1
|
Sarveswaran N, Pamela Y, Reddy AAN, Mustari AP, Parthasarathi A, Mancini AJ, Bishnoi A, Inamadar AC, Olabi B, Browne F, Deshmukh GN, McWilliam K, Vinay K, Srinivas S, Ibbs S, Natarajan S, Rao V, Zawar V, Gowda VK, Shaikh SS, Moss C, Woods CG, Drissi I. Midfacial toddler excoriation syndrome (MiTES): case series, diagnostic criteria and evidence for a pathogenic mechanism. Br J Dermatol 2024; 191:437-446. [PMID: 38591490 PMCID: PMC11324070 DOI: 10.1093/bjd/ljae151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 04/02/2024] [Accepted: 04/04/2024] [Indexed: 04/10/2024]
Abstract
BACKGROUND PRDM12 polyalanine tract expansions cause two different disorders: midfacial toddler excoriation syndrome (MiTES; itch with normal pain sensation associated with 18 homozygous alanines (18A); and congenital insensitivity to pain (CIP) with normal itch associated with 19 homozygous alanines (19A). Knowledge of the phenotype, genotype and disease mechanism of MiTES is incomplete. Why 18A vs. 19A PRDM12 can cause almost opposite phenotypes is unknown; no other polyalanine or polyglutamine tract expansion disease causes two such disparate phenotypes. OBJECTIVES To assess the genotype and phenotype of nine new, nine atypical and six previously reported patients diagnosed with MiTES. METHODS Using cell lines with homozygous PR domain zinc finger protein 12 (PRDM12) containing 12 alanines (12A; normal), 18A (MiTES) and 19A (CIP), we examined PRDM12 aggregation and subcellular localization by image-separation confocal microscopy and subcellular fractionation Western blotting. RESULTS MiTES presents in the first year of life; in all cases the condition regresses over the first decade, leaving scarring. The MiTES phenotype is highly distinctive. Features overlapping with PRDM12 CIP are rarely found. The genotype-phenotype study of the PRDM12 polyalanine tract shows that having 7-15 alanines is normal; 16-18 alanines is associated with MiTES; 19 alanines leads to CIP; and no clinically atypical cases of MiTES had a polyalanine tract expansion. PRDM12 aggregation and subcellular localization differed significantly between 18A and normal 12A cell lines and between 18A and 19A cell lines. MiTES is a new protein-aggregation disease. CONCLUSIONS We provide diagnostic criteria for MiTES and improved longitudinal data. MiTES and CIP are distinct phenotypes, despite their genotypes varying by a single alanine in the PRDM12 polyalanine tract. We found clear distinctions between the cellular phenotypes of normal, MiTES and CIP cells. We hypothesize that the developmental environment of the trigeminal ganglion is unique and critically sensitive to pre- and postnatal levels of PRDM12.
Collapse
Affiliation(s)
- Nivedita Sarveswaran
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Yunisa Pamela
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Akhila A N Reddy
- Dr. Anchala Skin Institute and Research Center, Hyderabad, India
| | - Akash P Mustari
- Department of Dermatology, Venereology and Leprology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | | | - Anthony J Mancini
- Division of Dermatology, Department of Pediatrics, Ann & Robert H. Lurie Children’s Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Anuradha Bishnoi
- Department of Dermatology, Venereology and Leprology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Arun C Inamadar
- Department of Dermatology, Shri BM Patil Medical College & Hospital, BLDE University, Bijapur, India
| | - Bayanne Olabi
- Department of Dermatology, Edinburgh Royal Infirmary, UK
| | - Fiona Browne
- Department of Paediatric Dermatology, Children’s Health Ireland (CHI) at Crumlin, Crumlin, Ireland
| | | | - Kenneth McWilliam
- Paediatric Neurology, Neurosciences Department, Royal Hospital for Children and Young People, Edinburgh, UK
| | - Keshavamurthy Vinay
- Department of Dermatology, Venereology and Leprology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Sahana Srinivas
- Department of Pediatric Dermatology, Indira Gandhi Institute of Child Health, Bangalore, India
| | - Samantha Ibbs
- Department of Paediatric Dermatology, Birmingham Children’s Women’s and Children’s NHS Foundation Trust, Birmingham, UK
| | | | | | - Vijay Zawar
- Department of Dermatology, Dr. Vasantrao Pawar Medical College and Research Center, Nashik, India
| | - Vykuntaraju K Gowda
- Department of Pediatric Neurology, Indira Gandhi Institute of Child Health, Bangalore, Karnataka, India
| | - Samiha S Shaikh
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Celia Moss
- Department of Paediatric Dermatology, Birmingham Children’s Women’s and Children’s NHS Foundation Trust, Birmingham, UK
- College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Christopher G Woods
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Ichrak Drissi
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| |
Collapse
|
2
|
Tsimpos P, Desiderio S, Cabochette P, Poelvoorde P, Kricha S, Vanhamme L, Poulard C, Bellefroid EJ. Loss of G9a does not phenocopy the requirement for Prdm12 in the development of the nociceptive neuron lineage. Neural Dev 2024; 19:1. [PMID: 38167468 PMCID: PMC10759634 DOI: 10.1186/s13064-023-00179-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 12/12/2023] [Indexed: 01/05/2024] Open
Abstract
Prdm12 is an epigenetic regulator expressed in developing and mature nociceptive neurons, playing a key role in their specification during neurogenesis and modulating pain sensation at adulthood. In vitro studies suggested that Prdm12 recruits the methyltransferase G9a through its zinc finger domains to regulate target gene expression, but how Prdm12 interacts with G9a and whether G9a plays a role in Prdm12's functional properties in sensory ganglia remain unknown. Here we report that Prdm12-G9a interaction is likely direct and that it involves the SET domain of G9a. We show that both proteins are largely co-expressed in dorsal root ganglia during early murine development, opening the possibility that G9a plays a role in DRG and may act as a mediator of Prdm12's function in the development of nociceptive sensory neurons. To test this hypothesis, we conditionally inactivated G9a in neural crest using a Wnt1-Cre transgenic mouse line. We found that the specific loss of G9a in the neural crest lineage does not lead to dorsal root ganglia hypoplasia due to the loss of somatic nociceptive neurons nor to the ectopic expression of the visceral determinant Phox2b as observed upon Prdm12 ablation. These findings suggest that Prdm12 function in the initiation of the nociceptive lineage does not critically involves its interaction with G9a.
Collapse
Affiliation(s)
- Panagiotis Tsimpos
- ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), Gosselies, B- 6041, Belgium
| | - Simon Desiderio
- ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), Gosselies, B- 6041, Belgium
| | - Pauline Cabochette
- ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), Gosselies, B- 6041, Belgium
| | - Philippe Poelvoorde
- Department of Molecular Biology, Institute of Biology and Molecular Medicine, IBMM, Université Libre de Bruxelles, Bruxelles, Belgium
| | - Sadia Kricha
- ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), Gosselies, B- 6041, Belgium
| | - Luc Vanhamme
- Department of Molecular Biology, Institute of Biology and Molecular Medicine, IBMM, Université Libre de Bruxelles, Bruxelles, Belgium
| | - Coralie Poulard
- Cancer Research Cancer of Lyon, Université de Lyon, Lyon, F-69000, France
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, F-69000, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, F-69000, France
| | - Eric J Bellefroid
- ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), Gosselies, B- 6041, Belgium.
| |
Collapse
|
3
|
Yamamoto T, Kambayashi Y, Tsukano K, Michiue T. Ndst1, a heparan sulfate modification enzyme, regulates neuroectodermal patterning by enhancing Wnt signaling in Xenopus. Dev Growth Differ 2023; 65:153-160. [PMID: 36726238 PMCID: PMC11520968 DOI: 10.1111/dgd.12843] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 01/05/2023] [Accepted: 01/10/2023] [Indexed: 02/03/2023]
Abstract
Neural tissue is derived from three precursor regions: neural plate, neural crest, and preplacodal ectoderm. These regions are determined by morphogen-mediated signaling. Morphogen distribution is generally regulated by binding to an extracellular matrix component, heparan sulfate (HS) proteoglycan. HS is modified by many enzymes, such as N-deacetyl sulfotransferase 1 (Ndst1), which is highly expressed in early development. However, functions of HS modifications in ectodermal patterning are largely unknown. In this study, we analyzed the role of Ndst1 using Xenopus embryos. We found that ndst1 was expressed in anterior neural plate and the trigeminal region at the neurula stage. ndst1 overexpression expanded the neural crest (NC) region, whereas translational inhibition reduced not only the trigeminal region, but also the adjacent NC region, especially the anterior part. At a later stage, ndst1 knocked-down embryos showed defects in cranial ganglion formation. We also found that Ndst1 activates Wnt signaling pathway at the neurula stage. Taken together, our results suggest that N-sulfonated HS accumulates Wnt ligand and activates Wnt signaling in ndst1-expressing cells, but that it inhibits signaling in non-ndst1-expressing cells, leading to proper neuroectodermal patterning.
