Targeting Fbpase Is an Emerging Novel Approach for Cancer Therapy Gao‑Min Liu and Yao‑Ming Zhang*

Total Page:16

File Type:pdf, Size:1020Kb

Targeting Fbpase Is an Emerging Novel Approach for Cancer Therapy Gao‑Min Liu and Yao‑Ming Zhang* Liu and Zhang Cancer Cell Int (2018) 18:36 https://doi.org/10.1186/s12935-018-0533-z Cancer Cell International REVIEW Open Access Targeting FBPase is an emerging novel approach for cancer therapy Gao‑Min Liu and Yao‑Ming Zhang* Abstract Cancer is a leading cause of death in both developed and developing countries. Metabolic reprogramming is an emerging hallmark of cancer. Glucose homeostasis is reciprocally controlled by the catabolic glycolysis and anabolic gluconeogenesis pathways. Previous studies have mainly focused on catabolic glycolysis, but recently, FBPase, a rate- limiting enzyme in gluconeogenesis, was found to play critical roles in tumour initiation and progression in several cancer types. Here, we review recent ideas and discoveries that illustrate the clinical signifcance of FBPase expression in various cancers, the mechanism through which FBPase infuences cancer, and the mechanism of FBPase silencing. Furthermore, we summarize some of the drugs targeting FBPase and discuss their potential use in clinical applications and the problems that remain unsolved. Keywords: FBPase, Cancer, Clinical signifcance, Mechanism Background [8, 9]. Aerobic glycolysis leads to several advantages for Cancer is one of the leading causes of death worldwide tumour growth by increasing glucose intake, enhancing [1]. In the past decade, reprogramming of energy metab- lactate production and secretion, and diverting glycolytic olism has emerged as new hallmarks of cancer [2]. Tere intermediates to anabolic reactions such as ribose syn- is increasing epidemiological evidence that links the risk thesis, serine and glycine synthesis, phosphoglycerol syn- of cancer with metabolic disorders, such as diabetes and thesis, and protein glycosylation [10]. Moreover, recent obesity [3]. However, the complete regulatory network of studies have developed promising individualized thera- metabolism reprogramming remains to be elucidated. peutic strategies by targeting the altered energy metabo- Glucose homeostasis is reciprocally controlled by the lism for the treatment of cancer [11]. catabolic glycolysis and anabolic gluconeogenesis path- Tough our understanding of metabolic reprogram- ways. Under normoxic conditions, normal cells process ming in cancer is progressing at an unprecedented glucose frst to pyruvate via glycolysis in the cytosol and pace, previous studies have mainly focused on glyco- thereafter to carbon dioxide in the mitochondria (oxida- lysis. Recently, fructose-1,6-bisphosphatase (FBPase), a tive phosphorylation, OXPHOS); under anaerobic condi- rate-limiting enzyme responsible for gluconeogenesis, tions, glycolysis is favoured, and relatively little pyruvate was found to play critical roles in tumour initiation and is dispatched to the oxygen-consuming mitochondria [4, progression in several cancer types [12–28]. Here, we 5]. However, as frst observed by Otto Warburg in the search Pubmed, EMbase, Web of Science using keywords 1920s, some cancer cells preferentially rely on glycolysis, “FBPase” and “cancer” (the most recent published report even in conditions of high oxygen tension (‘‘aerobic gly- search date: 1 August 2017) and review recent ideas and colysis” or “the Warburg efect’’) [6, 7]. Aerobic glycolysis discoveries regarding the role of FBPase in cancer. was validated with the wide use of 18F-deoxyglucose posi- tron emission tomography (FDG-PET) in most cancers FBPase location and regulation FBPase (EC 3.1.3.11), which catalyses the hydrolysis of fructose 1,6-bisphosphate to fructose 