1
|
Hussain A, Mohammad T, Khan S, Alajmi MF, Yadav DK, Hassan MI. Seven Hub Genes Associated with Huntington's Disease and Diagnostic and Therapeutic Potentials Identified by Computational Biology. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2025; 29:154-163. [PMID: 40059764 DOI: 10.1089/omi.2025.0006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/03/2025]
Abstract
Huntington's disease (HD) is characterized by progressive motor dysfunction and cognitive decline. Early diagnosis and new therapeutic targets are essential for effective interventions. We performed integrative analyses of mRNA profiles from three microarrays and one RNA-seq dataset from the Gene Expression Omnibus database. The datasets included were GSE8762, GSE24250, GSE45516, and GSE64810. Data pre-processing included background correction, normalization, log2 transformation, probe-to-gene symbol mapping, and differential expression analysis. We identified 80 differentially expressed genes (DEGs) based on a significance threshold (p < 0.05) and absolute log fold change (logFC) >0.65. Additionally, we conducted Gene Ontology (GO) and pathway analyses of the identified genes. Protein-protein interactions among DEGs revealed a network from which seven hub genes (VIM, COL1A1, COL3A1, COL1A2, DCN, CXCR2, and S100A9) were identified using the cytoHubba plugin in Cytoscape software. Two top DEGs, IGHG1 (up-regulated) and PITX1 (up-regulated), also hold potential as therapeutic targets. Insofar as biological contextualization of the findings is concerned, the top enriched GO terms were skeletal system development, blood vessel development, and vasculature development. Molecular function terms highlighted signaling receptor binding, extracellular matrix structural constituent, and platelet-derived growth factor binding. Notably, the significant KEGG pathways included amoebiasis, the AGE-RAGE signaling pathway in diabetic complications, and the relaxin signaling pathway. In conclusion, the present computational biology integrative analyses of multiple datasets discovered new DEGs and seven hub genes, shedding light on molecular mechanisms of HD. These findings call for translational clinical omics research and may potentially lead to future precision medicine interventions and novel diagnostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Afzal Hussain
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Taj Mohammad
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Shumayila Khan
- International Health Division, Indian Council of Medical Research, New Delhi, India
| | - Mohamed F Alajmi
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Dharmendra Kumar Yadav
- Department of Biologics, College of Pharmacy, Gachon University, Yeonsu-gu Incheon, Republic of Korea
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
2
|
Buzdin AA, Heydarov RN, Golounina OO, Suntsova MV, Matrosova AV, Bondarenko EV, Roumiantsev SA, Sorokin MI, Kholodenko RV, Kholodenko IV, Chekhonin VP, Plaksina EV, Rozhinskaya LY, Melnichenko GA, Belaya ZE. Transcriptome-Wide Analysis of Pituitary and Ectopic Adrenocorticotropic Hormone-Secreting Tumors. Cancers (Basel) 2025; 17:658. [PMID: 40002253 PMCID: PMC11852724 DOI: 10.3390/cancers17040658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/22/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Endogenous Cushing's syndrome (CS) is a rare neuroendocrine disorder characterized by either secondary cortisol increases due to an adrenocorticotropic hormone (ACTH)-secreting pituitary tumor (Cushing's disease (CD)), an ACTH-secreting neuroendocrine tumor (NET) of non-pituitary origin (ectopic ACTH syndrome (EAS)), or by the primarily adrenal autonomous overproduction of cortisol [...].
Collapse
Affiliation(s)
- Anton A. Buzdin
- Endocrinology Research Center, Moscow 117036, Russia; (R.N.H.); (O.O.G.); (M.V.S.); (A.V.M.); (E.V.B.); (S.A.R.); (M.I.S.); (V.P.C.); (E.V.P.); (L.Y.R.); (G.A.M.); (Z.E.B.)
- Institute of Personalized Oncology, I.M. Sechenov First Moscow State Medical University, Moscow 119991, Russia
- Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia;
| | - Rustam N. Heydarov
- Endocrinology Research Center, Moscow 117036, Russia; (R.N.H.); (O.O.G.); (M.V.S.); (A.V.M.); (E.V.B.); (S.A.R.); (M.I.S.); (V.P.C.); (E.V.P.); (L.Y.R.); (G.A.M.); (Z.E.B.)
- Institute of Personalized Oncology, I.M. Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Olga O. Golounina
- Endocrinology Research Center, Moscow 117036, Russia; (R.N.H.); (O.O.G.); (M.V.S.); (A.V.M.); (E.V.B.); (S.A.R.); (M.I.S.); (V.P.C.); (E.V.P.); (L.Y.R.); (G.A.M.); (Z.E.B.)
| | - Maria V. Suntsova
- Endocrinology Research Center, Moscow 117036, Russia; (R.N.H.); (O.O.G.); (M.V.S.); (A.V.M.); (E.V.B.); (S.A.R.); (M.I.S.); (V.P.C.); (E.V.P.); (L.Y.R.); (G.A.M.); (Z.E.B.)
- Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia
| | - Alina V. Matrosova
- Endocrinology Research Center, Moscow 117036, Russia; (R.N.H.); (O.O.G.); (M.V.S.); (A.V.M.); (E.V.B.); (S.A.R.); (M.I.S.); (V.P.C.); (E.V.P.); (L.Y.R.); (G.A.M.); (Z.E.B.)
- Institute of Personalized Oncology, I.M. Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Ekaterina V. Bondarenko
- Endocrinology Research Center, Moscow 117036, Russia; (R.N.H.); (O.O.G.); (M.V.S.); (A.V.M.); (E.V.B.); (S.A.R.); (M.I.S.); (V.P.C.); (E.V.P.); (L.Y.R.); (G.A.M.); (Z.E.B.)
| | - Sergey A. Roumiantsev
- Endocrinology Research Center, Moscow 117036, Russia; (R.N.H.); (O.O.G.); (M.V.S.); (A.V.M.); (E.V.B.); (S.A.R.); (M.I.S.); (V.P.C.); (E.V.P.); (L.Y.R.); (G.A.M.); (Z.E.B.)
| | - Maksim I. Sorokin
- Endocrinology Research Center, Moscow 117036, Russia; (R.N.H.); (O.O.G.); (M.V.S.); (A.V.M.); (E.V.B.); (S.A.R.); (M.I.S.); (V.P.C.); (E.V.P.); (L.Y.R.); (G.A.M.); (Z.E.B.)
- Oncobox LLC, Moscow 119991, Russia
| | - Roman V. Kholodenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia;
| | - Irina V. Kholodenko
- Laboratory of Cell Biology, Orekhovich Institute of Biomedical Chemistry, Moscow 119121, Russia;
| | - Vladimir P. Chekhonin
- Endocrinology Research Center, Moscow 117036, Russia; (R.N.H.); (O.O.G.); (M.V.S.); (A.V.M.); (E.V.B.); (S.A.R.); (M.I.S.); (V.P.C.); (E.V.P.); (L.Y.R.); (G.A.M.); (Z.E.B.)
| | - Evgeniya V. Plaksina
- Endocrinology Research Center, Moscow 117036, Russia; (R.N.H.); (O.O.G.); (M.V.S.); (A.V.M.); (E.V.B.); (S.A.R.); (M.I.S.); (V.P.C.); (E.V.P.); (L.Y.R.); (G.A.M.); (Z.E.B.)
| | - Liudmila Y. Rozhinskaya
- Endocrinology Research Center, Moscow 117036, Russia; (R.N.H.); (O.O.G.); (M.V.S.); (A.V.M.); (E.V.B.); (S.A.R.); (M.I.S.); (V.P.C.); (E.V.P.); (L.Y.R.); (G.A.M.); (Z.E.B.)
| | - Galina A. Melnichenko
- Endocrinology Research Center, Moscow 117036, Russia; (R.N.H.); (O.O.G.); (M.V.S.); (A.V.M.); (E.V.B.); (S.A.R.); (M.I.S.); (V.P.C.); (E.V.P.); (L.Y.R.); (G.A.M.); (Z.E.B.)
| | - Zhanna E. Belaya
- Endocrinology Research Center, Moscow 117036, Russia; (R.N.H.); (O.O.G.); (M.V.S.); (A.V.M.); (E.V.B.); (S.A.R.); (M.I.S.); (V.P.C.); (E.V.P.); (L.Y.R.); (G.A.M.); (Z.E.B.)
| |
Collapse
|
3
|
Aguilar GR, Vidal B, Ji H, Evenblij J, Liao CP, Ji H, Valperga G, Fang-Yen C, Hobert O. Functional analysis of conserved C. elegans bHLH family members uncovers lifespan control by a peptidergic hub neuron. PLoS Biol 2025; 23:e3002979. [PMID: 39761329 PMCID: PMC11703107 DOI: 10.1371/journal.pbio.3002979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 12/11/2024] [Indexed: 01/15/2025] Open
Abstract
Throughout the animal kingdom, several members of the basic helix-loop-helix (bHLH) family act as proneural genes during early steps of nervous system development. Roles of bHLH genes in specifying terminal differentiation of postmitotic neurons have been less extensively studied. We analyze here the function of 5 Caenorhabditis elegans bHLH genes, falling into 3 phylogenetically conserved subfamilies, which are continuously expressed in a very small number of postmitotic neurons in the central nervous system. We show (a) that 2 orthologs of the vertebrate bHLHe22/e23 genes, called hlh-17 and hlh-32, function redundantly to specify the identity of a single head interneuron class (AUA), as well as an individual motor neuron (VB2); (b) that the PTF1a ortholog hlh-13 acts as a terminal selector to control terminal differentiation and function of the sole octopaminergic neuron class in C. elegans, RIC; and (c) that the NHLH1/2 ortholog hlh-15 controls terminal differentiation and function of the peptidergic AVK head interneuron class, a known neuropeptidergic signaling hub in the animal. Strikingly, through null mutant analysis and cell-specific rescue experiments, we find that loss of hlh-15/NHLH in the peptidergic AVK neurons and the resulting abrogation of neuropeptide secretion from these neurons causes a substantially extended lifespan of the animal, which we propose to be akin to hypothalamic control of lifespan in vertebrates. Our functional analysis reveals themes of bHLH gene function during terminal differentiation that are complementary to the earlier lineage specification roles of other bHLH family members. However, such late functions are much more sparsely employed by members of the bHLH transcription factor family, compared to the function of the much more broadly employed homeodomain transcription factor family.
Collapse
Affiliation(s)
- G. Robert Aguilar
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, New York, United States of America
| | - Berta Vidal
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, New York, United States of America
| | - Hongzhu Ji
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, New York, United States of America
| | - Joke Evenblij
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, New York, United States of America
- Technische Universität, Braunschweig, Germany
| | - Chien-Po Liao
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, New York, United States of America
| | - Hongfei Ji
- Department of Biomedical Engineering, Ohio State University, Columbus, Ohio, United States of America
| | - Giulio Valperga
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, New York, United States of America
| | - Christopher Fang-Yen
- Department of Biomedical Engineering, Ohio State University, Columbus, Ohio, United States of America
| | - Oliver Hobert
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, New York, United States of America
| |
Collapse
|
4
|
Le Ciclé C, Cohen-Tannoudji J, L'Hôte D. Recent Advances in the Understanding of Gonadotrope Lineage Differentiation in the Developing Pituitary. Neuroendocrinology 2024; 115:195-210. [PMID: 39527929 PMCID: PMC11924211 DOI: 10.1159/000542513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND The pituitary gland is a vital endocrine organ regulating body homoeostasis through six hormone-secreting cell types. Among these, pituitary gonadotrope cells are essential for reproductive function. Throughout pituitary ontogenesis, gonadotrope cells differentiate in a stepwise process, involving both morphogenic cues and transcription factors, which drives specification of progenitor cells into specialised endocrine cells. It is crucial to understand the mechanisms underlying gonadotrope differentiation, as developmental defects and abnormalities in this process can lead to many reproductive pathologies. SUMMARY This review offers a detailed overview of the latest advances in gonadotrope cell differentiation. We addressed this question with a specific focus on three important aspects of gonadotrope differentiation: the identification of the progenitor population giving rise to gonadotrope cells, the early mechanisms that initiate Nr5a1 expression and thus gonadotrope fate commitment, and finally, the mechanisms driving the formation of physical and functional gonadotrope networks. KEY MESSAGES Overall, this review aimed to provide new insights into three aspects of the gonadotrope differentiation process by reconsidering pioneering studies in the light of data gained from latest technological developments. Firstly, we re-investigated the long debated developmental trajectory of pituitary gonadotrope cells. Secondly, we reported new regulatory mechanisms of Nr5a1 expression, focusing on the involvement of ERα. Finally, we highlighted the molecular and cellular mechanisms driving gonadotrope network formation during embryogenesis, a process that seems essential for regulation of gonadotrope activity. BACKGROUND The pituitary gland is a vital endocrine organ regulating body homoeostasis through six hormone-secreting cell types. Among these, pituitary gonadotrope cells are essential for reproductive function. Throughout pituitary ontogenesis, gonadotrope cells differentiate in a stepwise process, involving both morphogenic cues and transcription factors, which drives specification of progenitor cells into specialised endocrine cells. It is crucial to understand the mechanisms underlying gonadotrope differentiation, as developmental defects and abnormalities in this process can lead to many reproductive pathologies. SUMMARY This review offers a detailed overview of the latest advances in gonadotrope cell differentiation. We addressed this question with a specific focus on three important aspects of gonadotrope differentiation: the identification of the progenitor population giving rise to gonadotrope cells, the early mechanisms that initiate Nr5a1 expression and thus gonadotrope fate commitment, and finally, the mechanisms driving the formation of physical and functional gonadotrope networks. KEY MESSAGES Overall, this review aimed to provide new insights into three aspects of the gonadotrope differentiation process by reconsidering pioneering studies in the light of data gained from latest technological developments. Firstly, we re-investigated the long debated developmental trajectory of pituitary gonadotrope cells. Secondly, we reported new regulatory mechanisms of Nr5a1 expression, focusing on the involvement of ERα. Finally, we highlighted the molecular and cellular mechanisms driving gonadotrope network formation during embryogenesis, a process that seems essential for regulation of gonadotrope activity.