Collapse
Grants
- JPMJSP2108 Japan Science and Technology Agency
- 18K06244 Ministry of Education, Culture, Sports, Science and Technology
- 19K16138 Ministry of Education, Culture, Sports, Science and Technology
- 21K06183 Ministry of Education, Culture, Sports, Science and Technology
- Narishige Zoological Science Award
- Japan Science and Technology Agency
- Ministry of Education, Culture, Sports, Science and Technology
Collapse
Affiliation(s)
- Takayoshi Yamamoto
- Department of Life Sciences, Graduate School of Arts and SciencesThe University of TokyoTokyoJapan
| | - Yuta Kambayashi
- Department of Life Sciences, Graduate School of Arts and SciencesThe University of TokyoTokyoJapan
| | - Kohei Tsukano
- Department of Life Sciences, Graduate School of Arts and SciencesThe University of TokyoTokyoJapan
| | - Tatsuo Michiue
- Department of Life Sciences, Graduate School of Arts and SciencesThe University of TokyoTokyoJapan
- Department of Biological Sciences, Graduate School of ScienceThe University of TokyoTokyoJapan
| |
Collapse
|
4
|
Yamamoto T, Kaneshima T, Tsukano K, Michiue T. The heparan sulfate modification enzyme, Hs6st1, governs Xenopus neuroectodermal patterning by regulating distributions of Fgf and Noggin. Dev Biol 2023; 496:87-94. [PMID: 36739958 DOI: 10.1016/j.ydbio.2023.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 01/20/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023]
Abstract
The nervous system has various types of cells derived from three neuroectodermal regions: neural plate (NP), neural crest (NC), and preplacodal ectoderm (PPE). Differentiation of these regions is regulated by various morphogens. However, regulatory mechanisms of morphogen distribution in neural patterning are still debated. In general, an extracellular component, heparan sulfate (HS), is essential to regulate morphogen gradients by modulating morphogen binding. The present study focused on an HS modification enzyme, heparan sulfate 6-O-sulfotransferase 1 (Hs6st1), which is highly expressed during the neurula stage in Xenopus. Our present in situ hybridization analysis revealed that Hs6st1 is expressed in the lateral sensorial layer of neuroectoderm. Overexpression of Hs6st1 expands Sox3 (NP marker gene) expression, and slightly dampens FoxD3 (NC marker) expression. Hs6st1 knockout using the CRISPR/Cas9 system also expands the neural plate region, followed by retinal malformation. These results imply that 6-O sulfation, mediated by Hs6st1, selectively regulates morphogen distribution required for neuroectodermal patterning. Among morphogens required for patterning, Fgf8a accumulates on Hs6st1-expressing cells, whereas a secreted BMP antagonist, Noggin, diffuses away from those cells. Thus, cell-autonomous 6-O sulfation of HS at the sensorial layer of neuroectoderm also affects neuroectodermal patterning in neighboring regions, including neural plate and neural crest, not only through accumulation, but also through dispersal of specific morphogens.
Collapse
Affiliation(s)
- Takayoshi Yamamoto
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo, 153-8902, Japan.
| | - Toki Kaneshima
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo, 153-8902, Japan
| | - Kohei Tsukano
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo, 153-8902, Japan
| | - Tatsuo Michiue
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo, 153-8902, Japan; Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| |
Collapse
|
5
|
Donoso V, Whitson J, Lom B. Developmental exposure to the pesticide malathion enhances expression of Prdm12, a regulator of nociceptor development, in Xenopus laevis. MICROPUBLICATION BIOLOGY 2023; 2023:10.17912/micropub.biology.000786. [PMID: 37033708 PMCID: PMC10074175 DOI: 10.17912/micropub.biology.000786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 02/18/2023] [Accepted: 02/28/2023] [Indexed: 04/11/2023]
Abstract
The transcription factor Prdm12 exerts important influences on the development of nociceptors, peripheral touch and pain-sensing neurons, and has been implicated in human pain sensation disorders. We examined the consequences of exposing developing Xenopus laevis embryos to the commonly used pesticide malathion on Prdm12 expression. Using qPCR and western blot analysis we observed that malathion treatment for the first six days of tadpole development significantly increased both prdm12 mRNA levels and Prdm12 protein levels compared to controls. Consequently, early exposure to this pesticide has potential to alter nociceptor development.
Collapse
Affiliation(s)
- Valeria Donoso
- Biology, Davidson College, Davidson, North Carolina, United States
- Preventative Medicine, Northwestern University, Chicago, Illinois, United States
| | - Jeremy Whitson
- Biology, Davidson College, Davidson, North Carolina, United States
- Biology, High Point University, High Point, North Carolina, United States
| | - Barbara Lom
- Biology, Davidson College, Davidson, North Carolina, United States
- Correspondence to: Barbara Lom (
)
| |
Collapse
|
6
|
Li W, Zhang Y, Lv J, Zhang Y, Bai J, Zhen L, He X. MicroRNA-137-mediated lysine demethylase 4A regulates the recovery of spinal cord injury via the SFRP4-Wnt/β-Catenin axis. Int J Neurosci 2023; 133:37-50. [PMID: 33499717 DOI: 10.1080/00207454.2021.1881093] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
OBJECTIVE Spinal cord injury (SCI) causes great harm to the normal life of patients. Histone demethylase is involved in many biological processes, including SCI. Hence, this study explored the role and mechanism of histone lysine demethylase 4A (KDM4A) in SCI. METHODS The acute SCI (ASCI) rat model was established after spinal compression and the SCI neuronal model was induced via treating PC12 cells with lipopolysaccharide (LPS). KDM4A expression during SCI was detected. The microRNA (miRNA) targeting KDM4A was predicted and verified. The miRNA and KDM4A expression patterns were intervened in LPS-stimulated PC12 cells to evaluate their combined effects on neuronal cells in SCI. The downstream pathways of KDM4A were predicted, and SFRP4 and H3K9me3 expressions were determined. After the intervention of SFRP4 in LPS-treated cells, β-Catenin expression and the effect of SFRP4 on neuronal cells in SCI were detected. Finally, the effectiveness of the miR-137/KDM4A/SFRP4/Wnt/β-Catenin axis was verified in vivo. RESULTS KDM4A was abnormally elevated in SCI. miR-137 targeted KDM4A. miR-137 effectively inhibited the apoptosis of LPS-challenged PC12 cells, which could be reversed after overexpressing KDM4A. KDM4A promoted SFRP4 expression through demethylation of H3K9me3. Overexpression of SFRP4 blocked the Wnt/β-Catenin pathway and promoted apoptosis of LPS-stimulated cells. In vivo, miR-137 overexpression remarkably improved SCI symptoms, accompanied by obviously increased β-Catenin expression and notably decreased KDM4A and SFRP4 expressions, while overexpressed KDM4A treatment showed the opposite trend in the presence of miR-137. CONCLUSION We demonstrated that miR-137 targeted KDM4A and then downregulated SFRP4 to ameliorate SCI in a Wnt/β-Catenin-dependent manner.
Collapse
Affiliation(s)
- Wei Li
- Department of Anesthesia, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P.R. China
| | - Ying Zhang
- Department of Anesthesia, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P.R. China
| | - Jianrui Lv
- Department of Anesthesia, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P.R. China
| | - Yong Zhang
- Department of Anesthesia, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P.R. China
| | - Jie Bai
- Department of Anesthesia, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P.R. China
| | - Luming Zhen
- Department of Anesthesia, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P.R. China
| | - Xijing He
- Department of Orthopaedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P.R. China
| |
Collapse
|
7
|
Ash2l, an obligatory component of H3K4 methylation complexes, regulates neural crest development. Dev Biol 2022; 492:14-24. [PMID: 36162552 DOI: 10.1016/j.ydbio.2022.09.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 08/29/2022] [Accepted: 09/20/2022] [Indexed: 02/05/2023]
Abstract
The vertebrate nervous system develops from embryonic neural plate and neural crest. Although genetic mechanisms governing vertebrate neural development have been investigated in depth, epigenetic regulation of this process remains less understood. Redundancy of epigenetic factors and early lethality of animals deficient in critical epigenetic components pose major challenges in characterization of epigenetic factors in vertebrate neural development. In this study, we use the amphibian model Xenopus laevis to investigate the roles of non-redundant, obligatory components of all histone H3K4 activating methylation complexes (COMPASS, also known as SET1/MLL complexes) in early neural development. The two genes that we focus on, Ash2l and Dpy30, regulate mesendodermal differentiation in mouse embryonic stem cells and cause early embryonic lethality when removed from mouse embryos. Using targeted knockdown of the genes in dorsal ectoderm of Xenopus that gives rise to future nervous system, we show here that ash2l and dpy30 are required for neural and neural crest marker expression in Xenopus late neurula embryos but are dispensable for early neural and neural plate border gene expression. Co-immunoprecipitation assays reveal that Dpy30 and Ash2L associate with the neural plate border transcription factors, such as Msx1 and Tfap2a. Chromatin immunoprecipitation (ChIP) assay further demonstrates that Ash2L and the H3K4me3 active histone mark accumulate at the promoter regions of the neural crest gene sox10 in a Tfap2a-dependent manner. Collectively, our data suggest that Ash2l and Dpy30 interact with specific transcription factors to recruit COMPASS complexes to the regulatory regions of neural crest specification genes to control their expression and influence development of the nervous system during vertebrate embryogenesis.
Collapse
|
8
|
Latragna A, Sabaté San José A, Tsimpos P, Vermeiren S, Gualdani R, Chakrabarti S, Callejo G, Desiderio S, Shomroni O, Sitte M, Kricha S, Luypaert M, Vanhollebeke B, Laumet G, Salinas G, Smith ESJ, Ris L, Bellefroid EJ. Prdm12 modulates pain-related behavior by remodeling gene expression in mature nociceptors. Pain 2022; 163:e927-e941. [PMID: 34961757 PMCID: PMC9341233 DOI: 10.1097/j.pain.0000000000002536] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 10/29/2021] [Accepted: 11/03/2021] [Indexed: 11/26/2022]
Abstract
ABSTRACT Prdm12 is a conserved epigenetic transcriptional regulator that displays restricted expression in nociceptors of the developing peripheral nervous system. In mice, Prdm12 is required for the development of the entire nociceptive lineage. In humans, PRDM12 mutations cause congenital insensitivity to pain, likely because of the loss of nociceptors. Prdm12 expression is maintained in mature nociceptors suggesting a yet-to-be explored functional role in adults. Using Prdm12 inducible conditional knockout mouse models, we report that in adult nociceptors Prdm12 is no longer required for cell survival but continues to play a role in the transcriptional control of a network of genes, many of them encoding ion channels and receptors. We found that disruption of Prdm12 alters the excitability of dorsal root ganglion neurons in culture. Phenotypically, we observed that mice lacking Prdm12 exhibit normal responses to thermal and mechanical nociceptive stimuli but a reduced response to capsaicin and hypersensitivity to formalin-induced inflammatory pain. Together, our data indicate that Prdm12 regulates pain-related behavior in a complex way by modulating gene expression in adult nociceptors and controlling their excitability. The results encourage further studies to assess the potential of Prdm12 as a target for analgesic development.