6-phosphate and *Correspondence: [email protected] inorganic phosphate, is a rate-limiting enzyme responsi- Department of Hepatobiliary Surgery, Meizhou People’s Hospital, No. 38 Huangtang Road, Meizhou 514000, China ble for gluconeogenesis and glyconeogenesis and, more © The Author(s) 2018. This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/ publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated. Liu and Zhang Cancer Cell Int (2018) 18:36 Page 2 of 16 generally, for the control of energy metabolism and glu- and kidneys, is located at chromosome region 9q22.3. cose homeostasis [29]. In mammals, two separate genes, FBP1 consists of seven exons which span more than FBP1 and FBP2, encode the liver and muscle isoforms 31 kb that encode 338 amino acids and six introns [30]. of FBPase, respectively. Te FBP1 gene, which is mainly Te FBP1 promoter has been well characterized since expressed in gluconeogenic organs, such as the liver 2000 [31]. Te FBP2 gene, which was initially isolated Fig. 1 Amino acid sequences and characteristic homotetrameric structure of human FBPase. This fgure provides the human FBP1 and FBP2 amino acid sequence. Human FBP2 is 77% identical to human FBP2 [34]. The sequence alignments were run through BLAST. The characteristic homotetra‑ meric structure of human FBPase was shown [35] Liu and Zhang Cancer Cell Int (2018) 18:36 Page 3 of 16 from muscle tissues and later found to be expressed in FBPase-2, leading to the dephosphorylation of F2,6BP all cells, is located at chromosome region 1p36.1 [32]. and concomitant increases in FBPase activity and glu- FBP2 encodes a 339-amino acid protein that shares 77% coneogenesis [45]. In addition, PKA activates cAMP identity with the FBP1 protein [33]. However, the pro- response element-binding protein (CREB), the CREB moter of the FBP2 gene has not yet been characterized. coactivator CRTC2 [52], and the class II histone deacety- All FBPases are homotetrameric proteins with a molecu- lases (HDACs)/FOXO pathways [53, 54] and promotes lar weight of approximately 37 kDa per subunit (Fig. 1). the expression of key gluconeogenic genes; however, it FBP1 has been identifed as the regulatory enzyme of suppresses the expression of glycolytic genes. Gluco- gluconeogenesis, but the physiological role of FBP2 is far corticoid hormones and glucagon plays essential syn- from clear. ergistic roles in the regulation of gluconeogenesis [55]. Acute regulation of FBPase is achieved mainly through Te response to glucocorticoids is mediated by the glu- its oscillation between two conformational states: the cocorticoid receptor (GR), which binds to glucocorti- inactive T form when in complex with AMP and the coid responsive elements (GREs) in the promoters of active R form [29, 36–40]. By modulating pyruvate kinase gluconeogenic genes [56]. In contrast, insulin acts to activity, fructose-1,6-bisphosphate (F1,6BP) afects repress the transcription of gluconeogenic enzymes. Tis substrate cycling in the pyruvatel/phosphoenolpyru- repression of the expression of gluconeogenic enzymes vate (PEP) substrate cycle [41]. Phosphofructokinase-1 is achieved via the activation of the insulin-phosphoi- (PFK1), the “gatekeeper” of glycolysis, converts fructose- nositide 3-kinase (PI3K)-Akt pathway or suppression of 6-phosphate (F6P) to F1,6BP and has the opposite efect the cAMP/PKA/CREB pathway [57] (Fig. 2). of FBPase in regulating F1,6BP [42]. Fructose-2,6-bi- sphosphate (F2,6BP) is a potent allosteric activator of FBPase and non‑cancerous diseases PFK1 and a competitive inhibitor of FBPase [5, 41–44]. FBP1 defciency When the level of F2,6BP is low, the rates of gluconeo- Mutations in the FBP1 gene cause FBP1 defciency, an genesis are high (such as in starvation and diabetes). inherited autosomal