Collapse
Affiliation(s)
- Charles Le Ciclé
- Université Paris Cité, CNRS, Inserm, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Joëlle Cohen-Tannoudji
- Université Paris Cité, CNRS, Inserm, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - David L'Hôte
- Université Paris Cité, CNRS, Inserm, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| |
Collapse
|
5
|
Aguilar GR, Vidal B, Ji H, Evenblij J, Ji H, Valperga G, Liao CP, Fang-Yen C, Hobert O. Functional analysis of conserved C. elegans bHLH family members uncovers lifespan control by a peptidergic hub neuron. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.12.603289. [PMID: 39071424 PMCID: PMC11275782 DOI: 10.1101/2024.07.12.603289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Throughout the animal kingdom, several members of the basic helix-loop-helix (bHLH) family act as proneural genes during early steps of nervous system development. Roles of bHLH genes in specifying terminal differentiation of postmitotic neurons have been less extensively studied. We analyze here the function of five C. elegans bHLH genes, falling into three phylogenetically conserved subfamilies, which are continuously expressed in a very small number of postmitotic neurons in the central nervous system. We show (a) that two orthologs of the vertebrate bHLHb4/b5 genes, called hlh-17 and hlh-32, function redundantly to specify the identity of a single head interneuron (AUA), as well as an individual motor neuron (VB2), (b) that the PTF1a ortholog hlh-13 acts as a terminal selector to control terminal differentiation and function of the sole octopaminergic neuron class in C. elegans, RIC, and (c) that the NHLH1/2 ortholog hlh-15 controls terminal differentiation and function of the peptidergic AVK head interneuron class, a known neuropeptidergic signaling hub in the animal. Strikingly, through null mutant analysis and cell-specific rescue experiments, we find that loss of hlh-15/NHLH in the peptidergic AVK neurons and the resulting abrogation of neuropeptide secretion causes a substantially expanded lifespan of the animal, revealing an unanticipated impact of a central, peptidergic hub neuron in regulating lifespan, which we propose to be akin to hypothalamic control of lifespan in vertebrates. Taken together, our functional analysis reveals themes of bHLH gene function during terminal differentiation that are complementary to the earlier lineage specification roles of other bHLH family members. However, such late functions are much more sparsely employed by members of the bHLH transcription factor family, compared to the function of the much more broadly employed homeodomain transcription factor family.
Collapse
Affiliation(s)
- G. Robert Aguilar
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, NY
| | - Berta Vidal
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, NY
| | - Hongzhu Ji
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, NY
| | - Joke Evenblij
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, NY
- Technische Universität, Braunschweig, Germany
| | - Hongfei Ji
- Department of Biomedical Engineering, Ohio State University, Columbus, OH
| | - Giulio Valperga
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, NY
| | - Chien-Po Liao
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, NY
| | | | - Oliver Hobert
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, NY
| |
Collapse
|
6
|
Zhao J, Xu Y. PITX1 plays essential functions in cancer. Front Oncol 2023; 13:1253238. [PMID: 37841446 PMCID: PMC10570508 DOI: 10.3389/fonc.2023.1253238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 09/15/2023] [Indexed: 10/17/2023] Open
Abstract
PITX1, also known as the pituitary homeobox 1 gene, has emerged as a key regulator in animal growth and development, attracting significant research attention. Recent investigations have revealed the implication of dysregulated PITX1 expression in tumorigenesis, highlighting its involvement in cancer development. Notably, PITX1 interacts with p53 and exerts control over crucial cellular processes including cell cycle progression, apoptosis, and chemotherapy resistance. Its influence extends to various tumors, such as esophageal, colorectal, gastric, and liver cancer, contributing to tumor progression and metastasis. Despite its significance, a comprehensive review examining PITX1's role in oncology remains lacking. This review aims to address this gap by providing a comprehensive overview of PITX1 in different cancer types, with a particular focus on its clinicopathological significance.
Collapse
Affiliation(s)
- Jingpu Zhao
- Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, Shandong, China
| | - Yongfeng Xu
- Abdominal Oncology Ward, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
7
|
Le Ciclé C, Pacini V, Rama N, Tauszig-Delamasure S, Airaud E, Petit F, de Beco S, Cohen-Tannoudji J, L'hôte D. The Neurod1/4-Ntrk3-Src pathway regulates gonadotrope cell adhesion and motility. Cell Death Discov 2023; 9:327. [PMID: 37658038 PMCID: PMC10474047 DOI: 10.1038/s41420-023-01615-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 08/08/2023] [Accepted: 08/16/2023] [Indexed: 09/03/2023] Open
Abstract
Pituitary gonadotrope cells are essential for the endocrine regulation of reproduction in vertebrates. These cells emerge early during embryogenesis, colonize the pituitary glands and organize in tridimensional networks, which are believed to be crucial to ensure proper regulation of fertility. However, the molecular mechanisms regulating the organization of gonadotrope cell population during embryogenesis remain poorly understood. In this work, we characterized the target genes of NEUROD1 and NEUROD4 transcription factors in the immature gonadotrope αT3-1 cell model by in silico functional genomic analyses. We demonstrated that NEUROD1/4 regulate genes belonging to the focal adhesion pathway. Using CRISPR/Cas9 knock-out approaches, we established a double NEUROD1/4 knock-out αT3-1 cell model and demonstrated that NEUROD1/4 regulate cell adhesion and cell motility. We then characterized, by immuno-fluorescence, focal adhesion number and signaling in the context of NEUROD1/4 insufficiency. We demonstrated that NEUROD1/4 knock-out leads to an increase in the number of focal adhesions associated with signaling abnormalities implicating the c-Src kinase. We further showed that the neurotrophin tyrosine kinase receptor 3 NTRK3, a target of NEUROD1/4, interacts physically with c-Src. Furthermore, using motility rescue experiments and time-lapse video microscopy, we demonstrated that NTRK3 is a major regulator of gonadotrope cell motility. Finally, using a Ntrk3 knock-out mouse model, we showed that NTRK3 regulates gonadotrope cells positioning in the developing pituitary, in vivo. Altogether our study demonstrates that the Neurod1/4-Ntrk3-cSrc pathway is a major actor of gonadotrope cell mobility, and thus provides new insights in the regulation of gonadotrope cell organization within the pituitary gland.
Collapse
Affiliation(s)
- Charles Le Ciclé
- Université Paris Cité, CNRS, Inserm, Unité de Biologie Fonctionnelle et Adaptative, F-75013, Paris, France
| | - Vincent Pacini
- Université Paris Cité, CNRS, Inserm, Unité de Biologie Fonctionnelle et Adaptative, F-75013, Paris, France
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013, Paris, France
| | - Nicolas Rama
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard, Université Lyon1, 69008, Lyon, France
| | - Servane Tauszig-Delamasure
- Institut NeuroMyoGène - CNRS UMR 5310 - Inserm U1217 de Lyon - UCBL Lyon 1, Faculté de Médecine et de Pharmacie, Lyon, France
| | - Eloïse Airaud
- Université Paris Cité, CNRS, Inserm, Unité de Biologie Fonctionnelle et Adaptative, F-75013, Paris, France
| | - Florence Petit
- Université Paris Cité, CNRS, Inserm, Unité de Biologie Fonctionnelle et Adaptative, F-75013, Paris, France
- Faculty of Pharmacy, Université de Montréal, Montréal, QC, H3T 1J4, Canada
| | - Simon de Beco
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013, Paris, France
| | - Joëlle Cohen-Tannoudji
- Université Paris Cité, CNRS, Inserm, Unité de Biologie Fonctionnelle et Adaptative, F-75013, Paris, France
| | - David L'hôte
- Université Paris Cité, CNRS, Inserm, Unité de Biologie Fonctionnelle et Adaptative, F-75013, Paris, France.
| |
Collapse
|
8
|
Abstract
Endogenous Cushing's syndrome (CS) is associated with morbidities (diabetes, hypertension, clotting disorders) and shortens life because of infections, pulmonary thromboembolism, and cardiovascular disease. Its clinical presentation is immensely variable, and diagnosis and treatment are often delayed. Thus, there are many opportunities for basic and clinical research leading to better tests, faster diagnosis, and optimized medical treatments. This review focuses on CS caused by excessive adrenocorticotropin (ACTH) production. It describes current concepts of the regulation of ACTH synthesis and secretion by normal corticotropes and mechanisms by which dysregulation occurs in corticotrope (termed "Cushing's disease") and noncorticotrope (so-called ectopic) ACTH-producing tumors. ACTH causes adrenal gland synthesis and pulsatile release of cortisol; the excess ACTH in these forms of CS leads to the hypercortisolism of endogenous CS. Again, the differences between healthy individuals and those with CS are highlighted. The clinical presentations and their use in the interpretation of CS screening tests are described. The tests used for screening and differential diagnosis of CS are presented, along with their relationship to cortisol dynamics, pathophysiology, and negative glucocorticoid feedback regulation in the two forms of ACTH-dependent CS. Finally, several gaps in current understanding are highlighted in the hope of stimulating additional research into this challenging disorder.
Collapse
Affiliation(s)
- Lynnette K Nieman
- Diabetes, Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
9
|
Yao H, Xie W, Dai Y, Liu Y, Gu W, Li J, Wu L, Xie J, Rui W, Ren B, Xue L, Cheng Y, Lin S, Li C, Tang H, Wang Y, Lou M, Zhang X, Hu R, Shang H, Huang J, Wu ZB. TRIM65 determines the fate of a novel subtype of pituitary neuroendocrine tumors via ubiquitination and degradation of TPIT. Neuro Oncol 2022; 24:1286-1297. [PMID: 35218667 PMCID: PMC9340636 DOI: 10.1093/neuonc/noac053] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Pituitary neuroendocrine tumors (PitNETs) are common intracranial tumors that are classified into seven histological subtypes, including lactotroph, somatotroph, corticotroph, thyrotroph, gonadotroph, null cell, and plurihormonal PitNETs. However, the molecular characteristics of these types of PitNETs are not completely clear. METHODS A total of 180 consecutive cases of PitNETs were collected to perform RNA sequencing. All subtypes of PitNETs were distinguished by unsupervised clustering analysis. We investigated the regulation of TPIT by TRIM65 and its effects on ACTH production and secretion in ACTH-secreting pituitary cell lines, as well as in murine models using biochemical analyses, confocal microscopy, and luciferase reporter assays. RESULTS A novel subtype of PitNETs derived from TPIT lineage cells was identified as with normal TPIT transcription but with lowered protein expression. Furthermore, for the first time, TRIM65 was identified as the E3 ubiquitin ligase of TPIT. Depending on the RING domain, TRIM65 ubiquitinated and degraded the TPIT protein at multiple Lys sites. In addition, TRIM65-mediated ubiquitination of TPIT inhibited POMC transcription and ACTH production to determine the fate of the novel subtype of PitNETs in vitro and in vivo. CONCLUSION Our studies provided a novel classification of PitNETs and revealed that the TRIM65-TPIT complex controlled the fate of the novel subtype of PitNETs, which provides a potential therapy target for Cushing's disease.