Collapse
Affiliation(s)
- Aurore Latragna
- ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), Gosselies, Belgium
- Laboratory of Neuroscience, UMONS Research Institute for Health Sciences and Technology, University of Mons, Mons, Belgium
| | - Alba Sabaté San José
- ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Panagiotis Tsimpos
- ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Simon Vermeiren
- ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Roberta Gualdani
- Laboratory of Neuroscience, UMONS Research Institute for Health Sciences and Technology, University of Mons, Mons, Belgium
| | | | - Gerard Callejo
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | - Simon Desiderio
- ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Orr Shomroni
- NGS Integrative Genomics, Department of Human Genetics at the University Medical Center Göttingen (UMG), Göttingen, Germany
| | - Maren Sitte
- NGS Integrative Genomics, Department of Human Genetics at the University Medical Center Göttingen (UMG), Göttingen, Germany
| | - Sadia Kricha
- ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Maëlle Luypaert
- ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Benoit Vanhollebeke
- ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Geoffroy Laumet
- Department of Physiology, Michigan State University, East Lansing, MI, United States
| | - Gabriela Salinas
- NGS Integrative Genomics, Department of Human Genetics at the University Medical Center Göttingen (UMG), Göttingen, Germany
| | - Ewan St. John Smith
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | - Laurence Ris
- Laboratory of Neuroscience, UMONS Research Institute for Health Sciences and Technology, University of Mons, Mons, Belgium
| | - Eric J. Bellefroid
- ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), Gosselies, Belgium
| |
Collapse
|
9
|
Han L, Luo J, Qu S, Shi X, Zhang J, Han B. kdm4aa is required for reproduction and development of zebrafish. AQUACULTURE AND FISHERIES 2022. [DOI: 10.1016/j.aaf.2022.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
10
|
Rienzo M, Di Zazzo E, Casamassimi A, Gazzerro P, Perini G, Bifulco M, Abbondanza C. PRDM12 in Health and Diseases. Int J Mol Sci 2021; 22:ijms222112030. [PMID: 34769459 PMCID: PMC8585061 DOI: 10.3390/ijms222112030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/03/2021] [Accepted: 11/04/2021] [Indexed: 11/18/2022] Open
Abstract
PRDM12 is a member of the PRDI-BF1 (positive regulatory domain I-binding factor 1) homologous domain (PRDM)-containing protein family, a subfamily of Kruppel-like zinc finger proteins, controlling key processes in the development of cancer. PRDM12 is expressed in a spatio-temporal manner in neuronal systems where it exerts multiple functions. PRDM12 is essential for the neurogenesis initiation and activation of a cascade of downstream pro-neuronal transcription factors in the nociceptive lineage. PRDM12 inactivation, indeed, results in a complete absence of the nociceptive lineage, which is essential for pain perception. Additionally, PRDM12 contributes to the early establishment of anorexigenic neuron identity and the maintenance of high expression levels of pro-opiomelanocortin, which impacts on the program bodyweight homeostasis. PRDMs are commonly involved in cancer, where they act as oncogenes/tumor suppressors in a “Yin and Yang” manner. PRDM12 is not usually expressed in adult normal tissues but its expression is re-activated in several cancer types. However, little information is currently available on PRDM12 expression in cancers and its mechanism of action has not been thoroughly described. In this review, we summarize the recent findings regarding PRDM12 by focusing on four main biological processes: neurogenesis, pain perception, oncogenesis and cell metabolism. Moreover, we wish to highlight the importance of future studies focusing on the PRDM12 signaling pathway(s) and its role in cancer onset and progression.
Collapse
Affiliation(s)
- Monica Rienzo
- Department of Environmental, Biological, and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, 81100 Caserta, Italy;
| | - Erika Di Zazzo
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy;
| | - Amelia Casamassimi
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via L. De Crecchio, 80138 Naples, Italy;
- Correspondence:
| | - Patrizia Gazzerro
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Salerno, Italy;
| | - Giovanni Perini
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy;
| | - Maurizio Bifulco
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, 80131 Naples, Italy;
| | - Ciro Abbondanza
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via L. De Crecchio, 80138 Naples, Italy;
| |
Collapse
|
11
|
Kokotović T, Langeslag M, Lenartowicz EM, Manion J, Fell CW, Alehabib E, Tafakhori A, Darvish H, Bellefroid EJ, Neely GG, Kress M, Penninger JM, Nagy V. PRDM12 Is Transcriptionally Active and Required for Nociceptor Function Throughout Life. Front Mol Neurosci 2021; 14:720973. [PMID: 34646120 PMCID: PMC8502974 DOI: 10.3389/fnmol.2021.720973] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 08/20/2021] [Indexed: 12/21/2022] Open
Abstract
PR domain-containing member 12 (PRDM12) is a key developmental transcription factor in sensory neuronal specification and survival. Patients with rare deleterious variants in PRDM12 are born with congenital insensitivity to pain (CIP) due to the complete absence of a subtype of peripheral neurons that detect pain. In this paper, we report two additional CIP cases with a novel homozygous PRDM12 variant. To elucidate the function of PRDM12 during mammalian development and adulthood, we generated temporal and spatial conditional mouse models. We find that PRDM12 is expressed throughout the adult nervous system. We observed that loss of PRDM12 during mid-sensory neurogenesis but not in the adult leads to reduced survival. Comparing cellular biophysical nociceptive properties in developmental and adult-onset PRDM12 deletion mouse models, we find that PRDM12 is necessary for proper nociceptive responses throughout life. However, we find that PRDM12 regulates distinct age-dependent transcriptional programs. Together, our results implicate PRDM12 as a viable therapeutic target for specific pain therapies even in adults.
Collapse
Affiliation(s)
- Tomislav Kokotović
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria.,CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.,Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Michiel Langeslag
- Department of Physiology and Medical Physics, Institute of Physiology, Medical University of Innsbruck, Innsbruck, Austria.,Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria.,Department of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Ewelina M Lenartowicz
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria.,Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - John Manion
- Charles Perkins Centre, Dr. John and Anne Chong Lab for Functional Genomics, Centenary Institute, and School of Life and Environmental Sciences, University of Sydney, Camperdown, NSW, Australia
| | - Christopher W Fell
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria.,CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.,Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Elham Alehabib
- Student Research Committee, Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Tafakhori
- Iranian Center of Neurological Research, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Darvish
- Neuroscience Research Center, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Eric J Bellefroid
- ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - G Gregory Neely
- Charles Perkins Centre, Dr. John and Anne Chong Lab for Functional Genomics, Centenary Institute, and School of Life and Environmental Sciences, University of Sydney, Camperdown, NSW, Australia
| | - Michaela Kress
- Department of Physiology and Medical Physics, Institute of Physiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, VBC - Vienna BioCenter, Vienna, Austria.,Department of Medical Genetics, Life Science Institute, University of British Columbia, Vancouver, BC, Canada
| | - Vanja Nagy
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria.,CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.,Department of Neurology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
12
|
Roffers-Agarwal J, Lidberg KA, Gammill LS. The lysine methyltransferase SETD2 is a dynamically expressed regulator of early neural crest development. Genesis 2021; 59:e23448. [PMID: 34498354 DOI: 10.1002/dvg.23448] [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: 12/22/2020] [Revised: 07/31/2021] [Accepted: 08/13/2021] [Indexed: 11/11/2022]
Abstract
SETD2 is a histone H3 lysine 36 (H3K36) tri-methylase that is upregulated in response to neural crest induction. Because the H3K36 di-methylase NSD3 and cytoplasmic non-histone protein methylation are necessary for neural crest development, we investigated the expression and requirement for SETD2 in the neural crest. SetD2 is expressed throughout the chick blastoderm beginning at gastrulation. Subsequently, SetD2 mRNA becomes restricted to the neural plate, where it is strongly and dynamically expressed as neural tissue is regionalized and cell fate decisions are made. This includes expression in premigratory neural crest cells, which is downregulated prior to migration. Likely due to the early onset of its expression, SETD2 morpholino knockdown does not significantly alter premigratory Sox10 expression or neural crest migration; however, both are disrupted by a methyltransferase mutant SETD2 construct. These results suggest that SETD2 activity is essential for early neural crest development, further demonstrating that lysine methylation is an important mechanism regulating the neural crest.