recessive disorder, which leads to Conversely, when the level of F2,6BP is high, the rate of the impairment of glucose synthesis from all gluconeo- gluconeogenesis is low (such as during refeeding and genic precursors [58]. Tis defciency was frst described insulin administration) [5]. Te synthesis and hydrolysis by Baker and Winegrad in 1970 [59]. Tis disorder is of this regulator are catalysed by the bifunctional enzyme characterized by recurrent episodes of hypoglycaemia 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase and metabolic acidosis during fasting, with symptoms (PFK-2/FBPase-2) [45]. Phosphorylation of this enzyme usually manifesting during the frst days of life [60–62]. by cAMP-dependent protein kinase results in inhibi- If not treated appropriately, FBP1 defciency leads to tion of the kinase and activation of the bisphosphatase, unexpected infant death [63]. However, with diet control whereas dephosphorylation has the opposite efect [36, and avoidance of prolonged fasting, most adult patients 40]. TIGAR (TP53-induced glycolysis and apoptosis exhibit relatively normal clinical profles. regulator), a recently identifed enzyme, shares similari- ties with the bisphosphatase domain of PFK-2/FBPase-2 FBPase and type 2 diabetes [46]. TIGAR acts to degrade intracellular F2,6BP [47]. Blood glucose levels are elevated in type 2 diabetes Interestingly, FBP1 and FBP2 difer signifcantly in their (T2DM) due to impaired insulin secretion resulting from kinetic properties. FBP2 is approximately 100 times declining β-cell function; decreased glucose uptake by tis- more susceptible to the allosteric inhibitors AMP and sues such as muscle, liver, and fat; and increased hepatic NAD+ [48] and approximately 1000 times more sensitive glucose production (HGP) [64].
Recommended publications
  • Mechanism of CREB Recognition and Coactivation by the CREB-Regulated Transcriptional Coactivator CRTC2
    Mechanism of CREB recognition and coactivation by the CREB-regulated transcriptional coactivator CRTC2 Qianyi Luoa, Kristin Visteb, Janny Concha Urday-Zaaa, Ganesan Senthil Kumara, Wen-Wei Tsaib, Afsaneh Talaia, Kelly E. Mayoa, Marc Montminyb,1, and Ishwar Radhakrishnana,1 aDepartment of Molecular Biosciences, Northwestern University, Evanston, IL 60208; and bClayton Foundation Laboratories for Peptide Biology, Salk Institute for Biological Studies, La Jolla, CA 92037 Contributed by Marc Montminy, November 2, 2012 (sent for review October 8, 2012) Basic leucine zipper (bZip) transcription factors regulate cellular program (10). Sequestered in the cytoplasm under feeding con- gene expression in response to a variety of extracellular signals ditions through phosphorylation by the Ser/Thr kinase SIK2, and nutrient cues. Although the bZip domain is widely known to CRTC2 is dephosphorylated in response to pancreatic glucagon play significant roles in DNA binding and dimerization, recent during fasting, when it translocates to the nucleus and stimulates studies point to an additional role for this motif in the recruitment gluconeogenic gene expression. of the transcriptional apparatus. For example, the cAMP response Recent studies support a conserved role for CRTCs through evolution. Deletion of TORC, the single CRTC homolog in Dro- element binding protein (CREB)-regulated transcriptional coacti- sophila, reduces fat stores and increases sensitivity to starvation vator (CRTC) family of transcriptional coactivators has been pro- (11). CRTCs also seem to be important in aging: RNAi-mediated posed to promote the expression of calcium and cAMP responsive depletion of CRTC1 in Caenorhabditis elegans extends lifespan, in genes, by binding to the CREB bZip in response to extracellular part through down-regulation of the CREB pathway (12).
