1
|
Wonkam A, Esoh K, Levine RM, Ngo Bitoungui VJ, Mnika K, Nimmagadda N, Dempsey EAD, Nkya S, Sangeda RZ, Nembaware V, Morrice J, Osman F, Beer MA, Makani J, Mulder N, Lettre G, Steinberg MH, Latanich R, Casella JF, Drehmer D, Arking DE, Chimusa ER, Yen JS, Newby GA, Antonarakis SE. FLT1 and other candidate fetal haemoglobin modifying loci in sickle cell disease in African ancestries. Nat Commun 2025; 16:2092. [PMID: 40025045 PMCID: PMC11873275 DOI: 10.1038/s41467-025-57413-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 02/20/2025] [Indexed: 03/04/2025] Open
Abstract
Known fetal haemoglobin (HbF)-modulating loci explain 10-24% variation of HbF level in Africans with Sickle Cell Disease (SCD), compared to 50% among Europeans. Here, we report fourteen candidate loci from a genome-wide association study (GWAS) of HbF level in patients with SCD from Cameroon, Tanzania, and the United States of America. We present results of cell-based experiments for FLT1 candidate, demonstrating expression in early haematopoiesis and a possible involvement in hypoxia associated HbF induction. Our study employed genotyping arrays that capture a broad range of African and non-African genetic variation and replicated known loci (BCL11A and HBS1L-MYB). We estimated the heritability of HbF level in SCD at 94%, higher than estimated in unselected Europeans, and suggesting a robust capture of HbF-associated loci by these arrays. Our approach, which involved genotype imputation against six reference haplotype panels and association analysis with each of the panels, proved superior over selecting a best-performing panel, evidenced by a substantial proportion of panel-specific (up to 18%) and a low proportion of shared (28%) imputed variants across the panels.
Collapse
Affiliation(s)
- Ambroise Wonkam
- McKusick-Nathans Institute and Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.
| | - Kevin Esoh
- McKusick-Nathans Institute and Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Rachel M Levine
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Khuthala Mnika
- Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Nikitha Nimmagadda
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Erin A D Dempsey
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Siana Nkya
- Department of Biochemistry and Molecular Biology, Muhimbili University of Health and Allied Sciences, Dar Es Salaam, Tanzania
| | - Raphael Z Sangeda
- Department of Pharmaceutical Microbiology, Muhimbili University of Health and Allied Sciences, Dar Es Salaam, Tanzania
| | - Victoria Nembaware
- Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Jack Morrice
- Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Fujr Osman
- McKusick-Nathans Institute and Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael A Beer
- McKusick-Nathans Institute and Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Julie Makani
- Sickle Cell Programme, Department of Haematology and Blood Transfusion, Muhimbili University of Health & Allied Sciences (MUHAS), Dar Es Salaam, Tanzania
- SickleInAfrica Clinical Coordinating Center, Muhimbili University of Health & Allied Sciences (MUHAS), Dar Es Salaam, Tanzania
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Nicola Mulder
- Computational Biology Division, Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, CIDRI-Africa Wellcome Trust Centre, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Guillaume Lettre
- Montreal Heart Institute, Université de Montréal, Montreal, QC, Canada
| | - Martin H Steinberg
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Rachel Latanich
- McKusick-Nathans Institute and Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - James F Casella
- Department of Pediatrics, Division of Hematology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Daiana Drehmer
- Armstrong Oxygen Biology Research Center, Institute for Cell Engineering, and Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dan E Arking
- McKusick-Nathans Institute and Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Emile R Chimusa
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle, Tyne and Wear, UK
| | - Jonathan S Yen
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Gregory A Newby
- McKusick-Nathans Institute and Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Stylianos E Antonarakis
- Department of Genetic Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
2
|
Zhang H, Hansen M, Di Summa F, Von Lindern M, Gillemans N, Van IJcken WFJ, Svendsen AF, Philipsen S, Van der Reijden B, Varga E, Van den Akker E. LSD1/KDM1A and GFI1B repress endothelial fate and induce hematopoietic fate in induced pluripotent stem cell-derived hemogenic endothelium. Haematologica 2024; 109:3975-3988. [PMID: 38961746 PMCID: PMC11609818 DOI: 10.3324/haematol.2024.285214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 06/24/2024] [Indexed: 07/05/2024] Open
Abstract
Differentiation of induced pluripotent stem cells (iPSC) into hematopoietic lineages offers great therapeutic potential. During embryogenesis, hemogenic endothelium (HE) gives rise to hematopoietic stem and progenitor cells through the endothelial- to-hematopoietic transition (EHT). Understanding this process using iPSC is key to generating functional hematopoietic stem cells (HSC), a currently unmet challenge. In this study, we examined the role of the transcriptional factor GFI1B and its co-factor LSD1/KDM1A in EHT. To this end, we employed patient-derived iPSC lines with a dominant-negative dysfunctional GFI1B Q287* and irreversible pharmacological LSD1/KDM1A inhibition in healthy iPSC lines. The formation of HE remained unaffected; however, hematopoietic output was severely reduced in both conditions. Single-cell RNA sequencing (scRNAseq) performed on the CD144+/CD31+ population derived from healthy iPSC revealed similar expression dynamics of genes associated with in vivo EHT. Interestingly, LSD1/KDM1A inhibition in healthy lines before EHT resulted in a complete absence of hematopoietic output. However, uncommitted HE cells did not display GFI1B expression, suggesting a timed transcriptional program. To test this hypothesis, we ectopically expressed GFI1B in uncommitted HE cells, leading to downregulation of endothelial genes and upregulation of hematopoietic genes, including GATA2, KIT, RUNX1, and SPI1. Thus, we demonstrate that LSD1/KDM1A and GFI1B can function at distinct temporal points in different cellular subsets during EHT. Although GFI1B is not detected in uncommitted HE cells, its ectopic expression allows for partial hematopoietic specification. These data indicate that precisely timed expression of specific transcriptional regulators during EHT is crucial to the eventual outcome of EHT.
Collapse
Affiliation(s)
- Huan Zhang
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam
| | - Marten Hansen
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam
| | - Franca Di Summa
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam
| | - Marieke Von Lindern
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam
| | | | | | | | | | - Bert Van der Reijden
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen
| | - Eszter Varga
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam
| | - Emile Van den Akker
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam.
| |
Collapse
|
3
|
Gosai SJ, Castro RI, Fuentes N, Butts JC, Mouri K, Alasoadura M, Kales S, Nguyen TTL, Noche RR, Rao AS, Joy MT, Sabeti PC, Reilly SK, Tewhey R. Machine-guided design of cell-type-targeting cis-regulatory elements. Nature 2024; 634:1211-1220. [PMID: 39443793 PMCID: PMC11525185 DOI: 10.1038/s41586-024-08070-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 09/18/2024] [Indexed: 10/25/2024]
Abstract
Cis-regulatory elements (CREs) control gene expression, orchestrating tissue identity, developmental timing and stimulus responses, which collectively define the thousands of unique cell types in the body1-3. While there is great potential for strategically incorporating CREs in therapeutic or biotechnology applications that require tissue specificity, there is no guarantee that an optimal CRE for these intended purposes has arisen naturally. Here we present a platform to engineer and validate synthetic CREs capable of driving gene expression with programmed cell-type specificity. We take advantage of innovations in deep neural network modelling of CRE activity across three cell types, efficient in silico optimization and massively parallel reporter assays to design and empirically test thousands of CREs4-8. Through large-scale in vitro validation, we show that synthetic sequences are more effective at driving cell-type-specific expression in three cell lines compared with natural sequences from the human genome and achieve specificity in analogous tissues when tested in vivo. Synthetic sequences exhibit distinct motif vocabulary associated with activity in the on-target cell type and a simultaneous reduction in the activity of off-target cells. Together, we provide a generalizable framework to prospectively engineer CREs from massively parallel reporter assay models and demonstrate the required literacy to write fit-for-purpose regulatory code.
Collapse
Affiliation(s)
- Sager J Gosai
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Harvard Graduate Program in Biological and Biomedical Science, Boston, MA, USA.
- Department Of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| | | | - Natalia Fuentes
- The Jackson Laboratory, Bar Harbor, ME, USA
- Harvard College, Harvard University, Cambridge, MA, USA
| | - John C Butts
- The Jackson Laboratory, Bar Harbor, ME, USA
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, USA
| | | | | | | | | | - Ramil R Noche
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT, USA
- Yale Zebrafish Research Core, Yale School of Medicine, New Haven, CT, USA
| | - Arya S Rao
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Mary T Joy
- The Jackson Laboratory, Bar Harbor, ME, USA
| | - Pardis C Sabeti
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department Of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
- Department of Immunology and Infectious Diseases, Harvard T H Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Steven K Reilly
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA.
- Wu Tsai Institute, Yale University, New Haven, CT, USA.
| | - Ryan Tewhey
- The Jackson Laboratory, Bar Harbor, ME, USA.
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, USA.
- Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA.
| |
Collapse
|
4
|
Ojo OA, Shen H, Ingram JT, Bonner JA, Welner RS, Lacaud G, Zajac AJ, Shi LZ. Gfi1 controls the formation of effector CD8 T cells during chronic infection and cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.18.579535. [PMID: 38659890 PMCID: PMC11042319 DOI: 10.1101/2024.04.18.579535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
During chronic infections and tumor progression, CD8 T cells gradually lose their effector functions and become exhausted. These exhausted CD8 T cells are heterogeneous and comprised of different subsets, including self-renewing progenitors that give rise to Ly108 - CX3CR1 + effector-like cells. Generation of these effector-like cells is essential for the control of chronic infections and tumors, albeit limited. However, the precise cues and mechanisms directing the formation and maintenance of exhausted effector-like are incompletely understood. Using genetic mouse models challenged with LCMV Clone 13 or syngeneic tumors, we show that the expression of a transcriptional repressor, growth factor independent 1 (Gfi1) is dynamically regulated in exhausted CD8 T cells, which in turn regulates the formation of exhausted effector-like cells. Gfi1 deletion in T cells dysregulates the chromatin accessibility and transcriptomic programs associated with the differentiation of LCMV Clone 13-specific CD8 T cell exhaustion, preventing the formation of effector-like and terminally exhausted cells while maintaining progenitors and a newly identified Ly108 + CX3CR1 + state. These Ly108 + CX3CR1 + cells have a distinct chromatin profile and may represent an alternative target for therapeutic interventions to combat chronic infections and cancer. In sum, we show that Gfi1 is a critical regulator of the formation of exhausted effector-like cells.
Collapse
|
5
|
Gosai SJ, Castro RI, Fuentes N, Butts JC, Kales S, Noche RR, Mouri K, Sabeti PC, Reilly SK, Tewhey R. Machine-guided design of synthetic cell type-specific cis-regulatory elements. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.08.552077. [PMID: 37609287 PMCID: PMC10441439 DOI: 10.1101/2023.08.08.552077] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Cis-regulatory elements (CREs) control gene expression, orchestrating tissue identity, developmental timing, and stimulus responses, which collectively define the thousands of unique cell types in the body. While there is great potential for strategically incorporating CREs in therapeutic or biotechnology applications that require tissue specificity, there is no guarantee that an optimal CRE for an intended purpose has arisen naturally through evolution. Here, we present a platform to engineer and validate synthetic CREs capable of driving gene expression with programmed cell type specificity. We leverage innovations in deep neural network modeling of CRE activity across three cell types, efficient in silico optimization, and massively parallel reporter assays (MPRAs) to design and empirically test thousands of CREs. Through in vitro and in vivo validation, we show that synthetic sequences outperform natural sequences from the human genome in driving cell type-specific expression. Synthetic sequences leverage unique sequence syntax to promote activity in the on-target cell type and simultaneously reduce activity in off-target cells. Together, we provide a generalizable framework to prospectively engineer CREs and demonstrate the required literacy to write regulatory code that is fit-for-purpose in vivo across vertebrates.