Collapse
Affiliation(s)
- Julaine Roffers-Agarwal
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, USA.,Developmental Biology Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Kevin A Lidberg
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, USA.,Developmental Biology Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Laura S Gammill
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, USA.,Developmental Biology Center, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
13
|
Landy MA, Goyal M, Casey KM, Liu C, Lai HC. Loss of Prdm12 during development, but not in mature nociceptors, causes defects in pain sensation. Cell Rep 2021; 34:108913. [PMID: 33789102 PMCID: PMC8048104 DOI: 10.1016/j.celrep.2021.108913] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 02/11/2021] [Accepted: 03/05/2021] [Indexed: 12/30/2022] Open
Abstract
Prdm12 is a key transcription factor in nociceptor neurogenesis. Mutations of Prdm12 cause congenital insensitivity to pain (CIP) from failure of nociceptor development. However, precisely how deletion of Prdm12 during development or adulthood affects nociception is unknown. Here, we employ tissue- and temporal-specific knockout mouse models to test the function of Prdm12 during development and in adulthood. We find that constitutive loss of Prdm12 causes deficiencies in proliferation during sensory neurogenesis. We also demonstrate that conditional knockout from dorsal root ganglia (DRGs) during embryogenesis causes defects in nociception. In contrast, we find that, in adult DRGs, Prdm12 is dispensable for most pain-sensation and injury-induced hypersensitivity. Using transcriptomic analysis, we find mostly unique changes in adult Prdm12 knockout DRGs compared with embryonic knockout and that PRDM12 is likely a transcriptional activator in the adult. Overall, we find that the function of PRDM12 changes over developmental time.
Collapse
Affiliation(s)
- Mark A Landy
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Megan Goyal
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Katherine M Casey
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Chen Liu
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA; Department of Internal Medicine, Hypothalamic Research Center, Dallas, TX 75390, USA
| | - Helen C Lai
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
14
|
Di Tullio F, Schwarz M, Zorgati H, Mzoughi S, Guccione E. The duality of PRDM proteins: epigenetic and structural perspectives. FEBS J 2021; 289:1256-1275. [PMID: 33774927 DOI: 10.1111/febs.15844] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 02/26/2021] [Accepted: 03/25/2021] [Indexed: 12/13/2022]
Abstract
PRDF1 and RIZ1 homology domain containing (PRDMs) are a subfamily of Krüppel-like zinc finger proteins controlling key processes in metazoan development and in cancer. PRDMs exhibit unique dualities: (a) PR domain/ZNF arrays-their structure combines a SET-like domain known as a PR domain, typically found in methyltransferases, with a variable array of C2H2 zinc fingers (ZNF) characteristic of DNA-binding transcription factors; (b) transcriptional activators/repressors-their physiological function is context- and cell-dependent; mechanistically, some PRDMs have a PKMT activity and directly catalyze histone lysine methylation, while others are rather pseudomethyltransferases and act by recruiting transcriptional cofactors; (c) oncogenes/tumor suppressors-their pathological function depends on the specific PRDM isoform expressed during tumorigenesis. This duality is well known as the 'Yin and Yang' of PRDMs and involves a complex regulation of alternative splicing or alternative promoter usage, to generate full-length or PR-deficient isoforms with opposing functions in cancer. In conclusion, once their dualities are fully appreciated, PRDMs represent a promising class of targets in oncology by virtue of their widespread upregulation across multiple tumor types and their somatic dispensability, conferring a broad therapeutic window and limited toxic side effects. The recent discovery of a first-in-class compound able to inhibit PRDM9 activity has paved the way for the identification of further small molecular inhibitors able to counteract PRDM oncogenic activity.
Collapse
Affiliation(s)
- Federico Di Tullio
- Department of Oncological Sciences and Pharmacological Sciences, Center for Therapeutics Discovery, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Megan Schwarz
- Department of Oncological Sciences and Pharmacological Sciences, Center for Therapeutics Discovery, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Habiba Zorgati
- Department of Oncological Sciences and Pharmacological Sciences, Center for Therapeutics Discovery, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Slim Mzoughi
- Department of Oncological Sciences and Pharmacological Sciences, Center for Therapeutics Discovery, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ernesto Guccione
- Department of Oncological Sciences and Pharmacological Sciences, Center for Therapeutics Discovery, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
15
|
Profiling NSD3-dependent neural crest gene expression reveals known and novel candidate regulatory factors. Dev Biol 2021; 475:118-130. [PMID: 33705737 DOI: 10.1016/j.ydbio.2021.02.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/17/2021] [Accepted: 02/21/2021] [Indexed: 12/17/2022]
Abstract
The lysine methyltransferase NSD3 is required for the expression of key neural crest transcription factors and the migration of neural crest cells. Nevertheless, a complete view of the genes dependent upon NSD3 for expression and the developmental processes impacted by NSD3 in the neural crest was lacking. We used RNA sequencing (RNA-seq) to profile transcripts differentially expressed after NSD3 knockdown in chick premigratory neural crest cells, identifying 674 genes. Gene Ontology and gene set enrichment analyses further support a requirement for NSD3 during neural crest development and show that NSD3 knockdown also upregulates ribosome biogenesis. To validate our results, we selected three genes not previously associated with neural crest development, Astrotactin 1 (Astn1), Dispatched 3 (Disp3), and Tropomyosin 1 (Tpm1). Using whole mount in situ hybridization, we show that premigratory neural crest cells express these genes and that NSD3 knockdown downregulates (Astn1 and Disp3) and upregulates (Tpm1) their expression, consistent with RNA-seq results. Altogether, this study identifies novel putative regulators of neural crest development and provides insight into the transcriptional consequences of NSD3 in the neural crest, with implications for cancer.
Collapse
|
16
|
Vermeiren S, Bellefroid EJ, Desiderio S. Vertebrate Sensory Ganglia: Common and Divergent Features of the Transcriptional Programs Generating Their Functional Specialization. Front Cell Dev Biol 2020; 8:587699. [PMID: 33195244 PMCID: PMC7649826 DOI: 10.3389/fcell.2020.587699] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 09/08/2020] [Indexed: 12/13/2022] Open
Abstract
Sensory fibers of the peripheral nervous system carry sensation from specific sense structures or use different tissues and organs as receptive fields, and convey this information to the central nervous system. In the head of vertebrates, each cranial sensory ganglia and associated nerves perform specific functions. Sensory ganglia are composed of different types of specialized neurons in which two broad categories can be distinguished, somatosensory neurons relaying all sensations that are felt and visceral sensory neurons sensing the internal milieu and controlling body homeostasis. While in the trunk somatosensory neurons composing the dorsal root ganglia are derived exclusively from neural crest cells, somato- and visceral sensory neurons of cranial sensory ganglia have a dual origin, with contributions from both neural crest and placodes. As most studies on sensory neurogenesis have focused on dorsal root ganglia, our understanding of the molecular mechanisms underlying the embryonic development of the different cranial sensory ganglia remains today rudimentary. However, using single-cell RNA sequencing, recent studies have made significant advances in the characterization of the neuronal diversity of most sensory ganglia. Here we summarize the general anatomy, function and neuronal diversity of cranial sensory ganglia. We then provide an overview of our current knowledge of the transcriptional networks controlling neurogenesis and neuronal diversification in the developing sensory system, focusing on cranial sensory ganglia, highlighting specific aspects of their development and comparing it to that of trunk sensory ganglia.
Collapse
Affiliation(s)
- Simon Vermeiren
- ULB Neuroscience Institute, Université Libre de Bruxelles, Gosselies, Belgium
| | - Eric J Bellefroid
- ULB Neuroscience Institute, Université Libre de Bruxelles, Gosselies, Belgium
| | - Simon Desiderio
- Institute for Neurosciences of Montpellier, INSERM U1051, University of Montpellier, Montpellier, France
| |
Collapse
|
17
|
Rahman MM, Kim IS, Ahn D, Tae HJ, Park BY. PR domaincontaining protein 12 (prdm12) is a downstream target of the transcription factor zic1 during cellular differentiation in the central nervous system: PR domain containing protein is the right form. Int J Dev Neurosci 2020; 80:528-537. [PMID: 32640092 DOI: 10.1002/jdn.10048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 06/27/2020] [Accepted: 06/29/2020] [Indexed: 11/05/2022] Open
Abstract
Transcription factor zic1 is an important regulator of neural plate patterning, formation of neural crest and cerebellar development, where its main function is neuronal cell differentiation. Among the genes identified, PR domain-containing 12 (prdm12) is a member of the prdm family and is expressed in the placode domain in the neurula stage. prdm12 is distinctly expressed in the dorsal part of the midbrain, trigeminal ganglion, and the motor neuron in the spinal cord. prdm12 knockdown results in the ventralization of the neural tube. zic1 knockdown results in the reduction of prdm12 expression in the midbrain, motor neuron and trigeminal ganglion, and overexpression of zic1 results in the expansion of prdm12 expression in the midbrain. zic1-activated wnt signaling is also a regulator of prdm12 expression in the midbrain. We propose that prdm12 is the downstream of zic1 and a novel player in the gene regulatory network controlling brain cell differentiation, along with some ganglions in Xenopus.
Collapse
Affiliation(s)
- Md Mahfujur Rahman
- College of Veterinary Medicine and Biosafety Research Institute, Jeonbuk National University, Iksan, Republic of Korea.,Department of Medicine, Faculty of Veterinary, Animal and Biomedical Sciences, Sylhet Agricultural University, Sylhet, Bangladesh
| | - In-Shik Kim
- College of Veterinary Medicine and Biosafety Research Institute, Jeonbuk National University, Iksan, Republic of Korea
| | - Dongchoon Ahn
- College of Veterinary Medicine and Biosafety Research Institute, Jeonbuk National University, Iksan, Republic of Korea
| | - Hyun-Jin Tae
- College of Veterinary Medicine and Biosafety Research Institute, Jeonbuk National University, Iksan, Republic of Korea
| | - Byung-Yong Park
- College of Veterinary Medicine and Biosafety Research Institute, Jeonbuk National University, Iksan, Republic of Korea
| |
Collapse
|
18
|
PRDM12: New Opportunity in Pain Research. Trends Mol Med 2020; 26:895-897. [PMID: 32828702 DOI: 10.1016/j.molmed.2020.07.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 07/18/2020] [Accepted: 07/21/2020] [Indexed: 01/09/2023]
Abstract
PRDM12 is a newly identified causative gene for a type of congenital insensitivity to pain disorder, which is characterized by the inability to perceive pain. Here, we discuss the (patho)physiology of PRDM12 function and the opportunities and challenges those data provide for novel therapeutic approaches in various pain disorders.
