pubmed_id
stringlengths
41
43
abstract
stringlengths
3
18.8k
http://www.ncbi.nlm.nih.gov/pubmed/30849537
1. Gene. 2019 Jun 15;701:161-168. doi: 10.1016/j.gene.2019.02.073. Epub 2019 Mar 5. Co-polysomy of 1p/19q in glial tumors: Retrospective analysis of 221 cases from single center. Kuskucu A(1), Tuysuz EC(2), Gurkan S(1), Demir Z(1), Yaltirik CK(3), Ozkan F(4), Ekici ID(4), Bayrak OF(5), Ture U(6). Author information: (1)Department of Medical Genetics, Yeditepe University Medical School, 34755 Istanbul, Turkey. (2)Department of Medical Genetics, Yeditepe University Medical School, 34755 Istanbul, Turkey; Department of Biotechnology, Institute of Science, Yeditepe University, 34755 Istanbul, Turkey. (3)Department of Neurosurgery, Yeditepe University Medical School, Yeditepe University, 34755 Istanbul, Turkey. (4)Department of Medical Pathology, Yeditepe University Medical School, Yeditepe University, 34755 Istanbul, Turkey. (5)Department of Medical Genetics, Yeditepe University Medical School, 34755 Istanbul, Turkey. Electronic address: [email protected]. (6)Department of Neurosurgery, Yeditepe University Medical School, Yeditepe University, 34755 Istanbul, Turkey. Electronic address: [email protected]. Glial tumors are malignant brain tumors that arise from glial cells of brain or spine and have genetic aberrations in their genome. 1p/19q co-deletion is associated with increased Overall Survival (OS) time with enhanced response to chemo- and radio-therapy in oligodendrogliomas. However, prognostic significance of 1p/19q co-polysomy is still unclear. We evaluated 1p/19q status of 221 patients with glial tumor by Fluorescent in situ Hybridization (FISH). Records of the patients were collected retrospectively. Our results demonstrated that 1p/19q co-polysomy was associated with decreased OS time, high P53 expression and frequently located in temporal lobe, whereas 1p/19q co-deletion was associated with increased overall survival time, low P53 expression and frontal lobe location. Furthermore, classification of patients based on both 1p/19q status and P53 expression revealed that patients with 1p/19q co-polysomy and high P53 expression had the worst prognosis. Lastly, our bioinformatic survival analysis revealed that high expression of SRM, ICMT, and FTL located in 1p36.13-p36.31 and 19q13.2-q13.33 region were related with decreased OS time in patients with Low Grade Glioma (LGG). The study demonstrated that 1p/19q co-polysomy is a poor prognostic marker for glial tumor. Copyright © 2019 Elsevier B.V. All rights reserved. DOI: 10.1016/j.gene.2019.02.073 PMID: 30849537 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/20446099
1. Acta Neurochir (Wien). 2010 Aug;152(8):1425-9. doi: 10.1007/s00701-010-0674-x. Epub 2010 May 6. Leptomeningeal dissemination of a pediatric neoplasm with 1p19q deletion showing mixed immunohistochemical features of an oligodendroglioma and neurocytoma. Rhiew RB(1), Manjila S, Lozen A, Guthikonda M, Sood S, Kupsky WJ. Author information: (1)Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA. Leptomeningeal dissemination of an oligodendroglioma is rarely reported in the neurosurgical literature, especially in cases with a classical 1p19q deletion. The authors describe a case wherein a 1p19q deletion in a disseminated tumor with mixed immunohistochemical features of oligodendroglioma and neurocytoma was encountered and treated. Stereotactic right frontal craniotomy was undertaken for obtaining definitive histological diagnosis. The results revealed a neuroectodermal neoplasm with histologic and immunohistochemical features of oligodendroglioma and neurocytoma. FISH analysis confirmed classical 1p19q deletion. The patient was treated postoperatively with chemotherapy and radiation therapy. He showed good clinical response and remains alive 16 months after diagnosis. DOI: 10.1007/s00701-010-0674-x PMID: 20446099 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/19624298
1. J Med Imaging Radiat Oncol. 2009 Jun;53(3):305-9. doi: 10.1111/j.1754-9485.2009.02074.x. Retrospective review of prognostic factors, including 1p19q deletion, in low-grade oligodendrogliomas and a review of recent published works. Capelle L(1), Oei P, Teoh H, Hamilton D, Palmer D, Low I, Campbell G. Author information: (1)Department of Radiation Oncology, Auckland City Hospital, Auckland, New Zealand. The purpose of the present study was to investigate potential prognostic factors in low-grade oligodendrogliomas (LGOs), particularly 1p19q deletion, due to its proven prognostic significance in anaplastic oligodendrogliomas. We carried out a retrospective review of patients with a histological diagnosis of LGO between 1990 and 2000 in Auckland and Wellington, New Zealand. All cases underwent central histopathological review and FISH testing for 1p19q status. Univariate analysis of potential prognostic factors including 1p19q status, age, tumour size, tumour crossing midline, tumour enhancement, extent of surgery and seizures at diagnosis was carried out. Thirty-one patients were eligible and FISH testing was successful in 28 specimens (90%). Twenty-three specimens (82%) had 1p19q deletion; four (14%) had no 1p19q deletion; and one (4%) had 1p deletion alone. At a median follow-up of 87 months (0-147 months), median survival had not been reached and no significant difference in overall survival (OS) based on 1p19q status was detected (1p19q deletion OS 56%; 1p19q intact OS 0%; 1p deletion alone 100% (P = 0.38)). None of the other prognostic factors investigated reached statistical significance. We confirmed the high incidence (82%) of combined 1p19q deletion in LGOs and the feasibility of successful FISH testing in paraffin embedded specimens up to 10-years-old. Analysis of potential prognostic factors was limited by the lack of events during the follow-up period. DOI: 10.1111/j.1754-9485.2009.02074.x PMID: 19624298 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/32906679
1. Cancers (Basel). 2020 Sep 7;12(9):2543. doi: 10.3390/cancers12092543. CeRNA Network Analysis Representing Characteristics of Different Tumor Environments Based on 1p/19q Codeletion in Oligodendrogliomas. Ahn JW(1), Park Y(1), Kang SJ(1), Hwang SJ(2), Cho KG(2), Lim J(2), Kwack K(1). Author information: (1)Department of Biomedical Science, College of Life Science, CHA University, Seongnam 13488, Korea. (2)Department of Neurosurgery, Bundang CHA Medical Center, CHA University School of Medicine, Seongnam 13496, Korea. Erratum in Cancers (Basel). 2021 Jan 27;13(3):478. doi: 10.3390/cancers13030478. Oligodendroglioma (OD) is a subtype of glioma occurring in the central nervous system. The 1p/19q codeletion is a prognostic marker of OD with an isocitrate dehydrogenase (IDH) mutation and is associated with a clinically favorable overall survival (OS); however, the exact underlying mechanism remains unclear. Long non-coding RNAs (lncRNAs) have recently been suggested to regulate carcinogenesis and prognosis in cancer patients. Here, we performed in silico analyses using low-grade gliomas from datasets obtained from The Cancer Genome Atlas to investigate the effects of ceRNA with 1p/19q codeletion on ODs. Thus, we selected modules of differentially expressed genes that were closely related to 1p/19q codeletion traits using weighted gene co-expression network analysis and constructed 16 coding RNA-miRNA-lncRNA networks. The ceRNA network participated in ion channel activity, insulin secretion, and collagen network and extracellular matrix (ECM) changes. In conclusion, ceRNAs with a 1p/19q codeletion can create different tumor microenvironments via potassium ion channels and ECM composition changes; furthermore, differences in OS may occur. Moreover, if extrapolated to gliomas, our results can provide insights into the consequences of identical gene expression, indicating the possibility of tracking different biological processes in different subtypes of glioma. DOI: 10.3390/cancers12092543 PMCID: PMC7564449 PMID: 32906679 Conflict of interest statement: The authors declare no conflict of interest.
http://www.ncbi.nlm.nih.gov/pubmed/23681562
1. J Neurooncol. 2013 Aug;114(1):85-91. doi: 10.1007/s11060-013-1152-0. Epub 2013 May 17. Combining two biomarkers, IDH1/2 mutations and 1p/19q codeletion, to stratify anaplastic oligodendroglioma in three groups: a single-center experience. Frenel JS(1), Leux C, Loussouarn D, Le Loupp AG, Leclair F, Aumont M, Mervoyer A, Martin S, Denis MG, Campone M. Author information: (1)Medical Oncology Department, Institut de Cancérologie de l'Ouest, Centre René Gauducheau, 11 Boulevard Jacques Monod, 44800, Saint-Herblain, Nantes, France. [email protected] IDH1/2 mutations and 1p/19q codeletion occur frequently in anaplastic gliomas and are prognostic factors. We combined these two biomarkers to stratify patients treated for anaplastic oligodendroglioma (AO). 43 consecutive WHO AO were selected. We combined immunohistochemistry (IHC) with the monoclonal antibody mIDH1R132H and DNA sequencing of IDH1 and IDH2 genes. Fluorescence in situ hybridization was carried out to evaluate 1p/19q codeletion. These biomarkers were correlated with progression-free survival (PFS) and overall survival (OS). IDH1/IDH2 mutations occurred in 23/43 (54 %) patients: 20/43 IDH1-R132H mutation in IHC, 2/43 IDH1-R132G mutation and 1/43 IDH2-R172K mutation identified by DNA sequencing. 1p/19q codeletion was detected for 23/43 patients. With median follow-up of 19 months (range 1.4-128), median PFS and OS were 22 and 35 months respectively. IDH1/IDH2 mutations were strongly associated with improved PFS and OS: 5-year PFS was 86 versus 6 % and 5-year OS was 91 versus 9 % for patients with IDH1/IDH2 mutations versus wild-type IDH respectively. In multivariate analyses, IDH1/IDH2 mutations and 1p/19q loss were independent prognostic factors. Three groups with distinct prognostic features were identified: patients with IDH1/2 mutations and 1p/19q loss (median PFS, median OS not reached), patients with IDH1/2 mutations or 1p/19q loss (median PFS: 22 months, median OS: 30 months), and patients without IDH1/2 mutations nor 1p/19q loss with a bad prognosis (median PFS: 8.6 months, median OS: 9.9 months). Combining two biomarkers, IDH1/2 and 1p/19q codeletion, makes it possible to stratify AO in three groups with very distinct prognostic features. DOI: 10.1007/s11060-013-1152-0 PMID: 23681562 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/12429693
1. Annu Rev Genet. 2002;36:233-78. doi: 10.1146/annurev.genet.36.042902.092433. Epub 2002 Jun 11. Xist RNA and the mechanism of X chromosome inactivation. Plath K(1), Mlynarczyk-Evans S, Nusinow DA, Panning B. Author information: (1)Department of Biochemistry & Biophysics, University of California San Francisco, San Francisco, California 94143, USA. [email protected] Dosage compensation in mammals is achieved by the transcriptional inactivation of one X chromosome in female cells. From the time X chromosome inactivation was initially described, it was clear that several mechanisms must be precisely integrated to achieve correct regulation of this complex process. X-inactivation appears to be triggered upon differentiation, suggesting its regulation by developmental cues. Whereas any number of X chromosomes greater than one is silenced, only one X chromosome remains active. Silencing on the inactive X chromosome coincides with the acquisition of a multitude of chromatin modifications, resulting in the formation of extraordinarily stable facultative heterochromatin that is faithfully propagated through subsequent cell divisions. The integration of all these processes requires a region of the X chromosome known as the X-inactivation center, which contains the Xist gene and its cis-regulatory elements. Xist encodes an RNA molecule that plays critical roles in the choice of which X chromosome remains active, and in the initial spread and establishment of silencing on the inactive X chromosome. We are now on the threshold of discovering the factors that regulate and interact with Xist to control X-inactivation, and closer to an understanding of the molecular mechanisms that underlie this complex process. DOI: 10.1146/annurev.genet.36.042902.092433 PMID: 12429693 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/12900550
1. Cytogenet Genome Res. 2002;99(1-4):92-8. doi: 10.1159/000071579. Ectopic XIST transcripts in human somatic cells show variable expression and localization. Chow JC(1), Hall LL, Lawrence JB, Brown CJ. Author information: (1)Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada. XIST encodes a functional RNA that is expressed exclusively from the inactive X in female mammals and is required for the silencing of most of the genes on the chromosome. XIST transcripts remain in the nucleus, and their specific localization to the inactive X is important for silencing; however, it is not known how these transcripts localize to the inactive X chromosome. Expression of mouse and human XIST from ectopic sites has suggested that localization to the chromosome from which the gene is expressed may be dependent upon either the copy number of the integrated constructs or the level of ectopic XIST expression. To further examine the behavior of XIST transgenes when expressed from ectopic sites, we introduced an XIST-containing PAC into the human male somatic cell line HT-1080. In five different transformant clones, the degree of localization and associated DNA condensation of the surrounding chromatin varied within nuclei of the same clone, as well as among different clones. Comparing the number of integrated transgenes and the levels of XIST expression revealed that neither factor was sufficient for a tight localization of the XIST signal. Therefore, the extent of expression and localization of XIST transcripts from ectopic transgenes is likely dependent upon many interacting factors, including the number of integrated transgenes, the level of XIST expression, and the site of integration. Copyright 2002 S. Karger AG, Basel DOI: 10.1159/000071579 PMID: 12900550 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/23689617
1. Acta Neuropathol. 2013 Aug;126(2):277-89. doi: 10.1007/s00401-013-1130-9. Epub 2013 May 21. Codeletion of 1p and 19q determines distinct gene methylation and expression profiles in IDH-mutated oligodendroglial tumors. Mur P(1), Mollejo M, Ruano Y, de Lope ÁR, Fiaño C, García JF, Castresana JS, Hernández-Laín A, Rey JA, Meléndez B. Author information: (1)Molecular Pathology Research Unit, Department of Pathology, Virgen de la Salud Hospital, Avda. Barber 30, 45004, Toledo, Spain. Oligodendroglial tumors (OTs) are primary brain tumors that show variable clinical and biological behavior. The 1p/19q codeletion is frequent in these tumors, indicating a better prognosis and/or treatment response. Recently, the prognostically favorable CpG island methylator phenotype (CIMP) in gliomas (G-CIMP+) was associated with mutations in the isocitrate dehydrogenase 1 and isocitrate dehydrogenase 2 (IDH) genes, as opposed to G-CIMP- tumors, highlighting the relevance of epigenetic mechanisms. We performed a whole-genome methylation study in 46 OTs, and a gene expression study of 25 tumors, correlating the methylation and transcriptomic profiles with molecular and clinical variables. Here, we identified two different epigenetic patterns within the previously described main G-CIMP+ profile. Both IDH mutation-associated methylation profiles featured one group of OTs with 1p/19q loss (CD-CIMP+), most of which were pure oligodendrogliomas, and a second group with intact 1p/19q and frequent TP53 mutation (CIMP+), most of which exhibited a mixed histopathology. A third group of OTs lacking the CIMP profile (CIMP-), and with a wild-type IDH and an intact 1p/19q, similar to the G-CIMP- subgroup, was also observed. The three CIMP groups presented a significantly better (CD-CIMP+), intermediate (CIMP+) or worse (CIMP-) prognosis. Furthermore, transcriptomic analyses revealed CIMP-specific gene expression signatures, indicating the impact of genetic status (IDH mutation, 1p/19q codeletion, TP53 mutation) on gene expression, and pointing to candidate biomarkers. Therefore, the CIMP profiles contributed to the identification of subgroups of OTs characterized by different prognoses, histopathologies, molecular features and gene expression signatures, which may help in the classification of OTs. DOI: 10.1007/s00401-013-1130-9 PMID: 23689617 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/29237010
1. Nucleic Acids Res. 2018 Mar 16;46(5):e26. doi: 10.1093/nar/gkx1227. CRISPR/Cas9-mediated modulation of splicing efficiency reveals short splicing isoform of Xist RNA is sufficient to induce X-chromosome inactivation. Yue M(1)(2), Ogawa Y(1)(2). Author information: (1)Division of Reproductive Sciences, Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA. (2)Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA. Alternative splicing of mRNA precursors results in multiple protein variants from a single gene and is critical for diverse cellular processes and development. Xist encodes a long noncoding RNA which is a central player to induce X-chromosome inactivation in female mammals and has two major splicing variants: long and short isoforms of Xist RNA. Although a differentiation-specific and a female-specific expression of Xist isoforms have been reported, the functional role of each Xist RNA isoform is largely unexplored. Using CRISPR/Cas9-mediated targeted modification of the 5' splice site in Xist intron 7, we create mutant female ES cell lines which dominantly express the long- or short-splicing isoform of Xist RNA from the inactive X-chromosome (Xi) upon differentiation. Successful execution of CRISPR/Cas-based splicing modulation indicates that our CRISPR/Cas-based targeted modification of splicing sites is a useful approach to study specific isoforms of a transcript generated by alternative splicing. Upon differentiation of splicing-mutant Xist female ES cells, we find that both long and short Xist isoforms can induce X-chromosome inactivation normally during ES cell differentiation, suggesting that the short splicing isoform of Xist RNA is sufficient to induce X-chromosome inactivation. DOI: 10.1093/nar/gkx1227 PMCID: PMC5861412 PMID: 29237010 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/25636755
1. Int J Radiat Oncol Biol Phys. 2015 Feb 1;91(2):268-76. doi: 10.1016/j.ijrobp.2014.10.027. Impact of 1p/19q codeletion and histology on outcomes of anaplastic gliomas treated with radiation therapy and temozolomide. Speirs CK(1), Simpson JR(1), Robinson CG(1), DeWees TA(1), Tran DD(2), Linette G(2), Chicoine MR(3), Dacey RG(3), Rich KM(3), Dowling JL(3), Leuthardt EC(3), Zipfel GJ(3), Kim AH(3), Huang J(4). Author information: (1)Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri. (2)Department of Medicine, Division of Medical Oncology, Washington University School of Medicine, St. Louis, Missouri. (3)Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri. (4)Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri. Electronic address: [email protected]. PURPOSE: Anaplastic gliomas represent a heterogeneous group of primary high-grade brain tumors, and the optimal postoperative treatment remains controversial. In this report, we present our institutional data on the clinical outcomes of radiation therapy (RT) plus temozolomide (RT + TMZ) for anaplastic gliomas, stratified by histology and 1p/19q codeletion. METHODS AND MATERIALS: A single-institution retrospective review was conducted of patients with supratentorial anaplastic oligodendroglioma (AO), mixed anaplastic oligoastrocytoma (AOA), and anaplastic astrocytoma (AA). After surgery, RT was delivered at a median total dose of 60 Gy (range, 31.6-63 Gy) in daily fractions. All patients received standard concurrent TMZ, with or without adjuvant TMZ. Histological/molecular subtypes were defined as codeleted AO/AOA, non-codeleted AO/AOA, and AA. RESULTS: From 2000 to 2012, 111 cases met study criteria and were evaluable. Codeleted AO/AOA had superior overall survival (OS) to non-codeleted AO/AOA (91% vs 68% at 5 years, respectively, P=.02), whereas progression-free survival (PFS) was not significantly different (70% vs 46% at 5 years, respectively, P=.10). AA had inferior OS to non-codeleted AO/AOA (37% vs 68% at 5 years, respectively, P=.007) and inferior PFS (27% vs 46%, respectively, P=.03). On multivariate analysis, age, performance status, and histological or molecular subtype were independent predictors for both PFS and OS. Compared to historical controls, RT + TMZ provided comparable OS to RT with procarbazine, lomustine, and vincristine (RT + PCV) for codeleted AO/AOA, superior OS to RT alone for non-codeleted AO/AOA, and similar OS to RT alone for AA. CONCLUSIONS: RT + TMZ may be a promising treatment for both codeleted and non-codeleted AO/AOA, but its role for AA remains unclear. Copyright © 2015 Elsevier Inc. All rights reserved. DOI: 10.1016/j.ijrobp.2014.10.027 PMID: 25636755 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/12492109
1. Mol Cancer Ther. 2002 Aug;1(10):769-76. Relationship of XIST expression and responses of ovarian cancer to chemotherapy. Huang KC(1), Rao PH, Lau CC, Heard E, Ng SK, Brown C, Mok SC, Berkowitz RS, Ng SW. Author information: (1)Laboratory of Gynecologic Oncology, Department of Obstetrics and Gynecology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA. Expression profiling to characterize cancer pharmacology has become a new approach to discover novel molecular targets for prognostic markers and cancer therapy. In a study to compare the global RNA expression profiles between primary and recurrent ovarian tumors from the same patient, we have identified XIST (inactive X chromosome-specific transcripts) as the most differentially expressed gene that was down-regulated in the recurrent tumor. XIST encodes a spliced noncoding polyadenylated transcript that is unique in being expressed exclusively from the inactive X chromosome and is involved in the X-inactivation process. Subsequent characterization of XIST expression in a panel of female cancer cell lines showed that the expression level of XIST correlates significantly with Taxol sensitivity. The clinical relevance of this observation is demonstrated by the strong association between XIST RNA levels and disease-free periods of ovarian cancer patients in a group of 21 ovarian cancer cases with Taxol in the therapeutic regiments. Cytogenetic studies on ovarian cancer cell lines indicated that loss of inactive X chromosome is one mechanism for the loss of XIST transcripts in the cell lines. Our data suggest that XIST expression may be a potential marker for chemotherapeutic responses in ovarian cancer. PMID: 12492109 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/25200388
1. BMC Genet. 2014 Sep 9;15:89. doi: 10.1186/s12863-014-0089-4. Differentially methylated CpG island within human XIST mediates alternative P2 transcription and YY1 binding. Chapman AG(1), Cotton AM(2), Kelsey AD(3), Brown CJ(4). Author information: (1)Department of Medical Genetics, Molecular Epigenetics Group, Life Sciences Center, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada. [email protected]. (2)Department of Medical Genetics, Molecular Epigenetics Group, Life Sciences Center, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada. [email protected]. (3)Department of Medical Genetics, Molecular Epigenetics Group, Life Sciences Center, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada. [email protected]. (4)Department of Medical Genetics, Molecular Epigenetics Group, Life Sciences Center, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada. [email protected]. BACKGROUND: X-chromosome inactivation silences one X chromosome in females to achieve dosage compensation with the single X chromosome in males. While most genes are silenced on the inactive X chromosome, the gene for the long non-coding RNA XIST is silenced on the active X chromosome and expressed from the inactive X chromosome with which the XIST RNA associates, triggering silencing of the chromosome. In mouse, an alternative Xist promoter, P2 is also the site of YY1 binding, which has been shown to serve as a tether between the Xist RNA and the DNA of the chromosome. In humans there are many differences from the initial events of mouse Xist activation, including absence of a functional antisense regulator Tsix, and absence of strictly paternal inactivation in extraembryonic tissues, prompting us to examine regulatory regions for the human XIST gene. RESULTS: We demonstrate that the female-specific DNase hypersensitivity site within XIST is specific to the inactive X chromosome and correlates with transcription from an internal P2 promoter. P2 is located within a CpG island that is differentially methylated between males and females and overlaps conserved YY1 binding sites that are only bound on the inactive X chromosome where the sites are unmethylated. However, YY1 binding is insufficient to drive P2 expression or establish the DHS, which may require a development-specific factor. Furthermore, reduction of YY1 reduces XIST transcription in addition to causing delocalization of XIST. CONCLUSIONS: The differentially methylated DNase hypersensitive site within XIST marks the location of an alternative promoter, P2, that generates a transcript of unknown function as it lacks the A repeats that are critical for silencing. In addition, this region binds YY1 on the unmethylated inactive X chromosome, and depletion of YY1 untethers the XIST RNA as well as decreasing transcription of XIST. DOI: 10.1186/s12863-014-0089-4 PMCID: PMC4363909 PMID: 25200388 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/26282267
1. Genome Biol. 2015 Aug 15;16(1):166. doi: 10.1186/s13059-015-0733-y. Xist localization and function: new insights from multiple levels. Cerase A(1), Pintacuda G(2), Tattermusch A(2), Avner P(3)(4). Author information: (1)EMBL Mouse Biology Unit, Monterotondo, 00015 (RM), Italy. [email protected]. (2)Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK. (3)EMBL Mouse Biology Unit, Monterotondo, 00015 (RM), Italy. [email protected]. (4)Institut Pasteur, Unite de Genetique Moleculaire Murine, CNRS, URA2578, Paris, France. [email protected]. In female mammals, one of the two X chromosomes in each cell is transcriptionally silenced in order to achieve dosage compensation between the genders in a process called X chromosome inactivation. The master regulator of this process is the long non-coding RNA Xist. During X-inactivation, Xist accumulates in cis on the future inactive X chromosome, triggering a cascade of events that provoke the stable silencing of the entire chromosome, with relatively few genes remaining active. How Xist spreads, what are its binding sites, how it recruits silencing factors and how it induces a specific topological and nuclear organization of the chromatin all remain largely unanswered questions. Recent studies have improved our understanding of Xist localization and the proteins with which it interacts, allowing a reappraisal of ideas about Xist function. We discuss recent advances in our knowledge of Xist-mediated silencing, focusing on Xist spreading, the nuclear organization of the inactive X chromosome, recruitment of the polycomb complex and the role of the nuclear matrix in the process of X chromosome inactivation. DOI: 10.1186/s13059-015-0733-y PMCID: PMC4539689 PMID: 26282267 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/26477563
1. Nat Commun. 2015 Oct 19;6:8564. doi: 10.1038/ncomms9564. An Xist-activating antisense RNA required for X-chromosome inactivation. Sarkar MK(1), Gayen S(1), Kumar S(1), Maclary E(1), Buttigieg E(1), Hinten M(1), Kumari A(1), Harris C(1), Sado T(2), Kalantry S(1). Author information: (1)Department of Human Genetics, University of Michigan Medical School, Ann Arbor, Michigan 48109, USA. (2)Department of Advanced Bioscience, Graduate School of Agriculture, Kinki University, 3327-204 Nakamachi, Nara 631-8505, Japan. The transcriptional imbalance due to the difference in the number of X chromosomes between male and female mammals is remedied through X-chromosome inactivation, the epigenetic transcriptional silencing of one of the two X chromosomes in females. The X-linked Xist long non-coding RNA functions as an X inactivation master regulator; Xist is selectively upregulated from the prospective inactive X chromosome and is required in cis for X inactivation. Here we discover an Xist antisense long non-coding RNA, XistAR (Xist Activating RNA), which is encoded within exon 1 of the mouse Xist gene and is transcribed only from the inactive X chromosome. Selective truncation of XistAR, while sparing the overlapping Xist RNA, leads to a deficiency in Xist RNA expression in cis during the initiation of X inactivation. Thus, the Xist gene carries within its coding sequence an antisense RNA that drives Xist expression. DOI: 10.1038/ncomms9564 PMCID: PMC4616153 PMID: 26477563 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/20950563
1. Biol Aujourdhui. 2010;204(3):199-204. doi: 10.1051/jbio/2010013. Epub 2010 Oct 13. [Establishing transcriptional silencing of the X chromosome during early embryogenesis]. [Article in French] Nora EP(1), Heard E. Author information: (1)Epigenese et developpement des mammiferes, Institut Curie, Paris, France. Early development of female mammals is accompanied by transcriptional inactivation of one of their two X chromosomes. This process, known as X-chromosome inactivation, relies on monoallelic activation of the Xist gene. Xist produces a non-coding RNA that can coat the chromosome from which it is transcribed in cis and trigger its silencing. How Xist expression is controlled and how it initiates transcriptional repression are central questions for our understanding of how this chromosome-wide monoallelic program is expressed. Several trans-acting factors have been identified as regulators of Xist expression. Interestingly, some Xist activators are encoded by the X chromosome itself, thereby efficiently promoting Xist expression in females (XX) but not in males (XY). Female cells also display transient physical pairing between their two X chromosomes at the level of their Xics (X inactivation centers) during the time window when X inactivation is initiated. It has been proposed that these pairing events may play a role in Xist activation and its monoallelic regulation. Xist RNA accumulates over the X chromosome from which it is expressed and rapidly triggers the exclusion of the transcription machinery. Genic sequences are initially located outside of this Xist RNA coated domain but as they become progressively silenced they are relocated into this silent nuclear compartment created by Xist. However genes are not all silenced with the same kinetics. Furthermore, some genes can escape X inactivation and remain located outside the Xist-coated compartment. Recent findings have revealed that young, active LINE-1 retrotransposons are expressed from the inactive X chromosome and may facilitate X inactivation, particularly in regions of the X that would otherwise be prone to escape. © Société de Biologie, 2010. DOI: 10.1051/jbio/2010013 PMID: 20950563 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/26739568
1. Proc Natl Acad Sci U S A. 2016 Jan 19;113(3):E309-18. doi: 10.1073/pnas.1515971113. Epub 2016 Jan 6. Sex-specific silencing of X-linked genes by Xist RNA. Gayen S(1), Maclary E(1), Hinten M(1), Kalantry S(2). Author information: (1)Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI 48109. (2)Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI 48109 [email protected]. X-inactive specific transcript (Xist) long noncoding RNA (lncRNA) is thought to catalyze silencing of X-linked genes in cis during X-chromosome inactivation, which equalizes X-linked gene dosage between male and female mammals. To test the impact of Xist RNA on X-linked gene silencing, we ectopically induced endogenous Xist by ablating the antisense repressor Tsix in mice. We find that ectopic Xist RNA induction and subsequent X-linked gene silencing is sex specific in embryos and in differentiating embryonic stem cells (ESCs) and epiblast stem cells (EpiSCs). A higher frequency of X(ΔTsix)Y male cells displayed ectopic Xist RNA coating compared with X(ΔTsix)X female cells. This increase reflected the inability of X(ΔTsix)Y cells to efficiently silence X-linked genes compared with X(ΔTsix)X cells, despite equivalent Xist RNA induction and coating. Silencing of genes on both Xs resulted in significantly reduced proliferation and increased cell death in X(ΔTsix)X female cells relative to X(ΔTsix)Y male cells. Thus, whereas Xist RNA can inactivate the X chromosome in females it may not do so in males. We further found comparable silencing in differentiating X(ΔTsix)Y and 39,X(ΔTsix) (X(ΔTsix)O) ESCs, excluding the Y chromosome and instead implicating the X-chromosome dose as the source of the sex-specific differences. Because X(ΔTsix)X female embryonic epiblast cells and EpiSCs harbor an inactivated X chromosome prior to ectopic inactivation of the active X(ΔTsix) X chromosome, we propose that the increased expression of one or more X-inactivation escapees activates Xist and, separately, helps trigger X-linked gene silencing. DOI: 10.1073/pnas.1515971113 PMCID: PMC4725534 PMID: 26739568 [Indexed for MEDLINE] Conflict of interest statement: The authors declare no conflict of interest.
