learning, from ATP. Genetic studies with candidate gene and genome-wide association study Informatics, Management and Ethics, approaches have demonstrated that ADCY3 genetic polymorphisms are associated Karolinska Institutet, Stockholm 17177, with obesity in European and Chinese populations. Epigenetic studies have indi- Sweden, 4Rolf Luft Center for Diabetes cated that increased DNA methylation levels in the ADCY3 gene are involved in Research and Endocrinology, Department of the pathogenesis of obesity. Furthermore, biological analyses with animal models Molecular Medicine and Surgery, Karolinska have implicated that ADCY3 dysfunction resulted in increased body weight and Institutet, Karolinska University Hospital, Solna, fat mass, while reduction of body weight is partially explained by ADCY3 activa- Stockholm 17176, Sweden, 5Department of tion. In this review, we describe genomic and biological features of ADCY3, sum- Clinical Science, Intervention and marize genetic and epigenetic association studies of the ADCY3 gene with obesity Technologies, Karolinska Institutet, Karolinska and discuss dysfunction and activation of ADCY3.
    [Show full text]
  • Supplemental Table S1
    Entrez Gene Symbol Gene Name Affymetrix EST Glomchip SAGE Stanford Literature HPA confirmed Gene ID Profiling profiling Profiling Profiling array profiling confirmed 1 2 A2M alpha-2-macroglobulin 0 0 0 1 0 2 10347 ABCA7 ATP-binding cassette, sub-family A (ABC1), member 7 1 0 0 0 0 3 10350 ABCA9 ATP-binding cassette, sub-family A (ABC1), member 9 1 0 0 0 0 4 10057 ABCC5 ATP-binding cassette, sub-family C (CFTR/MRP), member 5 1 0 0 0 0 5 10060 ABCC9 ATP-binding cassette, sub-family C (CFTR/MRP), member 9 1 0 0 0 0 6 79575 ABHD8 abhydrolase domain containing 8 1 0 0 0 0 7 51225 ABI3 ABI gene family, member 3 1 0 1 0 0 8 29 ABR active BCR-related gene 1 0 0 0 0 9 25841 ABTB2 ankyrin repeat and BTB (POZ) domain containing 2 1 0 1 0 0 10 30 ACAA1 acetyl-Coenzyme A acyltransferase 1 (peroxisomal 3-oxoacyl-Coenzyme A thiol 0 1 0 0 0 11 43 ACHE acetylcholinesterase (Yt blood group) 1 0 0 0 0 12 58 ACTA1 actin, alpha 1, skeletal muscle 0 1 0 0 0 13 60 ACTB actin, beta 01000 1 14 71 ACTG1 actin, gamma 1 0 1 0 0 0 15 81 ACTN4 actinin, alpha 4 0 0 1 1 1 10700177 16 10096 ACTR3 ARP3 actin-related protein 3 homolog (yeast) 0 1 0 0 0 17 94 ACVRL1 activin A receptor type II-like 1 1 0 1 0 0 18 8038 ADAM12 ADAM metallopeptidase domain 12 (meltrin alpha) 1 0 0 0 0 19 8751 ADAM15 ADAM metallopeptidase domain 15 (metargidin) 1 0 0 0 0 20 8728 ADAM19 ADAM metallopeptidase domain 19 (meltrin beta) 1 0 0 0 0 21 81792 ADAMTS12 ADAM metallopeptidase with thrombospondin type 1 motif, 12 1 0 0 0 0 22 9507 ADAMTS4 ADAM metallopeptidase with thrombospondin type 1
    [Show full text]
  • Exosome-Mediated MIR211 Modulates Tumor Microenvironment Via the DUSP6-ERK5 Axis and Contributes to BRAFV600E Inhibitor Resistan
    bioRxiv preprint doi: https://doi.org/10.1101/548818; this version posted February 13, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Exosome-mediated MIR211 modulates tumor microenvironment via the DUSP6-ERK5 axis and contributes to BRAFV600E inhibitor resistance in melanoma. Bongyong Lee1,3, Anupama Sahoo3, Junko Sawada1,3, John Marchica1,3, Sanjay Sahoo3, Fabiana I. A. L. Layng4, Darren Finlay4, Joseph Mazar5, Piyush Joshi1, Masanobu Komatsu1,3, Kristiina Vuori4, Garth Powis4, Petrus R. de Jong4, Animesh Ray6,7, and Ranjan J. Perera 1,2,3* 1 Department of Cancer and Blood Disorders Institute, Johns Hopkins All Children’s Hospital, St. Petersburg, FL 33701 USA 2 Department of Oncology, Sydney Kimmel Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287 USA 3 Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL 32827 USA 4 Sanford Burnham Prebys Medical Discovery Institute, NCI-Designated Cancer Center, La Jolla, CA 92037, USA 5 Nemours Children Hospital, Orlando, FL 32827 USA 6 Keck Graduate Institute, Claremont CA 91711 USA, 7 Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125 USA. *Correspondence: Ranjan Perera Tel: 1-727-767-3491 Email: [email protected] 1 bioRxiv preprint doi: https://doi.org/10.1101/548818; this version posted February 13, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. ABSTRACT The microRNA MIR211 is an important regulator of melanoma tumor cell behavior.