Collapse
|
19
|
Esmaeili M, Blythe SA, Tobias JW, Zhang K, Yang J, Klein PS. Chromatin accessibility and histone acetylation in the regulation of competence in early development. Dev Biol 2020; 462:20-35. [PMID: 32119833 PMCID: PMC7225061 DOI: 10.1016/j.ydbio.2020.02.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 01/29/2020] [Accepted: 02/25/2020] [Indexed: 02/06/2023]
Abstract
As development proceeds, inductive cues are interpreted by competent tissues in a spatially and temporally restricted manner. While key inductive signaling pathways within competent cells are well-described at a molecular level, the mechanisms by which tissues lose responsiveness to inductive signals are not well understood. Localized activation of Wnt signaling before zygotic gene activation in Xenopus laevis leads to dorsal development, but competence to induce dorsal genes in response to Wnts is lost by the late blastula stage. We hypothesize that loss of competence is mediated by changes in histone modifications leading to a loss of chromatin accessibility at the promoters of Wnt target genes. We use ATAC-seq to evaluate genome-wide changes in chromatin accessibility across several developmental stages. Based on overlap with p300 binding, we identify thousands of putative cis-regulatory elements at the gastrula stage, including sites that lose accessibility by the end of gastrulation and are enriched for pluripotency factor binding motifs. Dorsal Wnt target gene promoters are not accessible after the loss of competence in the early gastrula while genes involved in mesoderm and neural crest development maintain accessibility at their promoters. Inhibition of histone deacetylases increases acetylation at the promoters of dorsal Wnt target genes and extends competence for dorsal gene induction by Wnt signaling. Histone deacetylase inhibition, however, is not sufficient to extend competence for mesoderm or neural crest induction. These data suggest that chromatin state regulates the loss of competence to inductive signals in a context-dependent manner.
Collapse
Affiliation(s)
- Melody Esmaeili
- Cell and Molecular Biology Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Shelby A Blythe
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
| | - John W Tobias
- Penn Genomic Analysis Core and Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kai Zhang
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Jing Yang
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| | - Peter S Klein
- Cell and Molecular Biology Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Departments of Medicine (Hematology-Oncology) and Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
20
|
Bartesaghi L, Wang Y, Fontanet P, Wanderoy S, Berger F, Wu H, Akkuratova N, Bouçanova F, Médard JJ, Petitpré C, Landy MA, Zhang MD, Harrer P, Stendel C, Stucka R, Dusl M, Kastriti ME, Croci L, Lai HC, Consalez GG, Pattyn A, Ernfors P, Senderek J, Adameyko I, Lallemend F, Hadjab S, Chrast R. PRDM12 Is Required for Initiation of the Nociceptive Neuron Lineage during Neurogenesis. Cell Rep 2020; 26:3484-3492.e4. [PMID: 30917305 PMCID: PMC7676307 DOI: 10.1016/j.celrep.2019.02.098] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 12/06/2018] [Accepted: 02/25/2019] [Indexed: 12/21/2022] Open
Abstract
The sensation of pain is essential for the preservation of the functional integrity of the body. However, the key molecular regulators necessary for the initiation of the development of pain-sensing neurons have remained largely unknown. Here, we report that, in mice, inactivation of the transcriptional regulator PRDM12, which is essential for pain perception in humans, results in a complete absence of the nociceptive lineage, while proprioceptive and touch-sensitive neurons remain. Mechanistically, our data reveal that PRDM12 is required for initiation of neurogenesis and activation of a cascade of downstream pro-neuronal transcription factors, including NEUROD1, BRN3A, and ISL1, in the nociceptive lineage while it represses alternative fates other than nociceptors in progenitor cells. Our results thus demonstrate that PRDM12 is necessary for the generation of the entire lineage of pain-initiating neurons. The sensation of pain, temperature, and itch by neurons of the nociceptive lineage is essential for animal survival. Bartesaghi et al. report that the transcriptional regulator PRDM12 is indispensable in neural crest cells (NCCs) for the initiation of the sensory neuronal differentiation program that generates the entire nociceptive lineage.
Collapse
Affiliation(s)
- Luca Bartesaghi
- Department of Neuroscience, Karolinska Institutet, Stockholm, 17165, Sweden; Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, 17165, Sweden
| | - Yiqiao Wang
- Department of Neuroscience, Karolinska Institutet, Stockholm, 17165, Sweden
| | - Paula Fontanet
- Department of Neuroscience, Karolinska Institutet, Stockholm, 17165, Sweden
| | - Simone Wanderoy
- Department of Neuroscience, Karolinska Institutet, Stockholm, 17165, Sweden
| | - Finja Berger
- Department of Neuroscience, Karolinska Institutet, Stockholm, 17165, Sweden; Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, 17165, Sweden
| | - Haohao Wu
- Department of Neuroscience, Karolinska Institutet, Stockholm, 17165, Sweden
| | - Natalia Akkuratova
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, 17165, Sweden; Institute of Translational Biomedicine, Saint Petersburg State University, St. Petersburg, 199034, Russia
| | - Filipa Bouçanova
- Department of Neuroscience, Karolinska Institutet, Stockholm, 17165, Sweden; Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, 17165, Sweden
| | - Jean-Jacques Médard
- Department of Neuroscience, Karolinska Institutet, Stockholm, 17165, Sweden; Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, 17165, Sweden
| | - Charles Petitpré
- Department of Neuroscience, Karolinska Institutet, Stockholm, 17165, Sweden
| | - Mark A Landy
- Department of Neuroscience, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Ming-Dong Zhang
- Department of Medical Biochemistry and Biophysics, Division of Molecular Neurobiology, Karolinska Institutet, Stockholm, 17165, Sweden
| | - Philip Harrer
- Friedrich-Baur-Institute at the Department of Neurology, University Hospital, LMU Munich, Munich, Germany
| | - Claudia Stendel
- Friedrich-Baur-Institute at the Department of Neurology, University Hospital, LMU Munich, Munich, Germany
| | - Rolf Stucka
- Friedrich-Baur-Institute at the Department of Neurology, University Hospital, LMU Munich, Munich, Germany
| | - Marina Dusl
- Friedrich-Baur-Institute at the Department of Neurology, University Hospital, LMU Munich, Munich, Germany
| | - Maria Eleni Kastriti
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, 17165, Sweden; Center for Brain Research, Medical University Vienna, Vienna, Austria
| | - Laura Croci
- Center for Brain Research, Medical University Vienna, Vienna, Austria
| | - Helen C Lai
- Department of Neuroscience, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | | | - Alexandre Pattyn
- Institute for Neurosciences of Montpellier, INSERM, UMR1051, Hôpital Saint-Eloi, Montpellier, 34000, France
| | - Patrik Ernfors
- Department of Medical Biochemistry and Biophysics, Division of Molecular Neurobiology, Karolinska Institutet, Stockholm, 17165, Sweden
| | - Jan Senderek
- Friedrich-Baur-Institute at the Department of Neurology, University Hospital, LMU Munich, Munich, Germany
| | - Igor Adameyko
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, 17165, Sweden; Center for Brain Research, Medical University Vienna, Vienna, Austria
| | - Francois Lallemend
- Department of Neuroscience, Karolinska Institutet, Stockholm, 17165, Sweden.
| | - Saida Hadjab
- Department of Neuroscience, Karolinska Institutet, Stockholm, 17165, Sweden.
| | - Roman Chrast
- Department of Neuroscience, Karolinska Institutet, Stockholm, 17165, Sweden; Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, 17165, Sweden.
| |
Collapse
|
21
|
Meyer DN, Crofts EJ, Akemann C, Gurdziel K, Farr R, Baker BB, Weber D, Baker TR. Developmental exposure to Pb 2+ induces transgenerational changes to zebrafish brain transcriptome. CHEMOSPHERE 2020; 244:125527. [PMID: 31816550 PMCID: PMC7015790 DOI: 10.1016/j.chemosphere.2019.125527] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 11/27/2019] [Accepted: 11/30/2019] [Indexed: 05/24/2023]
Abstract
Lead (Pb2+) is a major public health hazard for urban children, with profound and well-characterized developmental and behavioral implications across the lifespan. The ability of early Pb2+ exposure to induce epigenetic changes is well-established, suggesting that Pb2+-induced neurobehavioral deficits may be heritable across generations. Understanding the long-term and multigenerational repercussions of lead exposure is crucial for clarifying both the genotypic alterations behind these behavioral outcomes and the potential mechanism of heritability. To study this, zebrafish (Danio rerio) embryos (<2 h post fertilization; EK strain) were exposed for 24 h to waterborne Pb2+ at a concentration of 10 μM. This exposed F0 generation was raised to adulthood and spawned to produce the F1 generation, which was subsequently spawned to produce the F2 generation. Previous avoidance conditioning studies determined that a 10 μM Pb2+ dose resulted in learning impairments persisting through the F2 generation. RNA was extracted from control- and 10 μM Pb2+-lineage F2 brains, (n = 10 for each group), sequenced, and transcript expression was quantified utilizing Quant-Seq. 648 genes were differentially expressed in the brains of F2 lead-lineage fish versus F2 control-lineage fish. Pathway analysis revealed altered genes in processes including synaptic function and plasticity, neurogenesis, endocrine homeostasis, and epigenetic modification, all of which are implicated in lead-induced neurobehavioral deficits and/or their inheritance. These data will inform future investigations to elucidate the mechanism of adult-onset and transgenerational health effects of developmental lead exposure.