http://www.ncbi.nlm.nih.gov/pubmed/32535328
1. Curr Opin Cell Biol. 2020 Jun;64:139-147. doi: 10.1016/j.ceb.2020.04.009. Epub 2020 Jun 11. Xist drives spatial compartmentalization of DNA and protein to orchestrate initiation and maintenance of X inactivation. Strehle M(1), Guttman M(2). Author information: (1)Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA. (2)Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA. Electronic address: [email protected]. X chromosome inactivation (XCI) is the process whereby one of the X chromosomes in female mammalian cells is silenced to equalize X-linked gene expression with males. XCI depends on the long noncoding RNA Xist, which coats the inactive X chromosome in cis and triggers a cascade of events that ultimately lead to chromosome-wide transcriptional silencing that is stable for the lifetime of an organism. In recent years, the discovery of proteins that interact with Xist have led to new insights into how the initiation of XCI occurs. Nevertheless, there are still various unknowns about the mechanisms by which Xist orchestrates and maintains stable X-linked silencing. Here, we review recent work elucidating the role of Xist and its protein partners in mediating chromosome-wide transcriptional repression, as well as discuss a model by which Xist may compartmentalize proteins across the inactive X chromosome to enable both the initiation and maintenance of XCI. Copyright © 2020 Elsevier Ltd. All rights reserved. DOI: 10.1016/j.ceb.2020.04.009 PMID: 32535328 [Indexed for MEDLINE] Conflict of interest statement: Conflict of interest statement Nothing declared.
http://www.ncbi.nlm.nih.gov/pubmed/29910081
1. Trends Cell Biol. 2018 Dec;28(12):999-1013. doi: 10.1016/j.tcb.2018.05.005. Epub 2018 Jun 14. The Role of Xist in X-Chromosome Dosage Compensation. Sahakyan A(1), Yang Y(1), Plath K(2). Author information: (1)David Geffen School of Medicine, Department of Biological Chemistry, Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, Jonsson Comprehensive Cancer Center, Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA 90095, USA. (2)David Geffen School of Medicine, Department of Biological Chemistry, Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, Jonsson Comprehensive Cancer Center, Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA 90095, USA. Electronic address: [email protected]. In each somatic cell of a female mammal one X chromosome is transcriptionally silenced via X-chromosome inactivation (XCI), initiating early in development. Although XCI events are conserved in mouse and human postimplantation development, regulation of X-chromosome dosage in preimplantation development occurs differently. In preimplantation development, mouse embryos undergo imprinted form of XCI, yet humans lack imprinted XCI and instead regulate gene expression of both X chromosomes by dampening transcription. The long non-coding RNA Xist/XIST is expressed in mouse and human preimplantation and postimplantation development to orchestrate XCI, but its role in dampening is unclear. In this review, we discuss recent advances in our understanding of the role of Xist in X chromosome dosage compensation in mouse and human. Copyright © 2018 Elsevier Ltd. All rights reserved. DOI: 10.1016/j.tcb.2018.05.005 PMCID: PMC6249047 PMID: 29910081 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/28236732
1. Curr Opin Cell Biol. 2017 Jun;46:54-61. doi: 10.1016/j.ceb.2017.01.007. Epub 2017 Feb 23. X chromosome inactivation: silencing, topology and reactivation. Robert Finestra T(1), Gribnau J(2). Author information: (1)Department of Developmental Biology, Erasmus MC, Wytemaweg 80, Rotterdam CN 3015, The Netherlands. (2)Department of Developmental Biology, Erasmus MC, Wytemaweg 80, Rotterdam CN 3015, The Netherlands. Electronic address: [email protected]. To ensure X-linked gene dosage compensation between females (XX) and males (XY), one X chromosome undergoes X chromosome inactivation (XCI) in female cells. This process is tightly regulated throughout development by many different factors, with Xist as a key regulator, encoding a long non-coding RNA, involved in establishment of several layers of repressive epigenetic modifications. Several recent studies on XCI focusing on identification and characterization of Xist RNA-protein interactors, revealed new factors involved in gene silencing, genome topology and nuclear membrane attachment, amongst others. Also, new insights in higher order chromatin organization have been presented, revealing differences between the topological organization of active and inactive X chromosomes (Xa and Xi), with associated differences in gene expression. Finally, further evidence indicates that the inactive state of the Xi can be (partially) reversed, and that this X chromosome reactivation (XCR) might be associated with disease. Copyright © 2017. Published by Elsevier Ltd. DOI: 10.1016/j.ceb.2017.01.007 PMID: 28236732 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/31537017
1. PLoS Genet. 2019 Sep 19;15(9):e1008333. doi: 10.1371/journal.pgen.1008333. eCollection 2019 Sep. Xist RNA in action: Past, present, and future. Loda A(1), Heard E(1)(2). Author information: (1)Directors' research, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany. (2)Collège de France, Paris, France. In mammals, dosage compensation of sex chromosomal genes between females (XX) and males (XY) is achieved through X-chromosome inactivation (XCI). The X-linked X-inactive-specific transcript (Xist) long noncoding RNA is indispensable for XCI and initiates the process early during development by spreading in cis across the X chromosome from which it is transcribed. During XCI, Xist RNA triggers gene silencing, recruits a plethora of chromatin modifying factors, and drives a major structural reorganization of the X chromosome. Here, we review our knowledge of the multitude of epigenetic events orchestrated by Xist RNA to allow female mammals to survive through embryonic development by establishing and maintaining proper dosage compensation. In particular, we focus on recent studies characterizing the interaction partners of Xist RNA, and we discuss how they have affected the field by addressing long-standing controversies or by giving rise to new research perspectives that are currently being explored. This review is dedicated to the memory of Denise Barlow, pioneer of genomic imprinting and functional long noncoding RNAs (lncRNAs), whose work has revolutionized the epigenetics field and continues to inspire generations of scientists. DOI: 10.1371/journal.pgen.1008333 PMCID: PMC6752956 PMID: 31537017 [Indexed for MEDLINE] Conflict of interest statement: The authors have declared that no competing interests exist.
http://www.ncbi.nlm.nih.gov/pubmed/29701779
1. Hum Mol Genet. 2018 Aug 1;27(R2):R242-R249. doi: 10.1093/hmg/ddy148. The eXceptional nature of the X chromosome. Balaton BP(1), Dixon-McDougall T(1), Peeters SB(1), Brown CJ(1). Author information: (1)Molecular Epigenetics Group, Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada. The X chromosome is unique in the genome. In this review we discuss recent advances in our understanding of the genetics and epigenetics of the X chromosome. The X chromosome shares limited conservation with its ancestral homologue the Y chromosome and the resulting difference in X-chromosome dosage between males and females is largely compensated for by X-chromosome inactivation. The process of inactivation is initiated by the long non-coding RNA X-inactive specific transcript (XIST) and achieved through interaction with multiple synergistic silencing pathways. Identification of Xist-interacting proteins has given insight into these processes yet the cascade of events from initiation to maintenance have still to be resolved. In particular, the initiation of inactivation in humans has been challenging to study as: it occurs very early in development; most human embryonic stem cell lines already have an inactive X; and the process seems to differ from mouse. Another difference between human and mouse X inactivation is the larger number of human genes that escape silencing. In humans over 20% of X-linked genes continue to be expressed from the otherwise inactive X chromosome. We are only beginning to understand how such escape occurs but there is growing recognition that escapees contribute to sexually dimorphic traits. The unique biology and epigenetics of the X chromosome have often led to its exclusion from disease studies, yet the X constitutes 5% of the genome and is an important contributor to disease, often in a sex-specific manner. DOI: 10.1093/hmg/ddy148 PMCID: PMC6061837 PMID: 29701779 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/32482714
1. Genes Dev. 2020 Jun 1;34(11-12):733-744. doi: 10.1101/gad.337196.120. Progress toward understanding chromosome silencing by Xist RNA. Brockdorff N(1), Bowness JS(1), Wei G(1). Author information: (1)Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom. The X inactive-specific transcript (Xist) gene is the master regulator of X chromosome inactivation in mammals. Xist produces a long noncoding (lnc)RNA that accumulates over the entire length of the chromosome from which it is transcribed, recruiting factors to modify underlying chromatin and silence X-linked genes in cis Recent years have seen significant progress in identifying important functional elements in Xist RNA, their associated RNA-binding proteins (RBPs), and the downstream pathways for chromatin modification and gene silencing. In this review, we summarize progress in understanding both how these pathways function in Xist-mediated silencing and the complex interplay between them. © 2020 Brockdorff et al.; Published by Cold Spring Harbor Laboratory Press. DOI: 10.1101/gad.337196.120 PMCID: PMC7263139 PMID: 32482714 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/28947655
1. Philos Trans R Soc Lond B Biol Sci. 2017 Nov 5;372(1733):20160356. doi: 10.1098/rstb.2016.0356. Mechanistic insights in X-chromosome inactivation. Lu Z(1), Carter AC(1), Chang HY(2). Author information: (1)Center for Dynamic Personal Regulomes, Stanford University, Stanford, CA 94305, USA. (2)Center for Dynamic Personal Regulomes, Stanford University, Stanford, CA 94305, USA [email protected]. X-chromosome inactivation (XCI) is a critical epigenetic mechanism for balancing gene dosage between XY males and XX females in eutherian mammals. A long non-coding RNA (lncRNA), XIST, and its associated proteins orchestrate this multi-step process, resulting in the inheritable silencing of one of the two X-chromosomes in females. The XIST RNA is large and complex, exemplifying the unique challenges associated with the structural and functional analysis of lncRNAs. Recent technological advances in the analysis of macromolecular structure and interactions have enabled us to systematically dissect the XIST ribonucleoprotein complex, which is larger than the ribosome, and its place of action, the inactive X-chromosome. These studies shed light on key mechanisms of XCI, such as XIST coating of the X-chromosome, recruitment of DNA, RNA and histone modification enzymes, and compaction and compartmentalization of the inactive X. Here, we summarize recent studies on XCI, highlight the critical contributions of new technologies and propose a unifying model for XIST function in XCI where modular domains serve as the structural and functional units in both lncRNA-protein complexes and DNA-protein complexes in chromatin.This article is part of the themed issue 'X-chromosome inactivation: a tribute to Mary Lyon'. © 2017 The Author(s). DOI: 10.1098/rstb.2016.0356 PMCID: PMC5627158 PMID: 28947655 [Indexed for MEDLINE] Conflict of interest statement: We have no competing interests.
http://www.ncbi.nlm.nih.gov/pubmed/23166390
1. Philos Trans R Soc Lond B Biol Sci. 2013 Jan 5;368(1609):20110325. doi: 10.1098/rstb.2011.0325. Advances in understanding chromosome silencing by the long non-coding RNA Xist. Sado T(1), Brockdorff N. Author information: (1)Division of Epigenomics, Medical Institute of Bioregulation, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan. [email protected] In female mammals, one of the two X chromosomes becomes genetically silenced to compensate for dosage imbalance of X-linked genes between XX females and XY males. X chromosome inactivation (X-inactivation) is a classical model for epigenetic gene regulation in mammals and has been studied for half a century. In the last two decades, efforts have been focused on the X inactive-specific transcript (Xist) locus, discovered to be the master regulator of X-inactivation. The Xist gene produces a non-coding RNA that functions as the primary switch for X-inactivation, coating the X chromosome from which it is transcribed in cis. Significant progress has been made towards understanding how Xist is regulated at the onset of X-inactivation, but our understanding of the molecular basis of silencing mediated by Xist RNA has progressed more slowly. A picture has, however, begun to emerge, and new tools and resources hold out the promise of further advances to come. Here, we provide an overview of the current state of our knowledge, what is known about Xist RNA and how it may trigger chromosome silencing. DOI: 10.1098/rstb.2011.0325 PMCID: PMC3539355 PMID: 23166390 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/17333537
1. Chromosome Res. 2007;15(2):127-36. doi: 10.1007/s10577-006-1115-9. Epub 2007 Mar 5. Genes flanking Xist in mouse and human are separated on the X chromosome in American marsupials. Shevchenko AI(1), Zakharova IS, Elisaphenko EA, Kolesnikov NN, Whitehead S, Bird C, Ross M, Weidman JR, Jirtle RL, Karamysheva TV, Rubtsov NB, VandeBerg JL, Mazurok NA, Nesterova TB, Brockdorff N, Zakian SM. Author information: (1)Institute of Cytology and Genetics, Russian Academy of Sciences, Siberian Department, Novosibirsk, Russia. X inactivation, the transcriptional silencing of one of the two X chromosomes in female mammals, achieves dosage compensation of X-linked genes relative to XY males. In eutherian mammals X inactivation is regulated by the X-inactive specific transcript (Xist), a cis-acting non-coding RNA that triggers silencing of the chromosome from which it is transcribed. Marsupial mammals also undergo X inactivation but the mechanism is relatively poorly understood. We set out to analyse the X chromosome in Monodelphis domestica and Didelphis virginiana, focusing on characterizing the interval defined by the Chic1 and Slc16a2 genes that in eutherians flank the Xist locus. The synteny of this region is retained on chicken chromosome 4 where other loci belonging to the evolutionarily ancient stratum of the human X chromosome, the so-called X conserved region (XCR), are also located. We show that in both M. domestica and D. virginiana an evolutionary breakpoint has separated the Chic1 and Slc16a2 loci. Detailed analysis of opossum genomic sequences revealed linkage of Chic1 with the Lnx3 gene, recently proposed to be the evolutionary precursor of Xist, and Fip1, the evolutionary precursor of Tsx, a gene located immediately downstream of Xist in eutherians. We discuss these findings in relation to the evolution of Xist and X inactivation in mammals. DOI: 10.1007/s10577-006-1115-9 PMCID: PMC2797855 PMID: 17333537 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/30496473
1. Nucleic Acids Res. 2019 Feb 20;47(3):1532-1543. doi: 10.1093/nar/gky1198. Developmental Xist induction is mediated by enhanced splicing. Stork C(1), Li Z(2), Lin L(3), Zheng S(1)(2)(3). Author information: (1)Graduate Program in Cell, Molecular and Developmental Biology, University of California, Riverside, Riverside, CA 92521, USA. (2)Graduate Program in Genetics, Genomics and Bioinformatics, University of California, Riverside, Riverside, CA 92521, USA. (3)Division of Biomedical Sciences, University of California, Riverside, Riverside, CA 92521, USA. X-inactive-specific transcript (Xist) is a long noncoding RNA (lncRNA) essential for inactivating one of the two X chromosomes in mammalian females. Random X chromosome inactivation is mediated by Xist RNA expressed from the inactive X chromosome. We found that Xist RNA is unspliced in naïve embryonic stem (ES) cells. Upon differentiation, Xist splicing becomes efficient across all exons independent of transcription, suggesting interdependent or coordinated removal of Xist introns. In female cells with mutated polypyrimidine tract binding protein 1 (Ptbp1), differentiation fails to substantially upregulate mature Xist RNA because of a defect in Xist splicing. We further found both Xist129 and XistCAS RNA are unspliced in Mus musculus 129SvJ/Mus castaneous (CAS) hybrid female ES cells. Upon differentiation, Xist129 exhibits a higher splicing efficiency than XistCAS, likely contributing to preferential inhibition of the X129 chromosome. Single cell analysis shows that the allelic choice of Xist splicing is linked to the inactive X chromosome. We conclude post-transcriptional control of Xist RNA splicing is an essential regulatory step of Xist induction. Our studies shed light on the developmental roles of splicing for nuclear-retained Xist lncRNA and suggest inefficient Xist splicing is an additional fail-safe mechanism to prevent Xist activity in ES cells. © The Author(s) 2018. Published by Oxford University Press on behalf of Nucleic Acids Research. DOI: 10.1093/nar/gky1198 PMCID: PMC6379716 PMID: 30496473 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/26489649
1. Proc Natl Acad Sci U S A. 2015 Nov 24;112(47):14415-22. doi: 10.1073/pnas.1519528112. Epub 2015 Oct 21. Xist imprinting is promoted by the hemizygous (unpaired) state in the male germ line. Sun S(1), Payer B(2), Namekawa S(3), An JY(2), Press W(2), Catalan-Dibene J(4), Sunwoo H(2), Lee JT(5). Author information: (1)Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114; Department of Genetics, Harvard Medical School, Boston, MA 02114; Department of Developmental and Cell Biology, School of Biological Sciences, University of California, Irvine, CA 92697; (2)Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114; Department of Genetics, Harvard Medical School, Boston, MA 02114; (3)Division of Reproductive Sciences, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229; (4)Department of Developmental and Cell Biology, School of Biological Sciences, University of California, Irvine, CA 92697; (5)Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114; Department of Genetics, Harvard Medical School, Boston, MA 02114; Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02114 [email protected]. Comment in Proc Natl Acad Sci U S A. 2015 Nov 24;112(47):14408-9. doi: 10.1073/pnas.1520097112. The long noncoding X-inactivation-specific transcript (Xist gene) is responsible for mammalian X-chromosome dosage compensation between the sexes, the process by which one of the two X chromosomes is inactivated in the female soma. Xist is essential for both the random and imprinted forms of X-chromosome inactivation. In the imprinted form, Xist is paternally marked to be expressed in female embryos. To investigate the mechanism of Xist imprinting, we introduce Xist transgenes (Tg) into the male germ line. Although ectopic high-level Xist expression on autosomes can be compatible with viability, transgenic animals demonstrate reduced fitness, subfertility, defective meiotic pairing, and other germ-cell abnormalities. In the progeny, paternal-specific expression is recapitulated by the 200-kb Xist Tg. However, Xist imprinting occurs efficiently only when it is in an unpaired or unpartnered state during male meiosis. When transmitted from a hemizygous father (+/Tg), the Xist Tg demonstrates paternal-specific expression in the early embryo. When transmitted by a homozygous father (Tg/Tg), the Tg fails to show imprinted expression. Thus, Xist imprinting is directed by sequences within a 200-kb X-linked region, and the hemizygous (unpaired) state of the Xist region promotes its imprinting in the male germ line. DOI: 10.1073/pnas.1519528112 PMCID: PMC4664331 PMID: 26489649 [Indexed for MEDLINE] Conflict of interest statement: The authors declare no conflict of interest.
http://www.ncbi.nlm.nih.gov/pubmed/14973270
1. Development. 2004 Mar;131(5):975-82. doi: 10.1242/dev.00995. De novo DNA methylation is dispensable for the initiation and propagation of X chromosome inactivation. Sado T(1), Okano M, Li E, Sasaki H. Author information: (1)Division of Human Genetics, National Institute of Genetics, 1111 Yata, Mishima 411-8540, Japan. [email protected] Xist (X-inactive specific transcript) plays a crucial role in X-inactivation. This non-coding RNA becomes upregulated on the X chromosome that is to be inactivated upon differentiation. Previous studies have revealed that although maintenance-type DNA methylation is not essential for X-inactivation to occur, it is required for the stable repression of Xist in differentiated cells. However, it is unknown whether differential de novo methylation at the Xist promoter, which is mediated by Dnmt3a and/or Dnmt3b, is a cause or a consequence of monoallelic expression of Xist. We show that Xist expression is appropriately regulated in the absence of Dnmt3a and Dnmt3b and that a single X chromosome undergoes proper inactivation in mutant females. Our results indicate that a mechanism(s) other than DNA methylation plays a principal role in initiating X-inactivation. We also demonstrate that delayed upregulation of Xist does not induce X-inactivation, consistent with a crucial developmental window for the chromosomal silencing. DOI: 10.1242/dev.00995 PMID: 14973270 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/21212949
1. World J Surg. 2011 Aug;35(8):1751-6. doi: 10.1007/s00268-010-0951-0. Expression and function of a large non-coding RNA gene XIST in human cancer. Weakley SM(1), Wang H, Yao Q, Chen C. Author information: (1)Molecular Surgeon Research Center, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Baylor Plaza, Mail stop: BCM391, Houston, TX 77030, USA. BACKGROUND: X inactive-specific transcript (XIST) RNA is involved in X chromosome silencing in female cells and allows X chromosome equilibration with males. X inactive-specific transcript expression has been found to be dysregulated in a variety of human cancers when compared to normal cells; meanwhile, the inactivated X chromosome has been noted to be conspicuously absent in human cancer specimens, whereas X chromosome duplications are widely noted. The specific pathways whereby changes in X chromosome status and XIST expression occur in cancer remain incompletely described. Nevertheless, a role for XIST in BRCA1-mediated epigenetic activity has been proposed. METHODS: Here we review the data regarding XIST expression and X chromosome status in a variety of female, male, and non-sex-related human cancers. CONCLUSIONS: It is not yet known whether X chromosome duplication, XIST dysregulation, and over-expression of X-linked genes represent important factors in tumorgenesis or are simply a consequence of overall epigenetic instability in these cancers. DOI: 10.1007/s00268-010-0951-0 PMCID: PMC3275083 PMID: 21212949 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/33348832
1. Cells. 2020 Dec 17;9(12):2706. doi: 10.3390/cells9122706. New Insights into X-Chromosome Reactivation during Reprogramming to Pluripotency. Panda A(1), Zylicz JJ(2), Pasque V(1). Author information: (1)Laboratory of Cellular Reprogramming and Epigenetic Regulation, Department of Development and Regeneration, Leuven Stem Cell Institute, KU Leuven-University of Leuven, 3000 Leuven, Belgium. (2)The Novo Nordisk Foundation Center for Stem Cell Biology, University of Copenhagen, 2200 Copenhagen, Denmark. Dosage compensation between the sexes results in one X chromosome being inactivated during female mammalian development. Chromosome-wide transcriptional silencing from the inactive X chromosome (Xi) in mammalian cells is erased in a process termed X-chromosome reactivation (XCR), which has emerged as a paradigm for studying the reversal of chromatin silencing. XCR is linked with germline development and induction of naive pluripotency in the epiblast, and also takes place upon reprogramming somatic cells to induced pluripotency. XCR depends on silencing of the long non-coding RNA (lncRNA) X inactive specific transcript (Xist) and is linked with the erasure of chromatin silencing. Over the past years, the advent of transcriptomics and epigenomics has provided new insights into the transcriptional and chromatin dynamics with which XCR takes place. However, multiple questions remain unanswered about how chromatin and transcription related processes enable XCR. Here, we review recent work on establishing the transcriptional and chromatin kinetics of XCR, as well as discuss a model by which transcription factors mediate XCR not only via Xist repression, but also by direct targeting of X-linked genes. DOI: 10.3390/cells9122706 PMCID: PMC7766869 PMID: 33348832 [Indexed for MEDLINE] Conflict of interest statement: The authors declare no known conflict of interest. The funders had no role the writing of the manuscript.