Collapse
Affiliation(s)
- Hong Yao
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wanqun Xie
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuting Dai
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanting Liu
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiting Gu
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianfeng Li
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liang Wu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Xie
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiwei Rui
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bohan Ren
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Li Xue
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yijun Cheng
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shaojian Lin
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Changsheng Li
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hao Tang
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Wang
- Department of Neurosurgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Meiqing Lou
- Department of Neurosurgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaobiao Zhang
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ronggui Hu
- State Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai,China
| | - Hanbing Shang
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinyan Huang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhe Bao Wu
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
10
|
Drouin J. The corticotroph cells from early development to tumorigenesis. J Neuroendocrinol 2022; 34:e13147. [PMID: 35524583 DOI: 10.1111/jne.13147] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/06/2022] [Accepted: 04/15/2022] [Indexed: 11/27/2022]
Abstract
During development, highly specialized differentiated cells, such as pituitary secretory cells, acquire their identity and properties through a series of specification events exerted by transcription factors to implement a unique gene expression program and epigenomic state. The investigation of these developmental processes informs us on the unique features of a cell lineage, both to explain these features and also to outline where these processes may fail and cause disease. This review summarizes present knowledge on the developmental origin of pituitary corticotroph and melanotroph cells and on the underlying molecular mechanisms. At the onset, comparison of gene expression programs active in pituitary progenitors compared to those active in differentiated corticotrophs or melanotrophs indicated dramatic differences in the control of, for example, the cell cycle. Tpit is the transcription factor that determines terminal differentiation of pro-opiomelanocortin (POMC) lineages, both corticotrophs and melanotrophs, and its action involves this switch in cell cycle control in parallel with activation of cell-specific gene expression. There is thus far more to making a corticotroph cell than just activating transcription of the POMC gene. Indeed, Tpit also controls implementation of mechanisms for enhanced protein translation capacity and development of extensive secretory organelles. The corticotroph cell identity also includes mechanisms responsible for homotypic cell-cell interactions between corticotrophs and for privileged heterotypic cell interactions with pituitary cells of other lineages. The review also summarizes current knowledge on how a pioneer transcription factor, Pax7, remodels the epigenome such that the same determination transcription factor, Tpit, will implement the melanotroph program of gene expression. Finally, this canvas of regulatory mechanisms implementing POMC lineage identities constitutes the background to understand alterations that characterize corticotroph adenomas of Cushing's disease patients. The integration of all these data into a unified scheme will likely yield a scheme to globally understand pathogenic mechanisms in Cushing's disease.
Collapse
Affiliation(s)
- Jacques Drouin
- Institut de recherches cliniques de Montréal, Laboratoire de génétique moléculaire, Montréal, Québec, Canada
| |
Collapse
|
11
|
Sipani R, Joshi R. Hox genes collaborate with helix-loop-helix factor Grainyhead to promote neuroblast apoptosis along the anterior-posterior axis of the Drosophila larval central nervous system. Genetics 2022; 222:6632667. [DOI: 10.1093/genetics/iyac101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 06/21/2022] [Indexed: 11/14/2022] Open
Abstract
Abstract
Hox genes code for a family of a homeodomain (HD) containing transcription factors that use TALE-HD containing factors Pbx/Exd and Meis/Hth to specify the development of the anterior-posterior (AP) axis of an organism. However, the absence of TALE-HD containing factors from specific tissues emphasizes the need to identify and validate new Hox cofactors. In Drosophila central nervous system (CNS), Hox execute segment-specific apoptosis of neural stem cells (neuroblasts-NBs) and neurons. In abdominal segments of larval CNS, Hox gene Abdominal-A (AbdA) mediates NB apoptosis with the help of Exd and bHLH factor Grainyhead (Grh) using a 717 bp apoptotic enhancer. In this study, we show that this enhancer is critical for abdominal NB apoptosis and relies on two separable set of DNA binding motifs responsible for its initiation and maintenance. Our results also show that AbdA and Grh interact through their highly conserved DNA binding domains, and the DNA binding specificity of AbdA-HD is important for it to interact with Grh and essential for it to execute NB apoptosis in CNS. We also establish that Grh is required for Hox-dependent NB apoptosis in Labial and Sex Combs Reduced (Scr) expressing regions of the CNS, and it can physically interact with all the Hox proteins in vitro. Our biochemical and functional data collectively support the idea that Grh can function as a Hox cofactor and help them carry out their in vivo roles during development.
Collapse
Affiliation(s)
- Rashmi Sipani
- Laboratory of Drosophila Neural Development, Centre for DNA Fingerprinting and Diagnostics (CDFD) , Inner Ring Road, Uppal, Hyderabad-500039. India
- Graduate Studies, Manipal Academy of Higher Education , Manipal 576104, India
| | - Rohit Joshi
- Laboratory of Drosophila Neural Development, Centre for DNA Fingerprinting and Diagnostics (CDFD) , Inner Ring Road, Uppal, Hyderabad-500039. India
| |
Collapse
|
12
|
Singh A, Mahesh A, Noack F, Cardoso de Toledo B, Calegari F, Tiwari VK. Tcf12 and NeuroD1 cooperatively drive neuronal migration during cortical development. Development 2022; 149:dev200250. [PMID: 35147187 PMCID: PMC8918803 DOI: 10.1242/dev.200250] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 12/31/2021] [Indexed: 01/06/2023]
Abstract
Corticogenesis consists of a series of synchronised events, including fate transition of cortical progenitors, neuronal migration, specification and connectivity. NeuroD1, a basic helix-loop-helix (bHLH) transcription factor (TF), contributes to all of these events, but how it coordinates these independently is still unknown. Here, we demonstrate that NeuroD1 expression is accompanied by a gain of active chromatin at a large number of genomic loci. Interestingly, transcriptional activation of these loci relied on a high local density of adjacent bHLH TFs motifs, including, predominantly, Tcf12. We found that activity and expression levels of Tcf12 were high in cells with induced levels of NeuroD1 that spanned the transition of cortical progenitors from proliferative to neurogenic divisions. Moreover, Tcf12 forms a complex with NeuroD1 and co-occupies a subset of NeuroD1 target loci. This Tcf12-NeuroD1 cooperativity is essential for gaining active chromatin and targeted expression of genes involved in cell migration. By functional manipulation in vivo, we further show that Tcf12 is essential during cortical development for the correct migration of newborn neurons and, hence, for proper cortical lamination.
Collapse
Affiliation(s)
- Aditi Singh
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queens University Belfast, Belfast BT9 7BL, UK
| | - Arun Mahesh
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queens University Belfast, Belfast BT9 7BL, UK
| | - Florian Noack
- CRTD-Center for Regenerative Therapies, School of Medicine, Technische Universität Dresden, 01307 Dresden, Germany
| | - Beatriz Cardoso de Toledo
- CRTD-Center for Regenerative Therapies, School of Medicine, Technische Universität Dresden, 01307 Dresden, Germany
| | - Federico Calegari
- CRTD-Center for Regenerative Therapies, School of Medicine, Technische Universität Dresden, 01307 Dresden, Germany
| | - Vijay K. Tiwari
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queens University Belfast, Belfast BT9 7BL, UK
| |
Collapse
|
13
|
Mechanisms of Binding Specificity among bHLH Transcription Factors. Int J Mol Sci 2021; 22:ijms22179150. [PMID: 34502060 PMCID: PMC8431614 DOI: 10.3390/ijms22179150] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/14/2021] [Accepted: 08/18/2021] [Indexed: 12/25/2022] Open
Abstract
The transcriptome of every cell is orchestrated by the complex network of interaction between transcription factors (TFs) and their binding sites on DNA. Disruption of this network can result in many forms of organism malfunction but also can be the substrate of positive natural selection. However, understanding the specific determinants of each of these individual TF-DNA interactions is a challenging task as it requires integrating the multiple possible mechanisms by which a given TF ends up interacting with a specific genomic region. These mechanisms include DNA motif preferences, which can be determined by nucleotide sequence but also by DNA’s shape; post-translational modifications of the TF, such as phosphorylation; and dimerization partners and co-factors, which can mediate multiple forms of direct or indirect cooperative binding. Binding can also be affected by epigenetic modifications of putative target regions, including DNA methylation and nucleosome occupancy. In this review, we describe how all these mechanisms have a role and crosstalk in one specific family of TFs, the basic helix-loop-helix (bHLH), with a very conserved DNA binding domain and a similar DNA preferred motif, the E-box. Here, we compile and discuss a rich catalog of strategies used by bHLH to acquire TF-specific genome-wide landscapes of binding sites.
Collapse
|
14
|
Transcription Factors of the bHLH Family Delineate Vertebrate Landmarks in the Nervous System of a Simple Chordate. Genes (Basel) 2020; 11:genes11111262. [PMID: 33114624 PMCID: PMC7693978 DOI: 10.3390/genes11111262] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 10/12/2020] [Indexed: 02/07/2023] Open
Abstract
Tunicates are marine invertebrates whose tadpole-like larvae feature a highly simplified version of the chordate body plan. Similar to their distant vertebrate relatives, tunicate larvae develop a regionalized central nervous system and form distinct neural structures, which include a rostral sensory vesicle, a motor ganglion, and a caudal nerve cord. The sensory vesicle contains a photoreceptive complex and a statocyst, and based on the comparable expression patterns of evolutionarily conserved marker genes, it is believed to include proto-hypothalamic and proto-retinal territories. The evolutionarily conserved molecular fingerprints of these landmarks of the vertebrate brain consist of genes encoding for different transcription factors, and of the gene batteries that they control, and include several members of the bHLH family. Here we review the complement of bHLH genes present in the streamlined genome of the tunicate Ciona robusta and their current classification, and summarize recent studies on proneural bHLH transcription factors and their expression territories. We discuss the possible roles of bHLH genes in establishing the molecular compartmentalization of the enticing nervous system of this unassuming chordate.
Collapse
|
15
|
Noy EB, Watanabe Y, Grommen SVH, De Groef B. Transcriptional regulation of the chicken CRHR2 gene by pituitary transcription factors. Gen Comp Endocrinol 2019; 284:113263. [PMID: 31454497 DOI: 10.1016/j.ygcen.2019.113263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 08/15/2019] [Accepted: 08/23/2019] [Indexed: 11/22/2022]
Abstract
Corticotropin-releasing hormone (CRH) is known to act as a potent thyrotropin-releasing factor in non-mammalian species such as chicken and bullfrog. This interaction is mediated by type 2 CRH receptors (CRHR2) expressed by the thyrotropes in the pituitary gland. However, the response elements (REs) and their corresponding transcription factors (TFs) that control CRHR2 expression in thyrotropes are not known. Since thyrotrope-specific expression of the β-subunit of thyrotropin is synergistically stimulated by the co-expression of POU1F1 and GATA2, we hypothesised that in non-mammalian vertebrates like chicken, CRHR2 expression is controlled by the same TFs and that their REs are present in the chicken CRHR2 gene promoter. In situ hybridisation and immunohistochemistry suggest that chicken thyrotropes, like those of mammals, express the mRNAs for the TFs GATA2, POU1F1 and PITX1, but not NR5A1. Using luciferase reporter assays, we show that both GATA2 and PITX1 can activate the promoter of CRHR2, but PITX1 requires a functional GATA2 RE to be present. POU1F1 alone did not affect promoter activity, but synergistically increased the effect of GATA2. Promoter deletion analysis and mutagenesis showed that essential GATA2 and PITX1 REs are located between 116 and 198 bp upstream of the start codon. These REs are highly conserved in non-mammalian species. Additionally, NR5A1 (steroidogenic factor 1) suppressed both GATA2- and PITX1-induced promoter activity and may therefore play a role in restricting CRHR2 expression in gonadotropes. We conclude that the expression of CRHR2 in chicken thyrotropes is stimulated by GATA2 with interactions with POU1F1 and PITX1, in the absence of NR5A1.