    [Show full text]
  • Dual-Specificity Phosphatase 3 Deletion Protects Female, but Not
    Published August 28, 2017, doi:10.4049/jimmunol.1602092 The Journal of Immunology Dual-Specificity Phosphatase 3 Deletion Protects Female, but Not Male, Mice from Endotoxemia-Induced and Polymicrobial-Induced Septic Shock Maud M. Vandereyken,*,1 Pratibha Singh,*,1 Caroline P. Wathieu,* Sophie Jacques,* Tinatin Zurashvilli,* Lien Dejager,†,‡ Mathieu Amand,* Lucia Musumeci,* Maneesh Singh,* Michel P. Moutschen,* Claude R. F. Libert,†,‡ and Souad Rahmouni* Dual-specificity phosphatase 3 (DUSP3) is a small phosphatase with poorly known physiological functions and for which only a few substrates are known. Using knockout mice, we recently reported that DUSP3 deficiency confers resistance to endotoxin- and polymicrobial-induced septic shock. We showed that this protection was macrophage dependent. In this study, we further investigated the role of DUSP3 in sepsis tolerance and showed that the resistance is sex dependent. Using adoptive-transfer experiments and ovariectomized mice, we highlighted the role of female sex hormones in the phenotype. Indeed, in ovariec- tomized females and in male mice, the dominance of M2-like macrophages observed in DUSP32/2 female mice was reduced, suggesting a role for this cell subset in sepsis tolerance. At the molecular level, DUSP3 deletion was associated with estrogen- dependent decreased phosphorylation of ERK1/2 and Akt in peritoneal macrophages stimulated ex vivo by LPS. Our results demonstrate that estrogens may modulate M2-like responses during endotoxemia in a DUSP3-dependent manner. The Journal of Immunology, 2017, 199: 000–000. epsis and septic shock are complex clinical syndromes that ally, death (4). Sepsis occurrence and outcome depend on arise when the local body response to pathogens becomes pathogen characteristics, as well as on risk factors, such as age S systemic and injures its own tissues and organs (1).
    [Show full text]
  • De Novo EIF2AK1 and EIF2AK2 Variants Are Associated with Developmental Delay, Leukoencephalopathy, and Neurologic Decompensation
    bioRxiv preprint doi: https://doi.org/10.1101/757039; this version posted September 16, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. De novo EIF2AK1 and EIF2AK2 variants are associated with developmental delay, leukoencephalopathy, and neurologic decompensation Dongxue Mao1,2, Chloe M. Reuter3,4, Maura R.Z. Ruzhnikov5,6, Anita E. Beck7, Emily G. Farrow8,9,10, Lisa T. Emrick1,11,12,13, Jill A. Rosenfeld12, Katherine M. Mackenzie5, Laurie Robak2,12,13, Matthew T. Wheeler3,14, Lindsay C. Burrage12,13, Mahim Jain15, Pengfei Liu12, Daniel Calame11,13, Sebastien Küry17,18, Martin Sillesen19, Klaus Schmitz-Abe20, Davide Tonduti21, Luigina Spaccini22, Maria Iascone23, Casie A. Genetti20, Madeline Graf16, Alyssa Tran12, Mercedes Alejandro12, Undiagnosed Diseases Network, Brendan H. Lee12,13, Isabelle Thiffault8,9,24, Pankaj B. Agrawal#,20, Jonathan A. Bernstein#,3,25, Hugo J. Bellen#,2,12,26,27,28, Hsiao- Tuan Chao#,1,2,11,12,13,28,27,29 #Correspondence should be addressed: [email protected] (P.A.), [email protected] (J.A.B.), [email protected] (H.J.B.), [email protected] (H.T.C.) 1Department of Pediatrics, Baylor College of Medicine (BCM), Houston, TX 2Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX 3Stanford Center for Undiagnosed Diseases, Stanford University, Stanford, CA 4Stanford Center for Inherited Cardiovascular Disease, Division of Cardiovascular Medicine,
    [Show full text]
  • CXCR4 Pathway Retards Muscle Atrophy During Cancer Cachexia
    Oncogene (2016) 35, 6212–6222 © 2016 Macmillan Publishers Limited, part of Springer Nature. All rights reserved 0950-9232/16 www.nature.com/onc ORIGINAL ARTICLE Activation of the SDF1/CXCR4 pathway retards muscle atrophy during cancer cachexia GB Martinelli1, D Olivari1, AD Re Cecconi1, L Talamini1, L Ottoboni2, SH Lecker3, C Stretch4, VE Baracos4, OF Bathe5, A Resovi6, R Giavazzi1, L Cervo7 and R Piccirillo1 Cancer cachexia is a life-threatening syndrome that affects most patients with advanced cancers and causes severe body weight loss, with rapid depletion of skeletal muscle. No treatment is available. We analyzed microarray data sets to identify a subset of genes whose expression is specifically altered in cachectic muscles of Yoshida hepatoma-bearing rodents but not in those with diabetes, disuse, uremia or fasting. Ingenuity Pathways Analysis indicated that three genes belonging to the C-X-C motif chemokine receptor 4 (CXCR4) pathway were downregulated only in muscles atrophying because of cancer: stromal cell-derived factor 1 (SDF1), adenylate cyclase 7 (ADCY7), and p21 protein-activated kinase 1 (PAK1). Notably, we found that, in the Rectus Abdominis muscle of cancer patients, the expression of SDF1 and CXCR4 was inversely correlated with that of two ubiquitin ligases induced in muscle wasting, atrogin-1 and MuRF1, suggesting a possible clinical relevance of this pathway. The expression of all main SDF1 isoforms (α, β, γ) also declined in Tibialis Anterior muscle from cachectic mice bearing murine colon adenocarcinoma or human renal cancer and drugs with anticachexia properties restored their expression. Overexpressing genes of this pathway (that is, SDF1 or CXCR4) in cachectic muscles increased the fiber area by 20%, protecting them from wasting.