Collapse
Affiliation(s)
- SJ Gosai
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Graduate Program in Biological and Biomedical Science, Boston MA
- Department Of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - RI Castro
- The Jackson Laboratory, Bar Harbor, ME, USA
| | - N Fuentes
- The Jackson Laboratory, Bar Harbor, ME, USA
- Harvard College, Harvard University, Cambridge, MA, USA
| | - JC Butts
- The Jackson Laboratory, Bar Harbor, ME, USA
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, USA
| | - S Kales
- The Jackson Laboratory, Bar Harbor, ME, USA
| | - RR Noche
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT, USA
- Yale Zebrafish Research Core, Yale School of Medicine, New Haven, CT, USA
| | - K Mouri
- The Jackson Laboratory, Bar Harbor, ME, USA
| | - PC Sabeti
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department Of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - SK Reilly
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
- Wu Tsai Institute, Yale University, New Haven, CT, USA
| | - R Tewhey
- The Jackson Laboratory, Bar Harbor, ME, USA
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, USA
- Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
| |
Collapse
|
6
|
Dehdilani N, Goshayeshi L, Yousefi Taemeh S, Bahrami AR, Rival Gervier S, Pain B, Dehghani H. Integrating Omics and CRISPR Technology for Identification and Verification of Genomic Safe Harbor Loci in the Chicken Genome. Biol Proced Online 2023; 25:18. [PMID: 37355580 DOI: 10.1186/s12575-023-00210-5] [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: 02/10/2023] [Accepted: 06/02/2023] [Indexed: 06/26/2023] Open
Abstract
BACKGROUND One of the most prominent questions in the field of transgenesis is 'Where in the genome to integrate a transgene?'. Escape from epigenetic silencing and promoter shutdown of the transgene needs reliable genomic safe harbor (GSH) loci. Advances in genome engineering technologies combined with multi-omics bioinformatics data have enabled rational evaluation of GSH loci in the host genome. Currently, no validated GSH loci have been evaluated in the chicken genome. RESULTS Here, we analyzed and experimentally examined two GSH loci in the genome of chicken cells. To this end, putative GSH loci including chicken HIPP-like (cHIPP; between DRG1 and EIF4ENIF1 genes) and chicken ROSA-like (cROSA; upstream of the THUMPD3 gene) were predicted using multi-omics bioinformatics data. Then, the durable expression of the transgene was validated by experimental characterization of continuously-cultured isogenous cell clones harboring DsRed2-ΔCMV-EGFP cassette in the predicted loci. The weakened form of the CMV promoter (ΔCMV) allowed the precise evaluation of GSH loci in a locus-dependent manner compared to the full-length CMV promoter. CONCLUSIONS cHIPP and cROSA loci introduced in this study can be reliably exploited for consistent bio-manufacturing of recombinant proteins in the genetically-engineered chickens. Also, results showed that the genomic context dictates the expression of transgene controlled by ΔCMV in GSH loci.
Collapse
Affiliation(s)
- Nima Dehdilani
- Stem Cell Biology and Regenerative Medicine Research Group, Research Institute of Biotechnology, Ferdowsi University of Mashhad, Azadi Square, Mashhad, Iran
| | - Lena Goshayeshi
- Stem Cell Biology and Regenerative Medicine Research Group, Research Institute of Biotechnology, Ferdowsi University of Mashhad, Azadi Square, Mashhad, Iran
- Division of Biotechnology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Sara Yousefi Taemeh
- Stem Cell Biology and Regenerative Medicine Research Group, Research Institute of Biotechnology, Ferdowsi University of Mashhad, Azadi Square, Mashhad, Iran
- Division of Biotechnology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Ahmad Reza Bahrami
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
- Industrial Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Sylvie Rival Gervier
- Stem Cell and Brain Research Institute, University of Lyon, Université Lyon 1, INSERM, INRAE, U1208, USC1361, 69500, Bron, France
| | - Bertrand Pain
- Stem Cell and Brain Research Institute, University of Lyon, Université Lyon 1, INSERM, INRAE, U1208, USC1361, 69500, Bron, France
| | - Hesam Dehghani
- Stem Cell Biology and Regenerative Medicine Research Group, Research Institute of Biotechnology, Ferdowsi University of Mashhad, Azadi Square, Mashhad, Iran.
- Division of Biotechnology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran.
- Department of Basic Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran.
| |
Collapse
|
7
|
Albright ER, Walter RM, Saffert RT, Kalejta RF. NFκB and Cyclic AMP Response Element Sites Mediate the Valproic Acid and UL138 Responsiveness of the Human Cytomegalovirus Major Immediate Early Enhancer and Promoter. J Virol 2023; 97:e0002923. [PMID: 36856444 PMCID: PMC10062163 DOI: 10.1128/jvi.00029-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 02/08/2023] [Indexed: 03/02/2023] Open
Abstract
The major immediate early enhancer and promoter (MIEP) of human cytomegalovirus (HCMV) drives the transcription of the immediate early one (IE1) and IE2 genes, whose encoded proteins stimulate productive, lytic replication. The MIEP is activated by the virally encoded and tegument-delivered pp71 protein at the start of de novo lytic infections of fully differentiated cells. Conversely, the MIEP is silenced at the start of de novo latent infections within incompletely differentiated myeloid cells in part because tegument-delivered pp71 is sequestered in the cytoplasm in these cells, but also by viral factors that repress transcription from this locus, including the UL138 protein. During both modes of infection, MIEP activity can be increased by the histone deacetylase inhibitor valproic acid (VPA); however, UL138 inhibits the VPA-responsiveness of the MIEP. Here, we show that two families of cellular transcription factors, NF-κB and cAMP response element-binding protein (CREB), together control the VPA-mediated activation and UL138-mediated repression of the HCMV MIEP. IMPORTANCE Artificial regulation of the HCMV MIEP, either activation or repression, is an attractive potential means to target the latent reservoirs of virus for which there is currently no available intervention. The MIEP could be repressed to prevent latency reactivation or induced to drive the virus into the lytic stage that is visible to the immune system and inhibited by multiple small-molecule antiviral drugs. Understanding how the MIEP is regulated is a critical part of designing and implementing either strategy. Our revelation here that NF-κB and CREB control the responsiveness of the MIEP to the viral UL138 protein and the FDA-approved drug VPA could help in the formulation and execution of promoter regulatory strategies against latent HCMV.
Collapse
Affiliation(s)
- Emily R. Albright
- Institute for Molecular Virology and McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Ryan M. Walter
- Institute for Molecular Virology and McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Ryan T. Saffert
- Institute for Molecular Virology and McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Robert F. Kalejta
- Institute for Molecular Virology and McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
8
|
Molecular detection of mixed infection with peste des petits ruminants and retroviruses in Egyptian sheep and goats. Trop Anim Health Prod 2023; 55:102. [PMID: 36849557 DOI: 10.1007/s11250-023-03504-8] [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: 08/22/2022] [Accepted: 02/11/2023] [Indexed: 03/01/2023]
Abstract
Peste des petits ruminants (PPR) is a contagious viral disease causing massive economic loss to animal industries in endemic countries including Egypt. Although a vaccine is available, coinfections can overwhelm the animal immune system and interfere with vaccine protection. Small ruminant retrovirus (SRR), including enzootic nasal tumor virus (ENTV) and Jaagsiekte sheep retrovirus (JSRV), is responsible for coinfections with PPR. Investigation of clinical cases in this study confirmed the presence of PPR virus by RT-PCR among four flocks. Sequence of five PPR amplicons revealed that all strains had 100% aa similarity and belonged to lineage IV. In addition, these strains had 98-99% nt similarity with all previous Egyptian and African strains from Sudan (MK371449) and Ethiopia (MK371449). Illumina sequencing of a representative sample showed a genome of 5753 nt compatible with ENT-2 virus with 98.42% similarity with the Chinese strain (MN564750.1). Four ORFs representing gag, pro, pol, and env genes were identified and annotated. Pro gene was highly stable while gag, pol, and env showed eight, two, and three aa differences with the reference strains. Sanger sequencing revealed that two amplicons were ENT-2 virus, and one was JSRV. ENT-2 sequences had 100% similarity with KU258870 and KU258871 reference strains while JSRV was 100% similar to the EF68031 reference strain. The phylogenetic tree showed a close relationship between the ENT of goats and the JSRV of sheep. This study highlights the complexity of PPR molecular epidemiology, with SRR that was not molecularly characterized previously in Egypt.
Collapse
|
9
|
Costa A, Powell LM, Malaguti M, Soufi A, Lowell S, Jarman AP. Repurposing the lineage-determining transcription factor Atoh1 without redistributing its genomic binding sites. Front Cell Dev Biol 2022; 10:1016367. [PMID: 36420143 PMCID: PMC9676683 DOI: 10.3389/fcell.2022.1016367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 10/17/2022] [Indexed: 11/09/2022] Open
Abstract
Although the lineage-determining ability of transcription factors is often modulated according to cellular context, the mechanisms by which such switching occurs are not well known. Using a transcriptional programming model, we found that Atoh1 is repurposed from a neuronal to an inner ear hair cell (HC) determinant by the combined activities of Gfi1 and Pou4f3. In this process, Atoh1 maintains its regulation of neuronal genes but gains ability to regulate HC genes. Pou4f3 enables Atoh1 access to genomic locations controlling the expression of sensory (including HC) genes, but Atoh1 + Pou4f3 are not sufficient for HC differentiation. Gfi1 is key to the Atoh1-induced lineage switch, but surprisingly does not alter Atoh1's binding profile. Gfi1 acts in two divergent ways. It represses the induction by Atoh1 of genes that antagonise HC differentiation, a function in keeping with its well-known repressor role in haematopoiesis. Remarkably, we find that Gfi1 also acts as a co-activator: it binds directly to Atoh1 at existing target genes to enhance its activity. These findings highlight the diversity of mechanisms by which one TF can redirect the activity of another to enable combinatorial control of cell identity.