Collapse
Affiliation(s)
- Danielle N Meyer
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI, USA; Institute of Environmental Health Sciences, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Emily J Crofts
- Institute of Environmental Health Sciences, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Camille Akemann
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI, USA; Institute of Environmental Health Sciences, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Katherine Gurdziel
- Applied Genome Technology Center, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Rebecca Farr
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Bridget B Baker
- Institute of Environmental Health Sciences, School of Medicine, Wayne State University, Detroit, MI, USA; Division of Laboratory Animal Resources, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Daniel Weber
- Children's Environmental Health Sciences Core Center, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Tracie R Baker
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI, USA; Institute of Environmental Health Sciences, School of Medicine, Wayne State University, Detroit, MI, USA.
| |
Collapse
|
22
|
Prajapati RS, Hintze M, Streit A. PRDM1 controls the sequential activation of neural, neural crest and sensory progenitor determinants. Development 2019; 146:dev.181107. [PMID: 31806661 DOI: 10.1242/dev.181107] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 11/27/2019] [Indexed: 12/25/2022]
Abstract
During early embryogenesis, the ectoderm is rapidly subdivided into neural, neural crest and sensory progenitors. How the onset of lineage determinants and the loss of pluripotency markers are temporally and spatially coordinated in vivo is still debated. Here, we identify a crucial role for the transcription factor PRDM1 in the orderly transition from epiblast to defined neural lineages in chick. PRDM1 is initially expressed broadly in the entire epiblast, but becomes gradually restricted as cell fates are specified. We find that PRDM1 is required for the loss of some pluripotency markers and the onset of neural, neural crest and sensory progenitor specifier genes. PRDM1 directly activates their expression by binding to their promoter regions and recruiting the histone demethylase Kdm4a to remove repressive histone marks. However, once neural lineage determinants become expressed, they in turn repress PRDM1, whereas prolonged PRDM1 expression inhibits neural, neural crest and sensory progenitor genes, suggesting that its downregulation is necessary for cells to maintain their identity. Therefore, PRDM1 plays multiple roles during ectodermal cell fate allocation.
Collapse
Affiliation(s)
- Ravindra S Prajapati
- Centre for Craniofacial & Regenerative Biology, Faculty of Dental, Oral and Craniofacial Sciences, King's College London, London SE1 9RT, UK
| | - Mark Hintze
- Centre for Craniofacial & Regenerative Biology, Faculty of Dental, Oral and Craniofacial Sciences, King's College London, London SE1 9RT, UK
| | - Andrea Streit
- Centre for Craniofacial & Regenerative Biology, Faculty of Dental, Oral and Craniofacial Sciences, King's College London, London SE1 9RT, UK
| |
Collapse
|
23
|
Yildiz O, Downes GB, Sagerström CG. Zebrafish prdm12b acts independently of nkx6.1 repression to promote eng1b expression in the neural tube p1 domain. Neural Dev 2019; 14:5. [PMID: 30813944 PMCID: PMC6391800 DOI: 10.1186/s13064-019-0129-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 02/14/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Functioning of the adult nervous system depends on the establishment of neural circuits during embryogenesis. In vertebrates, neurons that make up motor circuits form in distinct domains along the dorsoventral axis of the neural tube. Each domain is characterized by a unique combination of transcription factors (TFs) that promote a specific fate, while repressing fates of adjacent domains. The prdm12 TF is required for the expression of eng1b and the generation of V1 interneurons in the p1 domain, but the details of its function remain unclear. METHODS We used CRISPR/Cas9 to generate the first germline mutants for prdm12 and employed this resource, together with classical luciferase reporter assays and co-immunoprecipitation experiments, to study prdm12b function in zebrafish. We also generated germline mutants for bhlhe22 and nkx6.1 to examine how these TFs act with prdm12b to control p1 formation. RESULTS We find that prdm12b mutants lack eng1b expression in the p1 domain and also possess an abnormal touch-evoked escape response. Using luciferase reporter assays, we demonstrate that Prdm12b acts as a transcriptional repressor. We also show that the Bhlhe22 TF binds via the Prdm12b zinc finger domain to form a complex. However, bhlhe22 mutants display normal eng1b expression in the p1 domain. While prdm12 has been proposed to promote p1 fates by repressing expression of the nkx6.1 TF, we do not observe an expansion of the nkx6.1 domain upon loss of prdm12b function, nor is eng1b expression restored upon simultaneous loss of prdm12b and nkx6.1. CONCLUSIONS We conclude that prdm12b germline mutations produce a phenotype that is indistinguishable from that of morpholino-mediated loss of prdm12 function. In terms of prdm12b function, our results indicate that Prdm12b acts as transcriptional repressor and interacts with both EHMT2/G9a and Bhlhe22. However, bhlhe22 function is not required for eng1b expression in vivo, perhaps indicating that other bhlh genes can compensate during embryogenesis. Lastly, we do not find evidence for nkx6.1 and prdm12b acting as a repressive pair in formation of the p1 domain - suggesting that prdm12b is not solely required to repress non-p1 fates, but is specifically needed to promote p1 fates.
Collapse
Affiliation(s)
- Ozge Yildiz
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, 364 Plantation St/LRB815, Worcester, MA 01605 USA
| | - Gerald B. Downes
- Department of Biology, University of Massachusetts, Amherst, MA 01003 USA
| | - Charles G. Sagerström
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, 364 Plantation St/LRB815, Worcester, MA 01605 USA
| |
Collapse
|
24
|
Rogers CD, Nie S. Specifying neural crest cells: From chromatin to morphogens and factors in between. WILEY INTERDISCIPLINARY REVIEWS. DEVELOPMENTAL BIOLOGY 2018; 7:e322. [PMID: 29722151 PMCID: PMC6215528 DOI: 10.1002/wdev.322] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 03/26/2018] [Accepted: 03/27/2018] [Indexed: 12/16/2022]
Abstract
Neural crest (NC) cells are a stem-like multipotent population of progenitor cells that are present in vertebrate embryos, traveling to various regions in the developing organism. Known as the "fourth germ layer," these cells originate in the ectoderm between the neural plate (NP), which will become the brain and spinal cord, and nonneural tissues that will become the skin and the sensory organs. NC cells can differentiate into more than 30 different derivatives in response to the appropriate signals including, but not limited to, craniofacial bone and cartilage, sensory nerves and ganglia, pigment cells, and connective tissue. The molecular and cellular mechanisms that control the induction and specification of NC cells include epigenetic control, multiple interactive and redundant transcriptional pathways, secreted signaling molecules, and adhesion molecules. NC cells are important not only because they transform into a wide variety of tissue types, but also because their ability to detach from their epithelial neighbors and migrate throughout developing embryos utilizes mechanisms similar to those used by metastatic cancer cells. In this review, we discuss the mechanisms required for the induction and specification of NC cells in various vertebrate species, focusing on the roles of early morphogenesis, cell adhesion, signaling from adjacent tissues, and the massive transcriptional network that controls the formation of these amazing cells. This article is categorized under: Nervous System Development > Vertebrates: General Principles Gene Expression and Transcriptional Hierarchies > Regulatory Mechanisms Gene Expression and Transcriptional Hierarchies > Gene Networks and Genomics Signaling Pathways > Cell Fate Signaling.
Collapse
Affiliation(s)
- Crystal D. Rogers
- Department of Biology, College of Science and Mathematics, California State University Northridge, Northridge, California
| | - Shuyi Nie
- School of Biological Sciences and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia
| |
Collapse
|
25
|
Pla P, Monsoro-Burq AH. The neural border: Induction, specification and maturation of the territory that generates neural crest cells. Dev Biol 2018; 444 Suppl 1:S36-S46. [PMID: 29852131 DOI: 10.1016/j.ydbio.2018.05.018] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 05/23/2018] [Accepted: 05/23/2018] [Indexed: 11/17/2022]
Abstract
The neural crest is induced at the edge between the neural plate and the nonneural ectoderm, in an area called the neural (plate) border, during gastrulation and neurulation. In recent years, many studies have explored how this domain is patterned, and how the neural crest is induced within this territory, that also participates to the prospective dorsal neural tube, the dorsalmost nonneural ectoderm, as well as placode derivatives in the anterior area. This review highlights the tissue interactions, the cell-cell signaling and the molecular mechanisms involved in this dynamic spatiotemporal patterning, resulting in the induction of the premigratory neural crest. Collectively, these studies allow building a complex neural border and early neural crest gene regulatory network, mostly composed by transcriptional regulations but also, more recently, including novel signaling interactions.