Collapse
Affiliation(s)
- Ellyse B Noy
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria 3086, Australia.
| | - Yugo Watanabe
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria 3086, Australia.
| | - Sylvia V H Grommen
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria 3086, Australia.
| | - Bert De Groef
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria 3086, Australia.
| |
Collapse
|
16
|
Rajagopal N, Nangia S. Obtaining Protein Association Energy Landscape for Integral Membrane Proteins. J Chem Theory Comput 2019; 15:6444-6455. [DOI: 10.1021/acs.jctc.9b00626] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Nandhini Rajagopal
- Department of Biomedical and Chemical Engineering, Syracuse University, 343 Link Hall, Syracuse, New York 13244, United States
| | - Shikha Nangia
- Department of Biomedical and Chemical Engineering, Syracuse University, 343 Link Hall, Syracuse, New York 13244, United States
| |
Collapse
|
17
|
Edwards W, Raetzman LT. Complex integration of intrinsic and peripheral signaling is required for pituitary gland development. Biol Reprod 2019; 99:504-513. [PMID: 29757344 DOI: 10.1093/biolre/ioy081] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 04/03/2018] [Indexed: 12/17/2022] Open
Abstract
The coordination of pituitary development is complicated and requires input from multiple cellular processes. Recent research has provided insight into key molecular determinants that govern cell fate specification in the pituitary. Moreover, increasing research aimed to identify, characterize, and functionally describe the presumptive pituitary stem cell population has allowed for a better understanding of the processes that govern endocrine cell differentiation in the developing pituitary. The culmination of this research has led to the ability of investigators to recapitulate some of embryonic pituitary development in vitro, the first steps to developing novel regenerative therapies for pituitary diseases. In this current review, we cover the major players in pituitary stem/progenitor cell function and maintenance, and the key molecular determinants of endocrine cell specification. In addition, we discuss the contribution of peripheral hormonal regulation of pituitary gland development, an understudied area of research.
Collapse
Affiliation(s)
- Whitney Edwards
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Lori T Raetzman
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
18
|
Mayran A, Sochodolsky K, Khetchoumian K, Harris J, Gauthier Y, Bemmo A, Balsalobre A, Drouin J. Pioneer and nonpioneer factor cooperation drives lineage specific chromatin opening. Nat Commun 2019; 10:3807. [PMID: 31444346 PMCID: PMC6707328 DOI: 10.1038/s41467-019-11791-9] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 07/30/2019] [Indexed: 12/13/2022] Open
Abstract
Pioneer transcription factors are characterized by having the unique property of enabling the opening of closed chromatin sites, for implementation of cell fates. We previously found that the pioneer Pax7 specifies melanotrope cells through deployment of an enhancer repertoire, which allows binding of Tpit, a nonpioneer factor that determines the related lineages of melanotropes and corticotropes. Here, we investigate the relation between these two factors in the pioneer mechanism. Cell-specific gene expression and chromatin landscapes are defined by scRNAseq and chromatin accessibility profiling. We find that in vivo deployment of the melanotrope enhancer repertoire and chromatin opening requires both Pax7 and Tpit. In cells, binding of heterochromatin targets by Pax7 is independent of Tpit but Pax7-dependent chromatin opening requires Tpit. The present work shows that pioneer core properties are limited to the ability to recognize heterochromatin targets and facilitate nonpioneer binding. Chromatin opening per se may be provided through cooperation with nonpioneer factors.
Collapse
Affiliation(s)
- Alexandre Mayran
- Laboratoire de Génétique Moléculaire, Institut de Recherches Cliniques de Montréal (IRCM), Montreal, QC, Canada
- Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Kevin Sochodolsky
- Laboratoire de Génétique Moléculaire, Institut de Recherches Cliniques de Montréal (IRCM), Montreal, QC, Canada
| | - Konstantin Khetchoumian
- Laboratoire de Génétique Moléculaire, Institut de Recherches Cliniques de Montréal (IRCM), Montreal, QC, Canada
| | - Juliette Harris
- Laboratoire de Génétique Moléculaire, Institut de Recherches Cliniques de Montréal (IRCM), Montreal, QC, Canada
| | - Yves Gauthier
- Laboratoire de Génétique Moléculaire, Institut de Recherches Cliniques de Montréal (IRCM), Montreal, QC, Canada
| | - Amandine Bemmo
- Laboratoire de Génétique Moléculaire, Institut de Recherches Cliniques de Montréal (IRCM), Montreal, QC, Canada
| | - Aurelio Balsalobre
- Laboratoire de Génétique Moléculaire, Institut de Recherches Cliniques de Montréal (IRCM), Montreal, QC, Canada
| | - Jacques Drouin
- Laboratoire de Génétique Moléculaire, Institut de Recherches Cliniques de Montréal (IRCM), Montreal, QC, Canada.
- Department of Biochemistry, McGill University, Montreal, QC, Canada.
- Département de Biochimie, Université de Montréal, Montreal, QC, Canada.
| |
Collapse
|
19
|
Zhao H, Zhang S, Wu X, Pan C, Li X, Lei C, Chen H, Lan X. DNA methylation pattern of the goat PITX1 gene and its effects on milk performance. Arch Anim Breed 2019; 62:59-68. [PMID: 31807614 PMCID: PMC6852879 DOI: 10.5194/aab-62-59-2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 01/24/2019] [Indexed: 12/23/2022] Open
Abstract
Paired-like homeodomain transcription factor 1 (PITX1) is a pivotal
gene in the hypothalamic–pituitary–adrenal axis, which is a well-known
pathway affecting lactation performance. The aim of this study was to analyze
the DNA methylation profile of the PITX1 gene and its relevance to
milk performance in Xinong Saanen dairy goats; thus, potential epigenetic
markers of lactation performance were identified. A total of 267 goat blood
samples were divided into “low” and “high” groups according to two milk
traits: the average milk yield (AMY) and the average milk density (AMD). One
CpG island in the 3′-flanking region of the PITX1 gene was
identified as being related to milk performance. Fisher's exact test
demonstrated that the methylation rates of the overall CpG island and the 3rd
and 12th CpG-dinucleotide loci in the blood were significantly associated
with the AMY, and the overall methylation rate of the high AMY group was
relative hypomethylation compared with the low AMY group. The overall
methylation rates of this CpG island in mammary gland tissue from dry and
lactation periods again exhibited a significant difference: the lactation
period showed relative hypomethylation compared with the dry period.
Bioinformatic transcription factor binding site prediction identified some
lactation performance related transcription factors in this CpG island, such
as CTCF, STAT, SMAD, CDEF, SP1, and KLFS. Briefly, overall methylation
changes of the CpG island in the PITX1 gene are relevant to
lactation performance, which will be valuable for future studies and
epigenetic marker-assisted selection (eMAS) in the breeding of goats with
respect to lactation performance.
Collapse
Affiliation(s)
- Haiyu Zhao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Sihuan Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Xianfeng Wu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Chuanying Pan
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Xiangchen Li
- Institute of Beijing Animal Science and Veterinary, Chinese Academy of Agricultural Science, Beijing 100194, China
| | - Chuzhao Lei
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Hong Chen
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Xianyong Lan
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| |
Collapse
|
20
|
Khandelwal R, Sipani R, Govinda Rajan S, Kumar R, Joshi R. Combinatorial action of Grainyhead, Extradenticle and Notch in regulating Hox mediated apoptosis in Drosophila larval CNS. PLoS Genet 2017; 13:e1007043. [PMID: 29023471 PMCID: PMC5667929 DOI: 10.1371/journal.pgen.1007043] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Revised: 11/02/2017] [Accepted: 09/26/2017] [Indexed: 01/09/2023] Open
Abstract
Hox mediated neuroblast apoptosis is a prevalent way to pattern larval central nervous system (CNS) by different Hox genes, but the mechanism of this apoptosis is not understood. Our studies with Abdominal-A (Abd-A) mediated larval neuroblast (pNB) apoptosis suggests that AbdA, its cofactor Extradenticle (Exd), a helix-loop-helix transcription factor Grainyhead (Grh), and Notch signaling transcriptionally contribute to expression of RHG family of apoptotic genes. We find that Grh, AbdA, and Exd function together at multiple motifs on the apoptotic enhancer. In vivo mutagenesis of these motifs suggest that they are important for the maintenance of the activity of the enhancer rather than its initiation. We also find that Exd function is independent of its known partner homothorax in this apoptosis. We extend some of our findings to Deformed expressing region of sub-esophageal ganglia where pNBs undergo a similar Hox dependent apoptosis. We propose a mechanism where common players like Exd-Grh-Notch work with different Hox genes through region specific enhancers to pattern respective segments of larval central nervous system. Specification of the head to tail axis of the developing central nervous system is carried out by Hox genes. Hox mediated programmed cell death of the neural progenitor cells plays an important role in specification of this axis, but the molecular mechanism of this phenomenon is not well understood. We have studied this phenomenon in abdominal and subesophageal regions of larval central nervous system of Drosophila. We find that different Hox genes use a combination of common players (Extradenticle, Grainyhead and Notch) but employ region specific enhancers to cause progenitor cell death in different segments of developing central nervous system.
Collapse
Affiliation(s)
- Risha Khandelwal
- Laboratory of Drosophila Neural Development, Centre for DNA Fingerprinting and Diagnostics (CDFD), Tuljaguda Complex, Nampally, Hyderabad, India
- Graduate Studies, Manipal University, Manipal, India
| | - Rashmi Sipani
- Laboratory of Drosophila Neural Development, Centre for DNA Fingerprinting and Diagnostics (CDFD), Tuljaguda Complex, Nampally, Hyderabad, India
- Graduate Studies, Manipal University, Manipal, India
| | - Sriivatsan Govinda Rajan
- Laboratory of Drosophila Neural Development, Centre for DNA Fingerprinting and Diagnostics (CDFD), Tuljaguda Complex, Nampally, Hyderabad, India
| | - Raviranjan Kumar
- Laboratory of Drosophila Neural Development, Centre for DNA Fingerprinting and Diagnostics (CDFD), Tuljaguda Complex, Nampally, Hyderabad, India
- Graduate Studies, Manipal University, Manipal, India
| | - Rohit Joshi
- Laboratory of Drosophila Neural Development, Centre for DNA Fingerprinting and Diagnostics (CDFD), Tuljaguda Complex, Nampally, Hyderabad, India
- * E-mail: ,
| |
Collapse
|
21
|
Parvin R, Saito-Hakoda A, Shimada H, Shimizu K, Noro E, Iwasaki Y, Fujiwara K, Yokoyama A, Sugawara A. Role of NeuroD1 on the negative regulation of Pomc expression by glucocorticoid. PLoS One 2017; 12:e0175435. [PMID: 28406939 PMCID: PMC5391015 DOI: 10.1371/journal.pone.0175435] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 03/24/2017] [Indexed: 01/20/2023] Open
Abstract
The mechanism of the negative regulation of proopiomelanocortin gene (Pomc) by glucocorticoids (Gcs) is still unclear in many points. Here, we demonstrated the involvement of neurogenic differentiation factor 1 (NeuroD1) in the Gc-mediated negative regulation of Pomc. Murine pituitary adrenocorticotropic hormone (ACTH) producing corticotroph tumor-derived AtT20 cells were treated with dexamethasone (DEX) (1-100 nM) and cultured for 24 hrs. Thereafter, Pomc mRNA expression was studied by quantitative real-time PCR and rat Pomc promoter (-703/+58) activity was examined by luciferase assay. Both Pomc mRNA expression and Pomc promoter activity were inhibited by DEX in a dose-dependent manner. Deletion and point mutant analyses of Pomc promoter suggested that the DEX-mediated transcriptional repression was mediated via E-box that exists at -376/-371 in the promoter. Since NeuroD1 is known to bind to and activate E-box of the Pomc promoter, we next examined the effect of DEX on NeuroD1 expression. Interestingly, DEX dose-dependently inhibited NeuroD1 mRNA expression, mouse NeuroD1 promoter (-2.2-kb) activity, and NeuroD1 protein expression in AtT20 cells. In addition, we confirmed the inhibitory effect of DEX on the interaction of NeuroD1 and E-box on Pomc promoter by chromatin immunoprecipitation (ChIP) assay. Finally, overexpression of mouse NeuroD1 could rescue the DEX-mediated inhibition of Pomc mRNA expression and Pomc promoter activity. Taken together, it is suggested that the suppression of NeuroD1 expression and the inhibition of NeuroD1/E-box interaction may play an important role in the Gc-mediated negative regulation of Pomc.