Collapse
Affiliation(s)
- Aida Costa
- Centre for Discovery Brain Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh, United Kingdom
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Lynn M. Powell
- Centre for Discovery Brain Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh, United Kingdom
| | - Mattias Malaguti
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Abdenour Soufi
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Sally Lowell
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Andrew P. Jarman
- Centre for Discovery Brain Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
10
|
Vorwerk J, Sun K, Frank D, Neumann F, Hüve J, Budde PM, Liu L, Xie X, Patnana PK, Ahmed HMM, Opalka B, Lenz G, Jayavelu AK, Khandanpour C. Presence of the GFI1-36N single nucleotide polymorphism enhances the response of MLL-AF9 leukemic cells to CDK4/6 inhibition. Front Oncol 2022; 12:903691. [PMID: 36003783 PMCID: PMC9393725 DOI: 10.3389/fonc.2022.903691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
The zinc finger protein Growth Factor Independence 1 (GFI1) acts as a transcriptional repressor regulating differentiation of myeloid and lymphoid cells. A single nucleotide polymorphism of GFI1, GFI1-36N, has a prevalence of 7% in healthy Caucasians and 15% in acute myeloid leukemia (AML) patients, hence most probably predisposing to AML. One reason for this is that GFI1-36N differs from the wildtype form GFI1-36S regarding its ability to induce epigenetic changes resulting in a derepression of oncogenes. Using proteomics, immunofluorescence, and immunoblotting we have now gained evidence that murine GFI1-36N leukemic cells exhibit a higher protein level of the pro-proliferative protein arginine N-methyltransferase 5 (PRMT5) as well as increased levels of the cell cycle propagating cyclin-dependent kinases 4 (CDK4) and 6 (CDK6) leading to a faster proliferation of GFI1-36N leukemic cells in vitro. As a therapeutic approach, we subsequently treated leukemic GFI1-36S and GFI1-36N cells with the CDK4/6 inhibitor palbociclib and observed that GFI1-36N leukemic cells were more susceptible to this treatment. The findings suggest that presence of the GFI1-36N variant increases proliferation of leukemic cells and could possibly be a marker for a specific subset of AML patients sensitive to CDK4/6 inhibitors such as palbociclib.
Collapse
Affiliation(s)
- Jan Vorwerk
- Department of Medicine A, Hematology, Hemostaseology, Oncology, and Pneumology, University Hospital Münster, Münster, Germany
| | - Kaiyan Sun
- Department of Medicine A, Hematology, Hemostaseology, Oncology, and Pneumology, University Hospital Münster, Münster, Germany
| | - Daria Frank
- Department of Medicine A, Hematology, Hemostaseology, Oncology, and Pneumology, University Hospital Münster, Münster, Germany
| | - Felix Neumann
- Fluorescence Microscopy Facility Münster, Institute of Medical Physics and Biophysics, University of Münster, Münster, Germany
- Evorion Biotechnologies GmbH, Münster, Germany
| | - Jana Hüve
- Fluorescence Microscopy Facility Münster, Institute of Medical Physics and Biophysics, University of Münster, Münster, Germany
| | - Paulina Marie Budde
- Department of Medicine A, Hematology, Hemostaseology, Oncology, and Pneumology, University Hospital Münster, Münster, Germany
| | - Longlong Liu
- Department of Medicine A, Hematology, Hemostaseology, Oncology, and Pneumology, University Hospital Münster, Münster, Germany
| | - Xiaoqing Xie
- Department of Medicine A, Hematology, Hemostaseology, Oncology, and Pneumology, University Hospital Münster, Münster, Germany
| | - Pradeep Kumar Patnana
- Department of Medicine A, Hematology, Hemostaseology, Oncology, and Pneumology, University Hospital Münster, Münster, Germany
| | - Helal Mohammed Mohammed Ahmed
- Department of Medicine A, Hematology, Hemostaseology, Oncology, and Pneumology, University Hospital Münster, Münster, Germany
| | - Bertram Opalka
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center (WTZ), University Hospital Essen, Essen, Germany
| | - Georg Lenz
- Department of Medicine A, Hematology, Hemostaseology, Oncology, and Pneumology, University Hospital Münster, Münster, Germany
| | - Ashok Kumar Jayavelu
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Munich, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- Department of Pediatric Oncology, Hematology, and Immunology, Heidelberg University Hospital, Heidelberg, Germany
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Leukemia, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Cyrus Khandanpour
- Department of Medicine A, Hematology, Hemostaseology, Oncology, and Pneumology, University Hospital Münster, Münster, Germany
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, University of Lübeck, Lübeck, Germany
- *Correspondence: Cyrus Khandanpour,
| |
Collapse
|
11
|
Johari YB, Scarrott JM, Pohle TH, Liu P, Mayer A, Brown AJ, James DC. Engineering of the CMV promoter for controlled expression of recombinant genes in HEK293 cells. Biotechnol J 2022; 17:e2200062. [PMID: 35482470 DOI: 10.1002/biot.202200062] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 04/07/2022] [Accepted: 04/23/2022] [Indexed: 11/08/2022]
Abstract
Expression of recombinant genes in HEK293 cells is frequently utilized for production of recombinant proteins and viral vectors. These systems frequently employ the cytomegalovirus (CMV) promoter to drive recombinant gene transcription. However, the mechanistic basis of CMV-mediated transcriptional activation in HEK293 cells is unknown and consequently there are no strategies to engineer CMV for controlled expression of recombinant genes. Extensive bioinformatic analyses of transcription factor regulatory elements (TFREs) within the human CMV sequence and transcription factor mRNAs within the HEK293 transcriptome revealed 80 possible regulatory interactions. Through in vitro functional testing using reporter constructs harboring discrete TFREs or CMV deletion variants we identified key TFRE components and clusters of TFREs (cis-regulatory modules) within the CMV sequence. Our data reveal that CMV activity in HEK293 cells is a function of the promoters various constituent TFREs including AhR:ARNT, CREB, E4F, Sp1, ZBED1, JunB, c-Rel, and NF-κB. We also identified critical Sp1-dependent upstream activator elements near the transcriptional start site that were required for efficient transcription and YY1 and RBP-Jκ binding sites that mediate transrepression. Our study shows for the first time that novel, compact CMV-derived promoters can be engineered that exhibit up to 50% higher transcriptional efficiency (activity per unit DNA sequence) or 14% increase in total activity compared to the wild-type counterpart.
Collapse
Affiliation(s)
- Yusuf B Johari
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, UK
| | - Joseph M Scarrott
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, UK
| | - Thilo H Pohle
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, UK
| | - Ping Liu
- Cell Line Development, REGENXBIO Inc., Rockville, Maryland, USA
| | - Ayda Mayer
- Cell Line Development, REGENXBIO Inc., Rockville, Maryland, USA
| | - Adam J Brown
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, UK.,Syngensys Ltd., Sheffield, UK
| | - David C James
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, UK.,Syngensys Ltd., Sheffield, UK
| |
Collapse
|
12
|
Chakraborty M, Das RK, Samal S, Das S, Alone DP. Fuchs Endothelial Corneal Dystrophy associated risk variant, rs3768617 in LAMC1 shows allele specific binding of GFI1B. Gene 2022; 817:146179. [PMID: 35031421 DOI: 10.1016/j.gene.2021.146179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 11/24/2021] [Accepted: 12/06/2021] [Indexed: 01/07/2023]
Abstract
AIMS To investigate the genetic and functional association of an intronic variant of LAMC1, rs3768617 with Fuchs endothelial corneal dystrophy (FECD) in the Indian population. METHODS Blood samples were collected from age and sex matched 356 controls and 120 FECD patients after a detailed assessment via specular microscopy. Genomic DNA was extracted and genotyping was done by fluorescence based capillary electrophoresis. The genetic association of rs3768617 polymorphisms was computed by the chi-square (χ2) test. Bioinformatics studies were performed to find the allele specific binding of different transcription factors in the region of rs3768617 and functional evaluation assessed by luciferase assay followed by Electrophoretic Mobility Shift Assay (EMSA) and Chromatin Immunoprecipitation assay (ChIP). Immunofluorescence assay was carried out to check for any differential expression of GFI1B between control and FECD endothelium samples. RESULTS SNP rs3768617 {chr1:183123365 (GRCh38.p13)} was found to be genetically associated with FECD in Indian population (p = 2.646 × 10-8). Luciferase assay suggested that the rs3768617 locus has a regulatory role. In silico analysis showed that the transcription factor, GFI1B binds to the risk allele 'G' of rs3768617, but not to the protective allele 'A' which was also experimentally validated by EMSA. High enrichment of DNA flanking the surrounding region of rs3768617 was also found in presence of GFI1B specific antibody in ChIP assay. There was a 0.63 fold decrease in GFI1B expression in FECD affected corneal endothelium compared to control endothelium. CONCLUSIONS The genetic association of rs3768617 in LAMC1 with FECD pathogenesis is mediated by GFI1B, thus finding the functional role of LAMC1 in FECD pathogenesis.
Collapse
Affiliation(s)
- Maynak Chakraborty
- School of Biological Sciences, National Institute of Science Education and Research (NISER) Bhubaneswar, HBNI, P.O. Bhimpur-Padanpur, Jatni, Khurda, Odisha 752050, India; Homi Bhabha National Institute (HBNI), Training School Complex, Anushaktinagar, Mumbai 400094, India
| | - Rajesh Kumar Das
- School of Biological Sciences, National Institute of Science Education and Research (NISER) Bhubaneswar, HBNI, P.O. Bhimpur-Padanpur, Jatni, Khurda, Odisha 752050, India; Homi Bhabha National Institute (HBNI), Training School Complex, Anushaktinagar, Mumbai 400094, India
| | - Sujata Samal
- LV Prasad Eye Institute, Bhubaneswar, Odisha 751024, India
| | - Sujata Das
- LV Prasad Eye Institute, Bhubaneswar, Odisha 751024, India
| | - Debasmita Pankaj Alone
- School of Biological Sciences, National Institute of Science Education and Research (NISER) Bhubaneswar, HBNI, P.O. Bhimpur-Padanpur, Jatni, Khurda, Odisha 752050, India; Homi Bhabha National Institute (HBNI), Training School Complex, Anushaktinagar, Mumbai 400094, India.
| |
Collapse
|
13
|
Petrusca DN, Mulcrone PL, Macar DA, Bishop RT, Berdyshev E, Suvannasankha A, Anderson JL, Sun Q, Auron PE, Galson DL, Roodman GD. GFI1-Dependent Repression of SGPP1 Increases Multiple Myeloma Cell Survival. Cancers (Basel) 2022; 14:cancers14030772. [PMID: 35159039 PMCID: PMC8833953 DOI: 10.3390/cancers14030772] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/27/2022] [Accepted: 01/31/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary New therapies have greatly improved the progression-free and overall survival for patients with “standard risk” multiple myeloma (MM). However, patients with “high risk” MM, in particular patients whose MM cells harbor non-functional p53, have very short survival times because of the early relapse and rapid development of highly therapy-resistant MM. In this report, we identify a novel mechanism responsible for Growth Factor Independence-1 (GFI1) regulation of the growth and survival of MM cells through its modulation of sphingolipid metabolism, regardless of their p53 status. We identify the Sphingosine-1-Phosphate Phosphatase (SGPP1) gene as a novel direct target of GFI1 transcriptional repression in MM cells, thus increasing intracellular sphingosine-1-phosphate levels, which stabilizes c-Myc. Our results support GFI1 as an attractive therapeutic target for all types of MM, including the “high risk” patient population with non-functional p53, as well as a possible therapeutic approach for other types of cancers expressing high levels of c-Myc. Abstract Multiple myeloma (MM) remains incurable for most patients due to the emergence of drug resistant clones. Here we report a p53-independent mechanism responsible for Growth Factor Independence-1 (GFI1) support of MM cell survival by its modulation of sphingolipid metabolism to increase the sphingosine-1-phosphate (S1P) level regardless of the p53 status. We found that expression of enzymes that control S1P biosynthesis, SphK1, dephosphorylation, and SGPP1 were differentially correlated with GFI1 levels in MM cells. We detected GFI1 occupancy on the SGGP1 gene in MM cells in a predicted enhancer region at the 5’ end of intron 1, which correlated with decreased SGGP1 expression and increased S1P levels in GFI1 overexpressing cells, regardless of their p53 status. The high S1P:Ceramide intracellular ratio in MM cells protected c-Myc protein stability in a PP2A-dependent manner. The decreased MM viability by SphK1 inhibition was dependent on the induction of autophagy in both p53WT and p53mut MM. An autophagic blockade prevented GFI1 support for viability only in p53mut MM, demonstrating that GFI1 increases MM cell survival via both p53WT inhibition and upregulation of S1P independently. Therefore, GFI1 may be a key therapeutic target for all types of MM that may significantly benefit patients that are highly resistant to current therapies.