Collapse
Affiliation(s)
- Patrick Pla
- Univ. Paris Sud, Université Paris Saclay, CNRS UMR 3347, INSERM U1021, Centre Universitaire, 15, rue Georges Clémenceau, F-91405 Orsay, France; Institut Curie Research Division, PSL Research University, CNRS UMR 3347, INSERM U1021, F-91405 Orsay, France
| | - Anne H Monsoro-Burq
- Univ. Paris Sud, Université Paris Saclay, CNRS UMR 3347, INSERM U1021, Centre Universitaire, 15, rue Georges Clémenceau, F-91405 Orsay, France; Institut Curie Research Division, PSL Research University, CNRS UMR 3347, INSERM U1021, F-91405 Orsay, France; Institut Universitaire de France, F-75005, Paris.
| |
Collapse
|
26
|
Lin H, Zhu X, Chen G, Song L, Gao L, Khand AA, Chen Y, Lin G, Tao Q. KDM3A-mediated demethylation of histone H3 lysine 9 facilitates the chromatin binding of Neurog2 during neurogenesis. Development 2017; 144:3674-3685. [DOI: 10.1242/dev.144113] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Accepted: 08/25/2017] [Indexed: 12/26/2022]
Abstract
Neurog2 is a crucial regulator of neuronal fate specification and differentiation in vivo and in vitro. However, it remains unclear how Neurog2 transactivates neuronal genes that are silenced by repressive chromatin. Here, we provide evidence that the histone H3 lysine 9 demethylase KDM3A facilitates the Xenopus Neurog2 (formerly known as Xngnr1) chromatin accessibility during neuronal transcription. Loss-of-function analyses reveal that KDM3A is not required for the transition of naive ectoderm to neural progenitor cells but is essential for primary neuron formation. ChIP series followed by qPCR analyses reveal that Neurog2 promotes the removal of the repressive H3K9me2 marks and addition of active histone marks, including H3K27ac and H3K4me3, at the NeuroD1 and Tubb2b promoters; this activity depends on the presence of KDM3A because Neurog2, via its C-terminal domain, interacts with KDM3A. Interestingly, KDM3A is dispensable for the neuronal transcription initiated by Ascl1, a proneural factor related to neurogenin in the bHLH family. In summary, our findings uncover a crucial role for histone H3K9 demethylation during Neurog2-mediated neuronal transcription and help in the understanding of the different activities of Neurog2 and Ascl1 in initiating neuronal development.
Collapse
Affiliation(s)
- Hao Lin
- MOE Key Laboratory of Protein Sciences, Tsinghua University School of Life Sciences, Beijing, China 100084
| | - Xuechen Zhu
- MOE Key Laboratory of Protein Sciences, Tsinghua University School of Life Sciences, Beijing, China 100084
| | - Geng Chen
- MOE Key Laboratory of Protein Sciences, Tsinghua University School of Life Sciences, Beijing, China 100084
| | - Lei Song
- MOE Key Laboratory of Protein Sciences, Tsinghua University School of Life Sciences, Beijing, China 100084
| | - Li Gao
- MOE Key Laboratory of Protein Sciences, Tsinghua University School of Life Sciences, Beijing, China 100084
| | - Aftab A. Khand
- MOE Key Laboratory of Protein Sciences, Tsinghua University School of Life Sciences, Beijing, China 100084
| | - Ying Chen
- Tongji University School of Life Sciences and Technology, Shanghai, China 200092
| | - Gufa Lin
- Tongji University School of Life Sciences and Technology, Shanghai, China 200092
| | - Qinghua Tao
- MOE Key Laboratory of Protein Sciences, Tsinghua University School of Life Sciences, Beijing, China 100084
| |
Collapse
|
27
|
Torroglosa A, Alves MM, Fernández RM, Antiñolo G, Hofstra RM, Borrego S. Epigenetics in ENS development and Hirschsprung disease. Dev Biol 2016; 417:209-16. [PMID: 27321561 DOI: 10.1016/j.ydbio.2016.06.017] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Revised: 06/10/2016] [Accepted: 06/13/2016] [Indexed: 12/18/2022]
Abstract
Hirschsprung disease (HSCR, OMIM 142623) is a neurocristopathy caused by a failure of the enteric nervous system (ENS) progenitors derived from neural crest cells (NCCs), to migrate, proliferate, differentiate or survive to and within the gastrointestinal tract, resulting in aganglionosis in the distal colon. The formation of the ENS is a complex process, which is regulated by a large range of molecules and signalling pathways involving both the NCCs and the intestinal environment. This tightly regulated process needs correct regulation of the expression of ENS specific genes. Alterations in the expression of these genes can have dramatic consequences. Several mechanisms that control the expression of genes have been described, such as DNA modification (epigenetic mechanisms), regulation of transcription (transcription factor, enhancers, repressors and silencers), post-transcriptional regulation (3'UTR and miRNAs) and regulation of translation. In this review, we focus on the epigenetic DNA modifications that have been described so far in the context of the ENS development. Moreover we describe the changes that are found in relation to the onset of HSCR.
Collapse
Affiliation(s)
- A Torroglosa
- Department of Genetics, Reproduction and Fetal Medicine, Institute of Biomedicine of Seville (IBIS), University Hospital Virgen del Rocío/CSIC/University of Seville, Seville, Spain; Centre for Biomedical Network Research on Rare Diseases (CIBERER), Seville, Spain
| | - M M Alves
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - R M Fernández
- Department of Genetics, Reproduction and Fetal Medicine, Institute of Biomedicine of Seville (IBIS), University Hospital Virgen del Rocío/CSIC/University of Seville, Seville, Spain; Centre for Biomedical Network Research on Rare Diseases (CIBERER), Seville, Spain
| | - G Antiñolo
- Department of Genetics, Reproduction and Fetal Medicine, Institute of Biomedicine of Seville (IBIS), University Hospital Virgen del Rocío/CSIC/University of Seville, Seville, Spain; Centre for Biomedical Network Research on Rare Diseases (CIBERER), Seville, Spain
| | - R M Hofstra
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands; Stem Cells and Regenerative Medicine, Birth Defects Research Centre UCL Institute of Child Health, London, UK
| | - S Borrego
- Department of Genetics, Reproduction and Fetal Medicine, Institute of Biomedicine of Seville (IBIS), University Hospital Virgen del Rocío/CSIC/University of Seville, Seville, Spain; Centre for Biomedical Network Research on Rare Diseases (CIBERER), Seville, Spain.
| |
Collapse
|
28
|
Nagy V, Cole T, Van Campenhout C, Khoung TM, Leung C, Vermeiren S, Novatchkova M, Wenzel D, Cikes D, Polyansky AA, Kozieradzki I, Meixner A, Bellefroid EJ, Neely GG, Penninger JM. The evolutionarily conserved transcription factor PRDM12 controls sensory neuron development and pain perception. Cell Cycle 2016; 14:1799-808. [PMID: 25891934 DOI: 10.1080/15384101.2015.1036209] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
PR homology domain-containing member 12 (PRDM12) belongs to a family of conserved transcription factors implicated in cell fate decisions. Here we show that PRDM12 is a key regulator of sensory neuronal specification in Xenopus. Modeling of human PRDM12 mutations that cause hereditary sensory and autonomic neuropathy (HSAN) revealed remarkable conservation of the mutated residues in evolution. Expression of wild-type human PRDM12 in Xenopus induced the expression of sensory neuronal markers, which was reduced using various human PRDM12 mutants. In Drosophila, we identified Hamlet as the functional PRDM12 homolog that controls nociceptive behavior in sensory neurons. Furthermore, expression analysis of human patient fibroblasts with PRDM12 mutations uncovered possible downstream target genes. Knockdown of several of these target genes including thyrotropin-releasing hormone degrading enzyme (TRHDE) in Drosophila sensory neurons resulted in altered cellular morphology and impaired nociception. These data show that PRDM12 and its functional fly homolog Hamlet are evolutionary conserved master regulators of sensory neuronal specification and play a critical role in pain perception. Our data also uncover novel pathways in multiple species that regulate evolutionary conserved nociception.
Collapse
Key Words
- BSA, bovine serum albumin
- Brn3d, brain 3d
- CGNL1, cyclin L1
- ChIP, chromatin immunoprecipitation
- DAPI, 4′,6-diamidino-2-phenylindole
- DDK, DYKDDDDK epitope
- Drgx, dorsal root ganglia homeobox
- ECL, enhanced chemiluminescence
- En1, engrailed-1
- FDR, false discovery rate
- FPKM, fragments per kilobase exon
- GAPDH, glyceraldehyde 3-phospate dehydrogenase
- GEO, gene expression omnibus
- GFP, green fluorescent protein
- HEK293, human embryonic kidney cell 293
- HRP, horseraddish peroxidase
- HSAN, hereditary and sensory autonomic neuropathy
- Hamlet
- Hmx3, H6 family homeobox 3
- IL1R1, interleukin 1 receptor type 1
- MO, morpholino oligonucleotide
- NBT/BCIP, nitro blue tetrazolium / 5-bromo-4-chloro-3-indolyl-phosphate
- PBS, phosphate buffered saline
- PDB, protein data base
- PMID, pubmed identification.
- PRDM12
- PRDM12, PR homology domain-containing member 12
- RA, retinoic acid
- RT-qPCR, real-time quantitative polymerase chain reaction
- S1PR1, Sphi8ngosine-1-phosphate receptor 1
- SET, Su(var)3–9 and ‘Enhancer of zeste’
- Sncg, Synuclein Gamma (Breast Cancer-Specific Protein 1)
- TRH(DE), tryrotropin-releasing hormone degrading enzyme
- TRHDE
- TRHDE, tyrotropin-releasing hormone degrading enzyme
- Tlx3, T-cell leukemia homeobox 3
- nociception
- pCMV6, plasmid cytomegalovirus
- sensory neurons
Collapse
Affiliation(s)
- Vanja Nagy
- IMBA-Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria; UNSW Medicine, Sydney, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Thélie A, Desiderio S, Hanotel J, Quigley I, Van Driessche B, Rodari A, Borromeo MD, Kricha S, Lahaye F, Croce J, Cerda-Moya G, Ordoño Fernandez J, Bolle B, Lewis KE, Sander M, Pierani A, Schubert M, Johnson JE, Kintner CR, Pieler T, Van Lint C, Henningfeld KA, Bellefroid EJ, Van Campenhout C. Prdm12 specifies V1 interneurons through cross-repressive interactions with Dbx1 and Nkx6 genes in Xenopus. Development 2016; 142:3416-28. [PMID: 26443638 DOI: 10.1242/dev.121871] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
V1 interneurons are inhibitory neurons that play an essential role in vertebrate locomotion. The molecular mechanisms underlying their genesis remain, however, largely undefined. Here, we show that the transcription factor Prdm12 is selectively expressed in p1 progenitors of the hindbrain and spinal cord in the frog embryo, and that a similar restricted expression profile is observed in the nerve cord of other vertebrates as well as of the cephalochordate amphioxus. Using frog, chick and mice, we analyzed the regulation of Prdm12 and found that its expression in the caudal neural tube is dependent on retinoic acid and Pax6, and that it is restricted to p1 progenitors, due to the repressive action of Dbx1 and Nkx6-1/2 expressed in the adjacent p0 and p2 domains. Functional studies in the frog, including genome-wide identification of its targets by RNA-seq and ChIP-Seq, reveal that vertebrate Prdm12 proteins act as a general determinant of V1 cell fate, at least in part, by directly repressing Dbx1 and Nkx6 genes. This probably occurs by recruiting the methyltransferase G9a, an activity that is not displayed by the amphioxus Prdm12 protein. Together, these findings indicate that Prdm12 promotes V1 interneurons through cross-repressive interactions with Dbx1 and Nkx6 genes, and suggest that this function might have only been acquired after the split of the vertebrate and cephalochordate lineages.