    [Show full text]
  • Genome-Wide Regulatory Roles of the C2H2-Type Zinc Finger Protein
    www.nature.com/scientificreports OPEN Genome-wide Regulatory Roles of the C2H2-type Zinc Finger Protein ZNF764 on the Glucocorticoid Received: 09 June 2016 Accepted: 23 December 2016 Receptor Published: 31 January 2017 Abeer Fadda1, Najeeb Syed2, Rafah Mackeh1, Anna Papadopoulou3, Shigeru Suzuki3,4, Puthen V. Jithesh2 & Tomoshige Kino1,3 The C2H2-type zinc finger protein ZNF764 acts as an enhancer for several steroid hormone receptors, and haploinsufficiency of this gene may be responsible for tissue resistance to multiple steroid hormones including glucocorticoids observed in a patient with 16p11.2 microdeletion. We examined genome-wide regulatory actions of ZNF764 on the glucocorticoid receptor (GR) in HeLa cells as a model system. ZNF764- and GR-binding sites demonstrated similar distribution in various genomic features. They positioned predominantly around 50–500 kbs from the transcription start sites of their nearby genes, and were closely localized with each other, overlapping in ~37% of them. ZNF764 demonstrated differential on/off effects on GR-binding and subsequent mRNA expression: some genes were highly dependent on the presence/absence of ZNF764, but others were not. Pathway analysis revealed that these 3 gene groups were involved in distinct cellular activities. ZNF764 physically interacted with GR at ligand-binding domain through its KRAB domain, and both its physical interaction to GR and zinc finger domain appear to be required for ZNF764 to regulate GR transcriptional activity. Thus, ZNF764 is a cofactor directing GR transcriptional activity toward specific biologic pathways by changing GR binding and transcriptional activity on the glucocorticoid-responsive genes. Steroid hormones exert diverse physiologic functions and play central roles in human physiology1,2.
    [Show full text]
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • The Essential Role of the Crtc2-CREB Pathway in Β Cell Function And
    The Essential Role of the Crtc2-CREB Pathway in β cell Function and Survival by Chandra Eberhard A thesis submitted to the School of Graduate Studies and Research in partial fulfillment of the requirements for the degree of Masters of Science in Cellular and Molecular Medicine Department of Cellular and Molecular Medicine Faculty of Medicine University of Ottawa September 28, 2012 © Chandra Eberhard, Ottawa, Canada, 2013 Abstract: Immunosuppressants that target the serine/threonine phosphatase calcineurin are commonly administered following organ transplantation. Their chronic use is associated with reduced insulin secretion and new onset diabetes in a subset of patients, suggestive of pancreatic β cell dysfunction. Calcineurin plays a critical role in the activation of CREB, a key transcription factor required for β cell function and survival. CREB activity in the islet is activated by glucose and cAMP, in large part due to activation of Crtc2, a critical coactivator for CREB. Previous studies have demonstrated that Crtc2 activation is dependent on dephosphorylation regulated by calcineurin. In this study, we sought to evaluate the impact of calcineurin-inhibiting immunosuppressants on Crtc2-CREB activation in the primary β cell. In addition, we further characterized the role and regulation of Crtc2 in the β cell. We demonstrate that Crtc2 is required for glucose dependent up-regulation of CREB target genes. The phosphatase calcineurin and kinase regulation by LKB1 contribute to the phosphorylation status of Crtc2 in the β cell. CsA and FK506 block glucose-dependent dephosphorylation and nuclear translocation of Crtc2. Overexpression of a constitutively active mutant of Crtc2 that cannot be phosphorylated at Ser171 and Ser275 enables CREB activity under conditions of calcineurin inhibition.
    [Show full text]
  • PRMT5 Modulates the Metabolic Response to Fasting Signals
    PRMT5 modulates the metabolic response to fasting signals Wen-Wei Tsaia, Sherry Niessenb, Naomi Goebela, John R. Yates IIIb, Ernesto Guccionec,d, and Marc Montminya,1 aThe Clayton Foundation Laboratories for Peptide Biology, Salk Institute, La Jolla, CA 92037; bDepartment of Chemical Physiology, The Scripps Research Institute, La Jolla, CA 92037; cInstitute of Molecular and Cell Biology, Proteos, Singapore 138673; dDepartment of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119074 Contributed by Marc Montminy, March 11, 2013 (sent for review February 20, 2013) Under fasting conditions, increases in circulating glucagon maintain sites. The results provide a mechanism to explain how latent cy- glucose balance by promoting hepatic gluconeogenesis. Triggering toplasmic regulators such as CRTC2 may contribute to signaling of the cAMP pathway stimulates gluconeogenic gene expression in the nucleus through their association with chromatin mod- through the PKA-mediated phosphorylation of the cAMP response ifying enzymes. element binding (CREB) protein and via the dephosphorylation of the latent cytoplasmic CREB regulated transcriptional coactivator 2 Results (CRTC2). CREB and CRTC2 activities are increased in insulin resis- In mass spectroscopy studies using epitope-tagged CRTC2 to tance, in which they promote hyperglycemia because of constitu- identify relevant interacting proteins, we recovered the protein tive induction of the gluconeogenic program. The extent to which arginine methyltransferase 5 (PRMT5) (Fig. S1 A and B). We CREB and CRTC2 are coordinately up-regulated in response to glu- confirmed the CRTC2:PRMT5 interaction in coimmunopreci- cagon, however, remains unclear. Here we show that, following its pitation studies with epitope-tagged proteins (Fig.