Collapse
Affiliation(s)
- Daniela N. Petrusca
- Department of Medicine, Hematology/Oncology Division, Indiana University School of Medicine, 980 Walnut St., Indianapolis, IN 46202, USA; (P.L.M.); (A.S.); (J.L.A.); (G.D.R.)
- Correspondence: ; Tel.: +1-(317)-278-5548
| | - Patrick L. Mulcrone
- Department of Medicine, Hematology/Oncology Division, Indiana University School of Medicine, 980 Walnut St., Indianapolis, IN 46202, USA; (P.L.M.); (A.S.); (J.L.A.); (G.D.R.)
| | - David A. Macar
- Department of Biological Sciences, Duquesne University, 600 Forbes Ave., Pittsburgh, PA 15219, USA; (D.A.M.); (P.E.A.)
| | - Ryan T. Bishop
- Department of Tumor Biology, H. Lee Moffitt Cancer Research Center and Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA;
| | - Evgeny Berdyshev
- Department of Medicine, National Jewish Health, 1400 Jackson Street, Denver, CO 80206, USA;
| | - Attaya Suvannasankha
- Department of Medicine, Hematology/Oncology Division, Indiana University School of Medicine, 980 Walnut St., Indianapolis, IN 46202, USA; (P.L.M.); (A.S.); (J.L.A.); (G.D.R.)
- Richard L. Rodebush Veterans Affairs Medical Center, 1481 W 10th St., Indianapolis, IN 46202, USA
| | - Judith L. Anderson
- Department of Medicine, Hematology/Oncology Division, Indiana University School of Medicine, 980 Walnut St., Indianapolis, IN 46202, USA; (P.L.M.); (A.S.); (J.L.A.); (G.D.R.)
| | - Quanhong Sun
- Department of Medicine, Division of Hematology/Oncology, McGowan Institute for Regenerative Medicine, University of Pittsburgh, UPMC Hillman Cancer Center Research Pavilion, 5117 Centre Ave, Pittsburgh, PA 15213, USA; (Q.S.); (D.L.G.)
| | - Philip E. Auron
- Department of Biological Sciences, Duquesne University, 600 Forbes Ave., Pittsburgh, PA 15219, USA; (D.A.M.); (P.E.A.)
| | - Deborah L. Galson
- Department of Medicine, Division of Hematology/Oncology, McGowan Institute for Regenerative Medicine, University of Pittsburgh, UPMC Hillman Cancer Center Research Pavilion, 5117 Centre Ave, Pittsburgh, PA 15213, USA; (Q.S.); (D.L.G.)
| | - G. David Roodman
- Department of Medicine, Hematology/Oncology Division, Indiana University School of Medicine, 980 Walnut St., Indianapolis, IN 46202, USA; (P.L.M.); (A.S.); (J.L.A.); (G.D.R.)
- Richard L. Rodebush Veterans Affairs Medical Center, 1481 W 10th St., Indianapolis, IN 46202, USA
| |
Collapse
|
14
|
Napolitano T, Avolio F, Silvano S, Forcisi S, Pfeifer A, Vieira A, Navarro-Sanz S, Friano ME, Ayachi C, Garrido-Utrilla A, Atlija J, Hadzic B, Becam J, Sousa-De-Veiga A, Plaisant MD, Balaji S, Pisani DF, Mondin M, Schmitt-Kopplin P, Amri EZ, Collombat P. Gfi1 Loss Protects against Two Models of Induced Diabetes. Cells 2021; 10:cells10112805. [PMID: 34831029 PMCID: PMC8616283 DOI: 10.3390/cells10112805] [Citation(s) in RCA: 4] [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: 09/08/2021] [Revised: 10/07/2021] [Accepted: 10/14/2021] [Indexed: 12/29/2022] Open
Abstract
Background: Although several approaches have revealed much about individual factors that regulate pancreatic development, we have yet to fully understand their complicated interplay during pancreas morphogenesis. Gfi1 is transcription factor specifically expressed in pancreatic acinar cells, whose role in pancreas cells fate identity and specification is still elusive. Methods: In order to gain further insight into the function of this factor in the pancreas, we generated animals deficient for Gfi1 specifically in the pancreas. Gfi1 conditional knockout animals were phenotypically characterized by immunohistochemistry, RT-qPCR, and RNA scope. To assess the role of Gfi1 in the pathogenesis of diabetes, we challenged Gfi1-deficient mice with two models of induced hyperglycemia: long-term high-fat/high-sugar feeding and streptozotocin injections. Results: Interestingly, mutant mice did not show any obvious deleterious phenotype. However, in depth analyses demonstrated a significant decrease in pancreatic amylase expression, leading to a diminution in intestinal carbohydrates processing and thus glucose absorption. In fact, Gfi1-deficient mice were found resistant to diet-induced hyperglycemia, appearing normoglycemic even after long-term high-fat/high-sugar diet. Another feature observed in mutant acinar cells was the misexpression of ghrelin, a hormone previously suggested to exhibit anti-apoptotic effects on β-cells in vitro. Impressively, Gfi1 mutant mice were found to be resistant to the cytotoxic and diabetogenic effects of high-dose streptozotocin administrations, displaying a negligible loss of β-cells and an imperturbable normoglycemia. Conclusions: Together, these results demonstrate that Gfi1 could turn to be extremely valuable for the development of new therapies and could thus open new research avenues in the context of diabetes research.
Collapse
Affiliation(s)
- Tiziana Napolitano
- Faculté des Sciences, Université Côte d’Azur, CNRS, Inserm, iBV, Parc Valrose, 06108 Nice, France; (T.N.); (S.S.); (A.P.); (A.V.); (M.E.F.); (C.A.); (A.G.-U.); (J.B.); (A.S.-D.-V.); (M.D.P.); (E.-Z.A.)
| | - Fabio Avolio
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark;
| | - Serena Silvano
- Faculté des Sciences, Université Côte d’Azur, CNRS, Inserm, iBV, Parc Valrose, 06108 Nice, France; (T.N.); (S.S.); (A.P.); (A.V.); (M.E.F.); (C.A.); (A.G.-U.); (J.B.); (A.S.-D.-V.); (M.D.P.); (E.-Z.A.)
| | - Sara Forcisi
- Research Unit Analytical BioGeoChemistry, Helmholtz Zentrum München, German Research Center for Environment Health, 85764 Neuherberg, Germany; (S.F.); (P.S.-K.)
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Anja Pfeifer
- Faculté des Sciences, Université Côte d’Azur, CNRS, Inserm, iBV, Parc Valrose, 06108 Nice, France; (T.N.); (S.S.); (A.P.); (A.V.); (M.E.F.); (C.A.); (A.G.-U.); (J.B.); (A.S.-D.-V.); (M.D.P.); (E.-Z.A.)
| | - Andhira Vieira
- Faculté des Sciences, Université Côte d’Azur, CNRS, Inserm, iBV, Parc Valrose, 06108 Nice, France; (T.N.); (S.S.); (A.P.); (A.V.); (M.E.F.); (C.A.); (A.G.-U.); (J.B.); (A.S.-D.-V.); (M.D.P.); (E.-Z.A.)
| | | | - Marika Elsa Friano
- Faculté des Sciences, Université Côte d’Azur, CNRS, Inserm, iBV, Parc Valrose, 06108 Nice, France; (T.N.); (S.S.); (A.P.); (A.V.); (M.E.F.); (C.A.); (A.G.-U.); (J.B.); (A.S.-D.-V.); (M.D.P.); (E.-Z.A.)
| | - Chaïma Ayachi
- Faculté des Sciences, Université Côte d’Azur, CNRS, Inserm, iBV, Parc Valrose, 06108 Nice, France; (T.N.); (S.S.); (A.P.); (A.V.); (M.E.F.); (C.A.); (A.G.-U.); (J.B.); (A.S.-D.-V.); (M.D.P.); (E.-Z.A.)
| | - Anna Garrido-Utrilla
- Faculté des Sciences, Université Côte d’Azur, CNRS, Inserm, iBV, Parc Valrose, 06108 Nice, France; (T.N.); (S.S.); (A.P.); (A.V.); (M.E.F.); (C.A.); (A.G.-U.); (J.B.); (A.S.-D.-V.); (M.D.P.); (E.-Z.A.)
| | | | - Biljana Hadzic
- Pediatric Oncology & Hematology Department, Centre Hospitalier Universitaire de Nice, Hopital Archet 2, 06202 Nice, France;
| | - Jérôme Becam
- Faculté des Sciences, Université Côte d’Azur, CNRS, Inserm, iBV, Parc Valrose, 06108 Nice, France; (T.N.); (S.S.); (A.P.); (A.V.); (M.E.F.); (C.A.); (A.G.-U.); (J.B.); (A.S.-D.-V.); (M.D.P.); (E.-Z.A.)
| | - Anette Sousa-De-Veiga
- Faculté des Sciences, Université Côte d’Azur, CNRS, Inserm, iBV, Parc Valrose, 06108 Nice, France; (T.N.); (S.S.); (A.P.); (A.V.); (M.E.F.); (C.A.); (A.G.-U.); (J.B.); (A.S.-D.-V.); (M.D.P.); (E.-Z.A.)
| | - Magali Dodille Plaisant
- Faculté des Sciences, Université Côte d’Azur, CNRS, Inserm, iBV, Parc Valrose, 06108 Nice, France; (T.N.); (S.S.); (A.P.); (A.V.); (M.E.F.); (C.A.); (A.G.-U.); (J.B.); (A.S.-D.-V.); (M.D.P.); (E.-Z.A.)
| | | | - Didier F. Pisani
- Medicine Faculty, Université Côte d’Azur, CNRS, LP2M, 06003 Nice, France;
| | - Magali Mondin
- Pôle Imagerie Photonique, Bordeaux Imaging Center, Université de Bordeaux, UMS 3420 CNRS-US4 Inserm, 33076 Bordeaux, France;
| | - Philippe Schmitt-Kopplin
- Research Unit Analytical BioGeoChemistry, Helmholtz Zentrum München, German Research Center for Environment Health, 85764 Neuherberg, Germany; (S.F.); (P.S.-K.)
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Ez-Zoubir Amri
- Faculté des Sciences, Université Côte d’Azur, CNRS, Inserm, iBV, Parc Valrose, 06108 Nice, France; (T.N.); (S.S.); (A.P.); (A.V.); (M.E.F.); (C.A.); (A.G.-U.); (J.B.); (A.S.-D.-V.); (M.D.P.); (E.-Z.A.)
| | - Patrick Collombat
- Faculté des Sciences, Université Côte d’Azur, CNRS, Inserm, iBV, Parc Valrose, 06108 Nice, France; (T.N.); (S.S.); (A.P.); (A.V.); (M.E.F.); (C.A.); (A.G.-U.); (J.B.); (A.S.-D.-V.); (M.D.P.); (E.-Z.A.)