    [Show full text]
  • Genome-Wide Association and Transcriptome Studies Identify Candidate Genes and Pathways for Feed Conversion Ratio in Pigs
    Miao et al. BMC Genomics (2021) 22:294 https://doi.org/10.1186/s12864-021-07570-w RESEARCH ARTICLE Open Access Genome-wide association and transcriptome studies identify candidate genes and pathways for feed conversion ratio in pigs Yuanxin Miao1,2,3, Quanshun Mei1,2, Chuanke Fu1,2, Mingxing Liao1,2,4, Yan Liu1,2, Xuewen Xu1,2, Xinyun Li1,2, Shuhong Zhao1,2 and Tao Xiang1,2* Abstract Background: The feed conversion ratio (FCR) is an important productive trait that greatly affects profits in the pig industry. Elucidating the genetic mechanisms underpinning FCR may promote more efficient improvement of FCR through artificial selection. In this study, we integrated a genome-wide association study (GWAS) with transcriptome analyses of different tissues in Yorkshire pigs (YY) with the aim of identifying key genes and signalling pathways associated with FCR. Results: A total of 61 significant single nucleotide polymorphisms (SNPs) were detected by GWAS in YY. All of these SNPs were located on porcine chromosome (SSC) 5, and the covered region was considered a quantitative trait locus (QTL) region for FCR. Some genes distributed around these significant SNPs were considered as candidates for regulating FCR, including TPH2, FAR2, IRAK3, YARS2, GRIP1, FRS2, CNOT2 and TRHDE. According to transcriptome analyses in the hypothalamus, TPH2 exhibits the potential to regulate intestinal motility through serotonergic synapse and oxytocin signalling pathways. In addition, GRIP1 may be involved in glutamatergic and GABAergic signalling pathways, which regulate FCR by affecting appetite in pigs. Moreover, GRIP1, FRS2, CNOT2,andTRHDE may regulate metabolism in various tissues through a thyroid hormone signalling pathway.
    [Show full text]
  • Gene and Pathway-Based Second-Wave Analysis of Genome-Wide Association Studies
    European Journal of Human Genetics (2010) 18, 111–117 & 2010 Macmillan Publishers Limited All rights reserved 1018-4813/10 $32.00 www.nature.com/ejhg ARTICLE Gene and pathway-based second-wave analysis of genome-wide association studies Gang Peng1, Li Luo2, Hoicheong Siu1, Yun Zhu1, Pengfei Hu1, Shengjun Hong1, Jinying Zhao3, Xiaodong Zhou4, John D Reveille4, Li Jin1, Christopher I Amos5 and Momiao Xiong*,2 Despite the great success of genome-wide association studies (GWAS) in identification of the common genetic variants associated with complex diseases, the current GWAS have focused on single-SNP analysis. However, single-SNP analysis often identifies only a few of the most significant SNPs that account for a small proportion of the genetic variants and offers only a limited understanding of complex diseases. To overcome these limitations, we propose gene and pathway-based association analysis as a new paradigm for GWAS. As a proof of concept, we performed a comprehensive gene and pathway-based association analysis of 13 published GWAS. Our results showed that the proposed new paradigm for GWAS not only identified the genes that include significant SNPs found by single-SNP analysis, but also detected new genes in which each single SNP conferred a small disease risk; however, their joint actions were implicated in the development of diseases. The results also showed that the new paradigm for GWAS was able to identify biologically meaningful pathways associated with the diseases, which were confirmed by a gene-set-rich analysis using gene expression
    [Show full text]