    [Show full text]
  • Adiponectin Suppresses Gluconeogenic Gene Expression in Mouse Hepatocytes Independent of LKB1-AMPK Signaling
    Adiponectin suppresses gluconeogenic gene expression in mouse hepatocytes independent of LKB1-AMPK signaling Russell A. Miller, … , Benoit Viollet, Morris J. Birnbaum J Clin Invest. 2011;121(6):2518-2528. https://doi.org/10.1172/JCI45942. Research Article Metabolism The adipocyte-derived hormone adiponectin signals from the fat storage depot to regulate metabolism in peripheral tissues. Inversely correlated with body fat levels, adiponectin reduction in obese individuals may play a causal role in the symptoms of metabolic syndrome. Adiponectin lowers serum glucose through suppression of hepatic glucose production, an effect attributed to activation of AMPK. Here, we investigated the signaling pathways that mediate the effects of adiponectin by studying mice with inducible hepatic deletion of LKB1, an upstream regulator of AMPK. We found that loss of LKB1 in the liver partially impaired the ability of adiponectin to lower serum glucose, though other actions of the hormone were preserved, including reduction of gluconeogenic gene expression and hepatic glucose production as assessed by euglycemic hyperinsulinemic clamp. Furthermore, in primary mouse hepatocytes, the absence of LKB1, AMPK, or the transcriptional coactivator CRTC2 did not prevent adiponectin from inhibiting glucose output or reducing gluconeogenic gene expression. These results reveal that whereas some of the hormone’s actions in vivo may be LKB1 dependent, substantial LKB1-, AMPK-, and CRTC2-independent signaling pathways also mediate effects of adiponectin. Find the latest version: https://jci.me/45942/pdf Research article Adiponectin suppresses gluconeogenic gene expression in mouse hepatocytes independent of LKB1-AMPK signaling Russell A. Miller,1 Qingwei Chu,1 John Le Lay,1 Philipp E. Scherer,2 Rexford S.
    [Show full text]
  • Xo PANEL DNA GENE LIST
    xO PANEL DNA GENE LIST ~1700 gene comprehensive cancer panel enriched for clinically actionable genes with additional biologically relevant genes (at 400 -500x average coverage on tumor) Genes A-C Genes D-F Genes G-I Genes J-L AATK ATAD2B BTG1 CDH7 CREM DACH1 EPHA1 FES G6PC3 HGF IL18RAP JADE1 LMO1 ABCA1 ATF1 BTG2 CDK1 CRHR1 DACH2 EPHA2 FEV G6PD HIF1A IL1R1 JAK1 LMO2 ABCB1 ATM BTG3 CDK10 CRK DAXX EPHA3 FGF1 GAB1 HIF1AN IL1R2 JAK2 LMO7 ABCB11 ATR BTK CDK11A CRKL DBH EPHA4 FGF10 GAB2 HIST1H1E IL1RAP JAK3 LMTK2 ABCB4 ATRX BTRC CDK11B CRLF2 DCC EPHA5 FGF11 GABPA HIST1H3B IL20RA JARID2 LMTK3 ABCC1 AURKA BUB1 CDK12 CRTC1 DCUN1D1 EPHA6 FGF12 GALNT12 HIST1H4E IL20RB JAZF1 LPHN2 ABCC2 AURKB BUB1B CDK13 CRTC2 DCUN1D2 EPHA7 FGF13 GATA1 HLA-A IL21R JMJD1C LPHN3 ABCG1 AURKC BUB3 CDK14 CRTC3 DDB2 EPHA8 FGF14 GATA2 HLA-B IL22RA1 JMJD4 LPP ABCG2 AXIN1 C11orf30 CDK15 CSF1 DDIT3 EPHB1 FGF16 GATA3 HLF IL22RA2 JMJD6 LRP1B ABI1 AXIN2 CACNA1C CDK16 CSF1R DDR1 EPHB2 FGF17 GATA5 HLTF IL23R JMJD7 LRP5 ABL1 AXL CACNA1S CDK17 CSF2RA DDR2 EPHB3 FGF18 GATA6 HMGA1 IL2RA JMJD8 LRP6 ABL2 