- Correspondence:
| |
Collapse
|
15
|
Fraszczak J, Arman KM, Lacroix M, Vadnais C, Gaboury L, Möröy T. Severe Inflammatory Reactions in Mice Expressing a GFI1 P2A Mutant Defective in Binding to the Histone Demethylase KDM1A (LSD1). THE JOURNAL OF IMMUNOLOGY 2021; 207:1599-1615. [PMID: 34408010 DOI: 10.4049/jimmunol.2001146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 07/07/2021] [Indexed: 11/19/2022]
Abstract
GFI1 is a DNA-binding transcription factor that regulates hematopoiesis by repressing target genes through its association with complexes containing histone demethylases such as KDM1A (LSD1) and histone deacetylases (HDACs). To study the consequences of the disruption of the complex between GFI1 and histone-modifying enzymes, we have used knock-in mice harboring a P2A mutation in GFI1 coding region that renders it unable to bind LSD1 and associated histone-modifying enzymes such as HDACs. GFI1P2A mice die prematurely and show increased numbers of memory effector and regulatory T cells in the spleen accompanied by a severe systemic inflammation with high serum levels of IL-6, TNF-α, and IL-1β and overexpression of the gene encoding the cytokine oncostatin M (OSM). We identified lung alveolar macrophages, CD8 T cell from the spleen and thymic eosinophils, and monocytes as the sources of these cytokines in GFI1P2A mice. Chromatin immunoprecipitation showed that GFI1/LSD1 complexes occupy sites at the Osm promoter and an intragenic region of the Tnfα gene and that a GFI1P2A mutant still remains bound at these sites even without LSD1. Methylation and acetylation of histone H3 at these sites were enriched in cells from GFI1P2A mice, the H3K27 acetylation being the most significant. These data suggest that the histone modification facilitated by GFI1 is critical to control inflammatory pathways in different cell types, including monocytes and eosinophils, and that a disruption of GFI1-associated complexes can lead to systemic inflammation with fatal consequences.
Collapse
Affiliation(s)
| | - Kaifee Mohammad Arman
- Institut de Recherches Cliniques de Montréal, Montreal, Canada.,Division of Experimental Medicine, McGill University, Montreal, Canada
| | - Marion Lacroix
- Institut de Recherches Cliniques de Montréal, Montreal, Canada.,Division of Experimental Medicine, McGill University, Montreal, Canada
| | - Charles Vadnais
- Institut de Recherches Cliniques de Montréal, Montreal, Canada
| | - Louis Gaboury
- Unité de Recherche en Histologie et Pathologie Moléculaire, Institut de Recherche en Immunologie et en Cancérologie, Montreal, Canada.,Département de Pathologie et Biologie Cellulaire, Faculté de Médecine, Université de Montréal, Montreal, Canada; and
| | - Tarik Möröy
- Institut de Recherches Cliniques de Montréal, Montreal, Canada; .,Division of Experimental Medicine, McGill University, Montreal, Canada.,Département de Microbiologie Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, Montreal, Canada
| |
Collapse
|
16
|
Gao S, Wang Z, Wang L, Wang H, Yuan H, Liu X, Chen S, Chen Z, de Thé H, Zhang W, Zhang Y, Zhu J, Zhou J. Irf2bp2a regulates terminal granulopoiesis through proteasomal degradation of Gfi1aa in zebrafish. PLoS Genet 2021; 17:e1009693. [PMID: 34351909 PMCID: PMC8370619 DOI: 10.1371/journal.pgen.1009693] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 08/17/2021] [Accepted: 07/02/2021] [Indexed: 11/19/2022] Open
Abstract
The ubiquitin-proteasome system plays important roles in various biological processes as it degrades the majority of cellular proteins. Adequate proteasomal degradation of crucial transcription regulators ensures the proper development of neutrophils. The ubiquitin E3 ligase of Growth factor independent 1 (GFI1), a key transcription repressor governing terminal granulopoiesis, remains obscure. Here we report that the deficiency of the ring finger protein Interferon regulatory factor 2 binding protein 2a (Irf2bp2a) leads to an impairment of neutrophils differentiation in zebrafish. Mechanistically, Irf2bp2a functions as a ubiquitin E3 ligase targeting Gfi1aa for proteasomal degradation. Moreover, irf2bp2a gene is repressed by Gfi1aa, thus forming a negative feedback loop between Irf2bp2a and Gfi1aa during neutrophils maturation. Different levels of GFI1 may turn it into a tumor suppressor or an oncogene in malignant myelopoiesis. Therefore, discovery of certain drug targets GFI1 for proteasomal degradation by IRF2BP2 might be an effective anti-cancer strategy.
Collapse
Affiliation(s)
- Shuo Gao
- Shanghai Institute of Hematology, CNRS-LIA Hematology and Cancer, Sino-French Research Center for Life Sciences and Genomics, State Key Laboratory of Medical Genomics, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Zixuan Wang
- Shanghai Institute of Hematology, CNRS-LIA Hematology and Cancer, Sino-French Research Center for Life Sciences and Genomics, State Key Laboratory of Medical Genomics, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Luxiang Wang
- Shanghai Institute of Hematology, CNRS-LIA Hematology and Cancer, Sino-French Research Center for Life Sciences and Genomics, State Key Laboratory of Medical Genomics, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
- Department of hematology, Shanghai General Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Haihong Wang
- Shanghai Institute of Hematology, CNRS-LIA Hematology and Cancer, Sino-French Research Center for Life Sciences and Genomics, State Key Laboratory of Medical Genomics, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Hao Yuan
- Shanghai Institute of Hematology, CNRS-LIA Hematology and Cancer, Sino-French Research Center for Life Sciences and Genomics, State Key Laboratory of Medical Genomics, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Xiaohui Liu
- Shanghai Institute of Hematology, CNRS-LIA Hematology and Cancer, Sino-French Research Center for Life Sciences and Genomics, State Key Laboratory of Medical Genomics, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Saijuan Chen
- Shanghai Institute of Hematology, CNRS-LIA Hematology and Cancer, Sino-French Research Center for Life Sciences and Genomics, State Key Laboratory of Medical Genomics, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Zhu Chen
- Shanghai Institute of Hematology, CNRS-LIA Hematology and Cancer, Sino-French Research Center for Life Sciences and Genomics, State Key Laboratory of Medical Genomics, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Hugues de Thé
- Shanghai Institute of Hematology, CNRS-LIA Hematology and Cancer, Sino-French Research Center for Life Sciences and Genomics, State Key Laboratory of Medical Genomics, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
- Université de Paris 7/INSERM/CNRS UMR 944/7212, Equipe Labellisée No. 11 Ligue Nationale Contre le Cancer, Hôpital St. Louis, Paris, France
| | - Wenqing Zhang
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, P.R. China
| | - Yiyue Zhang
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, P.R. China
- * E-mail: (YZ); (JZ); (JZ)
| | - Jun Zhu
- Shanghai Institute of Hematology, CNRS-LIA Hematology and Cancer, Sino-French Research Center for Life Sciences and Genomics, State Key Laboratory of Medical Genomics, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
- Université de Paris 7/INSERM/CNRS UMR 944/7212, Equipe Labellisée No. 11 Ligue Nationale Contre le Cancer, Hôpital St. Louis, Paris, France
- * E-mail: (YZ); (JZ); (JZ)
| | - Jun Zhou
- Shanghai Institute of Hematology, CNRS-LIA Hematology and Cancer, Sino-French Research Center for Life Sciences and Genomics, State Key Laboratory of Medical Genomics, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
- * E-mail: (YZ); (JZ); (JZ)
| |
Collapse
|
17
|
O'Reilly E, Zeinabad HA, Szegezdi E. Hematopoietic versus leukemic stem cell quiescence: Challenges and therapeutic opportunities. Blood Rev 2021; 50:100850. [PMID: 34049731 DOI: 10.1016/j.blre.2021.100850] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 04/22/2021] [Accepted: 05/07/2021] [Indexed: 12/13/2022]
Abstract
Hematopoietic stem cells (HSC) are responsible for the production of mature blood cells. To ensure that the HSC pool does not get exhausted over the lifetime of an individual, most HSCs are in a state of quiescence with only a small proportion of HSCs dividing at any one time. HSC quiescence is carefully controlled by both intrinsic and extrinsic, niche-driven mechanisms. In acute myeloid leukemia (AML), the leukemic cells overtake the hematopoietic bone marrow niche where they acquire a quiescent state. These dormant AML cells are resistant to chemotherapeutics. Because they can re-establish the disease after therapy, they are often termed as quiescent leukemic stem cells (LSC) or leukemia-initiating cells. While advancements are being made to target particular driver mutations in AML, there is less focus on how to tackle the drug resistance of quiescent LSCs. This review summarises the current knowledge on the biochemical characteristics of quiescent HSCs and LSCs, the intracellular signaling pathways and the niche-driven mechanisms that control quiescence and the key differences between HSC- and LSC-quiescence that may be exploited for therapy.
Collapse
Affiliation(s)
- Eimear O'Reilly
- Apoptosis Research Centre, Department of Biochemistry, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| | - Hojjat Alizadeh Zeinabad
- Apoptosis Research Centre, Department of Biochemistry, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| | - Eva Szegezdi
- Apoptosis Research Centre, Department of Biochemistry, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland.
| |
Collapse
|
18
|
The transcription factors GFI1 and GFI1B as modulators of the innate and acquired immune response. Adv Immunol 2021; 149:35-94. [PMID: 33993920 DOI: 10.1016/bs.ai.2021.03.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
GFI1 and GFI1B are small nuclear proteins of 45 and 37kDa, respectively, that have a simple two-domain structure: The first consists of a group of six c-terminal C2H2 zinc finger motifs that are almost identical in sequence and bind to very similar, specific DNA sites. The second is an N-terminal 20 amino acid SNAG domain that can bind to the pocket of the histone demethylase KDM1A (LSD1) near its active site. When bound to DNA, both proteins act as bridging factors that bring LSD1 and associated proteins into the vicinity of methylated substrates, in particular histone H3 or TP53. GFI1 can also bring methyl transferases such as PRMT1 together with its substrates that include the DNA repair proteins MRE11 and 53BP1, thereby enabling their methylation and activation. While GFI1B is expressed almost exclusively in the erythroid and megakaryocytic lineage, GFI1 has clear biological roles in the development and differentiation of lymphoid and myeloid immune cells. GFI1 is required for lymphoid/myeloid and monocyte/granulocyte lineage decision as well as the correct nuclear interpretation of a number of important immune-signaling pathways that are initiated by NOTCH1, interleukins such as IL2, IL4, IL5 or IL7, by the pre TCR or -BCR receptors during early lymphoid differentiation or by T and B cell receptors during activation of lymphoid cells. Myeloid cells also depend on GFI1 at both stages of early differentiation as well as later stages in the process of activation of macrophages through Toll-like receptors in response to pathogen-associated molecular patterns. The knowledge gathered on these factors over the last decades puts GFI1 and GFI1B at the center of many biological processes that are critical for both the innate and acquired immune system.