http://www.ncbi.nlm.nih.gov/pubmed/11780141
1. Nat Genet. 2002 Feb;30(2):167-74. doi: 10.1038/ng820. Epub 2002 Jan 7. Chromosomal silencing and localization are mediated by different domains of Xist RNA. Wutz A(1), Rasmussen TP, Jaenisch R. Author information: (1)Whitehead Institute for Biomedical Research, Nine Cambridge Center, Cambridge, Massachusetts 02142, USA. The gene Xist initiates the chromosomal silencing process of X inactivation in mammals. Its product, a noncoding RNA, is expressed from and specifically associates with the inactive X chromosome in female cells. Here we use an inducible Xist expression system in mouse embryonic stem cells that recapitulates long-range chromosomal silencing to elucidate which Xist RNA sequences are necessary for chromosomal association and silencing. We show that chromosomal association and spreading of Xist RNA can be functionally separated from silencing by specific mutations. Silencing requires a conserved repeat sequence located at the 5' end of Xist. Deletion of this element results in Xist RNA that still associates with chromatin and spreads over the chromosome but does not effect transcriptional repression. Association of Xist RNA with chromatin is mediated by functionally redundant sequences that act cooperatively and are dispersed throughout the remainder of Xist but show little or no homology. DOI: 10.1038/ng820 PMID: 11780141 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/25489864
1. J Vis Exp. 2014 Nov 26;(93):e52053. doi: 10.3791/52053. Quick fluorescent in situ hybridization protocol for Xist RNA combined with immunofluorescence of histone modification in X-chromosome inactivation. Yue M(1), Charles Richard JL(1), Yamada N(1), Ogawa A(2), Ogawa Y(3). Author information: (1)Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center; Department of Pediatrics, University of Cincinnati College of Medicine. (2)Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center. (3)Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center; Department of Pediatrics, University of Cincinnati College of Medicine; [email protected]. Combining RNA fluorescent in situ hybridization (FISH) with immunofluorescence (immuno-FISH) creates a technique that can be employed at the single cell level to detect the spatial dynamics of RNA localization with simultaneous insight into the localization of proteins, epigenetic modifications and other details which can be highlighted by immunofluorescence. X-chromosome inactivation is a paradigm for long non-coding RNA (lncRNA)-mediated gene silencing. X-inactive specific transcript (Xist) lncRNA accumulation (called an Xist cloud) on one of the two X-chromosomes in mammalian females is a critical step to initiate X-chromosome inactivation. Xist RNA directly or indirectly interacts with various chromatin-modifying enzymes and introduces distinct epigenetic landscapes to the inactive X-chromosome (Xi). One known epigenetic hallmark of the Xi is the Histone H3 trimethyl-lysine 27 (H3K27me3) modification. Here, we describe a simple and quick immuno-FISH protocol for detecting Xist RNA using RNA FISH with multiple oligonucleotide probes coupled with immunofluorescence of H3K27me3 to examine the localization of Xist RNA and associated epigenetic modifications. Using oligonucleotide probes results in a shorter incubation time and more sensitive detection of Xist RNA compared to in vitro transcribed RNA probes (riboprobes). This protocol provides a powerful tool for understanding the dynamics of lncRNAs and its associated epigenetic modification, chromatin structure, nuclear organization and transcriptional regulation. DOI: 10.3791/52053 PMCID: PMC4354415 PMID: 25489864 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/16679409
1. Proc Natl Acad Sci U S A. 2006 May 16;103(20):7706-11. doi: 10.1073/pnas.0602021103. Epub 2006 May 5. Attenuated spread of X-inactivation in an X;autosome translocation. Popova BC(1), Tada T, Takagi N, Brockdorff N, Nesterova TB. Author information: (1)Developmental Epigenetics, Medical Research Council Clinical Sciences Center, Imperial College Faculty of Medicine, Hammersmith Hospital, DuCane Road, London W12 ONN, United Kingdom. X inactivation in female mammals involves transcriptional silencing of an entire chromosome in response to a cis-acting noncoding RNA, the X inactive-specific transcript (Xist). Xist can also inactivate autosomal sequences, for example, in X;autosome translocations; but here, silencing appears to be relatively inefficient. This variation has been attributed to either attenuated spreading of Xist RNA at the onset of X inactivation or inefficient maintenance of autosomal silencing. Evidence to date has favored the latter. Here, we demonstrate attenuated spreading of Xist RNA at the onset of X inactivation in the T(X;4)37H X;autosome translocation. Our findings provide direct evidence that underlying chromosome/chromatin features can disrupt spreading of the primary inactivating signal. DOI: 10.1073/pnas.0602021103 PMCID: PMC1472509 PMID: 16679409 [Indexed for MEDLINE] Conflict of interest statement: Conflict of interest statement: No conflicts declared.
http://www.ncbi.nlm.nih.gov/pubmed/30539545
1. Methods Mol Biol. 2019;1870:41-50. doi: 10.1007/978-1-4939-8808-2_3. Visualization of Xist Long Noncoding RNA with a Fluorescent CRISPR/Cas9 System. Waśko U(1), Zheng Z(1), Bhatnagar S(2). Author information: (1)Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, USA. (2)Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, USA. [email protected]. X-inactive specific transcript (Xist) is a long noncoding RNA that is essential for initiating and maintaining epigenetic silencing of one copy of the X chromosome in mammalian females. But the mechanism by which Xist localizes and spreads on the X chromosome and facilitates transcriptional silencing remains largely unknown. This limited understanding, at least in part, is due to the technical difficulties in the visualization and functional characterization of Xist. Development of a successful method for Xist tracking is a key to better understanding of the X chromosome silencing, as well as to gain insight into the regulatory role of other long noncoding RNAs. Here, we describe an alternative method for visualization of Xist lncRNA in cells using a CRISPR/Cas9-based approach. This strategy is relatively simple approach to track Xist at different stages of cell differentiation, providing mechanistic insights into the initiation, maintenance, and establishment of X inactivation. DOI: 10.1007/978-1-4939-8808-2_3 PMID: 30539545 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/22722828
1. Nature. 2012 Jul 12;487(7406):254-8. doi: 10.1038/nature11171. Rsx is a metatherian RNA with Xist-like properties in X-chromosome inactivation. Grant J(1), Mahadevaiah SK, Khil P, Sangrithi MN, Royo H, Duckworth J, McCarrey JR, VandeBerg JL, Renfree MB, Taylor W, Elgar G, Camerini-Otero RD, Gilchrist MJ, Turner JM. Author information: (1)MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London NW71AA, UK. In female (XX) mammals, one of the two X chromosomes is inactivated to ensure an equal dose of X-linked genes with males (XY). X-chromosome inactivation in eutherian mammals is mediated by the non-coding RNA Xist. Xist is not found in metatherians (marsupials), and how X-chromosome inactivation is initiated in these mammals has been the subject of speculation for decades. Using the marsupial Monodelphis domestica, here we identify Rsx (RNA-on-the-silent X), an RNA that has properties consistent with a role in X-chromosome inactivation. Rsx is a large, repeat-rich RNA that is expressed only in females and is transcribed from, and coats, the inactive X chromosome. In female germ cells, in which both X chromosomes are active, Rsx is silenced, linking Rsx expression to X-chromosome inactivation and reactivation. Integration of an Rsx transgene on an autosome in mouse embryonic stem cells leads to gene silencing in cis. Our findings permit comparative studies of X-chromosome inactivation in mammals and pose questions about the mechanisms by which X-chromosome inactivation is achieved in eutherians. DOI: 10.1038/nature11171 PMCID: PMC3484893 PMID: 22722828 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/30091314
1. Yonsei Med J. 2018 Sep;59(7):816-826. doi: 10.3349/ymj.2018.59.7.816. XIST Induced by JPX Suppresses Hepatocellular Carcinoma by Sponging miR-155-5p. Lin XQ(1), Huang ZM(1), Chen X(1), Wu F(1), Wu W(2). Author information: (1)Department of Gastroenterology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China. (2)Department of Gastroenterology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China. [email protected]. PURPOSE: The influence of X-inactive specific transcript (XIST) and X-chromosome inactivation associated long non-coding RNAs (lncRNAs) just proximal to XIST (JPX) on hepatocellular carcinoma (HCC) remains controversial in light of previous reports, which the present study aimed to verify. MATERIALS AND METHODS: The DIANA lncRNA-microRNA (miRNA) interaction database was used to explore miRNA interactions with JPX or XIST. JPX, XIST, and miR-155-5p expression levels in paired HCC specimens and adjacent normal tissue were analyzed by RT-qPCR. Interaction between XIST and miR-155-5p was verified by dual luciferase reporter assay. Expression levels of miR-155-5p and its known target genes, SOX6 and PTEN, were verified by RT-qPCR and Western blot in HepG2 cells with or without XIST knock-in. The potential suppressive role of XIST and JPX on HCC was verified by cell functional assays and tumor formation assay using a xenograft model. RESULTS: JPX and XIST expression was significantly decreased in HCC pathologic specimens, compared to adjacent tissue, which correlated with HCC progression and increased miR-155-5p expression. Dual luciferase reporter assay revealed XIST as a direct target of miR-155-5p. XIST knock-in significantly reduced miR-155-5p expression level and increased that of SOX6 and PTEN, while significantly inhibiting HepG2 cell growth in vitro, which was partially reversed by miR-155-5p mimic transfection. JPX knock-in significantly increased XIST expression and inhibited HepG2 cell growth in vitro or tumor formation in vivo in a XIST dependent manner. CONCLUSION: JPX and XIST play a suppressive role in HCC. JPX increases expression levels of XIST in HCC cells, which suppresses HCC development by sponging the cancer promoting miR-155-5p. © Copyright: Yonsei University College of Medicine 2018. DOI: 10.3349/ymj.2018.59.7.816 PMCID: PMC6082978 PMID: 30091314 [Indexed for MEDLINE] Conflict of interest statement: The authors have no financial conflicts of interest.
http://www.ncbi.nlm.nih.gov/pubmed/25000994
1. Annu Rev Cell Dev Biol. 2014;30:561-80. doi: 10.1146/annurev-cellbio-101512-122415. Epub 2014 Jun 27. Noncoding RNAs and epigenetic mechanisms during X-chromosome inactivation. Gendrel AV(1), Heard E. Author information: (1)Mammalian Developmental Epigenetics Group, Genetics and Developmental Biology Unit, Institut Curie, 75248 Paris, France; email: [email protected]. In mammals, the process of X-chromosome inactivation ensures equivalent levels of X-linked gene expression between males and females through the silencing of one of the two X chromosomes in female cells. The process is established early in development and is initiated by a unique locus, which produces a long noncoding RNA, Xist. The Xist transcript triggers gene silencing in cis by coating the future inactive X chromosome. It also induces a cascade of chromatin changes, including posttranslational histone modifications and DNA methylation, and leads to the stable repression of all X-linked genes throughout development and adult life. We review here recent progress in our understanding of the molecular mechanisms involved in the initiation of Xist expression, the propagation of the Xist RNA along the chromosome, and the cis-elements and trans-acting factors involved in the maintenance of the repressed state. We also describe the diverse strategies used by nonplacental mammals for X-chromosome dosage compensation and highlight the common features and differences between eutherians and metatherians, in particular regarding the involvement of long noncoding RNAs. DOI: 10.1146/annurev-cellbio-101512-122415 PMID: 25000994 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/20657585
1. Nat Struct Mol Biol. 2010 Aug;17(8):948-54. doi: 10.1038/nsmb.1877. Epub 2010 Jul 25. The A-repeat links ASF/SF2-dependent Xist RNA processing with random choice during X inactivation. Royce-Tolland ME(1), Andersen AA, Koyfman HR, Talbot DJ, Wutz A, Tonks ID, Kay GF, Panning B. Author information: (1)Department of Biochemistry and Biophysics, University of California, San Francisco, California, USA. One X chromosome, selected at random, is silenced in each female mammalian cell. Xist encodes a noncoding RNA that influences the probability that the cis-linked X chromosome will be silenced. We found that the A-repeat, a highly conserved element within Xist, is required for the accumulation of spliced Xist RNA. In addition, the A-repeat is necessary for X-inactivation to occur randomly. In combination, our data suggest that normal Xist RNA processing is important in the regulation of random X-inactivation. We propose that modulation of Xist RNA processing may be part of the stochastic process that determines which X chromosome will be inactivated. DOI: 10.1038/nsmb.1877 PMCID: PMC4336797 PMID: 20657585 [Indexed for MEDLINE] Conflict of interest statement: COMPETING FINANCIAL INTERESTS The authors declare no competing financial interests.
http://www.ncbi.nlm.nih.gov/pubmed/21626138
1. Hum Genet. 2011 Aug;130(2):223-36. doi: 10.1007/s00439-011-1008-7. Epub 2011 May 28. Xist regulation and function explored. Pontier DB(1), Gribnau J. Author information: (1)Department of Reproduction and Development, Erasmus MC, Dr. Molewaterplein 50, 3015 GE Rotterdam, The Netherlands. X chromosome inactivation (XCI) is a process in mammals that ensures equal transcript levels between males and females by genetic inactivation of one of the two X chromosomes in females. Central to XCI is the long non-coding RNA Xist, which is highly and specifically expressed from the inactive X chromosome. Xist covers the X chromosome in cis and triggers genetic silencing, but its working mechanism remains elusive. Here, we review current knowledge about Xist regulation, structure, function and conservation and speculate on possible mechanisms by which its action is restricted in cis. We also discuss dosage compensation mechanisms other than XCI and how knowledge from invertebrate species may help to provide a better understanding of the mechanisms of mammalian XCI. DOI: 10.1007/s00439-011-1008-7 PMCID: PMC3132428 PMID: 21626138 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/28408975
1. F1000Res. 2017 Mar 27;6:F1000 Faculty Rev-344. doi: 10.12688/f1000research.10707.1. eCollection 2017. X chromosome inactivation: new players in the initiation of gene silencing. Pinheiro I(1), Heard E(1). Author information: (1)Mammalian Developmental Epigenetics Group (équipe labellisée La Ligue), Institut Curie, PSL Research University, CNRS UMR3215, INSERM U934, 26 Rue d'Ulm, 11 75248 Paris Cedex 05, France. X chromosome inactivation (XCI) is a dosage compensation process that was adopted by female mammals to balance gene dosage between XX females and XY males. XCI starts with the upregulation of the non-coding RNA Xist, after which most X-linked genes are silenced and acquire a repressive chromatin state. Even though the chromatin marks of the inactive X have been fairly well described, the mechanisms responsible for the initiation of XCI remain largely unknown. In this review, we discuss recent developments that revealed unexpected factors playing a role in XCI and that might be of crucial importance to understand the mechanisms responsible for the very first steps of this chromosome-wide gene-silencing event. DOI: 10.12688/f1000research.10707.1 PMCID: PMC5373419 PMID: 28408975 Conflict of interest statement: Competing interests: The authors declare that they have no competing interests.No competing interests were disclosed.No competing interests were disclosed.
http://www.ncbi.nlm.nih.gov/pubmed/26004255
1. Curr Opin Genet Dev. 2015 Apr;31:57-66. doi: 10.1016/j.gde.2015.04.002. Epub 2015 May 22. X-chromosome inactivation: new insights into cis and trans regulation. Galupa R(1), Heard E(2). Author information: (1)Mammalian Developmental Epigenetics Group, Institut Curie, PSL University, CNRS UMR3215, INSERM U934, 26, rue d'Ulm, 75005 Paris, France. (2)Mammalian Developmental Epigenetics Group, Institut Curie, PSL University, CNRS UMR3215, INSERM U934, 26, rue d'Ulm, 75005 Paris, France. Electronic address: [email protected]. X-chromosome inactivation (XCI) is a developmentally associated process that evolved in mammals to enable gene dosage compensation between XX and XY individuals. In placental mammals, it is triggered by the long noncoding RNA Xist, which is produced from a complex regulatory locus, the X-inactivation centre (Xic). Recent insights into the regulatory landscape of the Xic, including its partitioning into topological associating domains (TADs) and its genetic dissection, have important implications for the monoallelic regulation of Xist. Here, we present some of the latest studies on X inactivation with a special focus on the regulation of Xist, its various functions and the putative role of chromosome conformation in regulating the dynamics of this locus during development and differentiation. Copyright © 2015 Elsevier Ltd. All rights reserved. DOI: 10.1016/j.gde.2015.04.002 PMID: 26004255 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/34178980
1. Front Cell Dev Biol. 2021 Jun 10;9:645647. doi: 10.3389/fcell.2021.645647. eCollection 2021. Biological Function of Long Non-coding RNA (LncRNA) Xist. Wang W(1), Min L(1), Qiu X(1), Wu X(1), Liu C(1), Ma J(1), Zhang D(1), Zhu L(1). Author information: (1)Department of Biology and Chemistry, College of Liberal Arts and Sciences, National University of Defense Technology, Changsha, China. Long non-coding RNAs (lncRNAs) regulate gene expression in a variety of ways at epigenetic, chromatin remodeling, transcriptional, and translational levels. Accumulating evidence suggests that lncRNA X-inactive specific transcript (lncRNA Xist) serves as an important regulator of cell growth and development. Despites its original roles in X-chromosome dosage compensation, lncRNA Xist also participates in the development of tumor and other human diseases by functioning as a competing endogenous RNA (ceRNA). In this review, we comprehensively summarized recent progress in understanding the cellular functions of lncRNA Xist in mammalian cells and discussed current knowledge regarding the ceRNA network of lncRNA Xist in various diseases. Long non-coding RNAs (lncRNAs) are transcripts that are more than 200 nt in length and without an apparent protein-coding capacity (Furlan and Rougeulle, 2016; Maduro et al., 2016). These RNAs are believed to be transcribed by the approximately 98-99% non-coding regions of the human genome (Derrien et al., 2012; Fu, 2014; Montalbano et al., 2017; Slack and Chinnaiyan, 2019), as well as a large variety of genomic regions, such as exonic, tronic, and intergenic regions. Hence, lncRNAs are also divided into eight categories: Intergenic lncRNAs, Intronic lncRNAs, Enhancer lncRNAs, Promoter lncRNAs, Natural antisense/sense lncRNAs, Small nucleolar RNA-ended lncRNAs (sno-lncRNAs), Bidirectional lncRNAs, and non-poly(A) lncRNAs (Ma et al., 2013; Devaux et al., 2015; St Laurent et al., 2015; Chen, 2016; Quinn and Chang, 2016; Richard and Eichhorn, 2018; Connerty et al., 2020). A range of evidence has suggested that lncRNAs function as key regulators in crucial cellular functions, including proliferation, differentiation, apoptosis, migration, and invasion, by regulating the expression level of target genes via epigenomic, transcriptional, or post-transcriptional approaches (Cao et al., 2018). Moreover, lncRNAs detected in body fluids were also believed to serve as potential biomarkers for the diagnosis, prognosis, and monitoring of disease progression, and act as novel and potential drug targets for therapeutic exploitation in human disease (Jiang W. et al., 2018; Zhou et al., 2019a). Long non-coding RNA X-inactive specific transcript (lncRNA Xist) are a set of 15,000-20,000 nt sequences localized in the X chromosome inactivation center (XIC) of chromosome Xq13.2 (Brown et al., 1992; Debrand et al., 1998; Kay, 1998; Lee et al., 2013; da Rocha and Heard, 2017; Yang Z. et al., 2018; Brockdorff, 2019). Previous studies have indicated that lncRNA Xist regulate X chromosome inactivation (XCI), resulting in the inheritable silencing of one of the X-chromosomes during female cell development. Also, it serves a vital regulatory function in the whole spectrum of human disease (notably cancer) and can be used as a novel diagnostic and prognostic biomarker and as a potential therapeutic target for human disease in the clinic (Liu et al., 2018b; Deng et al., 2019; Dinescu et al., 2019; Mutzel and Schulz, 2020; Patrat et al., 2020; Wang et al., 2020a). In particular, lncRNA Xist have been demonstrated to be involved in the development of multiple types of tumors including brain tumor, Leukemia, lung cancer, breast cancer, and liver cancer, with the prominent examples outlined in Table 1. It was also believed that lncRNA Xist (Chaligne and Heard, 2014; Yang Z. et al., 2018) contributed to other diseases, such as pulmonary fibrosis, inflammation, neuropathic pain, cardiomyocyte hypertrophy, and osteoarthritis chondrocytes, and more specific details can be found in Table 2. This review summarizes the current knowledge on the regulatory mechanisms of lncRNA Xist on both chromosome dosage compensation and pathogenesis (especially cancer) processes, with a focus on the regulatory network of lncRNA Xist in human disease. Copyright © 2021 Wang, Min, Qiu, Wu, Liu, Ma, Zhang and Zhu. DOI: 10.3389/fcell.2021.645647 PMCID: PMC8222981 PMID: 34178980 Conflict of interest statement: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
http://www.ncbi.nlm.nih.gov/pubmed/30171939
1. Gene. 2018 Dec 30;679:28-35. doi: 10.1016/j.gene.2018.08.071. Epub 2018 Aug 29. X-inactive-specific transcript: A long noncoding RNA with complex roles in human cancers. Yang Z(1), Jiang X(2), Jiang X(3), Zhao H(4). Author information: (1)Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, China. (2)Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang, China. (3)Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, China. Electronic address: [email protected]. (4)Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, China. Electronic address: [email protected]. The X-inactive-specific transcript (XIST/Xist) is one of the first long non-coding RNAs discovered in mammals and plays an essential role in X chromosome inactivation. XIST is dysregulated and acts as an oncogene or a tumor suppressor in different human malignancies. XIST is implicated in many aspects of carcinogenesis including tumor initiation, invasion, metastasis, apoptosis, cell cycle, stemness, autophagy, and drug resistance. This review focuses on research progress on the roles of XIST in tumor development. The multiple pathological functions of XIST in various cancers are systematically reviewed to elucidate the molecular basis of its biological roles and to provide new directions for future research. Copyright © 2018 Elsevier B.V. All rights reserved. DOI: 10.1016/j.gene.2018.08.071 PMID: 30171939 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/29302591
1. Front Mol Biosci. 2017 Dec 19;4:90. doi: 10.3389/fmolb.2017.00090. eCollection 2017. Function by Structure: Spotlights on Xist Long Non-coding RNA. Pintacuda G(1), Young AN(2), Cerase A(2). Author information: (1)Department of Biochemistry, University of Oxford, Oxford, United Kingdom. (2)European Molecular Biology Laboratory, Monterotondo, Italy. Recent experimental evidence indicates that lncRNAs can act as regulatory molecules in the context of development and disease. Xist, the master regulator of X chromosome inactivation, is a classic example of how lncRNAs can exert multi-layered and fine-tuned regulatory functions, by acting as a molecular scaffold for recruitment of distinct protein factors. In this review, we discuss the methodologies employed to define Xist RNA structures and the tight interplay between structural clues and functionality of lncRNAs. This model of modular function dictated by structure, can be also generalized to other lncRNAs, beyond the field of X chromosome inactivation, to explain common features of similarly folded RNAs. DOI: 10.3389/fmolb.2017.00090 PMCID: PMC5742192 PMID: 29302591
http://www.ncbi.nlm.nih.gov/pubmed/23816838
1. J Mol Biol. 2013 Oct 9;425(19):3698-706. doi: 10.1016/j.jmb.2013.06.031. Epub 2013 Jun 28. Guided by RNAs: X-inactivation as a model for lncRNA function. Froberg JE(1), Yang L, Lee JT. Author information: (1)Howard Hughes Medical Institute, Boston, MA 02114, USA; Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Genetics, Harvard Medical School, Boston, MA 02114, USA. The recent revolution in sequencing technology has helped to reveal a large transcriptome of long non-coding RNAs (lncRNAs). A major challenge in the years to come is to determine what biological functions, if any, they serve. Although the purpose of these transcripts is largely unknown at present, existing examples suggest that lncRNAs play roles in a wide variety of biological processes. Exemplary cases are lncRNAs within the X-inactivation center. Indeed, lncRNAs dominate control of random X-chromosome inactivation (XCI). The RNA-based regulatory mechanisms of XCI include recruitment of chromatin modifiers, formation of RNA-based subnuclear compartments, and regulation of transcription by antisense transcription. XCI and lncRNAs now also appear to be very relevant in the development and progression of cancer. This perspective focuses on new insights into lncRNA-dependent regulation of XCI, which we believe serve as paradigms for understanding lncRNA function more generally. © 2013. DOI: 10.1016/j.jmb.2013.06.031 PMCID: PMC3771680 PMID: 23816838 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/35895262
1. Methods Mol Biol. 2022;2537:129-147. doi: 10.1007/978-1-0716-2521-7_8. Identification and Quantification of Microexons Using Bulk and Single-Cell RNA-Seq Data. Parada GE(1)(2), Hemberg M(3)(4). Author information: (1)Wellcome Sanger Institute, Cambridge, UK. (2)Donnelly Centre for Cellular and Biomolecular Research and Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada. (3)Wellcome Sanger Institute, Cambridge, UK. [email protected]. (4)Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA. [email protected]. The analysis of RNA-seq has greatly improved the characterization and understanding of the transcriptome. In particular, RNA-seq experiments have extended catalogs of alternative splicing events. However, the analysis of RNAs-seq data for detection and quantification of microexons, extremely short exons of length up to 30 nt, require specialized computational workflows. Here, we describe MicroExonator, a reproducible computational workflow for microexon splicing analysis using bulk or single-cell RNA-seq data. © 2022. The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature. DOI: 10.1007/978-1-0716-2521-7_8 PMID: 35895262 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/35364798
1. Drugs. 2022 Apr;82(6):703-710. doi: 10.1007/s40265-022-01704-4. Tebentafusp: First Approval. Dhillon S(1). Author information: (1)Springer Nature, Private Bag 65901, Mairangi Bay, Auckland, 0754, New Zealand. [email protected]. Tebentafusp (tebentafusp-tebn; Kimmtrak®) is a first-in-class, bispecific gp100 peptide-HLA-A*02:01 directed T cell receptor (TCR) CD3 T cell engager being developed by Immunocore for the treatment of uveal melanoma and malignant melanoma. The TCR arm of tebentafusp binds to HLA-A*02:01-positive uveal melanoma cells and activates polyclonal T cells, through CD3, to release inflammatory cytokines and cytolytic proteins, resulting in the direct lysis of tumour cells. In January 2022, tebentafusp received its first approval in the USA for the treatment of HLA-A*02:01-positive adults with unresectable or metastatic uveal melanoma, and in February 2022 received a Positive Opinion from the EU Committee for Medicinal Products for Human Use for the treatment of uveal melanoma. Tebentafusp is under regulatory review for the treatment of metastatic uveal melanoma in the UK, Australia and Canada. Clinical studies of tebentafusp are underway for uveal melanoma and cutaneous melanoma in several countries worldwide. This article summarizes the milestones in the development of tebentafusp leading to this first approval for unresectable or metastatic uveal melanoma. © 2022. The Author(s), under exclusive licence to Springer Nature Switzerland AG. DOI: 10.1007/s40265-022-01704-4 PMID: 35364798 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/35364557
1. Transl Oncol. 2022 Jun;20:101408. doi: 10.1016/j.tranon.2022.101408. Epub 2022 Mar 29. Tebentafusp in first-line melanoma trials: An outperforming outlier. Olivier T(1), Prasad V(2). Author information: (1)Department of Oncology, Geneva University Hospital, 4 Gabrielle-Perret-Gentil Street, Geneva 1205, Switzerland; Department of Epidemiology and Biostatistics, University of California San Francisco, 550 16th St, 2nd Fl, San Francisco, CA 94158, USA. Electronic address: [email protected]. (2)Department of Epidemiology and Biostatistics, University of California San Francisco, 550 16th St, 2nd Fl, San Francisco, CA 94158, USA. Uveal melanoma is distinct from other melanomas. In the advanced and metastatic stages, little to no improvement have been seen over time. Tebentafusp is a novel mechanism of action bispecific gp100 peptide-HLA-directed CD3 T-cell engager fusion protein ("-fusp"). Tebentafusp was granted full approval on January 25th 2022 in the setting of HLA-A*02:01-positive adult patients with unresectable or metastatic uveal melanoma. The approval was based on the overall survival advantage of tebentafusp over physician's choice therapy, in previously untreated uveal melanoma patients, based on the IMCgp100-202 trial. While we welcome positive results for this unmet need, three issues are raised by the trial. First, the control arm was restricted, precluding important options. Second, post-progression treatment was provided to a smaller fraction of patients than in real-life, which raises the question of whether overall survival was negatively impacted by limited care after the trial ended. Finally, the discrepancy between overall survival and progression-free survival benefit is an outlier in the context of previous melanoma trials. While it is clear that tebentafusp has an important role to play in this tumor type, the exact line is not yet well known. Confirmatory trials are needed for this compound. Copyright © 2022. Published by Elsevier Inc. DOI: 10.1016/j.tranon.2022.101408 PMCID: PMC8968051 PMID: 35364557 Conflict of interest statement: Vinay Prasad's Disclosures: Research funding: Arnold Ventures; Royalties: Johns Hopkins Press, Medscape; Honoraria: Grand Rounds/lectures from universities, medical centers, non-profits, and professional societies; Consulting: UnitedHealthcare; Speaking fees: Evicore; Other: Plenary Session podcast has Patreon backers. Timothée Olivier have no financial nor non-financial conflicts of interest to report.