B2M CACNB2 CDK18 CSF2RB DDX3X EPHB4 FGF19 GDNF HMGA2 IL2RB JUN LRRK2 ACE BABAM1 CADM2 CDK19 CSF3R DDX5 EPHB6 FGF2 GFI1 HMGCR IL2RG JUNB LSM1 ACSL6 BACH1 CALR CDK2 CSK DDX6 EPOR FGF20 GFI1B HNF1A IL3 JUND LTK ACTA2 BACH2 CAMTA1 CDK20 CSNK1D DEK ERBB2 FGF21 GFRA4 HNF1B IL3RA JUP LYL1 ACTC1 BAG4 CAPRIN2 CDK3 CSNK1E DHFR ERBB3 FGF22 GGCX HNRNPA3 IL4R KAT2A LYN ACVR1 BAI3 CARD10 CDK4 CTCF DHH ERBB4 FGF23 GHR HOXA10 IL5RA KAT2B LZTR1 ACVR1B BAP1 CARD11 CDK5 CTCFL DIAPH1 ERCC1 FGF3 GID4 HOXA11
    [Show full text]
  • CREB-Dependent Transcription in Astrocytes: Signalling Pathways, Gene Profiles and Neuroprotective Role in Brain Injury
    CREB-dependent transcription in astrocytes: signalling pathways, gene profiles and neuroprotective role in brain injury. Tesis doctoral Luis Pardo Fernández Bellaterra, Septiembre 2015 Instituto de Neurociencias Departamento de Bioquímica i Biologia Molecular Unidad de Bioquímica y Biologia Molecular Facultad de Medicina CREB-dependent transcription in astrocytes: signalling pathways, gene profiles and neuroprotective role in brain injury. Memoria del trabajo experimental para optar al grado de doctor, correspondiente al Programa de Doctorado en Neurociencias del Instituto de Neurociencias de la Universidad Autónoma de Barcelona, llevado a cabo por Luis Pardo Fernández bajo la dirección de la Dra. Elena Galea Rodríguez de Velasco y la Dra. Roser Masgrau Juanola, en el Instituto de Neurociencias de la Universidad Autónoma de Barcelona. Doctorando Directoras de tesis Luis Pardo Fernández Dra. Elena Galea Dra. Roser Masgrau In memoriam María Dolores Álvarez Durán Abuela, eres la culpable de que haya decidido recorrer el camino de la ciencia. Que estas líneas ayuden a conservar tu recuerdo. A mis padres y hermanos, A Meri INDEX I Summary 1 II Introduction 3 1 Astrocytes: physiology and pathology 5 1.1 Anatomical organization 6 1.2 Origins and heterogeneity 6 1.3 Astrocyte functions 8 1.3.1 Developmental functions 8 1.3.2 Neurovascular functions 9 1.3.3 Metabolic support 11 1.3.4 Homeostatic functions 13 1.3.5 Antioxidant functions 15 1.3.6 Signalling functions 15 1.4 Astrocytes in brain pathology 20 1.5 Reactive astrogliosis 22 2 The transcription
    [Show full text]
  • Compromised Global Embryonic Transcriptome Associated with Advanced Maternal Age
    Journal of Assisted Reproduction and Genetics (2019) 36:915–924 https://doi.org/10.1007/s10815-019-01438-5 EMBRYO BIOLOGY Compromised global embryonic transcriptome associated with advanced maternal age Blair R. McCallie1,2 & Jason C. Parks1,2 & G. Devon Trahan3 & Kenneth L. Jones3 & Breanne D. Coate4 & Darren K. Griffin2 & William B. Schoolcraft1 & Mandy G. Katz-Jaffe1 Received: 4 February 2019 /Accepted: 12 March 2019 /Published online: 25 April 2019 # The Author(s) 2019 Abstract Purpose To investigate the global transcriptome and associated embryonic molecular networks impacted with advanced maternal age (AMA). Methods Blastocysts derived from donor oocyte IVF cycles with no male factor infertility (< 30 years of age) and AMA