Collapse
|
19
|
Chan WF, Coughlan HD, Zhou JHS, Keenan CR, Bediaga NG, Hodgkin PD, Smyth GK, Johanson TM, Allan RS. Pre-mitotic genome re-organisation bookends the B cell differentiation process. Nat Commun 2021; 12:1344. [PMID: 33637722 PMCID: PMC7910489 DOI: 10.1038/s41467-021-21536-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 02/02/2021] [Indexed: 01/08/2023] Open
Abstract
During cellular differentiation chromosome conformation is intricately remodelled to support the lineage-specific transcriptional programs required for initiating and maintaining lineage identity. When these changes occur in relation to cell cycle, division and time in response to cellular activation and differentiation signals has yet to be explored, although it has been proposed to occur during DNA synthesis or after mitosis. Here, we elucidate the chromosome conformational changes in B lymphocytes as they differentiate and expand from a naive, quiescent state into antibody secreting plasma cells. We find gene-regulatory chromosome reorganization in late G1 phase before the first division, and that this configuration is remarkably stable as the cells massively and rapidly clonally expand. A second wave of conformational change occurs as cells terminally differentiate into plasma cells, coincident with increased time in G1 phase. These results provide further explanation for how lymphocyte fate is imprinted prior to the first division. They also suggest that chromosome reconfiguration occurs prior to DNA replication and mitosis, and is linked to a gene expression program that controls the differentiation process required for the generation of immunity.
Collapse
Affiliation(s)
- Wing Fuk Chan
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Hannah D Coughlan
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Jie H S Zhou
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Christine R Keenan
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Naiara G Bediaga
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Philip D Hodgkin
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Gordon K Smyth
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,School of Mathematics and Statistics, The University of Melbourne, Parkville, VIC, Australia
| | - Timothy M Johanson
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Rhys S Allan
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia. .,Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
20
|
Silva CP, Kamens HM. Cigarette smoke-induced alterations in blood: A review of research on DNA methylation and gene expression. Exp Clin Psychopharmacol 2021; 29:116-135. [PMID: 32658533 PMCID: PMC7854868 DOI: 10.1037/pha0000382] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Worldwide, smoking remains a threat to public health, causing preventable diseases and premature mortality. Cigarette smoke is a powerful inducer of DNA methylation and gene expression alterations, which have been associated with negative health consequences. Here, we review the current knowledge on smoking-related changes in DNA methylation and gene expression in human blood samples. We identified 30 studies focused on the association between active smoking, DNA methylation modifications, and gene expression alterations. Overall, we identified 1,758 genes with differentially methylated sites (DMS) and differentially expressed genes (DEG) between smokers and nonsmokers, of which 261 were detected in multiple studies (≥4). The most frequently (≥10 studies) reported genes were AHRR, GPR15, GFI1, and RARA. Functional enrichment analysis of the 261 genes identified the aryl hydrocarbon receptor repressor and T cell pathways (T helpers 1 and 2) as influenced by smoking status. These results highlight specific genes for future mechanistic and translational research that may be associated with cigarette smoke exposure and smoking-related diseases. (PsycInfo Database Record (c) 2021 APA, all rights reserved).
Collapse
Affiliation(s)
- Constanza P. Silva
- Department of Biobehavioral Health, The Pennsylvania State University, University Park, Pennsylvania, 16802, United States of America
| | - Helen M. Kamens
- Department of Biobehavioral Health, The Pennsylvania State University, University Park, Pennsylvania, 16802, United States of America.,Correspondence concerning this article should be addressed to Helen M. Kamens, 228 Biobehavioral Health Building, The Pennsylvania State University, University Park, PA 16802; ; Phone number: 814-865-1269; Fax number: 814-863-7525
| |
Collapse
|
21
|
Faleschini M, Papa N, Morel-Kopp MC, Marconi C, Giangregorio T, Melazzini F, Bozzi V, Seri M, Noris P, Pecci A, Savoia A, Bottega R. Dysregulation of oncogenic factors by GFI1B p32: investigation of a novel GFI1B germline mutation. Haematologica 2021; 107:260-267. [PMID: 33472357 PMCID: PMC8719102 DOI: 10.3324/haematol.2020.267328] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Indexed: 11/10/2022] Open
Abstract
GFI1B is a transcription factor essential for the regulation of erythropoiesis and megakaryopoiesis, and pathogenic variants have been associated with thrombocytopenia and bleeding. Analysing thrombocytopenic families by whole exome sequencing, we identified a novel GFI1B variant (c.648+5G>A), which causes exon 9 skipping and overexpression of a shorter p32 isoform. We report the clinical data of our patients and critically review the phenotype observed in individuals with different GFI1B variants leading to the same effect on the p32 expression. Since p32 is increased in acute and chronic leukemia cells, we tested the expression level of genes playing a role in various type of cancers, including hematological tumors and found that they are significantly dysregulated, suggesting a potential role for GFI1B in carcinogenesis regulation. Increasing the detection of individuals with GFI1B variants will allow us to better characterize this rare disease and determine whether it is associated with an increased risk of developing malignancies.
Collapse
Affiliation(s)
| | - Nicole Papa
- Institute for Maternal and Child Health - IRCCS Burlo Garofolo, Trieste
| | - Marie-Christine Morel-Kopp
- Department of Haematology and Transfusion Medicine, Royal North Shore Hospital and Northern Blood Research Centre, Kolling Institute, University of Sydney, Sydney
| | - Caterina Marconi
- Department of Medical and Surgical Sciences, University of Bologna, Bologna
| | | | - Federica Melazzini
- Biotechnology Research Laboratories, IRCCS Policlinico San Matteo Foundation, Pavia
| | - Valeria Bozzi
- Biotechnology Research Laboratories, IRCCS Policlinico San Matteo Foundation, Pavia
| | - Marco Seri
- Department of Medical and Surgical Sciences, University of Bologna, Bologna
| | - Patrizia Noris
- Biotechnology Research Laboratories, IRCCS Policlinico San Matteo Foundation, Pavia
| | - Alessandro Pecci
- Biotechnology Research Laboratories, IRCCS Policlinico San Matteo Foundation, Pavia
| | - Anna Savoia
- Institute for Maternal and Child Health - IRCCS Burlo Garofolo, Trieste, Italy; Department of Medical Sciences, University of Trieste, Trieste.
| | - Roberta Bottega
- Institute for Maternal and Child Health - IRCCS Burlo Garofolo, Trieste
| |
Collapse
|
22
|
Regulation of the MIE Locus During HCMV Latency and Reactivation. Pathogens 2020; 9:pathogens9110869. [PMID: 33113934 PMCID: PMC7690695 DOI: 10.3390/pathogens9110869] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/19/2020] [Accepted: 10/20/2020] [Indexed: 12/20/2022] Open
Abstract
Human cytomegalovirus (HCMV) is a ubiquitous herpesviral pathogen that results in life-long infection. HCMV maintains a latent or quiescent infection in hematopoietic cells, which is broadly defined by transcriptional silencing and the absence of de novo virion production. However, upon cell differentiation coupled with immune dysfunction, the virus can reactivate, which leads to lytic replication in a variety of cell and tissue types. One of the mechanisms controlling the balance between latency and reactivation/lytic replication is the regulation of the major immediate-early (MIE) locus. This enhancer/promoter region is complex, and it is regulated by chromatinization and associated factors, as well as a variety of transcription factors. Herein, we discuss these factors and how they influence the MIE locus, which ultimately impacts the phase of HCMV infection.
Collapse
|
23
|
Ashour N, Angulo JC, González-Corpas A, Orea MJ, Lobo MVT, Colomer R, Colás B, Esteller M, Ropero S. Epigenetic Regulation of Gfi1 in Endocrine-Related Cancers: a Role Regulating Tumor Growth. Int J Mol Sci 2020; 21:ijms21134687. [PMID: 32630147 PMCID: PMC7370116 DOI: 10.3390/ijms21134687] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 06/25/2020] [Accepted: 06/29/2020] [Indexed: 02/06/2023] Open
Abstract
Prostate and breast cancer constitute the most common cancers among men and women worldwide. The aging population is one of the main risk factors for prostate and breast cancer development and accumulating studies link aging with epigenetic changes. Growth factor independence-1 (Gfi1) is a transcriptional repressor with an important role in human malignancies, including leukemia, colorectal carcinoma, and lung cancer, but its role in prostate and breast cancer is unknown. We have found that Gfi1 epigenetic silencing is a common event in prostate and breast cancer. Gfi1 re-expression in prostate and breast cancer cell lines displaying Gfi1 epigenetic silencing decreases cell proliferation, reduced colony formation density, and tumor growth in nude mice xenografts. In addition, we found that Gfi1 repress alpha 1-anti-trypsin (AAT) and alpha 1-anti-chymotrypsin (ACT) expression, two genes with important functions in cancer development, suggesting that Gfi1 silencing promotes tumor growth by increasing AAT and ACT expression in our system. Finally, Gfi1 epigenetic silencing could be a promising biomarker for prostate cancer progression because it is associated with shorter disease-free survival. In conclusion, our findings strongly indicate that Gfi1 epigenetic silencing in prostate and breast cancer could be a crucial step in the development of these two-well characterized endocrine related tumors.
Collapse
Affiliation(s)
- Nadia Ashour
- Departamento de Biología de Sistemas, Unidad Docente de Bioquímica y Biología Molecular, Universidad de Alcalá, Alcalá de Henares, 28054 Madrid, Spain; (N.A.); (A.G.-C.); (M.J.O.); (B.C.)
| | - Javier C. Angulo
- Servicio de Urología, Hospital Universitario de Getafe, Fundación para la Investigación Biomédica del Hospital Universitario de Getafe, Universidad Europea de Madrid, Getafe, 28905 Madrid, Spain;
| | - Ana González-Corpas
- Departamento de Biología de Sistemas, Unidad Docente de Bioquímica y Biología Molecular, Universidad de Alcalá, Alcalá de Henares, 28054 Madrid, Spain; (N.A.); (A.G.-C.); (M.J.O.); (B.C.)
| | - María J. Orea
- Departamento de Biología de Sistemas, Unidad Docente de Bioquímica y Biología Molecular, Universidad de Alcalá, Alcalá de Henares, 28054 Madrid, Spain; (N.A.); (A.G.-C.); (M.J.O.); (B.C.)
| | - María V. T. Lobo
- Departamento de Biomedicina y Biotecnología, Universidad de Alcalá; Instituto Ramón y Cajal de Investigaciones Sanitarias (IRYCIS), 28054 Madrid, Spain;
| | - Ramón Colomer
- Medical Oncology Department, Instituto De Investigación Sanitaria La Princesa, HU La Princesa, 28029 Madrid, Spain;
- Department of Medicine, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Begoña Colás
- Departamento de Biología de Sistemas, Unidad Docente de Bioquímica y Biología Molecular, Universidad de Alcalá, Alcalá de Henares, 28054 Madrid, Spain; (N.A.); (A.G.-C.); (M.J.O.); (B.C.)
| | - Manel Esteller
- Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Catalonia, Spain;
- Centro de Investigacion Biomedica en Red Cancer (CIBERONC), 28040 Madrid, Spain
- Institucio Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Catalonia, Spain
- Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona (UB), 08028 Barcelona, Catalonia, Spain
| | - Santiago Ropero
- Departamento de Biología de Sistemas, Unidad Docente de Bioquímica y Biología Molecular, Universidad de Alcalá, Alcalá de Henares, 28054 Madrid, Spain; (N.A.); (A.G.-C.); (M.J.O.); (B.C.)