http://www.ncbi.nlm.nih.gov/pubmed/34999237
1. Prog Retin Eye Res. 2022 Sep;90:101041. doi: 10.1016/j.preteyeres.2022.101041. Epub 2022 Jan 6. Metastatic uveal melanoma: The final frontier. Rantala ES(1), Hernberg MM(2), Piperno-Neumann S(3), Grossniklaus HE(4), Kivelä TT(5). Author information: (1)Ocular Oncology Service, Department of Ophthalmology, University of Helsinki and Helsinki University Hospital, Haartmaninkatu 4 C, PL 220, FI-00029, HUS, Helsinki, Finland. Electronic address: [email protected]. (2)Comprehensive Cancer Center, Department of Oncology, Helsinki University Hospital and University of Helsinki, Paciuksenkatu 3, PL 180, FI-00029, HUS, Helsinki, Finland. Electronic address: [email protected]. (3)Department of Medical Oncology, Institut Curie, 26, rue d'Ulm, 75005, Paris, France. Electronic address: [email protected]. (4)Section of Ocular Oncology, Emory Eye Center, 1365 Clifton Road B, Atlanta, GA, 30322, USA. Electronic address: [email protected]. (5)Ocular Oncology Service, Department of Ophthalmology, University of Helsinki and Helsinki University Hospital, Haartmaninkatu 4 C, PL 220, FI-00029, HUS, Helsinki, Finland. Electronic address: [email protected]. Treatment of primary intraocular uveal melanoma has developed considerably, its driver genes are largely unraveled, and the ways to assess its risk for metastases are very precise, being based on an international staging system and genetic data. Unfortunately, the risk of distant metastases, which emerge in approximately one half of all patients, is unaltered. Metastases are the leading single cause of death after uveal melanoma is diagnosed, yet no consensus exists regarding surveillance, staging, and treatment of disseminated disease, and survival has not improved until recently. The final frontier in conquering uveal melanoma lies in solving these issues to cure metastatic disease. Most studies on metastatic uveal melanoma are small, uncontrolled, retrospective, and do not report staging. Meta-analyses confirm a median overall survival of 10-13 months, and a cure rate that approaches nil, although survival exceeding 5 years is possible, estimated 2% either with first-line treatment or with best supportive care. Hepatic ultrasonography and magnetic resonance imaging as surveillance methods have a sensitivity of 95-100% and 83-100%, respectively, to detect metastases without radiation hazard according to prevailing evidence, but computed tomography is necessary for staging. No blood-based tests additional to liver function tests are generally accepted. Three validated staging systems predict, each in defined situations, overall survival after metastasis. Their essential components include measures of tumor burden, liver function, and performance status or metastasis free interval. Age and gender may additionally influence survival. Exceptional mutational events in metastases may make them susceptible to checkpoint inhibitors. In a large meta-analysis, surgical treatment was associated with 6 months longer median overall survival as compared to conventional chemotherapy and, recently, tebentafusp as first-line treatment at the first interim analysis of a randomized phase III trial likewise provided a 6 months longer median overall survival compared to investigator's choice, mostly pembrolizumab; these treatments currently apply to selected patients. Promoting dormancy of micrometastases, harmonizing surveillance protocols, promoting staging, identifying predictive factors, initiating controlled clinical trials, and standardizing reporting will be critical steppingstones in reaching the final frontier of curing metastatic uveal melanoma. Copyright © 2022. Published by Elsevier Ltd. DOI: 10.1016/j.preteyeres.2022.101041 PMID: 34999237 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/35257475
1. Aging Cell. 2022 Apr;21(4):e13579. doi: 10.1111/acel.13579. Epub 2022 Mar 8. Deuterated docosahexaenoic acid protects against oxidative stress and geographic atrophy-like retinal degeneration in a mouse model with iron overload. Liu Y(1), Bell BA(1), Song Y(1), Zhang K(1), Anderson B(1), Axelsen PH(2), Bohannan W(3), Agbaga MP(3), Park HG(4), James G(4), Brenna JT(4), Schmidt K(5), Dunaief JL(1), Shchepinov MS(5). Author information: (1)F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA. (2)Department of Pharmacology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA. (3)Departments of Cell Biology and Ophthalmology, University of Oklahoma Health Sciences Center and the Dean McGee Eye Institute, Oklahoma City, Oklahoma, USA. (4)Dell Pediatric Research Institute, University of Texas at Austin, Austin, Texas, USA. (5)Retrotope, Inc., Los Altos, California, USA. Oxidative stress plays a central role in age-related macular degeneration (AMD). Iron, a potent generator of hydroxyl radicals through the Fenton reaction, has been implicated in AMD. One easily oxidized molecule is docosahexaenoic acid (DHA), the most abundant polyunsaturated fatty acid in photoreceptor membranes. Oxidation of DHA produces toxic oxidation products including carboxyethylpyrrole (CEP) adducts, which are increased in the retinas of AMD patients. In this study, we hypothesized that deuterium substitution on the bis-allylic sites of DHA in photoreceptor membranes could prevent iron-induced retinal degeneration by inhibiting oxidative stress and lipid peroxidation. Mice were fed with either DHA deuterated at the oxidation-prone positions (D-DHA) or control natural DHA and then given an intravitreal injection of iron or control saline. Orally administered D-DHA caused a dose-dependent increase in D-DHA levels in the neural retina and retinal pigment epithelium (RPE) as measured by mass spectrometry. At 1 week after iron injection, D-DHA provided nearly complete protection against iron-induced retinal autofluorescence and retinal degeneration, as determined by in vivo imaging, electroretinography, and histology. Iron injection resulted in carboxyethylpyrrole conjugate immunoreactivity in photoreceptors and RPE in mice fed with natural DHA but not D-DHA. Quantitative PCR results were consistent with iron-induced oxidative stress, inflammation, and retinal cell death in mice fed with natural DHA but not D-DHA. Taken together, our findings suggest that DHA oxidation is central to the pathogenesis of iron-induced retinal degeneration. They also provide preclinical evidence that dosing with D-DHA could be a viable therapeutic strategy for retinal diseases involving oxidative stress. © 2022 The Authors. Aging Cell published by Anatomical Society and John Wiley & Sons Ltd. DOI: 10.1111/acel.13579 PMCID: PMC9009113 PMID: 35257475 [Indexed for MEDLINE] Conflict of interest statement: MSS and KS are employed by Retrotope, Inc. JTB is a shareholder in and receives research support from Retrotope, Inc. MPA receives research support from Retrotope, Inc. The other authors declare that they have no commercial or financial relationships that could be construed as a potential conflict of interest.
http://www.ncbi.nlm.nih.gov/pubmed/21280898
1. J Biomed Opt. 2011 Jan-Feb;16(1):011011. doi: 10.1117/1.3524304. Mechanism of ceroid formation in atherosclerotic plaque: in situ studies using a combination of Raman and fluorescence spectroscopy. Haka AS(1), Kramer JR, Dasari RR, Fitzmaurice M. Author information: (1)Massachusetts Institute of Technology, G. R. Harrison Spectroscopy Laboratory, Cambridge, Massachusetts 02139, USA. [email protected] Accumulation of the lipid-protein complex ceroid is a characteristic of atherosclerotic plaque. The mechanism of ceroid formation has been extensively studied, because the complex is postulated to contribute to plaque irreversibility. Despite intensive research, ceroid deposits are defined through their fluorescence and histochemical staining properties, while their composition remains unknown. Using Raman and fluorescence spectral microscopy, we examine the composition of ceroid in situ in aorta and coronary artery plaque. The synergy of these two types of spectroscopy allows for identification of ceroid via its fluorescence signature and elucidation of its chemical composition through the acquisition of a Raman spectrum. In accordance with in vitro predictions, low density lipoprotein (LDL) appears within the deposits primarily in its peroxidized form. The main forms of modified LDL detected in both coronary artery and aortic plaques are peroxidation products from the Fenton reaction and myeloperoxidase-hypochlorite pathway. These two peroxidation products occur in similar concentrations within the deposits and represent ∼40 and 30% of the total LDL (native and peroxidized) in the aorta and coronary artery deposits, respectively. To our knowledge, this study is the first to successfully employ Raman spectroscopy to unravel a metabolic pathway involved in disease pathogenesis: the formation of ceroid in atherosclerotic plaque. DOI: 10.1117/1.3524304 PMCID: PMC3041153 PMID: 21280898 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/29227084
1. Ukr Biochem J. 2016 Jan-Feb;88(1):79-87. doi: 10.15407/ubj88.01.079. Amine oxidases as important agents of pathological processes of rhabdomyolysis in rats. Gudkova OO, Latyshko NV, Shandrenko SG. In this study we have tested an idea on the important role of amine oxidases (semicarbazide-sensitive amine oxidase, diamine oxidase, polyamine oxidase) as an additional source of oxidative/carbonyl stress under glycerol-induced rhabdomyolysis, since the enhanced formation of reactive oxygen species and reactive carbonyl species in a variety of tissues is linked to various diseases. In our experiments we used the sensitive fluorescent method devised for estimation of amine oxidases activity in the rat kidney and thymus as targeted organs under rhabdomyolysis. We have found in vivo the multiple rises in activity of semicarbazide-sensitive amine oxidase, diamine oxidase, polyamine oxidase (2-4.5 times) in the corresponding cell fractions, whole cells or their lysates at the 3-6th day after glycerol injection. Aberrant antioxidant activities depended on rhabdomyolysis stage and had organ specificity. Additional treatment of animals with metal chelator ‘Unithiol’ adjusted only the activity of antioxidant enzymes but not amine oxidases in both organs. Furthermore the in vitro experiment showed that Fenton reaction (hydrogen peroxide in the presence of iron) products alone had no effect on semicarbazide-sensitive amine oxidase activity in rat liver cell fraction whereas supplementation with methylglyoxal resulted in its significant 2.5-fold enhancement. Combined action of the both agents had additive effect on semicarbazide-sensitive amine oxidase activity. We can assume that biogenic amine and polyamine catabolism by amine oxidases is upregulated by oxidative and carbonyl stress factors directly under rhabdomyolysis progression, and the increase in catabolic products concentration contributes to tissue damage in glycerol-induced acute renal failure and apoptosis stimulation in thymus. DOI: 10.15407/ubj88.01.079 PMID: 29227084 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/35775547
1. Int J Mycobacteriol. 2022 Apr-Jun;11(2):150-158. doi: 10.4103/ijmy.ijmy_58_22. Mycobacterium smegmatis strains genetically resistant to moxifloxacin emerge de novo from the moxifloxacin-surviving population containing high levels of superoxide, H(2)O(2), hydroxyl radical, and Fe (II). Paul A(1), Nair RR(1), Jakkala K(1), Ajitkumar P(1). Author information: (1)Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka, India. BACKGROUND: The antibiotic-exposed bacteria often contain the reactive oxygen species (ROS), hydroxyl radical, which inflicts genome-wide mutations, causing the de novo formation of antibiotic-resistant strains. Hydroxyl radical is generated by Fenton reaction of Fe (II) with the ROS, H2O2, which, in turn, is formed by the dismutation of the ROS, superoxide. Therefore, for the emergence of bacterial strains genetically resistant to antibiotics, increased levels of superoxide, H2O2, hydroxyl radical, and Fe (II) should be present in the antibiotic-exposed bacteria. Here, we verified this premise by finding out whether the in vitro cultures of M. smegmatis, exposed to MBC of moxifloxacin for a prolonged duration, contain significantly high levels of superoxide, H2O2, hydroxyl radical, and Fe (II). METHODS: Biological triplicate cultures of M. smegmatis, were exposed to MBC of moxifloxacin for 84 h. The colony-forming units (CFUs) of the cultures were determined on moxifloxacin-free and moxifloxacin-containing plates for the entire 84 h at a regular interval of 6 h. The cultures were analyzed at specific time points of killing phase (KP), antibiotic-surviving phase (ASP), and regrowth phase (RGP) for the presence of superoxide, H2O2, hydroxyl radical, and Fe (II) using the ROS- and Fe (II)-detecting fluorescence probes. The experimental cultures were grown in the presence of ROS and Fe (II) quenchers also and determined the levels of fluorescence corresponding to the ROS- and Fe (II)-specific probes. This was performed to establish the specificity of detection of ROS and Fe (II). Biological triplicate cultures, unexposed to moxifloxacin but cultured for 84 h, were used as the control for the measurement of ROS and Fe (II) levels. The CFUs of the cultures were determined on moxifloxacin-free and moxifloxacin-containing plates for the entire 84 h at regular intervals of 6 h. Flow cytometry analyses were performed for the detection and quantitation of the levels of fluorescence of the ROS-and Fe (II)-specific probes. The experimental cultures were grown in the presence of thiourea and bipyridyl as the ROS and Fe (II) quenchers, respectively, for the determination of the levels of fluorescence corresponding to the ROS- and Fe (II)-specific probes. Paired t-test was used to calculate statistical significance (n = 3). RESULTS: The moxifloxacin-exposed cultures, but not the cultures unexposed to moxifloxacin, showed a triphasic response with a KP, ASP, and RGP. The cells in the late KP and ASP contained significantly elevated levels of superoxide, H2O2, hydroxyl radical, and Fe (II). Thus, high levels of the ROS and Fe (II) were found in the small population (in the ASP) of M. smegmatis cells that survived the moxifloxacin-mediated killing. From this moxifloxacin-surviving population (in the ASP), moxifloxacin-resistant genetic resisters emerged de novo at high frequency, regrew, divided, and populated the cultures. The levels of these ROS, Fe (II), and the high moxifloxacin resister generation frequency were quenched in the cultures grown in the presence of the respective ROS and Fe (II) quenchers. The cultures unexposed to moxifloxacin did not show any of these responses, indicating that the whole response was specific to antibiotic exposure. CONCLUSIONS: Significantly high levels of superoxide, H2O2, hydroxyl radical, and Fe (II) were generated in the M. smegmatis cultures exposed to moxifloxacin for a prolonged duration. It promoted the de novo emergence of genetic resisters to moxifloxacin at high frequency. DOI: 10.4103/ijmy.ijmy_58_22 PMID: 35775547 [Indexed for MEDLINE] Conflict of interest statement: None
http://www.ncbi.nlm.nih.gov/pubmed/16433646
1. J Prosthodont. 2006 Jan-Feb;15(1):9-19. doi: 10.1111/j.1532-849X.2006.00063.x. Free radical damage in facsimile synovium: correlation with adhesion formation in osteoarthritic TMJs. Sheets DW Jr(1), Okamoto T, Dijkgraaf LC, Milam SB, Schmitz JP, Zardeneta G. Author information: (1)Department of Prosthodontics, Wilford Hall Medical Center, Lackland AFB, San Antonio, TX, USA. PURPOSE: The purpose of this study was to use the rat air pouch model of facsimile synovium to evaluate oxidative stress as a primary mechanism in the pathogenesis of degenerative temporomandibular joint (TMJ) disease. MATERIALS AND METHODS: Forty-nine Sprague-Dawley adult female rats were used to generate the standard rat air pouch model of facsimile synovium. This was accomplished by daily air injections (20 cc) subdermally through the dorsal skin. Hydrogen peroxide and ferrous iron (components of the Fenton reaction which generate free radicals) were introduced into the pouches of the 4-, 7-, and 14-day groups to generate oxidative stress. Control rats were injected with phosphate-buffered solution (PBS), pH 7.4. Either N-acetylcysteine (NAC), a powerful free radical scavenger, or ibuprofen were simultaneously injected with the Fenton reagents into the pouches of the 14-day treatment groups to modulate free radical-mediated protein damage to the synovium. Animals were euthanized at appropriate experimental intervals and biopsies obtained from specimens to analyze: (1) proteins' amino acid modification (carbonyl group formation), (2) protein hydrophobicity, (3) detection of low molecular weight protein degradation products, and (4) histological and gross anatomical observations. RESULTS: Free radicals introduced into the rat air pouch interacted with synovial tissues causing oxidation and breakdown of proteins. Clinical evidence of adhesion formation consistent with features found in osteoarthritis of the TMJ developed. The groups subjected to oxidative stress experienced statistically significant (p < 0.05) increases in carbonyl formation, carbonyls/protein, and low molecular weight protein fragments. These groups also showed significant (p < 0.05) hydrophobicity changes consistent with free radical attack. Control synovial tissues were statistically undamaged. The 14-day NAC and ibuprofen treatment groups experienced statistically significant (p < 0.05) decreases in total carbonyl formation, carbonyls/protein, and hydrophobicity. Histological and gross observations in free radical damaged synovium exhibited features consistent with known arthoscopic and arthrocentesis findings in diseased TMJs. CONCLUSIONS: This study suggests that the rat air pouch model of facsimile synovium develops clinical evidence of adhesions and biochemical signs of protein modification when subjected to free radical attack. NAC and ibuprofen prevented carbonyl formation as well as hydrophobicity changes indicative of oxidative stress damage in facsimile synovium. These findings are consistent with features of degenerative human TMJ disease. Future direction may be taken from this study to postulate new analysis techniques and treatment modalities for patients with degenerative TMJ disease. Copyright (c) 2006 by The American College of Prosthodontists. DOI: 10.1111/j.1532-849X.2006.00063.x PMID: 16433646 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/31143689
1. Avicenna J Phytomed. 2019 May-Jun;9(3):213-220. Protective effect of Berberis vulgaris on Fenton reaction-induced DNA cleavage. Sadat Asadi N(1), Heidari MM(1), Khatami M(1). Author information: (1)Department of Biology, Faculty of Science, Yazd University, Yazd, Iran. OBJECTIVE: Berberis vulgaris contains antioxidants that can inhibit DNA cleavage. The purpose of this study was to evaluate the antioxidant and protective activity of B. vulgaris on DNA cleavage. MATERIALS AND METHODS: In this study, the antioxidant capacity of B. vulgaris was investigated using DPPH and its protective effect was evaluated on pBR322 plasmid and lymphocyte genomic DNA cleavage induced by Fenton reaction, by DNA electrophoresis. RESULTS: Aqueous extract of B. vulgaris presented dual behavior with a potent antioxidant activity at 0.25and 0.75mg/ml for pBR322 plasmid and lymphocyte genomic DNA, respectively, but a pro-oxidant activity was observed at higher concentrations. CONCLUSION: Our results indicated that B. vulgaris extract an inhibit Fenton reaction-induced DNA cleavage and oxidative cleavage of double-stranded DNA assay is a powerful technique that can be used to determine the antioxidant and pro-oxidant properties of a compound on cellular components such as DNA. PMCID: PMC6526040 PMID: 31143689 Conflict of interest statement: The authors declare no conflict of interest.
http://www.ncbi.nlm.nih.gov/pubmed/8597169
1. Toxicol Lett. 1995 Dec;82-83:969-74. doi: 10.1016/0378-4274(95)03532-x. Toxicity of iron and hydrogen peroxide: the Fenton reaction. Winterbourn CC(1). Author information: (1)Department of Pathology, Christchurch School of Medicine, New Zealand. [email protected] Iron and hydrogen peroxide are capable of oxidizing a wide range of substrates and causing biological damage. The reaction, referred to as the Fenton reaction, is complex and capable of generating both hydroxyl radicals and higher oxidation states of the iron. The mechanism and how it is affected by different chelators, and the interpretation of results obtained in biological systems, are discussed. DOI: 10.1016/0378-4274(95)03532-x PMID: 8597169 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/27340836
1. J Phys Chem A. 2016 Jul 21;120(28):5435-45. doi: 10.1021/acs.jpca.6b03805. Epub 2016 Jul 12. New Insights of the Fenton Reaction Using Glycerol as the Experimental Model. Effect of O2, Inhibition by Mg(2+), and Oxidation State of Fe. Vitale AA(1), Bernatene EA(2), Vitale MG(3), Pomilio AB(1)(2). Author information: (1)Area Hematología, Departamento de Bioquímica Clínica, Hospital de Clínicas "José de San Martín", Universidad de Buenos Aires , Av. Córdoba 2351, C1120AAF Buenos Aires, Argentina. (2)Instituto de Bioquímica y Medicina Molecular (IBIMOL, CONICET-UBA) , Junín 956, C1113AAD Buenos Aires, Argentina. (3)Hospital Infanto Juvenil "Dra. Carolina Tobar García", Universidad de Buenos Aires , Doctor Ramón Carrillo 315, C1275AHG Buenos Aires, Argentina. The use of iron ions as catalyst of oxidation with hydrogen peroxide, known as the Fenton reaction, is important for industry and biological systems. It has been widely studied since its discovery in the 19th century, but important aspects of the reaction as which is the oxidant, the role of oxygen, and the oxidation state of Fe still remain unclear. In this work new mechanistic insights of the oxidation of carbohydrates by the Fenton reaction using glycerol as experimental model are described. The reaction was studied by means of oxidation reduction potential (ORP) measures. The stoichiometry was measured, showing the important role of oxygen for lowering H2O2 consumption under aerobic conditions. Evidence is provided to demonstrate that in this system Fe(2+) generates a catalyst by reacting with a substrate to produce a complex, which gives rise to singlet oxygen after reacting with H2O2. This is the first time that the inhibitor effect of Mg(2+) is reported in this reaction, and its participation in the mechanism is described. A rational mechanism for the oxidation of glycerol using the Fenton reaction under these specific conditions is proposed. The role of oxygen, the participation of Fe(2+), and the inhibition by Mg(2+) are fully demonstrated experimentally. DOI: 10.1021/acs.jpca.6b03805 PMID: 27340836
http://www.ncbi.nlm.nih.gov/pubmed/32668496
1. Water Environ Res. 2021 Apr;93(4):645-651. doi: 10.1002/wer.1397. Epub 2020 Nov 4. A sandwich model of Cr(VI) adsorption and detoxification by Fenton modified chitosan. Jia N(1), Yun L(1), Huang J(1)(2), Chen H(3), Shen C(2), Wen Y(1). Author information: (1)MOE Key Laboratory of Environmental Remediation & Ecosystem Health, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, China. (2)College of Environmental Science and Engineering, Donghua University, Shanghai, China. (3)College of Science and Technology, Ningbo University, Ningbo, China. The Fenton reaction has the advantages of short reaction time, low cost, no toxicity, and straightforward application and control. The Fenton reaction generates highly reactive HO•, which has been applied effectively. However, the effect of the generated Fe3+ has not been investigated widely. In this study, the Fenton reaction was used to improve the Cr(VI) adsorption and detoxification capacities of chitosan. After the Fenton modification, chitosan efficiently adsorbed Cr(VI) and transformed it into the less toxic Cr(III) in a wide pH range as a result of layer formation, which was described by a sandwich model. The adsorption of Cr(VI) onto the Fenton modified chitosan was in good agreement with the Freundlich adsorption model, and the adsorption capacity exceeded 120 mg/g. During the Fenton reaction, H2 O2 and HO• with high oxidative activity broke the hydrogen bonds in the chitosan structure, resulting in the release of free amine groups for Fe3+ to form metal-binding biopolymers. The distance between the chitosan polymers increased, and additional adsorption sites were created. HCrO4 - entered the gap between the chitosan polymer and was adsorbed on the newly created adsorption sites. The sandwich adsorption model indicated that the Fenton modified chitosan provided a high concentration of active sites for Cr(VI) capture and detoxification. PRACTITIONER POINTS: Fenton reaction was used to improve the adsorption ability of chitosan. The formed Fe3+ in Fenton reaction was utilized. HO· broke the hydrogen bonds and Fe3+ ions chelated with chitosan in modification. Cr(VI) could be adsorbed and reduced efficiently by Fenton modified chitosan. The Fenton modified chitosan provided a high concentration of active sites for Cr(VI) capture and detoxification. © 2020 Water Environment Federation. DOI: 10.1002/wer.1397 PMID: 32668496 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/36080218
1. Molecules. 2022 Aug 25;27(17):5451. doi: 10.3390/molecules27175451. Current Use of Fenton Reaction in Drugs and Food. Abe C(1), Miyazawa T(1), Miyazawa T(1). Author information: (1)New Industry Creation Hatchery Center (NICHe), Tohoku University, Sendai 980-8579, Japan. Iron is the most abundant mineral in the human body and plays essential roles in sustaining life, such as the transport of oxygen to systemic organs. The Fenton reaction is the reaction between iron and hydrogen peroxide, generating hydroxyl radical, which is highly reactive and highly toxic to living cells. "Ferroptosis", a programmed cell death in which the Fenton reaction is closely involved, has recently received much attention. Furthermore, various applications of the Fenton reaction have been reported in the medical and nutritional fields, such as cancer treatment or sterilization. Here, this review summarizes the recent growing interest in the usefulness of iron and its biological relevance through basic and practical information of the Fenton reaction and recent reports. DOI: 10.3390/molecules27175451 PMCID: PMC9457891 PMID: 36080218 [Indexed for MEDLINE] Conflict of interest statement: The authors declare no conflict of interest.