Collapse
Affiliation(s)
- Rehana Parvin
- Department of Molecular Endocrinology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Akiko Saito-Hakoda
- Department of Molecular Endocrinology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Hiroki Shimada
- Department of Molecular Endocrinology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Kyoko Shimizu
- Department of Molecular Endocrinology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Erika Noro
- Department of Molecular Endocrinology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | | | - Ken Fujiwara
- Division of Histology and Cell Biology, Department of Anatomy, Jichi Medical University School of Medicine, Tochigi, Japan
| | - Atsushi Yokoyama
- Department of Molecular Endocrinology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Akira Sugawara
- Department of Molecular Endocrinology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- * E-mail:
| |
Collapse
|
22
|
Holmes EE, Goltz D, Sailer V, Jung M, Meller S, Uhl B, Dietrich J, Röhler M, Ellinger J, Kristiansen G, Dietrich D. PITX3 promoter methylation is a prognostic biomarker for biochemical recurrence-free survival in prostate cancer patients after radical prostatectomy. Clin Epigenetics 2016; 8:104. [PMID: 27708722 PMCID: PMC5037587 DOI: 10.1186/s13148-016-0270-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 09/16/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Molecular biomarkers that might help to distinguish between more aggressive and clinically insignificant prostate cancers (PCa) are still urgently needed. Aberrant DNA methylation as a common molecular alteration in PCa seems to be a promising source for such biomarkers. In this study, PITX3 DNA methylation (mPITX3) and its potential role as a prognostic biomarker were investigated. Furthermore, mPITX3 was analyzed in combination with the established PCa methylation biomarker PITX2 (mPITX2). METHODS mPITX3 and mPITX2 were assessed by a quantitative real-time PCR and by means of the Infinium HumanMethylation450 BeadChip. BeadChip data were obtained from The Cancer Genome Atlas (TCGA) Research Network. DNA methylation differences between normal adjacent, benign hyperplastic, and carcinomatous prostate tissues were examined in the TCGA dataset as well as in prostatectomy specimens from the University Hospital Bonn. Retrospective analyses of biochemical recurrence (BCR) were conducted in a training cohort (n = 498) from the TCGA and an independent validation cohort (n = 300) from the University Hospital Bonn. All patients received radical prostatectomy. RESULTS In PCa tissue, mPITX3 was increased significantly compared to normal and benign hyperplastic tissue. In univariate Cox proportional hazards analyses, mPITX3 showed a significant prognostic value for BCR (training cohort: hazard ratio (HR) = 1.83 (95 % CI 1.07-3.11), p = 0.027; validation cohort: HR = 2.56 (95 % CI 1.44-4.54), p = 0.001). A combined evaluation with PITX2 methylation further revealed that hypermethylation of a single PITX gene member (either PITX2 or PITX3) identifies an intermediate risk group. CONCLUSIONS PITX3 DNA methylation alone and in combination with PITX2 is a promising biomarker for the risk stratification of PCa patients and adds relevant prognostic information to common clinically implemented parameters. Further studies are required to determine whether the results are transferable to a biopsy-based patient cohort. Trial registration: Patients for this unregistered study were enrolled retrospectively.
Collapse
Affiliation(s)
- Emily Eva Holmes
- Institute of Pathology, University Hospital Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | - Diane Goltz
- Institute of Pathology, University Hospital Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | - Verena Sailer
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine of Cornell University, New York, NY USA
- Englander Institute for Precision Medicine, Weill Cornell Medicine of Cornell University, New York, NY USA
| | - Maria Jung
- Institute of Pathology, University Hospital Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | - Sebastian Meller
- Institute of Pathology, University Hospital Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | - Barbara Uhl
- Institute of Pathology, University Hospital Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | - Jörn Dietrich
- Department of Otolaryngology, Head and Neck Surgery, University Hospital Bonn, Bonn, Germany
| | - Magda Röhler
- Institute of Pathology, University Hospital Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | - Jörg Ellinger
- Department of Urology, University Hospital Bonn, Bonn, Germany
| | - Glen Kristiansen
- Institute of Pathology, University Hospital Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | - Dimo Dietrich
- Institute of Pathology, University Hospital Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
- Department of Otolaryngology, Head and Neck Surgery, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
23
|
Bifsha P, Balsalobre A, Drouin J. Specificity of Pitx3-Dependent Gene Regulatory Networks in Subsets of Midbrain Dopamine Neurons. Mol Neurobiol 2016; 54:4921-4935. [PMID: 27514757 DOI: 10.1007/s12035-016-0040-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 08/05/2016] [Indexed: 01/16/2023]
Abstract
Dysfunction of midbrain dopaminergic (mDA) neurons is involved in Parkinson's disease (PD) and neuropsychiatric disorders. Pitx3 is expressed in mDA neuron subsets of the substantia nigra compacta (SNc) and of the ventral tegmental area (VTA) that are degeneration-sensitive in PD. The genetic network(s) and mode(s) of action of Pitx3 in these mDA neurons remain poorly characterized. We hypothesized that, given their distinct neuronal identities, Pitx3-expressing neurons of SNc and VTA should differ in their Pitx3-controlled gene expression networks and this may involve subset-specific co-regulators. Expression profiling of purified mDA neuronal subsets indicates that Pitx3 regulates different sets of genes in SNc and VTA, such as activating the expression of primary cilium gene products specifically in VTA neurons. Interaction network analysis pointed to the participation of differentially expressed Lhx/Lmo family members in the modulation of Pitx3 action in SNc and VTA mDA neurons. Conversely, global binding patterns of Pitx3 on genomic DNA of human dopaminergic cells revealed that Pitx3 is often co-recruited to regions that foster the formation of GATA-bHLH-BRN complexes, which usually involve Lmo co-regulatory proteins. We focused on Lmo3 for its preferential expression in SNc neurons and demonstrated that it functions as a transcriptional co-activator of Pitx3 by enhancing its activity on genomic regulatory elements. In summary, we defined the SN and VTA-specific programs of Pitx3-dependent gene expression and identified Lmo3 as a SN-specific co-regulator of Pitx3-dependent transcription.
Collapse
Affiliation(s)
- Panojot Bifsha
- Laboratoire de génétique moléculaire, Institut de recherches cliniques de Montréal (IRCM), 110, avenue des Pins Ouest, Montréal, Québec, H2W 1R7, Canada.,Division of Experimental Medicine, McGill University, Montréal, Quebec, H3A 1A3, Canada
| | - Aurelio Balsalobre
- Laboratoire de génétique moléculaire, Institut de recherches cliniques de Montréal (IRCM), 110, avenue des Pins Ouest, Montréal, Québec, H2W 1R7, Canada
| | - Jacques Drouin
- Laboratoire de génétique moléculaire, Institut de recherches cliniques de Montréal (IRCM), 110, avenue des Pins Ouest, Montréal, Québec, H2W 1R7, Canada. .,Division of Experimental Medicine, McGill University, Montréal, Quebec, H3A 1A3, Canada.
| |
Collapse
|
24
|
Drouin J. 60 YEARS OF POMC: Transcriptional and epigenetic regulation of POMC gene expression. J Mol Endocrinol 2016; 56:T99-T112. [PMID: 26792828 DOI: 10.1530/jme-15-0289] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 01/12/2016] [Indexed: 01/31/2023]
Abstract
Expression of the pro-opiomelanocortin (POMC) gene integrates numerous inputs that reflect the developmental history of POMC-expressing cells of the pituitary and hypothalamus, as well as their critical role in the endocrine system. These inputs are integrated at specific regulatory sequences within the promoter and pituitary or hypothalamic enhancers of the POMC locus. Investigations of developmental mechanisms and transcription factors (TFs) responsible for pituitary activation of POMC transcription led to the discovery of the Pitx factors that have critical roles in pituitary development and striking patterning functions in embryonic development. Terminal differentiation of the two pituitary POMC lineages, the corticotrophs and melanotrophs, is controlled by Tpit; mutations of the human TPIT gene cause isolated adrenocorticotrophic hormone deficiency. Intermediate lobe and melanotroph identity is provided by the pioneer TF Pax7 that remodels chromatin to reveal a new repertoire of enhancers for Tpit action. Many signaling pathways regulate POMC transcription including activation by hypothalamic corticotrophin-releasing hormone acting through the orphan nuclear receptors of the Nur family and feedback repression by glucocorticoids and their glucocorticoid receptor. TFs of the basic helix-loop-helix, Smad, Stat, Etv, and nuclear factor-B families also mediate signals for control of POMC transcription. Whereas most of these regulatory processes are conserved in different species, there are also notable differences between specific targets for regulation of the human compared with mouse POMC genes.
Collapse
Affiliation(s)
- Jacques Drouin
- Laboratoire de génétique moléculaireInstitut de recherches cliniques de Montréal (IRCM), Montréal, Québec, Canada
| |
Collapse
|
25
|
Pataskar A, Jung J, Smialowski P, Noack F, Calegari F, Straub T, Tiwari VK. NeuroD1 reprograms chromatin and transcription factor landscapes to induce the neuronal program. EMBO J 2015; 35:24-45. [PMID: 26516211 DOI: 10.15252/embj.201591206] [Citation(s) in RCA: 195] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 09/14/2015] [Indexed: 12/30/2022] Open
Abstract
Cell fate specification relies on the action of critical transcription factors that become available at distinct stages of embryonic development. One such factor is NeuroD1, which is essential for eliciting the neuronal development program and possesses the ability to reprogram other cell types into neurons. Given this capacity, it is important to understand its targets and the mechanism underlying neuronal specification. Here, we show that NeuroD1 directly binds regulatory elements of neuronal genes that are developmentally silenced by epigenetic mechanisms. This targeting is sufficient to initiate events that confer transcriptional competence, including reprogramming of transcription factor landscape, conversion of heterochromatin to euchromatin, and increased chromatin accessibility, indicating potential pioneer factor ability of NeuroD1. The transcriptional induction of neuronal fate genes is maintained via epigenetic memory despite a transient NeuroD1 induction during neurogenesis. NeuroD1 also induces genes involved in the epithelial-to-mesenchymal transition, thereby promoting neuronal migration. Our study not only reveals the NeuroD1-dependent gene regulatory program driving neurogenesis but also increases our understanding of how cell fate specification during development involves a concerted action of transcription factors and epigenetic mechanisms.
Collapse
Affiliation(s)
| | - Johannes Jung
- Institute of Molecular Biology (IMB), Mainz, Germany
| | - Pawel Smialowski
- Adolf Butenandt Institute and Center for Integrated Protein Science, Ludwig Maximilian University, Munich, Germany
| | - Florian Noack
- DFG-Research Center for Regenerative Therapies, Cluster of Excellence, TU-Dresden, Dresden, Germany
| | - Federico Calegari
- DFG-Research Center for Regenerative Therapies, Cluster of Excellence, TU-Dresden, Dresden, Germany
| | - Tobias Straub
- Adolf Butenandt Institute and Center for Integrated Protein Science, Ludwig Maximilian University, Munich, Germany
| | | |
Collapse
|
26
|
Mudie S, Bandarra D, Batie M, Biddlestone J, Moniz S, Ortmann B, Shmakova A, Rocha S. PITX1, a specificity determinant in the HIF-1α-mediated transcriptional response to hypoxia. Cell Cycle 2015; 13:3878-91. [PMID: 25558831 PMCID: PMC4614811 DOI: 10.4161/15384101.2014.972889] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Hypoxia is an important developmental cue for multicellular organisms but it is also a contributing factor for several human pathologies, such as stroke, cardiovascular diseases and cancer. In cells, hypoxia activates a major transcriptional program coordinated by the Hypoxia Inducible Factor (HIF) family. HIF can activate more than one hundred targets but not all of them are activated at the same time, and there is considerable cell type variability. In this report we identified the paired-like homeodomain pituitary transcription factor (PITX1), as a transcription factor that helps promote specificity in HIF-1α dependent target gene activation. Mechanistically, PITX1 associates with HIF-1β and it is important for the induction of certain HIF-1 dependent genes but not all. In particular, PITX1 controls the HIF-1α-dependent expression of the histone demethylases; JMJD2B, JMJD2A, JMJD2C and JMJD1B. Functionally, PITX1 is required for the survival and proliferation responses in hypoxia, as PITX1 depleted cells have higher levels of apoptotic markers and reduced proliferation. Overall, our study identified PITX1 as a key specificity factor in HIF-1α dependent responses, suggesting PITX1 as a protein to target in hypoxic cancers.