blastocysts (≥ 42 years) with no other significant female factor infertility or male factor infertility were collected with informed patient consent. RNA sequencing libraries were prepared using the SMARTer® Ultra® Low Kit (Clontech Laboratories) and sequenced on the Illumina HiSEQ 4000. Bioinformatics included Ingenuity® Pathway Analysis (Qiagen) with ViiA™ 7qPCR utilized for gene expression validation (Applied Biosystems). Results A total of 2688 significant differentially expressed transcripts were identified to distinguish the AMA blastocysts from young, donor controls. 2551 (95%) of these displayed decreased transcription in the blastocysts from older women. Pathway analysis revealed three altered molecular signaling networks known to be critical for embryo and fetal development: CREBBP, ESR1, and SP1. Validation of genes within these networks confirmed the global decreased transcription observed in AMA blastocysts (P <0.05). Conclusions A significant, overall decreased global transcriptome was observed in blastocysts from AMA women. The ESR1/SP1/CREBBP pathway, in particular, was found to be a highly significant upstream regulator impacting biological processes that are vital during embryonic patterning and pre-implantation development.
    [Show full text]
  • A Truncated NRIP1 Mutant Amplifies Microsatellite Instability Of
    cancers Article A Truncated NRIP1 Mutant Amplifies Microsatellite Instability of Colorectal Cancer by Regulating MSH2/MSH6 Expression, and Is a Prognostic Marker of Stage III Tumors Pascale Palassin 1 , Marion Lapierre 1 , Samuel Pyrdziak 1, Antoine Wagner 1 ,Régine Stehle 1, Carole Corsini 1, Jacqueline Duffour 1, Sandrine Bonnet 1, Abdelhay Boulahtouf 1, Carmen Rodriguez 1, Alexandre Ho-Pun-Cheung 2, Evelyne Lopez-Crapez 1,2 , Florence Boissière-Michot 2 , Frédéric Bibeau 1, Simon Thezenas 1 , Nabila Elarouci 3, Janick Selves 4, Jean-Sébastien Hoffmann 4 , Paul Roepman 5, Thibault Mazard 1 , Olivier Buhard 6, Alex Duval 6, Stéphan Jalaguier 1, Vincent Cavaillès 1,*,† and Audrey Castet-Nicolas 1,7,8,† 1 IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM, U1194, Institut Régional du Cancer de Montpellier, Université de Montpellier, F-34298 Montpellier, France; [email protected] (P.P.); [email protected] (M.L.); [email protected] (S.P.); [email protected] (A.W.); [email protected] (R.S.); [email protected] (C.C.); [email protected] (J.D.); [email protected] (S.B.); [email protected] (A.B.); [email protected] (C.R.); [email protected] (E.L.-C.); [email protected] (F.B.); [email protected] (S.T.); [email protected] (T.M.); [email protected] (S.J.); [email protected] (A.C.-N.) Citation: Palassin, P.; Lapierre, M.; 2 Translational Research Unit, Institut du Cancer de Montpellier, F-34298 Montpellier, France; Pyrdziak, S.; Wagner, A.; Stehle, R.; [email protected] (A.H.-P.-C.); fl[email protected] (F.B.-M.) Corsini, C.; Duffour, J.; Bonnet, S.; 3 Programme Cartes d’Identité des Tumeurs (CIT), Ligue Nationale Contre Le Cancer, F-75013 Paris, France; Boulahtouf, A.; Rodriguez, C.; et al.