http://www.ncbi.nlm.nih.gov/pubmed/30152511
1. Phys Chem Chem Phys. 2018 Sep 12;20(35):22890-22901. doi: 10.1039/c8cp04381g. A computational study of the Fenton reaction in different pH ranges. Lu HF (1), Chen HF , Kao CL , Chao I , Chen HY . Author information: (1)Institute of Chemistry, Academia Sinica, Taipei 11529, Taiwan. The reaction of iron(ii) and hydrogen peroxide, namely the Fenton reaction, is well-known for its strong oxidizing capability. While the Fenton reactions are ubiquitous and have wide applications in many areas, the detailed mechanism, especially the nature of the reactive intermediates responsible for oxidation, is not completely clear. In this work, the performances of various density functional theory (DFT) methods on the relative energies of key Fenton intermediates are evaluated. The DFT method selected from the benchmark study is then exploited to investigate the aqueous Fenton reactions in different pH conditions. The results show that at pH > 2.2, the major Fenton oxidants are high-valent oxoiron(iv) aquo complexes. However, depending on the pH conditions, these complexes can exist in three protonation states that display quite different oxidation reactivities. The oxidizing power of FeIV[double bond, length as m-dash]O is found to be principally determined by the total charge of the ligands and is less influenced by the axial ligand effect. Moreover, the calculations reveal that the presence of the hydronium ion can stabilize the intermediate of the hydroxyl radical and further inhibit oxoiron(iv) formation via proton transfer. The contribution of hydroxyl radicals could compete with the oxoiron(iv) species at pH below 2.2. In addition, high-level ab initio calculations question the existence of the iron(iv)-dihydroxo intermediate suggested in the literature. The implications of the computational results for the Fenton oxidation process, cytochrome P450, and catalyst design are discussed. DOI: 10.1039/c8cp04381g PMID: 30152511
http://www.ncbi.nlm.nih.gov/pubmed/28531840
1. Chemosphere. 2017 Sep;182:738-744. doi: 10.1016/j.chemosphere.2017.05.039. Epub 2017 May 7. Hydroxyl radical yields in the Fenton process under various pH, ligand concentrations and hydrogen peroxide/Fe(II) ratios. Fischbacher A(1), von Sonntag C(2), Schmidt TC(3). Author information: (1)Instrumental Analytical Chemistry, University of Duisburg-Essen, Universitätsstraße 5, 45141 Essen, Germany. (2)Instrumental Analytical Chemistry, University of Duisburg-Essen, Universitätsstraße 5, 45141 Essen, Germany; Max-Planck-Institut für Bioanorganische Chemie, Stiftstr. 34-36, 45413 Mülheim an der Ruhr, Germany. (3)Instrumental Analytical Chemistry, University of Duisburg-Essen, Universitätsstraße 5, 45141 Essen, Germany; Centre for Water and Environmental Research (ZWU), University of Duisburg-Essen, Universitätsstraße 2, 45141 Essen, Germany. Electronic address: [email protected]. The Fenton process, one of several advanced oxidation processes, describes the reaction of Fe(II) with hydrogen peroxide. Fe(II) is oxidized to Fe(III) that reacts with hydrogen peroxide to Fe(II) and again initiates the Fenton reaction. In the course of the reactions reactive species, e.g. hydroxyl radicals, are formed. Conditions such as pH, ligand concentrations and the hydrogen peroxide/Fe(II) ratio may influence the OH radical yield. It could be shown that at pH < 2.7 and >3.5 the OH radical yield decreases significantly. Two ligands were investigated, pyrophosphate and sulfate. It was found that pyrophosphate forms a complex with Fe(III) that does not react with hydrogen peroxide and thus, the Fenton reaction is terminated and the OH radical yields do not further increase. The influence of sulfate is not as strong as that of pyrophosphate. The OH radical yield is decreased when sulfate is added but even at higher concentrations the Fenton reaction is not terminated. Copyright © 2017 Elsevier Ltd. All rights reserved. DOI: 10.1016/j.chemosphere.2017.05.039 PMID: 28531840 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/31259456
1. Macromol Rapid Commun. 2019 Sep;40(18):e1900220. doi: 10.1002/marc.201900220. Epub 2019 Jul 1. Fenton-Chemistry-Mediated Radical Polymerization. Reyhani A(1), McKenzie TG(1), Fu Q(1), Qiao GG(1). Author information: (1)Polymer Science Group, Department of Chemical Engineering, The University of Melbourne, Parkville, VIC, 3010, Australia. In this review, the power of a classical chemical reaction, the Fenton reaction for initiating radical polymerizations, is demonstrated. The reaction between the Fenton reagents (i.e., Fe2+ and H2 O2 ) generates highly reactive hydroxyl radicals, which can act as radical initiators for the polymerization of vinyl monomers. Since the Fenton reaction is fast, easy to set up, cheap, and biocompatible, this unique chemistry is widely employed in various polymer synthesis studies via free radical polymerization or reversible addition-fragmentation chain transfer polymerization, and is utilized in a wide range of applications, such as the fabrication of biomaterials, hydrogels, and core-shell particles. Biologically activated Fenton-mediated radical polymerization, which can be performed in aerobic environments, are particularly useful for applications in biomedical systems. © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim. DOI: 10.1002/marc.201900220 PMID: 31259456 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/36113495
1. Lancet Neurol. 2022 Nov;21(11):994-1003. doi: 10.1016/S1474-4422(22)00294-0. Epub 2022 Sep 13. Safety and efficacy of erenumab in patients with trigeminal neuralgia in Denmark: a double-blind, randomised, placebo-controlled, proof-of-concept study. Schott Andersen AS(1), Maarbjerg S(1), Noory N(1), Heinskou TB(1), Forman JL(2), Cruccu G(3), Ashina M(1), Bendtsen L(4). Author information: (1)Danish Headache Centre, Department of Neurology, Copenhagen University Hospital, Glostrup, Denmark. (2)Section of Biostatistics, Department of Public Health, University of Copenhagen, Copenhagen, Denmark. (3)Department of Neurology and Psychiatry, University of Rome-La Sapienza, Rome, Italy. (4)Danish Headache Centre, Department of Neurology, Copenhagen University Hospital, Glostrup, Denmark. Electronic address: [email protected]. Comment in Lancet Neurol. 2022 Nov;21(11):951-953. doi: 10.1016/S1474-4422(22)00389-1. BACKGROUND: Trigeminal neuralgia is a severe facial pain disorder that is difficult to treat. Erenumab, a monoclonal antibody against the calcitonin gene-related peptide (CGRP) receptor, has proven efficacy in migraine. Erenumab modulates sensory processing in peripheral trigeminal pain pathways in mice and was reported to be effective for patients with trigeminal neuralgia in open-label studies. We aimed to evaluate the efficacy of erenumab in patients with trigeminal neuralgia. METHODS: We did a randomised, double-blind, placebo-controlled trial in adults (aged 18-85 years) with idiopathic or classic trigeminal neuralgia as defined by the 3rd edition of the International Classification of Headache Disorders. The trial was based at the Danish Headache Center, Copenhagen University Hospital. Eligible participants had no clinically significant cerebrovascular or cardiovascular disease, had self-reported pain intensity of at least 4 on the 11-point Numeric Rating Scale (0=no pain, 10=worst pain imaginable), and had at least three daily pain paroxysms. After a 1-week pre-screening period, patients entered a 4-week baseline period. Participants who met pain inclusion criteria at the end of the baseline period were randomly assigned (1:1) to receive subcutaneous injections of either erenumab 140 mg or placebo and entered the 4-week follow-up period. Randomisation was done in blocks of 10 using a computer-generated schedule by a third-party company. Participants and assessors were masked to treatment allocation, and erenumab and placebo were packed in identical prefilled syringes. The primary outcome was the number of responders, defined as patients who had a reduction of at least 30% in mean average daily pain intensity during the follow-up period compared with during the baseline period, analysed in the intention-to-treat population. This trial is registered with the European Union Drug Regulating Authorities Clinical Trials Database, EudraCT number 2019-000848-95. FINDINGS: We assessed 860 patients for suitability and excluded 741 between Oct 28, 2019, and Sept 13, 2021. 119 participants entered a 1-week pre-screening period and 26 were excluded, 93 participants entered a 4-week baseline period with 13 excluded before randomisation, and 80 participants were randomly assigned to erenumab 140 mg (n=40) or placebo (n=40). There was no difference between groups in the number of responders at 4 weeks in the intention-to-treat population (14 [35%] of 40 with erenumab vs 18 [45%] of 40 with placebo; estimated effect size -10% [95% CI -31 to 11]; p=0·36). 20 (50%) of 40 participants reported adverse events in each group. The most common adverse events were constipation (28%) and headache (10%) in the erenumab group, and headache (13%), constipation (10%), and abdominal pain (10%) in the placebo group. INTERPRETATION: Erenumab did not reduce pain intensity compared with placebo in patients with trigeminal neuralgia and CGRP probably does not have an important role in paroxysmal pain. Well tolerated, effective treatments in trigeminal neuralgia are still needed. FUNDING: Novartis. Copyright © 2022 Elsevier Ltd. All rights reserved. DOI: 10.1016/S1474-4422(22)00294-0 PMID: 36113495 [Indexed for MEDLINE] Conflict of interest statement: Declaration of interests LB has given lectures and served on the scientific advisory board for Novartis, Allergan, Teva, Lundbeck, and Eli Lilly. LB received a research grant from Novartis during this study. GC has given lectures for Grunenthal and served as a consultant for Ely Lilly and Angelini. MA received personal fees from AbbVie, Allergan, Amgen, Eli Lilly, Lundbeck, Novartis, and Teva Pharmaceuticals during this study and has served as associate editor of Cephalalgia. ASSA, SM, TH, NN, and JF declare no competing interests.
http://www.ncbi.nlm.nih.gov/pubmed/26771990
1. Ann N Y Acad Sci. 2015 Nov;1358:95-104. doi: 10.1111/nyas.12965. The development of lurasidone for bipolar depression. Loebel A(1), Xu J(1), Hsu J(1), Cucchiaro J(1), Pikalov A(1). Author information: (1)Sunovion Pharmaceuticals Inc., Fort Lee, New Jersey, and Marlborough, Massachusetts. Bipolar disorder is a chronic, recurrent illness that ranks among the top 10 causes of disability in the developed world. As the illness progresses, major depressive episodes increasingly predominate. However, few treatment options are available that have demonstrated efficacy in the treatment of bipolar depression, either as monotherapy or adjunctive therapy in combination with mood stabilizers. Lurasidone is an atypical antipsychotic drug that was initially developed for the treatment of schizophrenia. Since no previous atypical antipsychotic development program had proceeded directly from work on schizophrenia to bipolar depression, the decision to focus on this indication represented an innovation in central nervous system drug development and was designed to address a clinically significant unmet need. The current review summarizes key results of a clinical development program undertaken to characterize the efficacy and safety of lurasidone in patients diagnosed with bipolar depression. Lurasidone is currently the only treatment for bipolar depression approved in the United States as both a monotherapy and an adjunctive therapy with lithium or valproate. The approval of lurasidone expands available treatment options for patients with bipolar depression and provides a therapy with an overall favorable risk-benefit profile. © 2015 The Authors. Annals of the New York Academy of Sciences published by Wiley Periodicals Inc. on behalf of The New York Academy of Sciences. DOI: 10.1111/nyas.12965 PMID: 26771990 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/33572131
1. Children (Basel). 2021 Feb 9;8(2):121. doi: 10.3390/children8020121. The Effectiveness of Lurasidone Add-On for Residual Aggressive Behavior and Obsessive Symptoms in Antipsychotic-Treated Children and Adolescents with Tourette Syndrome: Preliminary Evidence from a Case Series. Colizzi M(1)(2)(3), Bortoletto R(3), Zoccante L(3). Author information: (1)Section of Psychiatry, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy. (2)Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK. (3)Child and Adolescent Neuropsychiatry Unit, Maternal-Child Integrated Care Department, Integrated University Hospital of Verona, 37126 Verona, Italy. Children and adolescents with Tourette syndrome may suffer from comorbid psychological and behavioral difficulties, primarily Attention-Deficit Hyperactivity Disorder-related manifestations including impulsive, aggressive, and disruptive behavior, and Obsessive-Compulsive Disorder-related disturbances. Often, such additional problems represent the major cause of disability, requiring their prioritization above the tic symptomatology. Here, we present six cases of children and adolescents with treatment-resistant Tourette syndrome aged 11-17 years, whose symptoms, especially the non-tic symptoms such as aggressive behavior and obsessive symptoms, failed to respond adequately to at least two different antipsychotics and, where deemed appropriate, to a combination with a medication with a different therapeutic indication or chemical class (e.g., antidepressant or anticonvulsant). Such symptomatic manifestations were significantly reduced by the time of the subsequent control visit planned 30 days later, by using lurasidone as an add-on therapy to risperidone or aripiprazole (all p ≤ 0.009). No significant neuromotor or metabolic side effects were reported in all cases in a follow-up period ranging from 4 months to 6 months, supporting the stability of the observed clinical improvement. While still investigational, the preliminary evidence presented here gives reason to hope that lurasidone could possibly be an effective option in Tourette syndrome, warranting further investigation of its potential benefits in neurodevelopmental conditions. DOI: 10.3390/children8020121 PMCID: PMC7915970 PMID: 33572131 Conflict of interest statement: M.C. has been a consultant/advisor to GW Pharma Limited, outside of this work. All the other authors declare no conflict of interest.
http://www.ncbi.nlm.nih.gov/pubmed/25698146
1. CNS Drugs. 2015 Mar;29(3):253-63. doi: 10.1007/s40263-015-0234-2. Lurasidone: a review of its use in adult patients with bipolar I depression. Sanford M(1), Dhillon S. Author information: (1)Springer, Private Bag 65901, Mairangi Bay 0754, Auckland, New Zealand, [email protected]. Lurasidone (Latuda(®)), a benzisothiazole derivative antipsychotic, is approved in the USA and Canada for the treatment of adults with major depressive episodes (MDE) associated with bipolar I disorder; this article reviews studies of lurasidone in this indication. In two 6-week, placebo-controlled trials in adults with bipolar I depression, lurasidone 20-120 mg/day reduced depressive symptoms, either as monotherapy or as an adjunct to lithium or valproate. Lurasidone reduced the mean Montgomery-Åsberg Depression Rating Scale (MADRS) total score from baseline (primary endpoint) by >50 %; the reductions in scores were significantly greater than with placebo. The treatment effects were small to medium and the numbers needed to treat to obtain an additional MDE response (≥50 % reduction from baseline in the MADRS total score) were ≤7 across the lurasidone groups. In a third, similarly designed trial of lurasidone 20-120 mg/day adjunctive to lithium or valproate, there was no significant between-group difference in the change in the mean MADRS total score at week 6 (primary endpoint), although significant differences favouring lurasidone were observed from week 2 to week 5. Across trials, the most frequently occurring adverse events included akathisia, extrapyramidal symptoms and somnolence. Lurasidone had a favourable profile with respect to weight gain and metabolic disturbances, known to occur with some other antipsychotics. Thus, lurasidone offers a valuable addition to the therapies available for adult patients with bipolar depression, either as monotherapy or as an adjunct to lithium or valproate. DOI: 10.1007/s40263-015-0234-2 PMID: 25698146 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/24955752
1. CNS Spectr. 2015 Apr;20(2):140-7. doi: 10.1017/S1092852914000285. Epub 2014 Jun 23. Lurasidone for the treatment of depressive symptoms in schizophrenia: analysis of 4 pooled, 6-week, placebo-controlled studies. Nasrallah HA(1), Cucchiaro JB(2), Mao Y(2), Pikalov AA(2), Loebel AD(2). Author information: (1)1Department of Neurology and Psychiatry,St. Louis University,St. Louis,Missouri,USA. (2)2Sunovion Pharmaceuticals Inc.,Fort Lee,New Jersey,USA. OBJECTIVE: Depressive symptoms are common in schizophrenia and can worsen outcomes and increase suicide risk. Lurasidone is an atypical antipsychotic agent indicated for the treatment of schizophrenia and for the treatment of major depressive episodes associated with bipolar I disorder. This post hoc analysis evaluated the effect of lurasidone on depressive symptoms in patients with schizophrenia. METHODS: Patient-level data were pooled from 4 similarly designed, double-blind, placebo-controlled, 6-week registration studies of lurasidone (40-160 mg/d) in adult patients with an acute exacerbation of schizophrenia. Changes in depressive symptoms, measured by the Montgomery-Åsberg Depression Rating Scale (MADRS), were analyzed for the overall sample and for subgroups of patients stratified by baseline MADRS scores. RESULTS: MADRS assessments at baseline and endpoint (day 42 or last observation carried forward [LOCF]) were available for 1330 patients. Patients receiving lurasidone experienced significantly greater decreases in MADRS score (-2.8, least-squares [LS] mean change, LOCF) compared with patients receiving placebo (-1.4, P < .001, effect size 0.24). Analysis of change in MADRS score (LOCF) by baseline symptom severity (MADRS score of ≥12, ≥14, ≥16, ≥18) showed significantly greater improvement for lurasidone-treated patients across all severity groups; effect sizes ranged from 0.25 to 0.34. Among patients with a baseline MADRS score of ≥12, depressive symptom remission (defined as MADRS score <10 at LOCF endpoint) was attained by 45.0% of lurasidone-treated patients and 36.3% of patients receiving placebo (P < .05). CONCLUSIONS: In a pooled analysis of short-term, placebo-controlled studies, lurasidone significantly improved depressive symptoms in patients with schizophrenia. DOI: 10.1017/S1092852914000285 PMCID: PMC4411643 PMID: 24955752 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/21446639
1. J Psychosoc Nurs Ment Health Serv. 2011 Apr;49(4):13-5. doi: 10.3928/02793695-20110311-99. Epub 2011 Mar 30. Update on newer antipsychotic drugs. Howland RH(1). Author information: (1)University of Pittsburgh School of Medicine, Western Psychiatric Institute and Clinic, Pittsburgh, PA 15213, USA. [email protected] This article briefly reviews the novel atypical second-generation antipsychotic drugs iloperidone (Fanapt®), asenapine (Saphris®), and lurasidone (Latuda®), all of which have been approved by the U.S. Food and Drug Administration since 2009. Each is indicated for the treatment of schizophrenia, and asenapine has an additional indication for bipolar disorder. Very little information is available on their use in other disorders, pediatric and geriatric patients, and during pregnancy and breastfeeding. Their overall efficacy is no different than other antipsychotic drugs, but they do have different side effect profiles. Because of their unique pharmacologies and different tolerability profiles, they may be a more effective alternative for patients who do not respond to or cannot tolerate other antipsychotic drugs. Copyright 2011, SLACK Incorporated. DOI: 10.3928/02793695-20110311-99 PMID: 21446639 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/22364325
1. Expert Rev Neurother. 2012 Mar;12(3):265-73. doi: 10.1586/ern.12.7. Lurasidone for schizophrenia: what's different? Kantrowitz JT(1), Citrome L. Author information: (1)Nathan S Kline Institute for Psychiatric Research, Orangeburg, NY, USA. [email protected] Lurasidone is one of several antipsychotics approved in the recent past by the US FDA for the treatment of schizophrenia. Several Phase II and III studies have established that lurasidone is more efficacious than placebo. There are no available adequately powered head-to-head comparisons of efficacy of lurasidone with other antipsychotics. However, in contrast to some other antipsychotics, lurasidone is associated with minimal weight gain and no clinically meaningful alterations in glucose, lipids, or the ECG QT interval. As per the product label, the recommended starting dose is 40 mg/day and the maximum recommended dose is 80 mg/day. Higher doses do not appear to be more efficacious, and may be associated with increases in adverse effects, such as somnolence and akathisia; however, this tolerability issue was not observed in one recently conducted 6-week study when lurasidone was administered at a dose of 160 mg/day. It is recommended that lurasidone be administered once daily with at least 350 calories of food. Additional studies are desirable to directly compare and contrast lurasidone with other antipsychotic agents. DOI: 10.1586/ern.12.7 PMID: 22364325 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/29962579
1. Indian J Psychol Med. 2018 Mar-Apr;40(2):191-192. doi: 10.4103/IJPSYM.IJPSYM_374_17. Lurasidone Induced Thrombocytopenia: Is it a Signal of Drug Induced Myelosuppression? Rafi M(1), Goyal C(1), Reddy P(1), Reddy S(2). Author information: (1)Department of Pharmacology, Sri Aurobindo Medical College and PGI, Indore, Madhya Pradesh, India. (2)Department of Psychiatry, Sri Aurobindo Medical College and PGI, Indore, Madhya Pradesh, India. The U.S. Food and Drug Administration (FDA) has approved a supplemental new drug application Lurasidone (Latuda, Sunovion Pharmaceuticals), an atypical antipsychotic, for the treatment of schizophrenia in adolescents 13-17 years of age. Lurasidone was previously indicated in the U.S. for the treatment of adults with schizophrenia and major depressive episodes with bipolar I disorder as monotherapy. We present a case of a 29-year-old male patient who was hospitalized with thrombocytopenia (WHO grade-3 toxicity) (unlabeled) along with extrapyramidal disorder, gastritis, and hyperprolactinemia within 2-3 months of initiation of tablet lurasidone 80 mg/day (Lurasid, Intas Pharmaceuticals) in bipolar depression. Dechallenge was found to be positive in three reactions except hyperprolactinemia (outcome unknown) during hospital stay. The terms anemia and leukopenia are well labeled/listed with the drug literatures of lurasidone. Thus, this case presents a strong probability of lurasidone to cause myelosuppression/bone marrow depression. DOI: 10.4103/IJPSYM.IJPSYM_374_17 PMCID: PMC6009006 PMID: 29962579 Conflict of interest statement: There are no conflicts of interest.