Collapse
Affiliation(s)
- Sharon Mudie
- a Centre for Gene Regulation and Expression; College of Life Sciences ; University of Dundee ; Dundee , UK
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Can the ‘neuron theory’ be complemented by a universal mechanism for generic neuronal differentiation. Cell Tissue Res 2014; 359:343-84. [DOI: 10.1007/s00441-014-2049-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 10/23/2014] [Indexed: 12/19/2022]
|
28
|
An amino terminal phosphorylation motif regulates intranuclear compartmentalization of Olig2 in neural progenitor cells. J Neurosci 2014; 34:8507-18. [PMID: 24948806 DOI: 10.1523/jneurosci.0309-14.2014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The bHLH transcription factor Olig2 is expressed in cycling neural progenitor cells but also in terminally differentiated, myelinating oligodendrocytes. Sustained expression of Olig2 is counterintuitive because all known functions of the protein in expansion of neural progenitors and specification of oligodendrocyte progenitors are completed with the formation of mature white matter. How are the biological functions of Olig2 suppressed in terminally differentiated oligodendrocytes? In previous studies, we have shown that a triple serine motif in the amino terminus of Olig2 is phosphorylated in cycling neural progenitors but not in their differentiated progeny. We now show that phosphorylation of the triple serine motif regulates intranuclear compartmentalization of murine Olig2. Phosphorylated Olig2 is preferentially localized to a transcriptionally active "open" chromatin compartment together with coregulator proteins essential for regulation of gene expression. Unphosphorylated Olig2, as seen in mature white matter, is localized mainly within a transcriptionally inactive, chromatin fraction characterized by condensed and inaccessible DNA. Of special note is the observation that the p53 tumor suppressor protein is confined to the open chromatin fraction. Proximity ligation assays show that phosphorylation brings Olig2 within 30 nm of p53 within the open chromatin compartment. The data thus shed light on previously noted promitogenic functions of phosphorylated Olig2, which reflect, at least in part, an oppositional relationship with p53 functions.
Collapse
|
29
|
|
30
|
Uruno A, Saito-Hakoda A, Yokoyama A, Kogure N, Matsuda K, Parvin R, Shimizu K, Sato I, Kudo M, Yoshikawa T, Kagechika H, Iwasaki Y, Ito S, Sugawara A. Retinoic acid receptor-α up-regulates proopiomelanocortin gene expression in AtT20 corticotroph cells. Endocr J 2014; 61:1105-14. [PMID: 25132258 DOI: 10.1507/endocrj.ej14-0115] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Cushing's disease is a disorder caused by excessive ACTH secretion from a corticotroph tumor of the pituitary gland. Although its standard therapy is a transsphenoidal surgery, innovation of novel medical treatments for the disease is urgently necessary. Retinoic acid (RA) has been reported to suppress adrenocorticotropic hormone (ACTH) secretion in Cushing's disease. However, the role of RA receptor (RAR) in proopiomelanocortin (Pomc) gene expression remains uncertain. We here examined the involvement of RARα in Pomc regulation using AtT20 corticotroph cells. Surprisingly, a synthetic RARα agonist Am80 increased Pomc mRNA expression, CRH-induced ACTH secretion, and Pomc promoter activity. Small interfering RNA-mediated RARα-knockdown suppressed both basal and Am80-induced Pomc promoter activity. RARα-overexpression dose-dependently increased Pomc promoter activity. Pomc promoter mutation analysis revealed that both Tpit and NeuroD1 binding elements were responsible for the Am80-mediated effect. Am80 increased Tpit expression while RAR antagonist LE540 suppressed the increase. Tpit-overexpression increased Pomc promoter activity. Mammalian two-hybrid assay revealed that Am80 induced NeuroD1-RARα interaction. NeuroD1-overexpression enhanced the Am80-induced Pomc promoter activity, which was suppressed by NeuroD1 truncated mutant-overexpression. RARα thus positively regulates ACTH secretion/Pomc gene expression through interaction with NeuroD1 and Tpit expression increase. The present observation will be useful for the future development of the RA/retinoid-derived therapeutics of the disease.
Collapse
Affiliation(s)
- Akira Uruno
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Knopp P, Figeac N, Fortier M, Moyle L, Zammit PS. Pitx genes are redeployed in adult myogenesis where they can act to promote myogenic differentiation in muscle satellite cells. Dev Biol 2013; 377:293-304. [PMID: 23438814 DOI: 10.1016/j.ydbio.2013.02.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Revised: 01/29/2013] [Accepted: 02/15/2013] [Indexed: 11/17/2022]
Abstract
Skeletal muscle retains a resident stem cell population called satellite cells. Although mitotically quiescent in mature muscle, satellite cells can be activated to produce myoblast progeny to generate myonuclei for skeletal muscle homoeostasis, hypertrophy and repair. Regulation of satellite cell function in adult requires redeployment of many of the regulatory networks fundamental to developmental myogenesis. Involved in such control of muscle stem cell fate in embryos are members of the Pitx gene family of bicoid-class homeodomain proteins. Here, we investigated the expression and function of all three Pitx genes in muscle satellite cells of adult mice. Endogenous Pitx1 was undetectable, whilst Pitx2a, Pitx2b and Pitx2c were at low levels in proliferating satellite cells, but increased during the early stages of myogenic differentiation. By contrast, proliferating satellite cells expressed robust amounts of Pitx3, with levels then decreasing as cells differentiated, although Pitx3 remained expressed in unfused myoblasts. To examine the role of Pitx genes in satellite cell function, retroviral-mediated expression of Pitx1, all Pitx2 isoforms or Pitx3, was used. Constitutive expression of any Pitx isoform suppressed satellite cell proliferation, with the cells undergoing enhanced myogenic differentiation. Conversely, myogenic differentiation into multinucleated myotubes was decreased when Pitx2 or Pitx3 levels were reduced using siRNA. Together, our results show that Pitx genes play a role in regulating satellite cell function during myogenesis in adult.
Collapse
Affiliation(s)
- Paul Knopp
- King's College London, Randall Division of Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, London, SE1 1UL, England, UK
| | | | | | | | | |
Collapse
|
32
|
Separated at birth? The functional and molecular divergence of OLIG1 and OLIG2. Nat Rev Neurosci 2013; 13:819-31. [PMID: 23165259 DOI: 10.1038/nrn3386] [Citation(s) in RCA: 135] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The basic helix-loop-helix transcription factors oligodendrocyte transcription factor 1 (OLIG1) and OLIG2 are structurally similar and, to a first approximation, coordinately expressed in the developing CNS and postnatal brain. Despite these similarities, it was apparent from early on after their discovery that OLIG1 and OLIG2 have non-overlapping developmental functions in patterning, neuron subtype specification and the formation of oligodendrocytes. Here, we summarize more recent insights into the separate roles of these transcription factors in the postnatal brain during repair processes and in neurological disease states, including multiple sclerosis and malignant glioma. We discuss how the unique functions of OLIG1 and OLIG2 may reflect their distinct genetic targets, co-regulator proteins and/or post-translational modifications.
Collapse
|
33
|
Proopiomelanocortin (POMC) expression and conditioned place aversion during protracted withdrawal from chronic intermittent escalating-dose heroin in POMC-EGFP promoter transgenic mice. Neuroscience 2013; 236:220-32. [PMID: 23337531 DOI: 10.1016/j.neuroscience.2012.12.071] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Revised: 12/07/2012] [Accepted: 12/27/2012] [Indexed: 01/31/2023]
Abstract
In heroin-dependent individuals, the drive to avoid or ameliorate the negative affective/emotional state associated with the discontinuation of heroin contributes to the chronic relapsing nature of the disease. Here, we investigate changes in proopiomelanocortin (POMC) expression at three time points across an extended period of heroin withdrawal in a clinically relevant rodent model of addiction using conditioned place aversion (CPA) in POMC-EGFP (POMC-enhanced green fluorescent protein) bacterial artificial chromosome (BAC) transgenic mice. Neurons expressing POMC-EGFP were found in the medial nucleus of the amygdala (MeA), basomedial amygdala (BMA) and dentate gyrus of hippocampus (DG), as well as the arcuate nucleus of hypothalamus (ARC). Heroin-treated mice displayed robust CPA after acute spontaneous withdrawal (12h), which persisted across the extended (14days) withdrawal period. After 12-h withdrawal, heroin-treated mice showed lower signal intensity of POMC-EGFP-positive cells in the ARC, higher levels of POMC mRNA in the amygdala but lower levels in the hippocampus than saline controls. After 7-d withdrawal, heroin-treated mice showed fewer POMC-EGFP-positive cells in the MeA and lower POMC mRNA in the amygdala than saline controls. After extended (14days) withdrawal, heroin-treated mice showed more POMC-EGFP-positive cells in BMA and DG, increased intensity of POMC-EGFP signal in DG, and higher POMC mRNA levels in the hippocampus compared to controls. Our results show dynamic changes in POMC in hypothalamic and extra-hypothalamic regions that may contribute to the negative affective/emotional state of heroin withdrawal shown by CPA from acute to extended periods of heroin withdrawal.
Collapse
|
34
|
Jumlongras D, Lachke SA, O’Connell DJ, Aboukhalil A, Li X, Choe SE, Ho JWK, Turbe-Doan A, Robertson EA, Olsen BR, Bulyk ML, Amendt BA, Maas RL. An evolutionarily conserved enhancer regulates Bmp4 expression in developing incisor and limb bud. PLoS One 2012; 7:e38568. [PMID: 22701669 PMCID: PMC3373496 DOI: 10.1371/journal.pone.0038568] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Accepted: 05/07/2012] [Indexed: 12/02/2022] Open
Abstract
To elucidate the transcriptional regulation of Bmp4 expression during organogenesis, we used phylogenetic footprinting and transgenic reporter analyses to identify Bmp4 cis-regulatory modules (CRMs). These analyses identified a regulatory region located ∼46 kb upstream of the mouse Bmp4 transcription start site that had previously been shown to direct expression in lateral plate mesoderm. We refined this regulatory region to a 396-bp minimal enhancer, and show that it recapitulates features of endogenous Bmp4 expression in developing mandibular arch ectoderm and incisor epithelium during the initiation-stage of tooth development. In addition, this enhancer directs expression in the apical ectodermal ridge (AER) of the developing limb and in anterior and posterior limb mesenchyme. Transcript profiling of E11.5 mouse incisor dental lamina, together with protein binding microarray (PBM) analyses, allowed identification of a conserved DNA binding motif in the Bmp4 enhancer for Pitx homeoproteins, which are also expressed in the developing mandibular and incisor epithelium. In vitro electrophoretic mobility shift assays (EMSA) and in vivo transgenic reporter mutational analyses revealed that this site supports Pitx binding and that the site is necessary to recapitulate aspects of endogenous Bmp4 expression in developing craniofacial and limb tissues. Finally, Pitx2 chromatin immunoprecipitation (ChIP) demonstrated direct binding of Pitx2 to this Bmp4 enhancer site in a dental epithelial cell line. These results establish a direct molecular regulatory link between Pitx family members and Bmp4 gene expression in developing incisor epithelium.
Collapse
Affiliation(s)
- Dolrudee Jumlongras
- Division of Genetics, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Boston Massachusetts, United States of America
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts, United States of America
| | - Salil A. Lachke
- Department of Biological Sciences, Center for Bioinformatics and Computational Biology, University of Delaware, Newark, Delaware, United States of America
| | - Daniel J. O’Connell
- Division of Genetics, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Boston Massachusetts, United States of America
| | - Anton Aboukhalil
- Division of Genetics, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Boston Massachusetts, United States of America
- Department of Aeronautics and Astronautics, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts, United States of America
| | - Xiao Li
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, United States of America
| | - Sung E. Choe
- Division of Genetics, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Boston Massachusetts, United States of America
| | - Joshua W. K. Ho
- Division of Genetics, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Boston Massachusetts, United States of America
- Center for Biomedical Informatics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Annick Turbe-Doan
- Division of Genetics, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Boston Massachusetts, United States of America
| | - Erin A. Robertson
- Division of Genetics, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Boston Massachusetts, United States of America
| | - Bjorn R. Olsen
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts, United States of America
| | - Martha L. Bulyk
- Division of Genetics, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Boston Massachusetts, United States of America
- Department of Pathology, Harvard Medical School, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
- Harvard-MIT Division of Health Sciences and Technology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Brad A. Amendt
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, United States of America
| | - Richard L. Maas
- Division of Genetics, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Boston Massachusetts, United States of America
| |
Collapse
|
35
|
Pshennikova ES, Voronina AS. Cement gland as the adhesion organ in Xenopus laevis embryos. Russ J Dev Biol 2012. [DOI: 10.1134/s1062360411040096] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
36
|
Stender JD, Stossi F, Funk CC, Charn TH, Barnett DH, Katzenellenbogen BS. The estrogen-regulated transcription factor PITX1 coordinates gene-specific regulation by estrogen receptor-alpha in breast cancer cells. Mol Endocrinol 2011; 25:1699-709. [PMID: 21868451 DOI: 10.1210/me.2011-0102] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The estrogen receptor α (ERα) is a master regulator of gene expression and works along with cooperating transcription factors in mediating the actions of the hormone estradiol (E2) in ER-positive tissues and breast tumors. Here, we report that expression of paired-like homeodomain transcription factor (PITX1), a tumor suppressor and member of the homeobox family of transcription factors, is robustly up-regulated by E2 in several ERα-positive breast cancer cell lines via ERα-dependent interaction between the proximal promoter and an enhancer region 5' upstream of the PITX1 gene. Overexpression of PITX1 selectively inhibited the transcriptional activity of ERα and ERβ, while enhancing the activities of the glucocorticoid receptor and progesterone receptor. Reduction of PITX1 by small interfering RNA enhanced ERα-dependent transcriptional regulation of a subset of ERα target genes. The consensus PITX1 binding motif was found to be present in 28% of genome-wide ERα binding sites and was in close proximity to estrogen response elements in a subset of ERα binding sites, and E2 treatment enhanced PITX1 as well as ERα recruitment to these binding sites. These studies identify PITX1 as a new ERα transcriptional target that acts as a repressor to coordinate and fine tune target-specific, ERα-mediated transcriptional activity in human breast cancer cells.