- Correspondence:
| |
Collapse
|
24
|
Jung S, Gies V, Korganow AS, Guffroy A. Primary Immunodeficiencies With Defects in Innate Immunity: Focus on Orofacial Manifestations. Front Immunol 2020; 11:1065. [PMID: 32625202 PMCID: PMC7314950 DOI: 10.3389/fimmu.2020.01065] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 05/04/2020] [Indexed: 12/23/2022] Open
Abstract
The field of primary immunodeficiencies (PIDs) is rapidly evolving. Indeed, the number of described diseases is constantly increasing thanks to the rapid identification of novel genetic defects by next-generation sequencing. PIDs are now rather referred to as “inborn errors of immunity” due to the association between a wide range of immune dysregulation-related clinical features and the “prototypic” increased infection susceptibility. The phenotypic spectrum of PIDs is therefore very large and includes several orofacial features. However, the latter are often overshadowed by severe systemic manifestations and remain underdiagnosed. Patients with impaired innate immunity are predisposed to a variety of oral manifestations including oral infections (e.g., candidiasis, herpes gingivostomatitis), aphthous ulcers, and severe periodontal diseases. Although less frequently, they can also show orofacial developmental abnormalities. Oral lesions can even represent the main clinical manifestation of some PIDs or be inaugural, being therefore one of the first features indicating the existence of an underlying immune defect. The aim of this review is to describe the orofacial features associated with the different PIDs of innate immunity based on the new 2019 classification from the International Union of Immunological Societies (IUIS) expert committee. This review highlights the important role played by the dentist, in close collaboration with the multidisciplinary medical team, in the management and the diagnostic of these conditions.
Collapse
Affiliation(s)
- Sophie Jung
- Université de Strasbourg, Faculté de Chirurgie Dentaire, Strasbourg, France.,Hôpitaux Universitaires de Strasbourg, Centre de Référence Maladies Rares Orales et Dentaires (O-Rares), Pôle de Médecine et de Chirurgie Bucco-Dentaires, Strasbourg, France.,Université de Strasbourg, INSERM UMR_S 1109 "Molecular ImmunoRheumatology", Strasbourg, France
| | - Vincent Gies
- Université de Strasbourg, INSERM UMR_S 1109 "Molecular ImmunoRheumatology", Strasbourg, France.,Université de Strasbourg, Faculté de Pharmacie, Illkirch-Graffenstaden, France.,Hôpitaux Universitaires de Strasbourg, Service d'Immunologie Clinique et de Médecine Interne, Centre de Référence des Maladies Auto-immunes Systémiques Rares (RESO), Centre de Compétences des Déficits Immunitaires Héréditaires, Strasbourg, France
| | - Anne-Sophie Korganow
- Université de Strasbourg, INSERM UMR_S 1109 "Molecular ImmunoRheumatology", Strasbourg, France.,Hôpitaux Universitaires de Strasbourg, Service d'Immunologie Clinique et de Médecine Interne, Centre de Référence des Maladies Auto-immunes Systémiques Rares (RESO), Centre de Compétences des Déficits Immunitaires Héréditaires, Strasbourg, France.,Université de Strasbourg, Faculté de Médecine, Strasbourg, France
| | - Aurélien Guffroy
- Université de Strasbourg, INSERM UMR_S 1109 "Molecular ImmunoRheumatology", Strasbourg, France.,Hôpitaux Universitaires de Strasbourg, Service d'Immunologie Clinique et de Médecine Interne, Centre de Référence des Maladies Auto-immunes Systémiques Rares (RESO), Centre de Compétences des Déficits Immunitaires Héréditaires, Strasbourg, France.,Université de Strasbourg, Faculté de Médecine, Strasbourg, France
| |
Collapse
|
25
|
Mouse models of neutropenia reveal progenitor-stage-specific defects. Nature 2020; 582:109-114. [PMID: 32494068 PMCID: PMC8041154 DOI: 10.1038/s41586-020-2227-7] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 02/24/2020] [Indexed: 02/07/2023]
Abstract
Advances in genetics and sequencing reveal a plethora of disease-associated and disease-causing genetic alterations. Resolving causality between genetics and disease requires generating accurate models for molecular dissection; however, the rapid expansion of single-cell landscapes presents a major challenge to accurate comparisons between mutants and their wild-type equivalents. Here, we generated mouse models of human severe congenital neutropenia (SCN) using patient-derived mutations in the Growth factor independent-1 (GFI1) transcription factor. To delineate the impact of SCN mutations, we generated single-cell references for granulopoietic genomic states with linked epitopes1, aligned mutant cells to their wild-type equivalent and identified differentially expressed genes and epigenetic loci. We find that Gfi1-target genes are altered sequentially, as cells traverse successive states during differentiation. These cell-state-specific insights facilitated genetic rescue of granulocytic specification but not post-commitment defects in innate-immune effector function; underscoring the importance of evaluating the impact of mutations and therapy within each relevant cell state.
Collapse
|
26
|
Forte E, Zhang Z, Thorp EB, Hummel M. Cytomegalovirus Latency and Reactivation: An Intricate Interplay With the Host Immune Response. Front Cell Infect Microbiol 2020; 10:130. [PMID: 32296651 PMCID: PMC7136410 DOI: 10.3389/fcimb.2020.00130] [Citation(s) in RCA: 129] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 03/10/2020] [Indexed: 12/16/2022] Open
Abstract
CMV is an ancient herpesvirus that has co-evolved with its host over millions of years. The 236 kbp genome encodes at least 165 genes, four non-coding RNAs and 14 miRNAs. Of the protein-coding genes, 43-44 are core replication genes common to all herpesviruses, while ~30 are unique to betaherpesviruses. Many CMV genes are involved in evading detection by the host immune response, and others have roles in cell tropism. CMV replicates systemically, and thus, has adapted to various biological niches within the host. Different biological niches may place competing demands on the virus, such that genes that are favorable in some contexts are unfavorable in others. The outcome of infection is dependent on the cell type. In fibroblasts, the virus replicates lytically to produce infectious virus. In other cell types, such as myeloid progenitor cells, there is an initial burst of lytic gene expression, which is subsequently silenced through epigenetic repression, leading to establishment of latency. Latently infected monocytes disseminate the virus to various organs. Latency is established through cell type specific mechanisms of transcriptional silencing. In contrast, reactivation is triggered through pathways activated by inflammation, infection, and injury that are common to many cell types, as well as differentiation of myeloid cells to dendritic cells. Thus, CMV has evolved a complex relationship with the host immune response, in which it exploits cell type specific mechanisms of gene regulation to establish latency and to disseminate infection systemically, and also uses the inflammatory response to infection as an early warning system which allows the virus to escape from situations in which its survival is threatened, either by cellular damage or infection of the host with another pathogen. Spontaneous reactivation induced by cellular aging/damage may explain why extensive expression of lytic genes has been observed in recent studies using highly sensitive transcriptome analyses of cells from latently infected individuals. Recent studies with animal models highlight the potential for harnessing the host immune response to blunt cellular injury induced by organ transplantation, and thus, prevent reactivation of CMV and its sequelae.
Collapse
Affiliation(s)
- Eleonora Forte
- Department of Surgery, Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Zheng Zhang
- Department of Surgery, Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Edward B. Thorp
- Department of Pathology and Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Mary Hummel
- Department of Surgery, Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
27
|
Stackowicz J, Jönsson F, Reber LL. Mouse Models and Tools for the in vivo Study of Neutrophils. Front Immunol 2020; 10:3130. [PMID: 32038641 PMCID: PMC6985372 DOI: 10.3389/fimmu.2019.03130] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 12/23/2019] [Indexed: 12/21/2022] Open
Abstract
Neutrophils are the most abundant leukocytes in human blood and critical actors of the immune system. Many neutrophil functions and facets of their activity in vivo were revealed by studying genetically modified mice or by tracking fluorescent neutrophils in animals using imaging approaches. Assessing the roles of neutrophils can be challenging, especially when exact molecular pathways are questioned or disease states are interrogated that alter normal neutrophil homeostasis. This review discusses the main in vivo models for the study of neutrophils, their advantages and limitations. The side-by-side comparison underlines the necessity to carefully choose the right model(s) to answer a given scientific question, and exhibit caveats that need to be taken into account when designing experimental procedures. Collectively, this review suggests that at least two models should be employed to legitimately conclude on neutrophil functions.
Collapse
Affiliation(s)
- Julien Stackowicz
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, UMR INSERM 1222, Paris, France.,Sorbonne Université, Collège Doctoral, Paris, France
| | - Friederike Jönsson
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, UMR INSERM 1222, Paris, France
| | - Laurent L Reber
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, UMR INSERM 1222, Paris, France.,Center for Pathophysiology Toulouse-Purpan (CPTP), UMR 1043, University of Toulouse, INSERM, CNRS, Toulouse, France
| |
Collapse
|
28
|
Ajuba: An emerging signal transducer in oncogenesis. Pharmacol Res 2019; 151:104546. [PMID: 31740385 DOI: 10.1016/j.phrs.2019.104546] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 11/05/2019] [Accepted: 11/13/2019] [Indexed: 12/12/2022]
Abstract
The LIM protein Ajuba contains an unstructured proline/glycine-rich preLIM region in the N terminus and conserved tandem LIM motifs in the C terminus. Additionally, Ajuba contains both nuclear export sequences (NES) and nuclear localization sequences (NLS), which enable Ajuba shuttle between the cytoplasm and the nucleus. Thus, Ajuba can act as a versatile scaffold participating in assembly of variety of protein complexes to execute multiple cellular functions including cell adhesion, motility, mitosis, survival, gene expression, microRNA processing and mechanical force sensing. Numerous studies have demonstrated that Ajuba plays important roles in oncogenesis and progression by regulating major signalling pathways such as Wnt, RAS/ERK, JAK/STAT and Hippo, and by acting as a co-regulator of key transcription factors such as Snail, Sp1 and nuclear hormone receptors. Clinically, Ajuba is highly expressed in various types of tumors and can be a marker for prognosis and diagnosis. In this review, we aim to summarize the up-to-date literatures on the signaling pathways mediated by Ajuba and its associated protein complexes in oncogenesis, and to discuss Ajuba as a potential target for new therapeutics to treat cancers.
Collapse
|
29
|
van Bergen MGJM, van der Reijden BA. Targeting the GFI1/1B-CoREST Complex in Acute Myeloid Leukemia. Front Oncol 2019; 9:1027. [PMID: 31649884 PMCID: PMC6794713 DOI: 10.3389/fonc.2019.01027] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 09/23/2019] [Indexed: 11/21/2022] Open
Abstract
One of the hallmarks of acute myeloid leukemia (AML) is a block in cellular differentiation. Recent studies have shown that small molecules targeting Lysine Specific Demethylase 1A (KDM1A) may force the malignant cells to terminally differentiate. KDM1A is a core component of the chromatin binding CoREST complex. Together with histone deacetylases CoREST regulates gene expression by histone 3 demethylation and deacetylation. The transcription factors GFI1 and GFI1B (for growth factor independence) are major interaction partners of KDM1A and recruit the CoREST complex to chromatin in myeloid cells. Recent studies show that the small molecules that target KDM1A disrupt the GFI1/1B-CoREST interaction and that this is key to inducing terminal differentiation of leukemia cells.