    [Show full text]
  • Effect of Exercise and Butyrate Supplementation on Microbiota Composition and Lipid Metabolism
    243 2 Journal of C Yu et al. Effect of exercise on gut 243:2 125–135 Endocrinology microbiota RESEARCH Effect of exercise and butyrate supplementation on microbiota composition and lipid metabolism Chunxia Yu1, Sujuan Liu2, Liqin Chen3, Jun Shen3, Yanmei Niu4, Tianyi Wang1, Wanqi Zhang3 and Li Fu1,4 1Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China 2Department of Anatomy and Embryology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China 3Tianjin Key Laboratory of Environment, Nutrition and Public Health, School of Public Health, Tianjin Medical University, Tianjin, China 4Department of Rehabilitation, School of Medical Technology, Tianjin Medical University, Tianjin, China Correspondence should be addressed to L Fu: [email protected] Abstract The composition and activity of the gut microbiota depend on the host genome, Key Words nutrition, and lifestyle. Exercise and sodium butyrate (NaB) exert metabolic benefits f microbiota in both mice and humans. However, the underlying mechanisms have not been fully f butyrate elucidated. This study aimed to examine the effect of exercise training and dietary f exercise supplementation of butyrate on the composition of gut microbiota and whether the f HFD altered gut microbiota can stimulate differential production of short-chain fatty acids f Sestrin2 (SCFAs), which promote the expression of SESN2 and CRTC2 to improve metabolic health f CRTC2 and protect against obesity. C57BL/6J mice were used to study the effect of exercise and high-fat diet (HFD) with or without NaB on gut microbiota. Bacterial communities were assayed in fecal samples using pyrosequencing of 16S rRNA gene amplicons.
    [Show full text]
  • Proteomic Profiling and Genome-Wide Mapping of O-Glcnac Chromatin
    ARTICLE https://doi.org/10.1038/s41467-020-19579-y OPEN Proteomic profiling and genome-wide mapping of O-GlcNAc chromatin-associated proteins reveal an O-GlcNAc-regulated genotoxic stress response Yubo Liu 1,3, Qiushi Chen 2,3, Nana Zhang1, Keren Zhang2, Tongyi Dou1, Yu Cao1, Yimin Liu1, Kun Li1, ✉ ✉ Xinya Hao1, Xueqin Xie1, Wenli Li1, Yan Ren 2 & Jianing Zhang 1 fi 1234567890():,; O-GlcNAc modi cation plays critical roles in regulating the stress response program and cellular homeostasis. However, systematic and multi-omics studies on the O-GlcNAc regu- lated mechanism have been limited. Here, comprehensive data are obtained by a chemical reporter-based method to survey O-GlcNAc function in human breast cancer cells stimulated with the genotoxic agent adriamycin. We identify 875 genotoxic stress-induced O-GlcNAc chromatin-associated proteins (OCPs), including 88 O-GlcNAc chromatin-associated tran- scription factors and cofactors (OCTFs), subsequently map their genomic loci, and construct a comprehensive transcriptional reprogramming network. Notably, genotoxicity-induced O- GlcNAc enhances the genome-wide interactions of OCPs with chromatin. The dynamic binding switch of hundreds of OCPs from enhancers to promoters is identified as a crucial feature in the specific transcriptional activation of genes involved in the adaptation of cancer cells to genotoxic stress. The OCTF nuclear factor erythroid 2-related factor-1 (NRF1) is found to be a key response regulator in O-GlcNAc-modulated cellular homeostasis. These results provide a valuable clue suggesting that OCPs act as stress sensors by regulating the expression of various genes to protect cancer cells from genotoxic stress.
    [Show full text]