Collapse
Affiliation(s)
- Joshua D Stender
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801-3704, USA
| | | | | | | | | | | |
Collapse
|
37
|
Budry L, Couture C, Balsalobre A, Drouin J. The Ets factor Etv1 interacts with Tpit protein for pituitary pro-opiomelanocortin (POMC) gene transcription. J Biol Chem 2011; 286:25387-96. [PMID: 21622576 DOI: 10.1074/jbc.m110.202788] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Pro-opiomelanocortin (POMC) is expressed in two lineages of the pituitary, the anterior lobe corticotrophs and the intermediate lobe melanotrophs. POMC expression in these two lineages is highly dependent on the cell-restricted transcription factor Tpit. As Tpit intervenes relatively late in differentiation of those lineages, we have been searching for other transcription factors that may participate in their gene expression program. On the basis of similarity with the Tpit expression profile, we identified Ets variant gene 1 (Etv1/Er81) as a putative POMC transcription factor. Using Etv1-lacZ knockin mice, we describe preferential Etv1 expression in pituitary POMC cells and also in posterior lobe pituicytes. We further show that Etv1 enhances POMC transcription on its own and in synergy with Tpit. The Ets-binding site located within the Tpit/Pitx regulatory element is necessary for Etv1 activity in POMC-expressing AtT-20 cells but dispensable for synergy with Tpit. Etv1 and Tpit interact together in coimmunoprecipitation experiments. Furthermore, Etv1 is present at the POMC promoter, and siRNA-mediated knockdown of Etv1 in AtT-20 cells produces a significant decrease in POMC expression. Etv1 knockout pituitaries show normal POMC cell distribution and normal POMC mRNA abundance, suggesting compensation by other factors. The coordinate expression of Etv1 with POMC cell differentiation and its interaction with the highly cell-restricted Tpit factor indicate that Etv1 participates in a combinatorial code for pituitary cell-specific gene expression.
Collapse
Affiliation(s)
- Lionel Budry
- Laboratoire de Génétique Moléculaire, Institut de Recherches Cliniques de Montréal, Montréal, Quebec H2W 1R7, Canada
| | | | | | | |
Collapse
|
38
|
Nagel S, Venturini L, Przybylski GK, Grabarczyk P, Schneider B, Meyer C, Kaufmann M, Schmidt CA, Scherr M, Drexler HG, Macleod RAF. Activation of Paired-homeobox gene PITX1 by del(5)(q31) in T-cell acute lymphoblastic leukemia. Leuk Lymphoma 2011; 52:1348-59. [PMID: 21425961 DOI: 10.3109/10428194.2011.566391] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
In T-cell acute lymphoblasic leukemia (T-ALL), neoplastic chromosomal rearrangements are known to deregulate members of the homeobox gene families NKL and HOXA. Here, analysis of T-ALL cell lines and primary cells identified aberrant expression of a third homeobox gene group, the Paired (PRD) class. LOUCY cells revealed chromosomal deletion at 5q31, which targets the downstream regulatory region of the PRD homeobox gene PITX1, removing a STAT1 binding site. STAT1 mediates repressive interleukin 2 (IL2)-STAT1 signaling, implicating IL2 pathway avoidance as a possible activation mechanism. Among primary T-ALL samples, 2/22 (9%) aberrantly expressed PITX1, highlighting the importance of this gene. Forced expression of PITX1 in JURKAT cells and subsequent target gene analysis prompted deregulation of genes involved in T-cell development including HES1, JUN, NKX3-1, RUNX1, RUNX2, and TRIB2. Taken together, our data show leukemic activation of PITX1, a novice PRD-class homeobox gene in a subset of early-staged T-ALL, which may promote leukemogenesis by inhibiting T-cell development.
Collapse
Affiliation(s)
- Stefan Nagel
- Department of Human and Animal Cell Lines, DSMZ-German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
L'honoré A, Ouimette JF, Lavertu-Jolin M, Drouin J. Pitx2 defines alternate pathways acting through MyoD during limb and somitic myogenesis. Development 2010; 137:3847-56. [PMID: 20978076 DOI: 10.1242/dev.053421] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The MyoD gene is part of the core regulatory network that governs skeletal myogenesis and acts as an essential determinant of the myogenic cell fate. Although generic regulatory networks converging on this gene have been described, the specific mechanisms leading to MyoD expression in muscles of different ontology remain misunderstood. We now show that the homeobox gene Pitx2 is required for initial activation of the MyoD gene in limb muscle precursors through direct binding of Pitx2 to the MyoD core enhancer. Whereas Myf5 and Mrf4 are dispensable for limb muscle progenitor fate, inactivation of Myf5 and Mrf4 in Pitx2 mutants results in a drastic decrease of limb MyoD expression. Thus, Pitx2 and Myf5 define parallel genetic pathways for limb myogenesis. We show a similar dependence on Pitx2 and Myf5(Mrf4) in myotome, where MyoD expression is initially activated by Myf5 and Mrf4. In their absence, MyoD expression is eventually rescued by a Pax3-dependent mechanism. We now provide evidence that Pitx2 contributes to the rescue of MyoD expression and that it acts downstream of Pax3. We thus propose that myogenic differentiation of somite-derived muscle cells relies on two parallel genetic pathways, with the Pitx2 pathway being of primary importance for limb myogenesis but the Myf5 and Mrf4 pathway predominating in myotome. Muscle-specific wiring of regulatory networks composed of similar transcription factors thus underlies development of distinct skeletal muscles.
Collapse
Affiliation(s)
- Aurore L'honoré
- Laboratoire de génétique moléculaire, Institut de recherches cliniques de Montréal (IRCM), QC, Canada
| | | | | | | |
Collapse
|
40
|
Davis SW, Castinetti F, Carvalho LR, Ellsworth BS, Potok MA, Lyons RH, Brinkmeier ML, Raetzman LT, Carninci P, Mortensen AH, Hayashizaki Y, Arnhold IJP, Mendonça BB, Brue T, Camper SA. Molecular mechanisms of pituitary organogenesis: In search of novel regulatory genes. Mol Cell Endocrinol 2010; 323:4-19. [PMID: 20025935 PMCID: PMC2909473 DOI: 10.1016/j.mce.2009.12.012] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Defects in pituitary gland organogenesis are sometimes associated with congenital anomalies that affect head development. Lesions in transcription factors and signaling pathways explain some of these developmental syndromes. Basic research studies, including the characterization of genetically engineered mice, provide a mechanistic framework for understanding how mutations create the clinical characteristics observed in patients. Defects in BMP, WNT, Notch, and FGF signaling pathways affect induction and growth of the pituitary primordium and other organ systems partly by altering the balance between signaling pathways. The PITX and LHX transcription factor families influence pituitary and head development and are clinically relevant. A few later-acting transcription factors have pituitary-specific effects, including PROP1, POU1F1 (PIT1), and TPIT (TBX19), while others, such as NeuroD1 and NR5A1 (SF1), are syndromic, influencing development of other endocrine organs. We conducted a survey of genes transcribed in developing mouse pituitary to find candidates for cases of pituitary hormone deficiency of unknown etiology. We identified numerous transcription factors that are members of gene families with roles in syndromic or non-syndromic pituitary hormone deficiency. This collection is a rich source for future basic and clinical studies.
Collapse
Affiliation(s)
- S W Davis
- University of Michigan Medical School, Ann Arbor, MI 41809-5618, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Abu-Elmagd M, Robson L, Sweetman D, Hadley J, Francis-West P, Münsterberg A. Wnt/Lef1 signaling acts via Pitx2 to regulate somite myogenesis. Dev Biol 2010; 337:211-9. [PMID: 19850024 DOI: 10.1016/j.ydbio.2009.10.023] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2009] [Revised: 09/18/2009] [Accepted: 10/14/2009] [Indexed: 11/17/2022]
Abstract
Wnt signaling has been implicated in somite, limb, and branchial arch myogenesis but the mechanisms and roles are not clear. We now show that Wnt signaling via Lef1 acts to regulate the number of premyogenic cells in somites but does not regulate myogenic initiation in the limb bud or maintenance in the first or second branchial arch. We have also analysed the function and regulation of a putative downstream transcriptional target of canonical Wnt signaling, Pitx2. We show that loss-of-function of Pitx2 decreases the number of myogenic cells in the somite, whereas overexpression increases myocyte number particularly in the epaxial region of the myotome. Increased numbers of mitotic cells were observed following overexpression of Pitx2 or an activated form of Lef1, suggesting an effect on cell proliferation. In addition, we show that Pitx2 expression is regulated by canonical Wnt signaling in the epaxial somite and second branchial arch, but not in the limb or the first branchial arch. These results suggest that Wnt/Lef1 signaling regulates epaxial myogenesis via Pitx2 but that this link is uncoupled in other regions of the body, emphasizing the unique molecular networks that control the development of various muscles in vertebrates.
Collapse
Affiliation(s)
- Muhammad Abu-Elmagd
- University of East Anglia, School of Biological Sciences, Norwich, NR4 7TJ Earlham Road, UK
| | | | | | | | | | | |
Collapse
|
42
|
Distinct developmental roles of cell cycle inhibitors p57Kip2 and p27Kip1 distinguish pituitary progenitor cell cycle exit from cell cycle reentry of differentiated cells. Mol Cell Biol 2009; 29:1895-908. [PMID: 19139274 DOI: 10.1128/mcb.01885-08] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Patterning and differentiation signals are often believed to drive the developmental program, including cell cycle exit of proliferating progenitors. Taking advantage of the spatial and temporal separation of proliferating and differentiated cells within the developing anterior pituitary gland, we investigated the control of cell proliferation during organogenesis. Thus, we identified a population of noncycling precursors that are uniquely marked by expression of the cell cycle inhibitor p57(Kip2) and by cyclin E. In p57(Kip2-/-) mice, the developing pituitary is hyperplastic due to accumulation of proliferating progenitors, whereas overexpression of p57(Kip2) leads to hypoplasia. p57(Kip2)-dependent cell cycle exit is not required for differentiation, and conversely, blockade of cell differentiation, as achieved in Tpit(-/-) pituitaries, does not prevent cell cycle exit but rather leads to accumulation of p57(Kip2)-positive precursors. Upon differentiation, p57(Kip2) is replaced by p27(Kip1). Accordingly, proliferating differentiated cells are readily detected in p27(Kip1-/-) pituitaries but not in wild-type or p57(Kip2-/-) pituitaries. Strikingly, all cells of p57(Kip2-/-);p27(Kip1-/-) pituitaries are proliferative. Thus, during normal development, progenitor cell cycle exit is controlled by p57(Kip2) followed by p27(Kip1) in differentiated cells; these sequential actions, taken together with different pituitary outcomes of their loss of function, suggest hierarchical controls of the cell cycle that are independent of differentiation.
Collapse
|
43
|
Cherrington BD, Bailey JS, Diaz AL, Mellon PL. NeuroD1 and Mash1 temporally regulate GnRH receptor gene expression in immortalized mouse gonadotrope cells. Mol Cell Endocrinol 2008; 295:106-14. [PMID: 18760324 PMCID: PMC2640340 DOI: 10.1016/j.mce.2008.07.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2008] [Revised: 07/08/2008] [Accepted: 07/25/2008] [Indexed: 01/23/2023]
Abstract
Accurate spatial and temporal expression of gonadotrope-specific genes, such as the gonadotropin-releasing hormone receptor (GnRHR) gene, is critical for gonadotrope maturation. Herein, we show that a specific E-box in the mouse GnRHR promoter binds two group A basic-helix-loop-helix (bHLH) transcription factors. Mutation of this E-box decreases expression in mouse gonadotrope-derived alphaT3-1 and LbetaT2 cell lines. Microarray and western blots show that the bHLH transcription factor NeuroD1 is strongly expressed in the gonadotrope progenitor, alphaT3-1, whereas Mash1 is strongly expressed in the more mature gonadotrope, LbetaT2. Over-expression of NeuroD1 or Mash1 increases expression of the GnRHR gene or a multimer of the E-box and this increase is lost upon mutation of the E-box. Electrophoretic mobility shift assays reveal that the GnRHR E-box binds NeuroD1 from alphaT3-1 cells, but binds Mash1 from LbetaT2 cells. The sequential binding of different members of the group A bHLH transcription factor family to mouse GnRHR E-box 3 as the gonadotrope differentiates may represent a mechanism necessary for proper spatial and temporal expression of the GnRHR during gonadotrope development.