Collapse
Affiliation(s)
- Aurore Thélie
- Laboratory of Developmental Genetics, Université Libre de Bruxelles (ULB), Institute of Molecular Biology and Medecine (IBMM) and ULB Neuroscience Institute, Gosselies B-6041, Belgium
| | - Simon Desiderio
- Laboratory of Developmental Genetics, Université Libre de Bruxelles (ULB), Institute of Molecular Biology and Medecine (IBMM) and ULB Neuroscience Institute, Gosselies B-6041, Belgium
| | - Julie Hanotel
- Laboratory of Developmental Genetics, Université Libre de Bruxelles (ULB), Institute of Molecular Biology and Medecine (IBMM) and ULB Neuroscience Institute, Gosselies B-6041, Belgium
| | - Ian Quigley
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | | | - Anthony Rodari
- Laboratory of Molecular Virology, ULB, IBMM, Gosselies B-6041, Belgium
| | - Mark D Borromeo
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sadia Kricha
- Laboratory of Developmental Genetics, Université Libre de Bruxelles (ULB), Institute of Molecular Biology and Medecine (IBMM) and ULB Neuroscience Institute, Gosselies B-6041, Belgium
| | - François Lahaye
- Sorbonne Universités, UPMC Université Paris 06, CNRS UMR 7009, Laboratoire de Biologie du Développement de Villefranche-sur-Mer (UMR 7009), Observatoire Océanologique de Villefranche-sur-Mer, Villefranche-sur-Mer 06230, France
| | - Jenifer Croce
- Sorbonne Universités, UPMC Université Paris 06, CNRS UMR 7009, Laboratoire de Biologie du Développement de Villefranche-sur-Mer (UMR 7009), Observatoire Océanologique de Villefranche-sur-Mer, Villefranche-sur-Mer 06230, France
| | - Gustavo Cerda-Moya
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Jesús Ordoño Fernandez
- Laboratory of Developmental Genetics, Université Libre de Bruxelles (ULB), Institute of Molecular Biology and Medecine (IBMM) and ULB Neuroscience Institute, Gosselies B-6041, Belgium
| | - Barbara Bolle
- Laboratory of Developmental Genetics, Université Libre de Bruxelles (ULB), Institute of Molecular Biology and Medecine (IBMM) and ULB Neuroscience Institute, Gosselies B-6041, Belgium
| | - Katharine E Lewis
- Department of Biology, Syracuse University, 107 College Place, Syracuse, NY 13244, USA
| | - Maike Sander
- Departments of Pediatrics and Cellular and Molecular Medicine, Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA 92093-0695, USA
| | - Alessandra Pierani
- Génétique et développement du cortex cerebral, Institut Jacques Monod, CNRS UMR 7592, Université Paris Diderot, Sorbonne Paris Cité, Paris Cedex 13 75205, France
| | - Michael Schubert
- Sorbonne Universités, UPMC Université Paris 06, CNRS UMR 7009, Laboratoire de Biologie du Développement de Villefranche-sur-Mer (UMR 7009), Observatoire Océanologique de Villefranche-sur-Mer, Villefranche-sur-Mer 06230, France
| | - Jane E Johnson
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Christopher R Kintner
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Tomas Pieler
- Department of Developmental Biochemistry, Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), University of Göttingen, 37077 Göttingen, Germany
| | - Carine Van Lint
- Laboratory of Molecular Virology, ULB, IBMM, Gosselies B-6041, Belgium
| | - Kristine A Henningfeld
- Department of Developmental Biochemistry, Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), University of Göttingen, 37077 Göttingen, Germany
| | - Eric J Bellefroid
- Laboratory of Developmental Genetics, Université Libre de Bruxelles (ULB), Institute of Molecular Biology and Medecine (IBMM) and ULB Neuroscience Institute, Gosselies B-6041, Belgium
| | - Claude Van Campenhout
- Laboratory of Developmental Genetics, Université Libre de Bruxelles (ULB), Institute of Molecular Biology and Medecine (IBMM) and ULB Neuroscience Institute, Gosselies B-6041, Belgium
| |
Collapse
|
30
|
Nahorski MS, Chen YC, Woods CG. New Mendelian Disorders of Painlessness. Trends Neurosci 2015; 38:712-724. [DOI: 10.1016/j.tins.2015.08.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 08/28/2015] [Accepted: 08/31/2015] [Indexed: 02/08/2023]
|
31
|
Zannino DA, Sagerström CG. An emerging role for prdm family genes in dorsoventral patterning of the vertebrate nervous system. Neural Dev 2015; 10:24. [PMID: 26499851 PMCID: PMC4620005 DOI: 10.1186/s13064-015-0052-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 10/13/2015] [Indexed: 12/13/2022] Open
Abstract
The embryonic vertebrate neural tube is divided along its dorsoventral (DV) axis into eleven molecularly discrete progenitor domains. Each of these domains gives rise to distinct neuronal cell types; the ventral-most six domains contribute to motor circuits, while the five dorsal domains contribute to sensory circuits. Following the initial neurogenesis step, these domains also generate glial cell types—either astrocytes or oligodendrocytes. This DV pattern is initiated by two morphogens—Sonic Hedgehog released from notochord and floor plate and Bone Morphogenetic Protein produced in the roof plate—that act in concentration gradients to induce expression of genes along the DV axis. Subsequently, these DV-restricted genes cooperate to define progenitor domains and to control neuronal cell fate specification and differentiation in each domain. Many genes involved in this process have been identified, but significant gaps remain in our understanding of the underlying genetic program. Here we review recent work identifying members of the Prdm gene family as novel regulators of DV patterning in the neural tube. Many Prdm proteins regulate transcription by controlling histone modifications (either via intrinsic histone methyltransferase activity, or by recruiting histone modifying enzymes). Prdm genes are expressed in spatially restricted domains along the DV axis of the neural tube and play important roles in the specification of progenitor domains, as well as in the subsequent differentiation of motor neurons and various types of interneurons. Strikingly, Prdm proteins appear to function by binding to, and modulating the activity of, other transcription factors (particularly bHLH proteins). The identity of key transcription factors in DV patterning of the neural tube has been elucidated previously (e.g. the nkx, bHLH and pax families), but it now appears that an additional family is also required and that it acts in a potentially novel manner.
Collapse
Affiliation(s)
- Denise A Zannino
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, 364 Plantation St./LRB815, Worcester, MA, 01605-2324, USA.
| | - Charles G Sagerström
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, 364 Plantation St./LRB815, Worcester, MA, 01605-2324, USA.
| |
Collapse
|
32
|
Gallagher SJ, Tiffen JC, Hersey P. Histone Modifications, Modifiers and Readers in Melanoma Resistance to Targeted and Immune Therapy. Cancers (Basel) 2015; 7:1959-82. [PMID: 26426052 PMCID: PMC4695870 DOI: 10.3390/cancers7040870] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 09/17/2015] [Accepted: 09/18/2015] [Indexed: 02/06/2023] Open
Abstract
The treatment of melanoma has been revolutionized by new therapies targeting MAPK signaling or the immune system. Unfortunately these therapies are hindered by either primary resistance or the development of acquired resistance. Resistance mechanisms involving somatic mutations in genes associated with resistance have been identified in some cases of melanoma, however, the cause of resistance remains largely unexplained in other cases. The importance of epigenetic factors targeting histones and histone modifiers in driving the behavior of melanoma is only starting to be unraveled and provides significant opportunity to combat the problems of therapy resistance. There is also an increasing ability to target these epigenetic changes with new drugs that inhibit these modifications to either prevent or overcome resistance to both MAPK inhibitors and immunotherapy. This review focuses on changes in histones, histone reader proteins and histone positioning, which can mediate resistance to new therapeutics and that can be targeted for future therapies.
Collapse
Affiliation(s)
- Stuart J Gallagher
- Melanoma Immunology and Oncology Group, Centenary Institute, University of Sydney, Camperdown 2050, Australia.
- Melanoma Institute Australia, Crow's Nest 2065, Sydney, Australia.
| | - Jessamy C Tiffen
- Melanoma Immunology and Oncology Group, Centenary Institute, University of Sydney, Camperdown 2050, Australia.
- Melanoma Institute Australia, Crow's Nest 2065, Sydney, Australia.
| | - Peter Hersey
- Melanoma Immunology and Oncology Group, Centenary Institute, University of Sydney, Camperdown 2050, Australia.
- Melanoma Institute Australia, Crow's Nest 2065, Sydney, Australia.
| |
Collapse
|