http://www.ncbi.nlm.nih.gov/pubmed/32124704
1. CNS Neurol Disord Drug Targets. 2020;19(2):109-114. doi: 10.2174/1871527319666200303120147. Risk Analysis of Lurasidone in Patients with Schizophrenia and Bipolar Depression. Modugula H(1), Kumar A(1). Author information: (1)Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow (UP) -226002, India. Lurasidone was approved by the United States Food and Drug Administration (FDA) for the treatment of schizophrenia, as well as for the treatment of bipolar depression. However, emerging reports have indicated various adverse drug reactions with the use of lurasidone. Thus, in this article, we have analyzed the risk profile of lurasidone in the established therapeutic indication. A total of 419 studies were published from October 2010-July 2019 regarding lurasidone. After the inclusion and exclusion criteria, 17 studies were selected for the analysis of risk. The adverse drug reactions (ADRs) of these studies were categorized as per the innovator summary of product characteristics (SmPC). Finally, the unlisted ADRs were analyzed by using the Naranjo probability algorithm. Telogen effluvium, thrombocytopenia, restless leg syndrome and hypersexuality were found with the use of lurasidone and fall under the unlisted category. The causality assessment has shown a probable correlation of lurasidone with hypersexuality, restless leg syndrome, thrombocytopenia and possible relation with telogen effluvium. In conclusion, lurasidone is a novel and efficacious pharmacological treatment for bipolar depression and schizophrenia. However, more data regarding the safety of this drug in a large population is needed. Copyright© Bentham Science Publishers; For any queries, please email at [email protected]. DOI: 10.2174/1871527319666200303120147 PMID: 32124704 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/26316760
1. Neuropsychiatr Dis Treat. 2015 Aug 19;11:2143-52. doi: 10.2147/NDT.S50961. eCollection 2015. Lurasidone for the treatment of bipolar depression: an evidence-based review. Franklin R(1), Zorowitz S(1), Corse AK(1), Widge AS(2), Deckersbach T(1). Author information: (1)Division of Neurotherapeutics, Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA. (2)Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA. Bipolar disorder (BD) is a debilitating and difficult-to-treat psychiatric disease that presents a serious burden to patients' lives as well as health care systems around the world. The essential diagnostic criterion for BD is episodes of mania or hypomania; however, the patients report that the majority of their time is spent in a depressive phase. Current treatment options for this component of BD have yet to achieve satisfactory remission rates. Lurasidone is a drug in the benzisothiazole class approved by the US Food and Drug Administration in June 2013 for the acute treatment of bipolar depression. Its pharmacological profile features high-affinity antagonism at D2, 5-HT2A, and 5-HT7 receptors; moderate-affinity antagonism at α2C-adrenergic receptors; low- to very low-affinity antagonism at α1A-adrenergic, α2A-adrenergic, H1, M1, and 5-HT2C receptors; and high-affinity partial agonism at 5-HT1A. Preliminary findings from two recent double-blinded clinical trials suggest that lurasidone is efficacious in treating bipolar I depression, with clinical effects manifesting as early as the first 2-3 weeks of treatment (as measured by the Montgomery-Åsberg Depression Rating Scale and Clinical Global Impressions Scale for use in bipolar illness). Its therapeutic benefit appears to be comparable to the current US Food and Drug Administration-indicated treatments: quetiapine and olanzapine-fluoxetine, according to a measure of effect size known as number needed to treat. These studies reported relatively limited extrapyramidal and metabolic side effects as a result of treatment with lurasidone, with the most common side effect being nausea. Safety data drawn from these studies, as well as a more extensive body of schizophrenia research, indicate that in comparison with other atypical antipsychotics, treatment with lurasidone is less likely to result in metabolic side effects such as weight gain or disturbances of serum glucose or lipid levels. Lurasidone holds clinical potential as a novel, efficacious pharmacological treatment for bipolar depression. However, current data on its use for the treatment of BD are limited, and more extensive research, both longer in duration as well as independently conducted, is needed. DOI: 10.2147/NDT.S50961 PMCID: PMC4547662 PMID: 26316760
http://www.ncbi.nlm.nih.gov/pubmed/22146224
1. Drugs Today (Barc). 2011 Nov;47(11):807-16. doi: 10.1358/dot.2011.47.11.1708832. Lurasidone: a new treatment option for schizophrenia. Owen RT(1). Author information: (1)Crewe, Cheshire, UK. [email protected] Lurasidone is a novel benzoisothiazol antipsychotic that has recently been approved for the treatment of schizophrenia in the U.S. Like many other second-generation antipsychotics, it has a high affinity for dopamine D(2) and serotonin 5-HT(2A) receptors as well as a high affinity for 5-HT(7) receptors. It has negligible affinity for α(1)-adrenoceptors, histamine H(1) receptors or muscarinic acetylcholine M(1) receptors. It has demonstrated efficacy in short-term trials versus placebo, two of which included an active comparator (olanzapine, quetiapine) assay arm. A short-term, head-to-head trial of lurasidone versus ziprasidone in chronic stable schizophrenia was also conducted. A long-term, 12-month risperidone-controlled study and open-label studies primarily investigated the safety and tolerability of lurasidone. Limited evidence of procognitive and antidepressant effects was seen although these need further corroboration. The incidence of extrapyramidal symptoms (excluding akathisia/restlessness) was greater with lurasidone (14.7%) than placebo (5.1%). Akathisia and somnolence were dose-related adverse events. Lurasidone appears to have relatively little effect on weight, plasma glucose or lipids to date. No evidence of QTc prolongation was seen and orthostatic hypotension was uncommon. Raised prolactin levels in short-term studies were dose-dependent, greater in females and occurred overall in 3.7 and 0.7% of lurasidone and placebo recipients, respectively. Copyright 2011 Prous Science, S.A.U. or its licensors. All rights reserved. DOI: 10.1358/dot.2011.47.11.1708832 PMID: 22146224 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/25852975
1. Innov Clin Neurosci. 2015 Jan-Feb;12(1-2):21-3. Lurasidone: a new treatment option for bipolar depression-a review. Bawa R(1), Scarff JR(1). Author information: (1)Dr. Bawa is with Lehman College, the City University of New York, Bronx, New York; and Dr. Scarff is with Veterans Affairs Outpatient Clinic, Spartanburg, South Carolina. Depressive episodes in bipolar disorder contribute to significant morbidity and mortality. Until recently, only quetiapine and an olanzapine-fluoxetine combination were approved to treat bipolar depression. Recently, lurasidone was approved to treat bipolar depression either as monotherapy or adjunctively with lithium or valproate. Lurasidone was well- tolerated, and commonly observed adverse reactions (incidence ≥5% and at least twice the rate for placebo) were akathisia, extrapyramidal symptoms, and somnolence. There were no significant metabolic or electrocardiogram abnormalities. It is taken with food to ensure maximal absorption, and dose should be adjusted in patients who receive moderate CYP450 inhibitors or inducers and in patients with renal disease. PMCID: PMC4382136 PMID: 25852975
http://www.ncbi.nlm.nih.gov/pubmed/34304240
1. Am J Case Rep. 2021 Jul 25;22:e933003. doi: 10.12659/AJCR.933003. BNT162b2 mRNA Vaccine Interference with Co-Administration of Tdap Vaccine. Chilimuri S(1), Mantri N(1), Shrestha E(1), Sun H(1), Gongati S(1), Zahid M(1), Kelly P(2). Author information: (1)Department of Medicine, Bronx Care Health System, Affiliated with Icahn School of Medicine at Mount Sinai, Bronx, NY, USA. (2)BronxCare Center for Travel Medicine, Bronx Care Health System, Affiliated with Icahn School of Medicine at Mount Sinai, Bronx, NY, USA. BACKGROUND It is unknown if the efficacy of the coronavirus disease-19 (COVID-19) vaccine is affected by the co-administration of other vaccines. The Centers for Disease Control and Prevention (CDC) has shifted their recommendations recently, allowing for the co-administration of the currently available COVID-19 vaccines with other vaccines. This is based on the experience with non-COVID-19 vaccines, where the immunogenicity and adverse event profiles were generally similar when vaccines are administered simultaneously or alone. CASE REPORT We present a case of a 29-year-old Asian woman who received the first dose of BNT162b2 mRNA vaccine and the tetanus toxoid, reduced diphtheria toxoid, and acellular pertussis (Tdap) vaccine at around the same time. BNT162b2 mRNA vaccine and Tdap vaccine were administered into the deltoid region of the left arm and right arm, respectively. We then monitored for immunogenicity. We observed a delay in the development of SARS-CoV-2 Spike (S1) protein antibodies at around 8 weeks after the second dose. CONCLUSIONS Unless warranted, it is important to adhere to current CDC recommendations with regards to the co-administration of vaccines. Although the administration of Tdap with COVID-19 vaccine in our case caused delay in immunogenicity, it did not negate the ability of the BNT162B2 mRNA vaccine to elicit an adequate immune response. The reason for delay in immune response with co-administration of COVID-19 vaccines with other vaccines is unknown and further studies are needed. DOI: 10.12659/AJCR.933003 PMCID: PMC8317582 PMID: 34304240 [Indexed for MEDLINE] Conflict of interest statement: Conflict of interest: None declared Conflict of Interest None declared.
http://www.ncbi.nlm.nih.gov/pubmed/21177242
1. Clin Schizophr Relat Psychoses. 2011 Jan;4(4):251-7. Lurasidone for schizophrenia: a brief review of a new second-generation antipsychotic. Citrome L(1). Author information: (1)Department of Psychiatry, New York University School of Medicine, New York, NY, USA. [email protected] Lurasidone is a second-generation antipsychotic newly approved by the U.S. Food and Drug Administration for the treatment of schizophrenia. Similar to most other second-generation antipsychotics, lurasidone is a full antagonist at dopamine D2 and serotonin 5HT2A receptors. Efficacy within the dose range of 40-120 mg/d was established in four 6-week, randomized, controlled trials. The recommended starting dose is 40 mg/d and the maximum recommended dose is 80 mg/d. Doses above 80 mg/d do not appear to confer added benefit and may be associated with a dose-related increase in certain adverse reactions such as somnolence and akathisia. Lurasidone is administered once daily with at least 350 calories of food in order to optimize bioavailability. Lurasidone is primarily metabolized in the liver through the CYP3A4 enzyme system, and coadministration with drugs that are strong inhibitors of CYP3A4 (such as ketoconazole) or strong inducers (such as rifampin) are contraindicated. Lurasidone is associated with minimal weight gain and no clinically meaningful alterations in glucose, lipids, or the ECG QT interval. PMID: 21177242 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/34534629
1. Int J Pharm. 2021 Oct 25;608:121098. doi: 10.1016/j.ijpharm.2021.121098. Epub 2021 Sep 14. Harnessing the potential of nanostructured formulations to mimic the food effect of lurasidone. Meola TR(1), Joyce P(1), Wignall A(1), Bremmell KE(1), Prestidge CA(2). Author information: (1)UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia; ARC Centre for Excellence in Bio-Nano Science and Technology, Adelaide, South Australia 5000, Australia. (2)UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia; ARC Centre for Excellence in Bio-Nano Science and Technology, Adelaide, South Australia 5000, Australia. Electronic address: [email protected]. Lurasidone is an important antipsychotic drug indicated for the treatment of schizophrenia and bipolar disorder, with an oral bioavailability of 9-19% owing to its poor aqueous solubility. Additionally, lurasidone exhibits a 2-fold positive food effect, such that patients must administer their medication with a meal, leading to significant non-compliance. The aim of this research was to evaluate the in vitro and in vivo performance of lurasidone when engineered as nanostructured systems. Specifically, a nanosuspension, nano-emulsion and silica-lipid hybrid (SLH) microparticles were formulated and the influence of composition and nanostructure on the mechanism of solubilisation was compared. Formulations were shown to enhance fasted state solubilisation levels in vitro by up to 5.9-fold, compared to pure drug. Fed- and fasted-state solubilisation profiles revealed that in contrast to the nanosuspension and nano-emulsion, lurasidone SLH mitigated the positive pharmaceutical effect of lurasidone. In vivo pharmacokinetic evaluations revealed that the nanosuspension, nano-emulsion and SLH enhanced the bioavailability of lurasidone by 3-fold, 2.4-fold and 8.8-fold, respectively, compared to pure drug after oral administration. For lurasidone, the combination of lipid-based nanostructure and porous silica nanostructure (SLH) led to optimal fasted state bioavailability which can ultimately result in enhanced treatment efficacy, easier dosing regimens and improved patient outcomes. Crown Copyright © 2021. Published by Elsevier B.V. All rights reserved. DOI: 10.1016/j.ijpharm.2021.121098 PMID: 34534629 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/26755968
1. BJPsych Bull. 2015 Oct;39(5):237-41. doi: 10.1192/pb.bp.114.048793. Lurasidone: a novel antipsychotic agent for the treatment of schizophrenia and bipolar depression. Loebel A(1), Citrome L(2). Author information: (1)Sunovion Pharmaceuticals, Fort Lee, New Jersey, USA. (2)New York Medical College, Valhalla, New York, USA. Lurasidone is a novel antipsychotic agent approved for the treatment of schizophrenia in a number of countries including the UK, and is also approved in the USA and Canada for the treatment of major depressive episodes associated with bipolar I disorder as either a monotherapy or adjunctive therapy with lithium or valproate. In addition to full antagonist activity at dopamine D2 (K i(D2) = 1 nM) and serotonin 5-HT2A (K i(5-HT2A) = 0.5 nM) receptors, the pharmacodynamic profile of lurasidone is notable for its high affinity for serotonin 5-HT7 receptors (K i(5-HT7) = 0.5 nM) and its partial agonist activity at 5-HT1A receptors (K i(5-HT1A) = 6.4 nM). Long-term treatment of schizophrenia with lurasidone has been shown to reduce the risk of relapse. Lurasidone appears associated with minimal effects on body weight and low risk for clinically meaningful alterations in glucose, lipids or electrocardiogram parameters. DOI: 10.1192/pb.bp.114.048793 PMCID: PMC4706192 PMID: 26755968 Conflict of interest statement: Declaration of interest A.L. is a full-time employee of Sunovion Pharmaceuticals. In the past 12 months, L.C. was a consultant for, has received honoraria from, or has conducted clinical research supported by: Actavis (Forest Laboratories), Alexza Pharmaceuticals, Alkermes, AstraZeneca, Bristol-Myers Squibb, Eli Lilly & Company, Forum Pharmaceuticals, Genentech, Janssen Pharmaceuticals, Jazz Pharmaceuticals, Lundbeck, Medivation, Merck Worldwide, Mylan Laboratories, Novartis Pharmaceuticals, Noven Pharmaceuticals, Otsuka Pharmaceutical Group, Pfizer, Reckitt Benckiser, Reviva Pharmaceuticals, Shire, Sunovion Pharmaceuticals, Takeda Pharmaceutical Company, and Teva Pharmaceutical Industries
http://www.ncbi.nlm.nih.gov/pubmed/35417663
1. Hum Vaccin Immunother. 2022 Nov 30;18(5):2049169. doi: 10.1080/21645515.2022.2049169. Epub 2022 Apr 13. Willingness toward COVID-19 vaccination, coadministration with other vaccines and receive a COVID-19 vaccine booster: a cross-sectional study on the guardians of children in China. Ma L(1)(2)(3), Yang J(1), Zhang T(1)(2), Han X(1), Huang Q(1), Yang Y(4), Feng L(1)(2), Yang W(1)(2), Wang C(5). Author information: (1)School of Population Medicine and Public Health, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China. (2)Institute of pharmaceutical and medical devices supervision, National Medical Products Administration-Chinese Academy of Medical Sciences, Beijing, China. (3)Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guilin Medical University, Guilin, China. (4)Division of Infectious Diseases, Chinese Center for Disease Control and Prevention, Beijing, China. (5)Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China. This study aimed to investigate the changes in the willingness of guardians to administer the COVID-19 vaccine to their children, allow the coadministration of other vaccines, and administer the COVID-19 vaccine booster dose. This was a follow-up study conducted 6 months after a similar previous study. The self-administered questionnaire was distributed through the "Xiao Dou Miao" app and 9424 guardians with access to this app participated in the survey that was conducted from September 15 to October 8, 2021. Of all the participating guardians, 86.68% were willing to vaccinate their children with the COVID-19 vaccine, which was approximately 16% more than those in our previous study. Guardians aged ≥40 years, healthcare workers, and those with children aged ≥3 years were more willing to vaccinate their children. Approximately 77% of the guardians were willing toward the coadministration of COVID-19 and influenza vaccines. Approximately 64% of the guardians were willing toward the coadministration of other nonimmunization program vaccines with the COVID-19 vaccine for their children. The primary reasons for reluctance toward the coadministration of vaccines were concerns about vaccine safety and effectiveness. If necessary, 92% of the guardians were willing to receive a COVID-19 vaccine booster and 82% were willing to vaccinate their children with a COVID-19 vaccine booster. We hope that this research will facilitate the formulation of successful strategies for the implementation of COVID-19 vaccinations, covaccinations, and COVID-19 booster doses, particularly for children aged <6 years. DOI: 10.1080/21645515.2022.2049169 PMCID: PMC9196675 PMID: 35417663 [Indexed for MEDLINE] Conflict of interest statement: No potential conflict of interest was reported by the author(s).
http://www.ncbi.nlm.nih.gov/pubmed/34774197
1. Lancet. 2021 Dec 18;398(10318):2277-2287. doi: 10.1016/S0140-6736(21)02329-1. Epub 2021 Nov 11. Safety and immunogenicity of concomitant administration of COVID-19 vaccines (ChAdOx1 or BNT162b2) with seasonal influenza vaccines in adults in the UK (ComFluCOV): a multicentre, randomised, controlled, phase 4 trial. Lazarus R(1), Baos S(2), Cappel-Porter H(2), Carson-Stevens A(3), Clout M(2), Culliford L(2), Emmett SR(4), Garstang J(5), Gbadamoshi L(4), Hallis B(6), Harris RA(2), Hutton D(2), Jacobsen N(7), Joyce K(2), Kaminski R(8), Libri V(9), Middleditch A(10), McCullagh L(10), Moran E(11), Phillipson A(12), Price E(13), Ryan J(14), Thirard R(2), Todd R(2), Snape MD(15), Tucker D(16), Williams RL(11), Nguyen-Van-Tam JS(17), Finn A(18), Rogers CA(2); ComfluCOV Trial Group. Collaborators: Adams K, Alaee S, Aley PK, Allum E, Anthony S, Ashton K, Awal T, Barnett L, Barratt A, Barron C, Baum H, Beard C, Bennett L, Bird S, Bishop S, Bisset J, Bodalia P, Bowles J, Bowyer C, Bradburn K, Bray JJH, Bressington C, Brimfield M, Broad L, Brown P, Brydon-Hill R, Burge S, Carmichael D, Chohan G, Clark T, Close A, Coleman T, Cowley C, Cranfield C, Cross E, D'Agostino A, D'Arcangelo S, David Otter A, Davies K, Davies C, Davies R, Davies L, Driver K, Eglinton C, Ekblad C, Eldridge E, Evans T, Evans M, Evans I, F Mujadidi Y, Farrow A, Faulkner B, Feltham S, Figueirido S, Ford J, Foxwell D, Frayling S, Gardiner S, Gooch KE, Goodwin J, Halliday A, Hamal S, Harrhy S, Harris A, Haxton L, Haynes M, Hazell M, Hembrough T, Hewson J, Hicks B, Higgins T, Hill M, Hills A, Hilton Z, Hitchings B, Hua C, Iftikhar H, Iqbal A, Jones L, Jones N, Kellett Wright J, Kidd S, Kirby A, Knibbs L, Lamb J, Langton H, Leach R, Lewis-Clarke P, Lloyd A, Maclean K, Manning N, Marriott A, McFadzean IJ, McLaughlin M, McQueen A, Mills B, Naik G, Nicholls L, Norman C, Northcott K, Nyland K, Oliver C, Oliver E, Oliver J, Owen D, Paterson J, Pearce L, Pegler S, Price Z, Pynsent WB, Ramos L, Rampling T, Rea D, Regan K, Riaz T, Ricamara M, Rice D, Rich M, Roots M, Ryan KA, Sakagami Y, Salem A, Salter-Hewitt J, Samuels M, Santoloce S, Seaman S, Seneviratne M, Shankland S, Silva L, Smart K, Smith RM, Smith J, Stringer R, Talbot H, Tarling T, Taylor SL, Thomas A, Tilzey G, Townley G, Trembath L, Turkentine K, Turner K, Tyler J, Ugoji J, Wale K, Walker-Smith T, Walton M, Warnes B, Whittley S, Williams J, Williams G, Williamson K, Yim YTN, Youlden N. Author information: (1)University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK. Electronic address: [email protected]. (2)Bristol Trials Centre, University of Bristol, Bristol, UK. (3)Division of Population Medicine, School of Medicine, Cardiff University, Cardiff, UK. (4)Royal United Hospitals NHS Foundation Trust, Bath, UK. (5)Knowle House Surgery, Plymouth, UK. (6)Porton Down, Public Health England, Salisbury, UK. (7)Newquay Health Centre, North Coast Medical, Newquay, UK. (8)Gloucestershire Hospitals NHS Foundation Trust, Gloucester, UK. (9)University College Hospitals NHS Foundation Trust, London, UK. (10)University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK. (11)North Bristol NHS Trust, Bristol, UK. (12)Rotherham Doncaster and South Humber NHS Foundation Trust, Doncaster, UK. (13)Great Western Hospitals NHS Foundation Trust, Swindon, UK. (14)The Alverton Practice, Atlantic Medical, Penzance, UK. (15)Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; Oxford NIHR-Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK. (16)Royal Cornwall Hospitals NHS Trust, Truro, UK. (17)Division of Epidemiology and Public Health, University of Nottingham School of Medicine, Nottingham, UK. (18)Bristol Vaccine Centre, Bristol Medical School, Bristol Population Health Sciences and School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK. BACKGROUND: Concomitant administration of COVID-19 and influenza vaccines could reduce burden on health-care systems. We aimed to assess the safety of concomitant administration of ChAdOx1 or BNT162b2 plus an age-appropriate influenza vaccine. METHODS: In this multicentre, randomised, controlled, phase 4 trial, adults in receipt of a single dose of ChAdOx1 or BNT162b2 were enrolled at 12 UK sites and randomly assigned (1:1) to receive concomitant administration of either an age-appropriate influenza vaccine or placebo alongside their second dose of COVID-19 vaccine. 3 weeks later the group who received placebo received the influenza vaccine, and vice versa. Participants were followed up for 6 weeks. The influenza vaccines were three seasonal, inactivated vaccines (trivalent, MF59C adjuvanted or a cellular or recombinant quadrivalent vaccine). Participants and investigators were masked to the allocation. The primary endpoint was one or more participant-reported solicited systemic reactions in the 7 days after first trial vaccination(s), with a difference of less than 25% considered non-inferior. Analyses were done on an intention-to-treat basis. Local and unsolicited systemic reactions and humoral responses were also assessed. The trial is registered with ISRCTN, ISRCTN14391248. FINDINGS: Between April 1 and June 26, 2021, 679 participants were recruited to one of six cohorts, as follows: 129 ChAdOx1 plus cellular quadrivalent influenza vaccine, 139 BNT162b2 plus cellular quadrivalent influenza vaccine, 146 ChAdOx1 plus MF59C adjuvanted, trivalent influenza vaccine, 79 BNT162b2 plus MF59C adjuvanted, trivalent influenza vaccine, 128 ChAdOx1 plus recombinant quadrivalent influenza vaccine, and 58 BNT162b2 plus recombinant quadrivalent influenza vaccine. 340 participants were assigned to concomitant administration of influenza and a second dose of COVID-19 vaccine at day 0 followed by placebo at day 21, and 339 participants were randomly assigned to concomitant administration of placebo and a second dose of COVID-19 vaccine at day 0 followed by influenza vaccine at day 21. Non-inferiority was indicated in four cohorts, as follows: ChAdOx1 plus cellular quadrivalent influenza vaccine (risk difference for influenza vaccine minus placebos -1·29%, 95% CI -14·7 to 12·1), BNT162b2 plus cellular quadrivalent influenza vaccine (6·17%, -6·27 to 18·6), BNT162b2 plus MF59C adjuvanted, trivalent influenza vaccine (-12·9%, -34·2 to 8·37), and ChAdOx1 plus recombinant quadrivalent influenza vaccine (2·53%, -13·3 to 18·3). In the other two cohorts, the upper limit of the 95% CI exceeded the 0·25 non-inferiority margin (ChAdOx1 plus MF59C adjuvanted, trivalent influenza vaccine 10·3%, -5·44 to 26·0; BNT162b2 plus recombinant quadrivalent influenza vaccine 6·75%, -11·8 to 25·3). Most systemic reactions to vaccination were mild or moderate. Rates of local and unsolicited systemic reactions were similar between the randomly assigned groups. One serious adverse event, hospitalisation with severe headache, was considered related to the trial intervention. Immune responses were not adversely affected. INTERPRETATION: Concomitant vaccination with ChAdOx1 or BNT162b2 plus an age-appropriate influenza vaccine raises no safety concerns and preserves antibody responses to both vaccines. Concomitant vaccination with both COVID-19 and influenza vaccines over the next immunisation season should reduce the burden on health-care services for vaccine delivery, allowing for timely vaccine administration and protection from COVID-19 and influenza for those in need. FUNDING: National Institute for Health Research Policy Research Programme. Copyright © 2021 The Author(s). Published by Elsevier Ltd. This is an Open Access article under the CC BY-NC-ND 4.0 license. Published by Elsevier Ltd.. All rights reserved. DOI: 10.1016/S0140-6736(21)02329-1 PMCID: PMC8585490 PMID: 34774197 [Indexed for MEDLINE] Conflict of interest statement: Declaration of interests RL reports grants from the National Institute for Health Research (NIHR) during the conduct of the trial, and grants from Elizabeth Blackwell Institute, AstraZeneca, Janssen, and Valneva outside the submitted work. CAR reports grants from NIHR, during the conduct of the trial. JSN-V-T reports that he is seconded to the Department of Health and Social Care, England. AF reports grants from Pfizer during the conduct of the trial, and grants from Elizabeth Blackwell Institute, Sanofi Pasteur, VBI Vaccines, Pfizer, Janssen, GSK, MedImmune, Novavax, and Valneva outside the submitted work. AM reports grants from NIHR during the conduct of the trial, and grants from AstraZeneca, Janssen, and Valneva outside the submitted work. MDS acts on behalf of the University of Oxford as an investigator on studies funded or sponsored by vaccine manufacturers, including AstraZeneca, GSK, Pfizer, Novavax, Pfizer, Janssen, Medimmune, and MCM. All other authors declare no competing interests.