Collapse
Affiliation(s)
| | | | - Alejandro L. Diaz
- Department of Reproductive Medicine and The Center for Reproductive Science and Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Pamela L. Mellon
- Department of Reproductive Medicine and The Center for Reproductive Science and Medicine, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
44
|
Mei S, Sun C, Liu X, Lu Q, Cai L, Li Y, Xiang H. The helix-loop-helix motif at the N terminus of HalI is essential for its immunity function against halocin C8. J Bacteriol 2008; 190:6501-8. [PMID: 18658263 PMCID: PMC2566001 DOI: 10.1128/jb.00665-08] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2008] [Accepted: 07/21/2008] [Indexed: 11/20/2022] Open
Abstract
Halocin C8 (HalC8) is a stable microhalocin exhibiting strong antimicrobial activity against a wide range of haloarchaea. HalI, a 207-amino-acid peptide derived from the N terminus of the HalC8 preproprotein, is the immunity protein of HalC8. In this study, the molecular mechanism of the immunity function of HalI was investigated. Both pull-down and surface plasmon resonance assays revealed that HalI directly interacted with HalC8, and a mixture of purified HalI and HalC8 readily formed a heterocomplex, which was verified by gel filtration. Interestingly, HalC8 tended to form a self-associated complex, and one immunity protein likely sequestered multiple halocins. Significantly, the helix-loop-helix (HLH) motif containing a 4-amino-acid repeat (RELA) at the N terminus of HalI played a key role in its immunity activity. Disruption of the HLH motif or mutagenesis of the key residues of the RELA repeat resulted in loss of both the immunity function and the ability of HalI to bind to HalC8. These results demonstrated that HalI sequestered the activity of HalC8 through specific and direct binding.
Collapse
Affiliation(s)
- Shuangshuang Mei
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Datun Road, Chaoyang District, Beijing 100101, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
45
|
Lavoie PL, Budry L, Balsalobre A, Drouin J. Developmental dependence on NurRE and EboxNeuro for expression of pituitary proopiomelanocortin. Mol Endocrinol 2008; 22:1647-57. [PMID: 18388149 DOI: 10.1210/me.2007-0567] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Cell-specific expression of the pituitary proopiomelanocortin (POMC) gene depends on the combinatorial action of a large number of DNA-binding transcription factors (TFs). These include general and cell-restricted factors, as well as factors that act as effectors of signaling pathways. We have previously defined in the distal POMC promoter a composite regulatory element that contains targets for basic helix-loop-helix TFs conferring cell specificity and for NGFI-B orphan nuclear receptors that are responsive to CRH signaling and to glucocorticoid negative feedback. These factors act on neighboring regulatory elements, the Ebox(Neuro) and NurRE, respectively. Currently, the Ebox(Neuro) is thought to be the target of NeuroD1 during fetal development, but this factor may not account for activity in the adult pituitary; it is also unknown whether the NurRE and NGFI-B-related factors are active before establishment of the hypothalamic-pituitary portal system. In order to assess the importance of these regulatory elements and their cognate TFs throughout pituitary organogenesis and in the adult, we have assessed the activity of mutant POMC promoters in transgenic mice throughout development. These experiments indicate that the Ebox(Neuro) and cognate basic helix-loop-helix factors are required throughout development and in the adult gland, beyond expression of NeuroD1. Similarly, the data reveal sustained importance of the NurRE and its cognate factors throughout pituitary development. These data contrast the sustained dependence throughout development on the same regulatory elements with the highly dynamic patterns of TF expression and the modulation of their activity in response to signaling pathways.
Collapse
Affiliation(s)
- Pierre-Luc Lavoie
- Laboratoire de Génétique Moléculaire, Institut de Recherches Cliniques de Montréal 110, Montréal, Québec, Canada
| | | | | | | |
Collapse
|
46
|
Suszko MI, Antenos M, Balkin DM, Woodruff TK. Smad3 and Pitx2 cooperate in stimulation of FSHbeta gene transcription. Mol Cell Endocrinol 2008; 281:27-36. [PMID: 18022758 DOI: 10.1016/j.mce.2007.10.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2007] [Revised: 10/03/2007] [Accepted: 10/04/2007] [Indexed: 10/22/2022]
Abstract
Activin is a member of the TGFbeta superfamily of growth and differentiation factors that control a variety of cellular and physiological functions. The canonical intracellular pathway of this ligand is well established and involves Smad signaling molecules. The tissue- and cell-specificity of activin action is achieved by Smad interaction with various transcriptional co-factors in the nucleus. In the reproductive axis, activin induces biosynthesis and secretion of follicle stimulating hormone (FSH) through transcriptional control of FSHbeta-subunit. Whereas it has been well demonstrated that this regulation is mediated by Smad pathway, the molecular mechanisms underlying gonadotrope-specific expression of the FSHbeta gene are not fully understood. Previously, we have identified Pitx2 as a pituitary-expressed transcription factor involved in activin-dependent induction of the FSHbeta promoter. Present data demonstrate that Pitx2 is not only sufficient, but also necessary for FSHbeta gene transcription, as a siRNA-mediated downregulation of Pitx2 protein expression abrogates both Smad3- and activin-mediated stimulation of the FSHbeta promoter. In addition, downregulation of Smad3 protein expression has a significant effect on Pitx2-dependent stimulation of the FSHbeta promoter, suggesting that cooperation between these factors is necessary for full transcriptional activation of the FSHbeta promoter. Furthermore, we show that Pitx2/Smad protein complexes assemble and can be co-immunoprecipitated. This interaction is mediated through the homeodomain of Pitx2 and is important for stimulation of FSHbeta gene transcription. Overall, these data contribute to the emerging molecular mechanism underlying both basal and activin-dependent FSHbeta gene regulation.
Collapse
Affiliation(s)
- Magdalena I Suszko
- Department of Obstetrics & Gynecology, Northwestern Medical School, Chicago, IL 60611, United States
| | | | | | | |
Collapse
|
47
|
Abstract
The vertebrate central nervous system contains a great diversity of neurons and glial cells, which are generated in the embryonic neural tube at specific times and positions. Several classes of transcription factors have been shown to control various steps in the differentiation of progenitor cells in the neural tube and to determine the identity of the cells produced. Recent evidence indicates that combinations of transcription factors of the homeodomain and basic helix-loop-helix families establish molecular codes that determine both where and when the different kinds of neurons and glial cells are generated.
Collapse
Affiliation(s)
- François Guillemot
- National Institute for Medical Research, The Ridgeway, Mill Hill, NW7 1AA London, UK.
| |
Collapse
|
48
|
Vallette-Kasic S, Couture C, Balsalobre A, Gauthier Y, Metherell L, Dattani M, Drouin J. The TPIT gene mutation M86R associated with isolated adrenocorticotropin deficiency interferes with protein: protein interactions. J Clin Endocrinol Metab 2007; 92:3991-9. [PMID: 17652218 DOI: 10.1210/jc.2007-0284] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Tpit is a T-box transcription factor important for terminal differentiation of pituitary proopiomelanocortin-expressing cells. We previously showed that human and murine mutations in the gene encoding this highly cortico/melanotrope-specific transcription factor cause a neonatal onset form of congenital isolated ACTH deficiency (IAD). We characterized the largest series of neonatal IAD patients caused by TPIT mutations, and this revealed a highly homogeneous clinical presentation. So far, 12 different loss-of-function TPIT mutations have been identified. The methionine 86 arginine (M86R) TPIT mutation was recently identified in compound heterozygosity with the 782delA frame-shift mutation in two siblings with early-onset IAD. OBJECTIVE We conducted a functional analysis of the missense M86R mutation to assess transcriptional activity, DNA binding activity, and nuclear location, as well as protein-protein interactions. RESULTS Although the M86 residue is located within the T-box DNA-binding domain, it did not affect monomer DNA-binding activity per se, but it impaired DNA binding with other DNA-bound proteins, including itself (homodimers) and pituitary homeobox 1 (Pitx1). The M86 residue is at the interface between T domains in the T dimers crystal structure, and it appears that the same residue is involved in heterodimer formation with pituitary Pitx1. Furthermore, TPIT M86R is deficient in the recruitment of the coactivator SRC2 that partly mediates the CRH stimulation of proopiomelanocortin transcription. CONCLUSION Thus, the M86R TPIT mutation is defining an important surface of the T domain for multiple protein interactions and for transcription.
Collapse
Affiliation(s)
- Sophie Vallette-Kasic
- Laboratoire de génétique moléculaire, Institut de recherches cliniques de Montréal, 110, avenue des Pins Ouest, Montréal, QC, Canada H2W 1R7
| | | | | | | | | | | | | |
Collapse
|
49
|
Ray SK, Leiter AB. The basic helix-loop-helix transcription factor NeuroD1 facilitates interaction of Sp1 with the secretin gene enhancer. Mol Cell Biol 2007; 27:7839-47. [PMID: 17875929 PMCID: PMC2169158 DOI: 10.1128/mcb.00438-07] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The basic helix-loop-helix transcription factor NeuroD1 is required for late events in neuronal differentiation, for maturation of pancreatic beta cells, and for terminal differentiation of enteroendocrine cells expressing the hormone secretin. NeuroD1-null mice demonstrated that this protein is essential for expression of the secretin gene in the murine intestine, and yet it is a relatively weak transcriptional activator by itself. The present study shows that Sp1 and NeuroD1 synergistically activate transcription of the secretin gene. NeuroD1, but not its widely expressed dimerization partner E12, physically interacts with the C-terminal 167 amino acids of Sp1, which include its DNA binding zinc fingers. NeuroD1 stabilizes Sp1 DNA binding to an adjacent Sp1 binding site on the promoter to generate a higher-order DNA-protein complex containing both proteins and facilitates Sp1 occupancy of the secretin promoter in vivo. NeuroD-dependent transcription of the genes encoding the hormones insulin and proopiomelanocortin is potentiated by lineage-specific homeodomain proteins. The stabilization of binding of the widely expressed transcription factor Sp1 to the secretin promoter by NeuroD represents a distinct mechanism from other NeuroD target genes for increasing NeuroD-dependent transcription.
Collapse
Affiliation(s)
- Subir K Ray
- Department of Medicine, Division of Gastroenterology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | | |
Collapse
|
50
|
Coulon V, L'Honoré A, Ouimette JF, Dumontier E, van den Munckhof P, Drouin J. A muscle-specific promoter directs Pitx3 gene expression in skeletal muscle cells. J Biol Chem 2007; 282:33192-200. [PMID: 17848564 DOI: 10.1074/jbc.m706119200] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The Pitx homeobox transcription factor genes have been implicated in different developmental processes, including determination of hind limb identity for Pitx1, left-right asymmetry for Pitx2, and eye development and survival of midbrain dopaminergic neurons for Pitx3. Pitx1 and Pitx2 have partly redundant activities in craniofacial development, including in pituitary organogenesis, as indicated by their names. These genes also exhibit redundant activities in the control of hind limb bud growth. Recent studies have shown expression of the three Pitx genes in muscle, with Pitx3 being the most widely expressed in all skeletal muscles. We now report the identification of a muscle-specific promoter within the Pitx3 gene that is situated between the first exon for eye and brain expression and exon 2 that contains the initiator ATG codon. Sequences proximal to this muscle-specific exon 1 are essential and sufficient to confer muscle-specific expression in transgenic mice, they are responsive to myogenic basic helix-loop-helix regulatory factors, and they recruit these factors in vivo. In agreement with exclusive use of the muscle-specific promoter in aphakia mice that are deleted of the brain promoter, the trimethyl-lysine 4 histone H3 promoter signature shifts to this promoter in embryonic day 13 ak limb bud muscle cells. Myogenic basic helix-loop-helix regulatory factor activation of Pitx3 transcription may be part of a positive feedback loop contributing to establishment of the myogenic program.
Collapse
Affiliation(s)
- Vincent Coulon
- Laboratoire de Génétique Moléculaire, Institut de Recherches Cliniques de Montréal, 110 Avenue des Pins Ouest, Montréal, Quebec, Canada
| | | | | | | | | | | |
Collapse
|