http://www.ncbi.nlm.nih.gov/pubmed/34919292
1. Acta Paediatr. 2022 Mar;111(3):478-489. doi: 10.1111/apa.16222. Epub 2022 Jan 31. Clinical benefits of music-based interventions on preterm infants' health: A systematic review of randomised trials. Costa VS(1), Bündchen DC(1), Sousa H(2), Pires LB(3), Felipetti FA(1). Author information: (1)Department of Health Sciences, Federal University of Santa Catarina (UFSC), Araranguá, Brazil. (2)Department of Education and Psychology, Center for Health Technology and Services Research (CINTESIS.UA), University of Aveiro, Aveiro, Portugal. (3)Espaço Envolver, Florianópolis, Brazil. AIM: This systematic review aimed to differentiate and isolate the results of different music-based interventions used with preterm infants in the neonatal intensive care unit and explore their clinical benefits. METHODS: The last search was performed on 5 July 2021 on Web of Science, Scopus, EMBASE, PsycINFO, CINAHL, LILACS and CENTRAL. Only randomised clinical trials that explored the health benefits of music-based interventions were considered. RESULTS: A total of 39 studies were included. All music-based interventions were divided into music medicine and music therapy. The overall results suggested that music medicine interventions were associated with a significant improvement in pain relief; in turn, improvements in cardiac and respiratory function, weight gain, eating behaviour, and quiet alert and sleep states were more consistent in studies that followed a music therapy approach with the presence of a music therapist. CONCLUSION: This review supports the beneficial effects of music-based interventions on the health of preterm infants in a neonatal intensive care unit; however, it also offers suggestions for future studies in order to increase the number of interventions with music therapists, since the results of music therapy approaches were more consistent for physiological and behavioural outcomes. © 2022 Foundation Acta Paediatrica. Published by John Wiley & Sons Ltd. DOI: 10.1111/apa.16222 PMID: 34919292 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/32499082
1. Therapie. 2020 Sep-Oct;75(5):397-406. doi: 10.1016/j.therap.2020.05.007. Epub 2020 May 18. Prolonged-release buprenorphine formulations: Perspectives for clinical practice. Chappuy M(1), Trojak B(2), Nubukpo P(3), Bachellier J(4), Bendimerad P(5), Brousse G(6), Rolland B(7). Author information: (1)Service universitaire d'addictologie de Lyon (SUAL), centre hospitalier Le Vinatier, 69500 Bron, France; Service d'addictologie, groupement hospitalier centre, Hospices Civils de Lyon, 69003 Lyon, France; Centre de soins, d'accompagnement et de prévention en addictologie, groupement hospitalier nord, Hospices Civils de Lyon, 69004 Lyon, France. Electronic address: [email protected]. (2)Service hospitalo-universitaire d'addictologie, CHU de Dijon, 21079 Dijon, France; INSERM U1093 cognition, action et plasticité sensorimotrice, UFR staps, université de Bourgogne Franche-Comté, 21078 Dijon, France. (3)Service universitaire d'addictologie, centre hospitalier Esquirol, 87000 Limoges, France; INSERM UMR 1094 neuroépidémiologie tropicale, université de Limoges, 87000 Limoges, France. (4)Service universitaire d'addictologie de Tours, CHU Bretonneau, 37000 Tours, France. (5)Service d'addictologie, groupe hospitalier de La Rochelle-Ré-Aunis, 17000 La Rochelle, France; Service de psychiatrie, groupe hospitalier de La Rochelle-Ré-Aunis, 17000 La Rochelle, France. (6)Service de psychiatrie B et d'addictologie, CHU de Clermont-Ferrand, 63000 Clermont-Ferrand, France; Équipe d'accueil 7280, unité de formation et de recherche de médecine, université Clermont Auvergne, Clermont-Ferrand, France. (7)Service universitaire d'addictologie de Lyon (SUAL), centre hospitalier Le Vinatier, 69500 Bron, France; Service d'addictologie, groupement hospitalier centre, Hospices Civils de Lyon, 69003 Lyon, France; Université de Lyon, UCBL1, INSERM, INSERM U1028, CNRS UMR 5292, CRNL, 69500 Bron, France. Buprenorphine and methadone are the two main opioid agonist treatments approved for opioid use disorder. Buprenorphine is a partial agonist of the mu opioid receptors, which has been merely available through sublingual form until now. In practice, the use of buprenorphine is smoother than that of methadone, and it induces reduced risks of overdose. However, sublingual buprenorphine also exposes to risks (e.g., withdrawal, misuse) and constraints (e.g., daily intake). Three new galenic formulations of prolonged-release buprenorphine (PRB) are being commercialized and should allow some improvements in patients' comfort and safety. This narrative review aims to describe the main technical features and efficacy and safety data of these PRBs, as well as patients' and professionals' expectancies and concerns, using data of the scientific literature and the regulatory texts. PRBs consist of one subcutaneous implant and two subcutaneous injection depots. Sixmo®/Probuphine® is a six-month-long implant which needs to be surgically placed and removed and is approved for subjects previously treated with a maximum daily dose of 8mg of sublingual buprenorphine, and can be used only for two successive periods of six months before the subject needs to be switched back to sublingual form. Sublocade® is a one-month-long depot formulation that is indicated in switch from sublingual buprenorphine, and which proposes only two dose schemes, i.e., 100 and 300mg monthly. Buvidal®/Brixadi® is a one-week- or one-month-long depot formulation with multiple dosages, which can be used in initiation or in switched from sublingual formulations. While opioid users report some concerns with a risk of coercive use of long-acting forms of buprenorphine, both users and professionals deem that these new specialties could be particularly appreciated in stabilized patients bothered with the daily intake of the treatments, or specific situations at risk of treatment dropout (e.g., following hospital discharge or prison release). Copyright © 2020 Société française de pharmacologie et de thérapeutique. Published by Elsevier Masson SAS. All rights reserved. DOI: 10.1016/j.therap.2020.05.007 PMID: 32499082 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/31532835
1. Acta Paediatr. 2020 Mar;109(3):511-517. doi: 10.1111/apa.15018. Epub 2019 Oct 18. No effect of a musical intervention on stress response to venepuncture in a neonatal population. Howard C(1), Powell AS(1), Pavlidis E(2), Pavel A(1)(2), Finn D(2), Allen A(3), Olavarria-Ramirez L(3), Clarke G(2)(3)(4), Livingstone V(2), Boylan GB(1)(2), Dempsey EM(1)(2). Author information: (1)Department of Paediatrics and Child Health, Neonatal Intensive Care Unit, Cork University Maternity Hospital, Cork, Ireland. (2)INFANT, Irish Centre for Fetal and Neonatal Translational Research, University College Cork, Cork, Ireland. (3)APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork, Ireland. (4)Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland. AIM: To investigate the effect of a musical intervention on neonatal stress response to venepuncture as measured by salivary cortisol levels and pain profile scores. METHODS: In a randomised control crossover trial, participants were randomised to both a control arm (sucrose) and intervention arm (sucrose and music) for routine venepuncture procedures. Salivary swabs were collected at baseline, 20 minutes post-venepuncture and 4 hours post-venepuncture. Pain levels were assessed using the Premature Infant Pain Profile (PIPP). A total of 16 preterm neonates participated in both arms to complete the study. RESULTS: Cortisol values were elevated at all timepoints in the intervention arm (baseline, 20 minutes, and 4 hours post-procedure) but not significantly so (P = .056, P = .3, and P = .575, respectively). Median change in cortisol values from baseline was +128.48 pg/mL (-47.66 to 517.02) at 20 minutes and +393.52 pg/mL (47.88-1221.34) at 4 hours post-procedure in the control arm compared to -69.564 pg/mL (-860.96 to 397.289) and +100.48 pg/mL (-560.46 to 842.99) at 20 minutes and 4 hours post-procedure in the intervention arm. There was no statistically significant difference observed between groups (P = .311 at 20 minutes, and P = .203 at 4 hours post-procedure). PIPP scores were not significantly different between study arms. CONCLUSION: Our findings did not support the additional benefit of music intervention on neonatal stress response to venepuncture in preterm infants. © 2019 Foundation Acta Paediatrica. Published by John Wiley & Sons Ltd. DOI: 10.1111/apa.15018 PMID: 31532835 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/25467598
1. Midwifery. 2015 Mar;31(3):365-72. doi: 10.1016/j.midw.2014.11.001. Epub 2014 Nov 11. Pain relief effect of breast feeding and music therapy during heel lance for healthy-term neonates in China: a randomized controlled trial. Zhu J(1), Hong-Gu H(2), Zhou X(3), Wei H(4), Gao Y(5), Ye B(6), Liu Z(7), Chan SW(8). Author information: (1)Department of Nursing, School of Medicine, Xiamen University, Xiamen, China. Electronic address: [email protected]. (2)Alice Lee Centre for Nursing Studies, Yong Loo Lin School of Medicine, National University of Singapore, Singapore. Electronic address: [email protected]. (3)Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, Xiamen, China. Electronic address: [email protected]. (4)BSc (Nursing) Programme, Department of Nursing, School of Medicine, Xiamen University, China. Electronic address: [email protected]. (5)BSc (Nursing) Programme, Department of Nursing, School of Medicine, Xiamen University, China. Electronic address: [email protected]. (6)Department of Nursing, School of Medicine, Xiamen University, Xiamen, China. Electronic address: [email protected]. (7)School of Medicine, Xiamen University, Xiangan Campus, Xiamen 361102, China. Electronic address: [email protected]. (8)School of Nursing and Midwifery, Faculty of Health and Medicine, The University of Newcastle, Australia. Electronic address: [email protected]. OBJECTIVES: to test the effectiveness of breast feeding (BF), music therapy (MT), and combined breast feeding and music therapy (BF+MT) on pain relief in healthy-term neonates during heel lance. DESIGN: randomised controlled trial. SETTING: in the postpartum unit of one university-affiliated hospital in China from August 2013 to February 2014. PARTICIPANTS: among 288 healthy-term neonates recruited, 250 completed the trial. All neonates were undergoing heel lancing for metabolic screening, were breast fed, and had not been fed for the previous 30 minutes. INTERVENTIONS: all participants were randomly assigned into four groups - BF, MT, BF+MT, and no intervention - with 72 neonates in each group. Neonates in the control group received routine care. Neonates in the other three intervention groups received corresponding interventions five minutes before the heel lancing and throughout the whole procedure. MEASUREMENTS: Neonatal Infant Pain Scale (NIPS), latency to first cry, and duration of first crying. FINDINGS: mean changes in NIPS scores from baseline over time was dependent on the interventions given. Neonates in the BF and combined BF+MT groups had significantly longer latency to first cry, shorter duration of first crying, and lower pain mean score during and one minute after heel lance, compared to the other two groups. No significant difference in pain response was found between BF groups with or without music therapy. The MT group did not achieve a significantly reduced pain response in all outcome measures. CONCLUSIONS: BF could significantly reduce pain response in healthy-term neonates during heel lance. MT did not enhance the effect of pain relief of BF. IMPLICATIONS FOR PRACTICE: healthy-term neonates should be breast fed to alleviate pain during heel lance. There is no need for the additional input of classical music on breast feeding in clinic to relieve procedural pain. Nurses should encourage breast feeding to relieve pain during heel lance. Copyright © 2014 Elsevier Ltd. All rights reserved. DOI: 10.1016/j.midw.2014.11.001 PMID: 25467598 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/35637252
1. Blood Cancer J. 2022 May 30;12(5):84. doi: 10.1038/s41408-022-00677-7. Clinical outcomes in patients with relapsed/refractory FLT3-mutated acute myeloid leukemia treated with gilteritinib who received prior midostaurin or sorafenib. Perl AE(1), Hosono N(2), Montesinos P(3), Podoltsev N(4), Martinelli G(5), Panoskaltsis N(6), Recher C(7), Smith CC(8), Levis MJ(9), Strickland S(10), Röllig C(11), Groß-Langenhoff M(12), Chou WC(13), Lee JH(14), Yokoyama H(15)(16), Hasabou N(17), Lu Q(17), Tiu RV(17), Altman JK(18). Author information: (1)Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA. [email protected]. (2)University of Fukui, Fukui, Japan. (3)Hospital Universitario y Politécnico La Fe, Valencia, Spain. (4)Yale School of Medicine, New Haven, CT, USA. (5)Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori" IRST S. r. l, Meldola, Italy. (6)Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA. (7)Cancer Research Center of Toulouse, Toulouse, France. (8)University of California-San Francisco, San Francisco, CA, USA. (9)The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA. (10)Vanderbilt Ingram Cancer Center, Nashville, TN, USA. (11)Universitätsklinikum Carl Gustav Carus, Dresden, Germany. (12)Astellas Pharma GmbH, Munich, Germany. (13)National Taiwan University Hospital, Taipei, Taiwan. (14)Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea. (15)Sendai Medical Center, National Hospital Organization, Sendai, Japan. (16)Tohoku University, National Hospital Organization, Sendai, Japan. (17)Astellas Pharma US, Northbrook, IL, USA. (18)Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA. The fms-like tyrosine kinase 3 (FLT3) inhibitor gilteritinib is indicated for relapsed or refractory (R/R) FLT3-mutated acute myeloid leukemia (AML), based on its observed superior response and survival outcomes compared with salvage chemotherapy (SC). Frontline use of FLT3 tyrosine kinase inhibitors (TKIs) midostaurin and sorafenib may contribute to cross-resistance to single-agent gilteritinib in the R/R AML setting but has not been well characterized. To clarify the potential clinical impact of prior TKI use, we retrospectively compared clinical outcomes in patients with R/R FLT3-mutated AML in the CHRYSALIS and ADMIRAL trials who received prior midostaurin or sorafenib against those without prior FLT3 TKI exposure. Similarly high rates of composite complete remission (CRc) were observed in patients who received a FLT3 TKI before gilteritinib (CHRYSALIS, 42%; ADMIRAL, 52%) and those without prior FLT3 TKI therapy (CHRYSALIS, 43%; ADMIRAL, 55%). Among patients who received a prior FLT3 TKI in ADMIRAL, a higher CRc rate (52%) and trend toward longer median overall survival was observed in the gilteritinib arm versus the SC arm (CRc = 20%; overall survival, 5.1 months; HR = 0.602; 95% CI: 0.299, 1.210). Remission duration was shorter with prior FLT3 TKI exposure. These findings support gilteritinib for FLT3-mutated R/R AML after prior sorafenib or midostaurin. © 2022. The Author(s). DOI: 10.1038/s41408-022-00677-7 PMCID: PMC9151663 PMID: 35637252 [Indexed for MEDLINE] Conflict of interest statement: AE Perl reports grants, personal fees and non-financial support from Astellas, during the conduct of the study; grants, personal fees, and non-financial support from FujiFilm; grants, personal fees, and non-financial support from Daiichi Sankyo; grants and personal fees from Abbvie and Actinium Pharmaceuticals; personal fees from Agios, Loxo, LLS/Beat AML, and Forma; non-financial support from Arog; personal fees and non-financial support from New Link Genetics, Novartis, Takeda, and Jazz; and grants from Bayer and Biomed Valley Discoveries, outside the submitted work. N Hosono reports no relevant conflicts of interest to disclose. P Montesinos reports research support from Pfizer, Abbvie, and Daiichi Sankyo; consultancy from Celgene, Pfizer, and Abbvie; and speakers bureau from Astellas, Novartis, and Janssen. N Podoltsev reports consultancy fees from Pfizer, Celgene, Agios Pharmaceuticals, Blueprint Medicines, Incyte, Novartis, Bristol-Myers Squibb, CTI Biopharma, PharmaEssentia, Constellation Pharmaceuticals, Cogent BioSciences, and AbbVie; and institutional research funding from Pfizer, Celgene, CTI Biopharma, Boehringer Ingelheim, Astellas, Daiichi Sankyo, Sunesis Pharmaceuticals, Jazz Pharmaceuticals, Astex Pharmaceuticals, Genentech, AI Therapeutics, Samus Therapeutics, Arog Therapeutics, and Kartos Therapeutics. G Martinelli reports grant funding and consultancy fees from Amgen, Ariad, Incyte, Pfizer, Roche, Celgene, Janssen, AbbVie, and Novartis. N Panoskaltsis reports no relevant conflicts of interest to disclose. C Recher reports grant funding and personal fees from Celgene, Sunesis, Amgen, and Novartis and personal fees from Incyte, Jazz Pharmaceuticals, AbbVie, Astellas, Macrogenics, and Otsuka. C Smith reports research funding from Abbvie, Revolution Medicines, Celgene, FujiFilm; consulting fees from Astellas, Daichi Sanyko, and Genentech to attend an advisory board meeting; and reports being a stockolder at Ligacept, LLC. MJ Levis reports grants and personal fees from Astellas and FujiFilm and personal fees from Daiichi Sankyo, Amgen, and Menarini. S Strickland reports consulting or advisory fees from Abbvie, Astellas Pharma, Jazz Pharmaceuticals, Kite, a Gilead company, Novartis, and Pfizer and research funding at an institutional level from Abbvie, Astellas Pharma, Inc, Celator/Jazz, Celgene, Daiichi Sankyo, Karyopharm Therapeutics, Menarini, Novartis, and Sunesis Pharmaceuticals. C Röllig has received grants from AbbVie, Novartis, and Pfizer; consulting fees from AbbVie, Amgen, Bristol-Myers Squibb, Celgene, Daiichi Sankyo, Janssen, Jazz, Novartis, Pfizer, and Roche; and honoraria for speaker engagements from AbbVie, Amgen, Bristol-Myers Squibb, Celgene, Daiichi Sankyo, Janssen, Jazz, Novartis, Pfizer, and Roche. WC Chou reports no relevant conflicts of interest to disclose. H Lee has received honoraria for speaker engagements from AbbVie Korea and Astellas Korea, participated in and advisory board for AbbVie and Astellas, and is President of The Korean Society of Hematology. H Yokoyama reports having received honoraria from Astellas for speaking engagements. Q Lu, N Hasabou, M Groß-Langenhoff are employees of Astellas. R Tiu is a former employee of Astellas. JK Altman reports advisory or consulting fees from AbbVie, Amgen, Astellas, Daiichi Sankyo, Kura Oncology, Syros, and Theradex; institutional research funding for trials conducted by ALX Oncology, Amgen, Aptos, Astellas, Aprea, BioSight, Bristol-Myers Squibb, Boehringer Ingelheim, Celgene, FujiFilm, Immunogen, Kartos, Kura Oncology, and Loxo; reimbursement for travel from BioSight; and serves on a data monitoring committee for GlycoMimetics.
http://www.ncbi.nlm.nih.gov/pubmed/35997897
1. BioDrugs. 2022 Sep;36(5):549-571. doi: 10.1007/s40259-022-00549-3. Epub 2022 Aug 23. Therapeutic siRNA: State-of-the-Art and Future Perspectives. Friedrich M(1)(2), Aigner A(3). Author information: (1)Faculty of Leipzig, Institute of Clinical Immunology, Max-Bürger-Forschungszentrum (MBFZ), University of Leipzig, Leipzig, Germany. (2)Department of Vaccines and Infection Models, Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany. (3)Rudolf-Boehm Institute for Pharmacology and Toxicology, Clinical Pharmacology, University of Leipzig, Haertelstrasse 16-18, 04107, Leipzig, Germany. [email protected]. The highly specific induction of RNA interference-mediated gene knockdown, based on the direct application of small interfering RNAs (siRNAs), opens novel avenues towards innovative therapies. Two decades after the discovery of the RNA interference mechanism, the first siRNA drugs received approval for clinical use by the US Food and Drug Administration and the European Medicines Agency between 2018 and 2022. These are mainly based on an siRNA conjugation with a targeting moiety for liver hepatocytes, N-acetylgalactosamine, and cover the treatment of acute hepatic porphyria, transthyretin-mediated amyloidosis, hypercholesterolemia, and primary hyperoxaluria type 1. Still, the development of siRNA therapeutics faces several challenges and issues, including the definition of optimal siRNAs in terms of target, sequence, and chemical modifications, siRNA delivery to its intended site of action, and the absence of unspecific off-target effects. Further siRNA drugs are in clinical studies, based on different delivery systems and covering a wide range of different pathologies including metabolic diseases, hematology, infectious diseases, oncology, ocular diseases, and others. This article reviews the knowledge on siRNA design and chemical modification, as well as issues related to siRNA delivery that may be addressed using different delivery systems. Details on the mode of action and clinical status of the various siRNA therapeutics are provided, before giving an outlook on issues regarding the future of siRNA drugs and on their potential as one emerging standard modality in pharmacotherapy. Notably, this may also cover otherwise un-druggable diseases, the definition of non-coding RNAs as targets, and novel concepts of personalized and combination treatment regimens. © 2022. The Author(s). DOI: 10.1007/s40259-022-00549-3 PMCID: PMC9396607 PMID: 35997897 [Indexed for MEDLINE] Conflict of interest statement: The authors have no conflicts of interest to declare.
http://www.ncbi.nlm.nih.gov/pubmed/35867041
1. Nucleic Acid Ther. 2022 Dec;32(6):507-512. doi: 10.1089/nat.2022.0010. Epub 2022 Jul 22. Cross-Species Translation of Biophase Half-Life and Potency of GalNAc-Conjugated siRNAs. Boianelli A(1), Aoki Y(1), Ivanov M(2), Dahlén A(3), Gennemark P(1)(4). Author information: (1)Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden. (2)Quantitative Biology SE, Data Sciences and Quantitative Biology, Discovery Sciences, AstraZeneca, Gothenburg, Sweden. (3)Oligonucleotide Discovery, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden. (4)Department of Biomedical Engineering, Linköping University, Linköping, Sweden. Small interfering RNAs (siRNAs) with N-acetylgalactosamine (GalNAc) conjugation for improved liver uptake represent an emerging class of drugs to treat liver diseases. Understanding how pharmacokinetics and pharmacodynamics translate is pivotal for in vivo study design and human dose prediction. However, the literature is sparse on translational data for this modality, and pharmacokinetics in the liver is seldom measured. To overcome these difficulties, we collected time-course biomarker data for 11 GalNAc-siRNAs in various species and applied the kinetic-pharmacodynamic modeling approach to estimate the biophase (liver) half-life and the potency. Our analysis indicates that the biophase half-life is 0.6-3 weeks in mouse, 1-8 weeks in monkey, and 1.5-14 weeks in human. For individual siRNAs, the biophase half-life is 1-8 times longer in human than in mouse, and generally 1-3 times longer in human than in monkey. The analysis indicates that the siRNAs are more potent in human than in mouse and monkey. DOI: 10.1089/nat.2022.0010 PMCID: PMC9784597 PMID: 35867041 [Indexed for MEDLINE] Conflict of interest statement: Authors are employed by AstraZeneca AB.
http://www.ncbi.nlm.nih.gov/pubmed/35625776
1. Biomedicines. 2022 Apr 30;10(5):1038. doi: 10.3390/biomedicines10051038. Targeting TKI-Activated NFKB2-MIF/CXCLs-CXCR2 Signaling Pathways in FLT3 Mutated Acute Myeloid Leukemia Reduced Blast Viability. Cao H(1)(2), Tadros V(3), Hiramoto B(3), Leeper K(3), Hino C(1), Xiao J(3), Pham B(1), Kim DH(3), Reeves ME(1)(2), Chen CS(1)(2), Zhong JF(4), Zhang KK(5)(6), Xie L(5), Wasnik S(3), Baylink DJ(3), Xu Y(1)(2)(3). Author information: (1)Division of Hematology and Oncology, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA. (2)Loma Linda University Cancer Center, Loma Linda, CA 92354, USA. (3)Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA. (4)Department of Basic Sciences, Loma Linda University, Loma Linda, CA 92354, USA. (5)Department of Nutrition, Texas A&M University, College Station, TX 77030, USA. (6)Center for Epigenetics & Disease Prevention, Institute of Biosciences & Technology, College of Medicine, Texas A&M University, Houston, TX 77030, USA. Disease relapse is a common cause of treatment failure in FMS-like tyrosine kinase 3 (FLT3) mutated acute myeloid leukemia (AML). In this study, to identify therapeutic targets responsible for the survival and proliferation of leukemic cells (blasts) with FLT3 mutations after gilteritinib (GILT, a 2nd generation tyrosine kinase inhibitor (TKI)) treatment, we performed proteomic screening of cytokine release and in vitro/ex vivo studies to investigate their associated signaling pathways and transcriptional regulation. Here, we report that macrophage migration inhibition factor (MIF) was significantly increased in the supernatant of GILT-treated blasts when compared to untreated controls. Additionally, the GILT-treated blasts that survived were found to exhibit higher expressions of the CXCR2 gene and protein, a common receptor for MIF and pro-inflammatory cytokines. The supplementation of exogenous MIF to GILT-treated blasts revealed a group of CD44High+ cells that might be responsible for the relapse. Furthermore, we identified the highly activated non-classical NFKB2 pathway after GILT-treatment. The siRNA transient knockdown of NFKB2 significantly reduced the gene expressions of MIF, CXCR2, and CXCL5. Finally, treatments of AML patient samples ex vivo demonstrated that the combination of a pharmaceutical inhibitor of the NFKB family and GILT can effectively suppress primary blasts' secretion of tumor-promoting cytokines, such as CXCL1/5/8. In summary, we provide the first evidence that targeting treatment-activated compensatory pathways, such as the NFKB2-MIF/CXCLs-CXCR2 axis could be a novel therapeutic strategy to overcome TKI-resistance and effectively treat AML patients with FLT3 mutations. DOI: 10.3390/biomedicines10051038 PMCID: PMC9138861 PMID: 35625776 Conflict of interest statement: The authors declare that they have no competing interest.
http://www.ncbi.nlm.nih.gov/pubmed/34350585
1. Br J Haematol. 2022 Jan;196(2):316-328. doi: 10.1111/bjh.17746. Epub 2021 Aug 4. Updates on targeted therapies for acute myeloid leukaemia. Kayser S(1)(2), Levis MJ(3). Author information: (1)Medical Clinic and Policlinic I, Hematology and Cellular Therapy, University Hospital Leipzig, Leipzig, Germany. (2)NCT Trial Center, National Center of Tumor Diseases, German Cancer Research Center (DKFZ), Heidelberg, Germany. (3)Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA. In the past few years research in the underlying pathogenic mechanisms of acute myeloid leukaemia (AML) has led to remarkable advances in our understanding of the disease. Cytogenetic and molecular aberrations are the most important factors in determining response to chemotherapy as well as long-term outcome, but beyond prognostication are potential therapeutic targets. Our increased understanding of the pathogenesis of AML facilitated by next-generation sequencing has spurred the development of new compounds in the treatment of AML, particularly the creation of small molecules that target the disease on a molecular level. Many of the hopeful predictions outlined in our AML review of 2018 are now therapeutic realities: gemtuzumab ozogamicin, venetoclax, FLT3 inhibitors (midostaurin, gilteritinib), IDH inhibitors (ivosidenib, enasidenib), CPX-351, glasdegib, oral decitabine, and oral azacitidine. Others may soon be (quizartinib, APR246 magrolimab, menin inhibitors). The wealth of positive data allows reconsideration of what might soon be new standards of care in younger and older patients with AML. In this review we give an overview of recently approved therapies in AML and address present and future research directions. © 2021 The Authors. British Journal of Haematology published by British Society for Haematology and John Wiley & Sons Ltd. DOI: 10.1111/bjh.17746 PMID: 34350585 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/34154993
1. Drug Metab Dispos. 2022 Jun;50(6):781-797. doi: 10.1124/dmd.121.000428. Epub 2021 Jun 21. The Nonclinical Disposition and Pharmacokinetic/Pharmacodynamic Properties of N-Acetylgalactosamine-Conjugated Small Interfering RNA Are Highly Predictable and Build Confidence in Translation to Human. McDougall R(1), Ramsden D(1), Agarwal S(1), Agarwal S(1), Aluri K(1), Arciprete M(1), Brown C(1), Castellanos-Rizaldos E(1), Charisse K(1), Chong S(1), Cichocki J(1), Fitzgerald K(1), Goel V(1), Gu Y(1), Guenther D(1), Habtemariam B(1), Jadhav V(1), Janas M(1), Jayaraman M(1), Kurz J(1), Li J(1), Liu J(1), Liu X(1), Liou S(1), Maclauchlin C(1), Maier M(1), Manoharan M(1), Nair JK(1), Robbie G(1), Schmidt K(1), Smith P(1), Theile C(1), Vaishnaw A(1), Waldron S(1), Xu Y(1), Zhang X(1), Zlatev I(1), Wu JT(2). Author information: (1)Alnylam Pharmaceuticals, Cambridge, Massachusetts. (2)Alnylam Pharmaceuticals, Cambridge, Massachusetts [email protected]. Conjugation of oligonucleotide therapeutics, including small interfering RNAs (siRNAs) or antisense oligonucleotides, to N-acetylgalactosamine (GalNAc) ligands has become the primary strategy for hepatocyte-targeted delivery, and with the recent approvals of GIVLAARI (givosiran) for the treatment of acute hepatic porphyria, OXLUMO (lumasiran) for the treatment of primary hyperoxaluria, and Leqvio (inclisiran) for the treatment of hypercholesterolemia, the technology has been well validated clinically. Although much knowledge has been gained over decades of development, there is a paucity of published literature on the drug metabolism and pharmacokinetic properties of GalNAc-siRNA. With this in mind, the goals of this minireview are to provide an aggregate analysis of these nonclinical absorption, distribution, metabolism, and excretion (ADME) data to build confidence on the translation of these properties to human. Upon subcutaneous administration, GalNAc-conjugated siRNAs are quickly distributed to the liver, resulting in plasma pharmacokinetic (PK) properties that reflect rapid elimination through asialoglycoprotein receptor-mediated uptake from circulation into hepatocytes. These studies confirm that liver PK, including half-life and, most importantly, siRNA levels in RNA-induced silencing complex in hepatocytes, are better predictors of pharmacodynamics (PD) than plasma PK. Several in vitro and in vivo nonclinical studies were conducted to characterize the ADME properties of GalNAc-conjugated siRNAs. These studies demonstrate that the PK/PD and ADME properties of GalNAc-conjugated siRNAs are highly conserved across species, are largely predictable, and can be accurately scaled to human, allowing us to identify efficacious and safe clinical dosing regimens in the absence of human liver PK profiles. SIGNIFICANCE STATEMENT: Several nonclinical ADME studies have been conducted in order to provide a comprehensive overview of the disposition and elimination of GalNAc-conjugated siRNAs and the pharmacokinetic/pharmacodynamic translation between species. These studies demonstrate that the ADME properties of GalNAc-conjugated siRNAs are well correlated and predictable across species, building confidence in the ability to extrapolate to human. Copyright © 2022 by The Author(s). DOI: 10.1124/dmd.121.000428 PMID: 34154993 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/36250252
1. Br J Pharmacol. 2023 Nov;180(21):2697-2720. doi: 10.1111/bph.15972. Epub 2022 Nov 13. Small interfering RNA: Discovery, pharmacology and clinical development-An introductory review. Ranasinghe P(1)(2), Addison ML(2), Dear JW(2), Webb DJ(2). Author information: (1)Department of Pharmacology, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka. (2)University/British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, UK. Post-transcriptional gene silencing targets and degrades mRNA transcripts, silencing the expression of specific genes. RNA interference technology, using synthetic structurally well-defined short double-stranded RNA (small interfering RNA [siRNA]), has advanced rapidly in recent years. This introductory review describes the utility of siRNA, by exploring the underpinning biology, pharmacology, recent advances and clinical developments, alongside potential limitations and ongoing challenges. Mediated by the RNA-induced silencing complex, siRNAs bind to specific complementary mRNAs, which are subsequently degraded. siRNA therapy offers advantages over other therapeutic approaches, including ability of specifically designed siRNAs to potentially target any mRNA and improved patient adherence through infrequent administration associated with a very long duration of action. Key pharmacokinetic and pharmacodynamic challenges include targeted administration, poor tissue penetration, nuclease inactivation, rapid renal elimination, immune activation and off-target effects. These have been overcome by chemical modification of siRNA and/or by utilising a range of delivery systems, increasing bioavailability and stability to allow successful clinical translation. Patisiran (hereditary transthyretin-mediated amyloidosis) was the first licensed siRNA, followed by givosiran (acute hepatic porphyria), lumasiran (primary hyperoxaluria type 1) and inclisiran (familial hypercholesterolaemia), which all use N-acetylgalactosamine (GalNAc) linkage for effective liver-directed delivery. Others are currently under development for indications varying from rare genetic diseases to common chronic non-communicable diseases (hypertension, cancer). Technological advances are paving the way for broader clinical use. Ongoing challenges remain in targeting organs beyond the liver and reaching special sites (e.g., brain). By overcoming these barriers, siRNA therapy has the potential to substantially widen its therapeutic impact. © 2022 The Authors. British Journal of Pharmacology published by John Wiley & Sons Ltd on behalf of British Pharmacological Society. DOI: 10.1111/bph.15972 PMID: 36250252 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/25434769
1. J Am Chem Soc. 2014 Dec 10;136(49):16958-61. doi: 10.1021/ja505986a. Epub 2014 Dec 1. Multivalent N-acetylgalactosamine-conjugated siRNA localizes in hepatocytes and elicits robust RNAi-mediated gene silencing. Nair JK(1), Willoughby JL, Chan A, Charisse K, Alam MR, Wang Q, Hoekstra M, Kandasamy P, Kel'in AV, Milstein S, Taneja N, O'Shea J, Shaikh S, Zhang L, van der Sluis RJ, Jung ME, Akinc A, Hutabarat R, Kuchimanchi S, Fitzgerald K, Zimmermann T, van Berkel TJ, Maier MA, Rajeev KG, Manoharan M. Author information: (1)Alnylam Pharmaceuticals , 300 Third Street, Cambridge, Massachusetts 02142, United States. Conjugation of small interfering RNA (siRNA) to an asialoglycoprotein receptor ligand derived from N-acetylgalactosamine (GalNAc) facilitates targeted delivery of the siRNA to hepatocytes in vitro and in vivo. The ligands derived from GalNAc are compatible with solid-phase oligonucleotide synthesis and deprotection conditions, with synthesis yields comparable to those of standard oligonucleotides. Subcutaneous (SC) administration of siRNA-GalNAc conjugates resulted in robust RNAi-mediated gene silencing in liver. Refinement of the siRNA chemistry achieved a 5-fold improvement in efficacy over the parent design in vivo with a median effective dose (ED50) of 1 mg/kg following a single dose. This enabled the SC administration of siRNA-GalNAc conjugates at therapeutically relevant doses and, importantly, at dose volumes of ≤1 mL. Chronic weekly dosing resulted in sustained dose-dependent gene silencing for over 9 months with no adverse effects in rodents. The optimally chemically modified siRNA-GalNAc conjugates are hepatotropic and long-acting and have the potential to treat a wide range of diseases involving liver-expressed genes. DOI: 10.1021/ja505986a PMID: 25434769 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/35505960
1. Mol Ther Nucleic Acids. 2022 Apr 2;28:423-434. doi: 10.1016/j.omtn.2022.04.003. eCollection 2022 Jun 14. RAB18 is a key regulator of GalNAc-conjugated siRNA-induced silencing in Hep3B cells. Lu J(1), Swearingen E(1), Hardy M(1), Collins P(1), Wu B(1), Yuan E(1), Lu D(1), Li CM(1), Wang S(1), Ollmann M(1). Author information: (1)Genome Analysis Unit, Amgen Global Research, 1120 Veteran Blvd, ASF1, South San Francisco, CA 94080, USA. Small interfering RNA (siRNA) therapeutics have developed rapidly in recent years, despite the challenges associated with delivery of large, highly charged nucleic acids. Delivery of siRNA therapeutics to the liver has been established, with conjugation of siRNA to N-acetylgalactosamine (GalNAc) providing durable gene knockdown in hepatocytes following subcutaneous injection. GalNAc binds the asialoglycoprotein receptor (ASGPR) that is highly expressed on hepatocytes and exploits this scavenger receptor to deliver siRNA across the plasma membrane by endocytosis. However, siRNA needs to access the RNA-induced silencing complex (RISC) in the cytoplasm to provide effective gene knockdown, and the entire siRNA delivery process is very inefficient, likely because of steps required for endosomal escape, intracellular trafficking, and stability of siRNA. To reveal the cellular factors limiting delivery of siRNA therapeutics, we performed a genome-wide pooled knockout screen on the basis of delivery of GalNAc-conjugated siRNA targeting the HPRT1 gene in the human hepatocellular carcinoma line Hep3B. Our primary genome-wide pooled knockout screen identified candidate genes that when knocked out significantly enhanced siRNA efficacy in Hep3B cells. Follow-up studies indicate that knockout of RAB18 improved the efficacy of siRNA delivered by GalNAc, cholesterol, or antibodies, but not siRNA delivered by Lipofectamine transfection, suggesting a role for RAB18 in siRNA delivery and intracellular trafficking. © 2022 The Authors. DOI: 10.1016/j.omtn.2022.04.003 PMCID: PMC9035644 PMID: 35505960 Conflict of interest statement: All authors have the following conflicts of interest to declare: J.L., E.S., B.W., E.Y., D.L., C.-M.L., and S.W. are employees of Amgen Inc. M.O., P.C., and M.H. were employed by Amgen Inc. while working on the study. All authors owned Amgen shares when the study was carried out. However, these do not alter the authors’ adherence to all journal policies on sharing data and materials. None of the authors serves as a current editorial team member for this journal.
http://www.ncbi.nlm.nih.gov/pubmed/25786782
1. Chembiochem. 2015 Apr 13;16(6):903-8. doi: 10.1002/cbic.201500023. Epub 2015 Mar 18. Hepatocyte-specific delivery of siRNAs conjugated to novel non-nucleosidic trivalent N-acetylgalactosamine elicits robust gene silencing in vivo. Rajeev KG(1), Nair JK, Jayaraman M, Charisse K, Taneja N, O'Shea J, Willoughby JL, Yucius K, Nguyen T, Shulga-Morskaya S, Milstein S, Liebow A, Querbes W, Borodovsky A, Fitzgerald K, Maier MA, Manoharan M. Author information: (1)Alnylam. Pharmaceuticals, 300 Third Street, Cambridge, Massachusetts 02142 (USA). [email protected]. We recently demonstrated that siRNAs conjugated to triantennary N-acetylgalactosamine (GalNAc) induce robust RNAi-mediated gene silencing in the liver, owing to uptake mediated by the asialoglycoprotein receptor (ASGPR). Novel monovalent GalNAc units, based on a non-nucleosidic linker, were developed to yield simplified trivalent GalNAc-conjugated oligonucleotides under solid-phase synthesis conditions. Synthesis of oligonucleotide conjugates using monovalent GalNAc building blocks required fewer synthetic steps compared to the previously optimized triantennary GalNAc construct. The redesigned trivalent GalNAc ligand maintained optimal valency, spatial orientation, and distance between the sugar moieties for proper recognition by ASGPR. siRNA conjugates were synthesized by sequential covalent attachment of the trivalent GalNAc to the 3'-end of the sense strand and resulted in a conjugate with in vitro and in vivo potency similar to that of the parent trivalent GalNAc conjugate design. © 2015 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim. DOI: 10.1002/cbic.201500023 PMID: 25786782 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/25730476
1. ACS Chem Biol. 2015 May 15;10(5):1181-7. doi: 10.1021/cb501028c. Epub 2015 Mar 2. siRNA conjugates carrying sequentially assembled trivalent N-acetylgalactosamine linked through nucleosides elicit robust gene silencing in vivo in hepatocytes. Matsuda S(1), Keiser K(1), Nair JK(1), Charisse K(1), Manoharan RM(1), Kretschmer P(1), Peng CG(1), V Kel'in A(1), Kandasamy P(1), Willoughby JL(1), Liebow A(1), Querbes W(1), Yucius K(1), Nguyen T(1), Milstein S(1), Maier MA(1), Rajeev KG(1), Manoharan M(1). Author information: (1)Alnylam Pharmaceuticals, 300 Third Street, Cambridge, Massachusetts 02142, United States. Asialoglycoprotein receptor (ASGPR) mediated delivery of triantennary N-acetylgalactosamine (GalNAc) conjugated short interfering RNAs (siRNAs) to hepatocytes is a promising paradigm for RNAi therapeutics. Robust and durable gene silencing upon subcutaneous administration at therapeutically acceptable dose levels resulted in the advancement of GalNAc-conjugated oligonucleotide-based drugs into preclinical and clinical developments. To systematically evaluate the effect of display and positioning of the GalNAc moiety within the siRNA duplex on ASGPR binding and RNAi activity, nucleotides carrying monovalent GalNAc were designed. Evaluation of clustered and dispersed incorporation of GalNAc units to the sense (S) strand indicated that sugar proximity is critical for ASGPR recognition, and location of the clustered ligand impacts the intrinsic potency of the siRNA. An array of nucleosidic GalNAc monomers resembling a trivalent ligand at or near the 3' end of the S strand retained in vitro and in vivo siRNA activity, similar to the parent conjugate design. This work demonstrates the utility of simple, nucleotide-based, cost-effective siRNA-GalNAc conjugation strategies. DOI: 10.1021/cb501028c PMID: 25730476 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/34413198
1. J Pharmacol Exp Ther. 2021 Nov;379(2):134-146. doi: 10.1124/jpet.121.000805. Epub 2021 Aug 19. Minimal Physiologically Based Pharmacokinetic-Pharmacodynamic (mPBPK-PD) Model of N-Acetylgalactosamine-Conjugated Small Interfering RNA Disposition and Gene Silencing in Preclinical Species and Humans. Ayyar VS(1), Song D(2), Zheng S(2), Carpenter T(2), Heald DL(2). Author information: (1)Clinical Pharmacology & Pharmacometrics (V.S.A., D.S.) and Janssen BioTherapeutics (V.S.A., S.Z., T.C., D.L.H.), Janssen Research and Development, Spring House, Pennsylvania [email protected]. (2)Clinical Pharmacology & Pharmacometrics (V.S.A., D.S.) and Janssen BioTherapeutics (V.S.A., S.Z., T.C., D.L.H.), Janssen Research and Development, Spring House, Pennsylvania. Conjugation of small interfering RNA (siRNA) to tris N-acetylgalactosamine [(GalNAc)3] can enable highly selective, potent, and durable knockdown of targeted proteins in the liver. However, potential knowledge gaps between in vitro experiments, preclinical species, and clinical scenarios remain. A minimal physiologically based pharmacokinetic-pharmacodynamic model for GalNAc-conjugated siRNA (GalNAc-siRNA) was developed using published data for fitusiran (ALN-AT3), an investigational compound targeting liver antithrombin (AT), to delineate putative determinants governing the whole-body-to-cellular pharmacokinetic (PK) and pharmacodynamic (PD) properties of GalNAc-siRNA and facilitate preclinical-to-clinical translation. The model mathematically linked relevant mechanisms: 1) hepatic biodistribution, 2) tris-GalNAc binding to asialoglycoprotein receptors (ASGPRs) on hepatocytes, 3) ASGPR endocytosis and recycling, 4) endosomal transport and escape of siRNA, 5) cytoplasmic RNA-induced silencing complex (RISC) loading, 6) degradation of target mRNA by bound RISC, and 7) knockdown of protein. Physiologic values for 36 out of 48 model parameters were obtained from the literature. Kinetic parameters governing (GalNAc)3-ASGPR binding and internalization were derived from published studies of uptake in hepatocytes. The proposed model well characterized reported pharmacokinetics, RISC dynamics, and knockdown of AT mRNA and protein by ALN-AT3 in mice. The model bridged multiple PK-PD data sets in preclinical species (mice, rat, monkey) and successfully captured reported plasma pharmacokinetics and AT knockdown in a phase I ascending-dose study. Estimates of in vivo potency were similar (∼2-fold) across species. Subcutaneous absorption and serum AT degradation rate constants scaled across species by body weight with allometric exponents of -0.29 and -0.22. The proposed mechanistic modeling framework characterizes the unique PK-PD properties of GalNAc-siRNA. SIGNIFICANCE STATEMENT: Tris N-acetylgalactosamine (GalNAc)3-conjugated small interfering RNA (siRNA) therapeutics enable liver-targeted gene therapy and precision medicine. Using a translational and systems-based minimal physiologically based pharmacokinetic-pharmacodynamic (mPBPK-PD) modeling approach, putative determinants influencing GalNAc-conjugated siRNA (GalNAc-siRNA) functionality in three preclinical species and humans were investigated. The developed model successfully integrated and characterized relevant published in vitro-derived biomeasures, mechanistic PK-PD profiles in animals, and observed clinical PK-PD responses for an investigational GalNAc-siRNA (fitusiran). This modeling effort delineates the disposition and liver-targeted pharmacodynamics of GalNAc-siRNA. Copyright © 2021 by The American Society for Pharmacology and Experimental Therapeutics. DOI: 10.1124/jpet.121.000805 PMID: 34413198 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/34091052
1. Mol Ther. 2021 Oct 6;29(10):2910-2919. doi: 10.1016/j.ymthe.2021.06.002. Epub 2021 Jun 4. Ligand conjugate SAR and enhanced delivery in NHP. Holland RJ(1), Lam K(1), Ye X(1), Martin AD(1), Wood MC(1), Palmer L(1), Fraser D(1), McClintock K(1), Majeski S(1), Jarosz A(1), Lee ACH(2), Thi EP(2), Judge A(1), Heyes J(3). Author information: (1)Genevant Sciences Corporation, Vancouver, BC V5T 4T5, Canada. (2)Arbutus Biopharma Corporation, Warminster, PA 18974, USA. (3)Genevant Sciences Corporation, Vancouver, BC V5T 4T5, Canada. Electronic address: [email protected]. Comment in Mol Ther. 2021 Oct 6;29(10):2893-2894. doi: 10.1016/j.ymthe.2021.09.006. N-Acetylgalactosamine (GalNAc) conjugated short interfering RNAs (siRNAs) are a leading RNA interference (RNAi) platform allowing targeted inhibition of disease-causing genes in hepatocytes. More than a decade of development has recently resulted in the first approvals for this class of drugs. While substantial effort has been made to improve nucleic acid modification patterns for better payload stability and efficacy, relatively little attention has been given to the GalNAc targeting ligand. In addition, the lack of an intrinsic endosomal release mechanism has limited potency. Here, we report a stepwise analysis of the structure activity relationships (SAR) of the components comprising these targeting ligands. We show that there is relatively little difference in biological performance between bi-, tri-, and tetravalent ligand structures while identifying other features that affect their biological activity more significantly. Further, we demonstrate that subcutaneous co-administration of a GalNAc-functionalized, pH responsive endosomal release agent markedly improved the activity and duration of effect for siRNA conjugates, without compromising tolerability, in non-human primates. These findings could address a significant bottleneck for future siRNA ligand conjugate development. Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved. DOI: 10.1016/j.ymthe.2021.06.002 PMCID: PMC8531135 PMID: 34091052 [Indexed for MEDLINE] Conflict of interest statement: Declaration of interests The authors are employees or consultants of Genevant Sciences Corporation or Arbutus Biopharma Corporation as noted in the author affiliations and own shares or stock options in their respective companies.
http://www.ncbi.nlm.nih.gov/pubmed/29792572
1. Nucleic Acid Ther. 2018 Jun;28(3):109-118. doi: 10.1089/nat.2018.0736. Epub 2018 May 24. GalNAc-siRNA Conjugates: Leading the Way for Delivery of RNAi Therapeutics. Springer AD(1), Dowdy SF(1). Author information: (1)Department of Cellular and Molecular Medicine, University of California San Diego , La Jolla, California. Short-interfering RNA (siRNA)-induced RNAi responses have great potential to treat a wide variety of human diseases from cancer to pandemic viral outbreaks to Parkinson's Disease. However, before siRNAs can become drugs, they must overcome a billion years of evolutionary defenses designed to keep invading RNAs on the outside cells from getting to the inside of cells. Not surprisingly, significant effort has been placed in developing a wide array of delivery technologies. Foremost of these has been the development of N-acetylgalactosamine (GalNAc) siRNA conjugates for delivery to liver. Tris-GalNAc binds to the Asialoglycoprotein receptor that is highly expressed on hepatocytes resulting in rapid endocytosis. While the exact mechanism of escape across the endosomal lipid bilayer membrane remains unknown, sufficient amounts of siRNAs enter the cytoplasm to induce robust, target selective RNAi responses in vivo. Multiple GalNAc-siRNA conjugate clinical trials, including two phase III trials, are currently underway by three biotech companies to treat a wide variety of diseases. GalNAc-siRNA conjugates are a simple solution to the siRNA delivery problem for liver hepatocytes and have shown the RNAi (and antisense oligonucleotide) field the path forward for targeting other tissue types. DOI: 10.1089/nat.2018.0736 PMCID: PMC5994659 PMID: 29792572 [Indexed for MEDLINE] Conflict of interest statement: S.F.D. is a founder of Solstice Biologics. A.D.S. declares no competing financial interests exist.
http://www.ncbi.nlm.nih.gov/pubmed/36281788
1. Clin Pharmacol Ther. 2023 Feb;113(2):328-338. doi: 10.1002/cpt.2774. Epub 2022 Nov 14. Translational Population-Pharmacodynamic Modeling of a Novel Long-Acting siRNA Therapy, Inclisiran, for the Treatment of Hypercholesterolemia. Gosselin NH(1), Schuck VJA(1), Barriere O(1), Kulmatycki K(2), Margolskee A(2), Smith P(1), He Y(2). Author information: (1)Certara, Cambridge, Massachusetts, USA. (2)Novartis Institute for Biomedical Research, Cambridge, Massachusetts, USA. Inclisiran is a novel N-acetylgalactosamine (GalNAc) conjugated small-interfering ribonucleic acid (siRNA) therapy designed to specifically target proprotein convertase subtilisin/kexin type 9 (PCSK9) mRNA in the liver for the treatment of hypercholesterolemia. Inclisiran's GalNAc attachment results in a rapid uptake into the liver, and thus a short plasma half-life, but long duration of effects on PCSK9 inhibition and low-density lipoprotein cholesterol (LDL-C) lowering. The effects on PCSK9 inhibition and consequent LDL-C reduction are sustained for more than 6 months following a single subcutaneous (s.c.) dose, despite inclisiran being detectable in the plasma only for up to 48 hours. A kinetic-pharmacodynamic (K-PD) model was developed to characterize inclisiran's dose-related LDL-C lowering effects and to evaluate the impact of intrinsic and extrinsic factors on LDL-C lowering. To accommodate the long duration of action, the K-PD model incorporated an effect compartment which represents the liver. Inclisiran concentration in the liver leads to decreased production of the PCSK9 protein and allow recycling of more LDL-C receptors on the hepatocyte cell surface, which results in a reduction of circulating LDL-C. The analysis of covariates identified PCSK9 and LDL-C baseline levels as important factors for the effects of LDL-C lowering. Observations and modeling and simulation results demonstrated that PCSK9 and LDL-C reductions are achieved rapidly after dosing and sustained when patients are treated with a 300 mg s.c. dose once every 6 months. © 2022 The Authors. Clinical Pharmacology & Therapeutics © 2022 American Society for Clinical Pharmacology and Therapeutics. DOI: 10.1002/cpt.2774 PMID: 36281788 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/32170309
1. Nucleic Acids Res. 2020 May 7;48(8):4028-4040. doi: 10.1093/nar/gkaa125. Chimeric siRNAs with chemically modified pentofuranose and hexopyranose nucleotides: altritol-nucleotide (ANA) containing GalNAc-siRNA conjugates: in vitro and in vivo RNAi activity and resistance to 5'-exonuclease. Kumar P(1), Degaonkar R(1), Guenther DC(1), Abramov M(2), Schepers G(2), Capobianco M(1), Jiang Y(1), Harp J(3), Kaittanis C(1), Janas MM(1), Castoreno A(1), Zlatev I(1), Schlegel MK(1), Herdewijn P(2), Egli M(3), Manoharan M(1). Author information: (1)Alnylam Pharmaceuticals, 300 Third Street, Cambridge, MA 02142, USA. (2)Medicinal Chemistry, Rega Institute for Medical Research, KU Leuven, Herestraat 49, 3000 Leuven, Belgium. (3)Department of Biochemistry, School of Medicine, Vanderbilt University, Nashville, TN 37232, USA. In this report, we investigated the hexopyranose chemical modification Altriol Nucleic Acid (ANA) within small interfering RNA (siRNA) duplexes that were otherwise fully modified with the 2'-deoxy-2'-fluoro and 2'-O-methyl pentofuranose chemical modifications. The siRNAs were designed to silence the transthyretin (Ttr) gene and were conjugated to a trivalent N-acetylgalactosamine (GalNAc) ligand for targeted delivery to hepatocytes. Sense and antisense strands of the parent duplex were synthesized with single ANA residues at each position on the strand, and the resulting siRNAs were evaluated for their ability to inhibit Ttr mRNA expression in vitro. Although ANA residues were detrimental at the 5' end of the antisense strand, the siRNAs with ANA at position 6 or 7 in the seed region had activity comparable to the parent. The siRNA with ANA at position 7 in the seed region was active in a mouse model. An Oligonucleotide with ANA at the 5' end was more stable in the presence of 5'-exonuclease than an oligonucleotide of the same sequence and chemical composition without the ANA modification. Modeling studies provide insight into the origins of regiospecific changes in potency of siRNAs and the increased protection against 5'-exonuclease degradation afforded by the ANA modification. © The Author(s) 2020. Published by Oxford University Press on behalf of Nucleic Acids Research. DOI: 10.1093/nar/gkaa125 PMCID: PMC7192627 PMID: 32170309 [Indexed for MEDLINE]