Collapse
Affiliation(s)
| | - Bert A. van der Reijden
- Laboratory of Hematology, Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
30
|
The regulatory elements of PLZF gene are not conserved as reveled by molecular cloning and functional characterization of PLZF gene promoter of Clarias batrachus. GENE REPORTS 2019. [DOI: 10.1016/j.genrep.2019.100402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
31
|
Cheng AN, Bao EL, Fiorini C, Sankaran VG. Macrothrombocytopenia associated with a rare GFI1B missense variant confounding the presentation of immune thrombocytopenia. Pediatr Blood Cancer 2019; 66:e27874. [PMID: 31207059 PMCID: PMC6646087 DOI: 10.1002/pbc.27874] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 05/03/2019] [Accepted: 05/25/2019] [Indexed: 11/05/2022]
Abstract
Growth factor-independent 1B (GFI1B) variants are a rare cause of thrombocytopenia. We report on a male child who was initially diagnosed with immune thrombocytopenia. However, subtle clinical signs led to suspicion of a genetic cause of thrombocytopenia. Gene panel sequencing revealed a rare variant in GFI1B (C168F), which has recently been reported in several families with thrombocytopenia. We demonstrate that this variant significantly alters platelet parameters in population studies. This case highlights how diagnoses of exclusion, such as immune thrombocytopenia, can be confounded by genetic variation. Our understanding of blood disorders will undoubtedly evolve from an increased knowledge of human genetic variation.
Collapse
Affiliation(s)
- Aaron N. Cheng
- Division of Hematology/Oncology, Boston Children’s Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School Boston, MA, USA,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Erik L. Bao
- Division of Hematology/Oncology, Boston Children’s Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School Boston, MA, USA,Broad Institute of MIT and Harvard, Cambridge, MA, USA,Harvard-MIT Health Sciences and Technology, Harvard Medical School, Boston, MA, USA
| | - Claudia Fiorini
- Division of Hematology/Oncology, Boston Children’s Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School Boston, MA, USA,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Vijay G. Sankaran
- Division of Hematology/Oncology, Boston Children’s Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School Boston, MA, USA,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
32
|
McClellan D, Casey MJ, Bareyan D, Lucente H, Ours C, Velinder M, Singer J, Lone MD, Sun W, Coria Y, Mason CC, Engel ME. Growth Factor Independence 1B-Mediated Transcriptional Repression and Lineage Allocation Require Lysine-Specific Demethylase 1-Dependent Recruitment of the BHC Complex. Mol Cell Biol 2019; 39:e00020-19. [PMID: 30988160 PMCID: PMC6580704 DOI: 10.1128/mcb.00020-19] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 01/30/2019] [Accepted: 04/09/2019] [Indexed: 12/16/2022] Open
Abstract
Growth factor independence 1B (GFI1B) coordinates assembly of transcriptional repressor complexes comprised of corepressors and histone-modifying enzymes to control gene expression programs governing lineage allocation in hematopoiesis. Enforced expression of GFI1B in K562 erythroleukemia cells favors erythroid over megakaryocytic differentiation, providing a platform to define molecular determinants of binary fate decisions triggered by GFI1B. We deployed proteome-wide proximity labeling to identify factors whose inclusion in GFI1B complexes depends upon GFI1B's obligate effector, lysine-specific demethylase 1 (LSD1). We show that GFI1B preferentially recruits core and putative elements of the BRAF-histone deacetylase (HDAC) (BHC) chromatin-remodeling complex (LSD1, RCOR1, HMG20A, HMG20B, HDAC1, HDAC2, PHF21A, GSE1, ZMYM2, and ZNF217) in an LSD1-dependent manner to control acquisition of erythroid traits by K562 cells. Among these elements, depletion of both HMG20A and HMG20B or of GSE1 blocks GFI1B-mediated erythroid differentiation, phenocopying impaired differentiation brought on by LSD1 depletion or disruption of GFI1B-LSD1 binding. These findings demonstrate the central role of the GFI1B-LSD1 interaction as a determinant of BHC complex recruitment to enable cell fate decisions driven by GFI1B.
Collapse
Affiliation(s)
- David McClellan
- Department of Oncological Sciences, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Mattie J Casey
- Department of Oncological Sciences, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Diana Bareyan
- Department of Oncological Sciences, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Helena Lucente
- Department of Oncological Sciences, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Christopher Ours
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah, USA
- Primary Children's Hospital, Salt Lake City, Utah, USA
| | - Matthew Velinder
- Department of Genetics, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Jason Singer
- Department of Genetics, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Mehraju Din Lone
- Department of Oncological Sciences, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Wenxiang Sun
- Department of Oncological Sciences, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Yunuen Coria
- Department of Oncological Sciences, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Clinton C Mason
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah, USA
- Primary Children's Hospital, Salt Lake City, Utah, USA
| | - Michael E Engel
- Department of Oncological Sciences, University of Utah School of Medicine, Salt Lake City, Utah, USA
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah, USA
- Primary Children's Hospital, Salt Lake City, Utah, USA
- Center for Investigational Therapeutics, Huntsman Cancer Institute, Salt Lake City, Utah, USA
- Nuclear Control of Cell Growth and Differentiation Program, Huntsman Cancer Institute, Salt Lake City, Utah, USA
| |
Collapse
|
33
|
Tang Q, Xu M, Xu J, Xie X, Yang H, Gan L. Gfi1-GCE inducible Cre line for hair cell-specific gene manipulation in mouse inner ear. Genesis 2019; 57:e23304. [PMID: 31077553 DOI: 10.1002/dvg.23304] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 04/29/2019] [Accepted: 04/30/2019] [Indexed: 01/16/2023]
Abstract
Tissue-specific inducible Cre recombinase mouse lines allow precise genetic manipulations in spatiotemporal manners and are pivotal for functional studies of genes during development and in adults. Growth factor independence 1 (GFI1) is an essential transcription factor expressed in the hair cells of mouse inner ear and Gfi1 locus serves as an excellent anchor site to drive the expression of inducible Cre recombinase in mouse inner hair cells. In this study, we have generated Gfi1-P2A-GFP-CreERT2 (Gfi1-GCE) knock-in mouse line by in-frame fusion of a self-cleaving GCE to the C-terminus of GFI1. We have shown that as predicted, the expression of GCE and GFI1 was detected specifically in the cytosol and nuclei of hair cells, respectively, of uninduced Gfi1-GCE mice, suggesting the successful cleavage and simultaneous expression of GFI1 and GCE. In addition, the in-frame fusion of the self-cleaving GCE does not interrupt the function of Gfi1 in the inner ear. Administration of tamoxifen leads to nuclear translocation of GCE and results in an efficient activation of tdTomato reporter gene expression specifically in most hair cells throughout development and in adults. Thus, this inducible Gfi1-GCE mouse line is a highly efficient Cre deleter and is suitable for gene manipulation in developing and adult inner ear hair cells.
Collapse
Affiliation(s)
- Qi Tang
- Department of Otolaryngology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Department of Ophthalmology and Flaum Eye Institute, University of Rochester, Rochester, New York
| | - Mei Xu
- Department of Ophthalmology and Flaum Eye Institute, University of Rochester, Rochester, New York.,Institute of Life Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Jiadong Xu
- Department of Ophthalmology and Flaum Eye Institute, University of Rochester, Rochester, New York.,Institute of Life Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Xiaoling Xie
- Department of Ophthalmology and Flaum Eye Institute, University of Rochester, Rochester, New York
| | - Hua Yang
- Department of Otolaryngology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lin Gan
- Department of Ophthalmology and Flaum Eye Institute, University of Rochester, Rochester, New York
| |
Collapse
|
34
|
Moore C, Richens JL, Hough Y, Ucanok D, Malla S, Sang F, Chen Y, Elworthy S, Wilkinson RN, Gering M. Gfi1aa and Gfi1b set the pace for primitive erythroblast differentiation from hemangioblasts in the zebrafish embryo. Blood Adv 2018; 2:2589-2606. [PMID: 30309860 PMCID: PMC6199651 DOI: 10.1182/bloodadvances.2018020156] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 09/07/2018] [Indexed: 12/14/2022] Open
Abstract
The transcriptional repressors Gfi1(a) and Gfi1b are epigenetic regulators with unique and overlapping roles in hematopoiesis. In different contexts, Gfi1 and Gfi1b restrict or promote cell proliferation, prevent apoptosis, influence cell fate decisions, and are essential for terminal differentiation. Here, we show in primitive red blood cells (prRBCs) that they can also set the pace for cellular differentiation. In zebrafish, prRBCs express 2 of 3 zebrafish Gfi1/1b paralogs, Gfi1aa and Gfi1b. The recently identified zebrafish gfi1aa gene trap allele qmc551 drives erythroid green fluorescent protein (GFP) instead of Gfi1aa expression, yet homozygous carriers have normal prRBCs. prRBCs display a maturation defect only after splice morpholino-mediated knockdown of Gfi1b in gfi1aa qmc551 homozygous embryos. To study the transcriptome of the Gfi1aa/1b double-depleted cells, we performed an RNA-Seq experiment on GFP-positive prRBCs sorted from 20-hour-old embryos that were heterozygous or homozygous for gfi1aa qmc551 , as well as wt or morphant for gfi1b We subsequently confirmed and extended these data in whole-mount in situ hybridization experiments on newly generated single- and double-mutant embryos. Combined, the data showed that in the absence of Gfi1aa, the synchronously developing prRBCs were delayed in activating late erythroid differentiation, as they struggled to suppress early erythroid and endothelial transcription programs. The latter highlighted the bipotent nature of the progenitors from which prRBCs arise. In the absence of Gfi1aa, Gfi1b promoted erythroid differentiation as stepwise loss of wt gfi1b copies progressively delayed Gfi1aa-depleted prRBCs even further, showing that Gfi1aa and Gfi1b together set the pace for prRBC differentiation from hemangioblasts.
Collapse
Affiliation(s)
| | | | | | | | - Sunir Malla
- Deep Seq, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom
| | - Fei Sang
- Deep Seq, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom
| | - Yan Chen
- Department of Infection, Immunity & Cardiovascular Disease, Medical School, and
- Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | - Stone Elworthy
- Department of Infection, Immunity & Cardiovascular Disease, Medical School, and
- Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | - Robert N Wilkinson
- Department of Infection, Immunity & Cardiovascular Disease, Medical School, and
- Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | | |
Collapse
|
35
|
Cai H, Zhang F, Li Z. Gfi-1 promotes proliferation of human cervical carcinoma via targeting of FBW7 ubiquitin ligase expression. Cancer Manag Res 2018; 10:2849-2857. [PMID: 30197537 PMCID: PMC6113912 DOI: 10.2147/cmar.s161130] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background The independent growth factor 1 (Gfi-1) is a transcription factor essential for several diverse hematopoietic functions and developments. However, the role and molecular mechanism of Gfi-1 in the development and progression of cervical cancer remains unclear. Purpose The present study investigates the relation of expression of Gfi-1 with prognoses in patients with cervical cancer. Methods We used Western blot and reverse transcription polymerase chain reaction (RT-PCR) and the inhibition of proliferation and metastasis of cervical cancer cells in vitro. Results This study confirms that the expression of Gfi-1 in cervical cancer tissues was higher than that in adjacent normal tissues. The level of Gfi-1 mRNA in human cervical cancer tissues was significantly higher than that in normal tissues adjacent to cancer. Furthermore, overexpression of Gfi-1 promoted cell proliferation, colony formation, and migration of cervical cancer cells. The increased expression of Gfi-1 promotes the proliferation of cervical cancer cells targeting the tumor suppressor F-box and WD repeat domain containing 7 (FBW7). Clinically, our data suggest that overexpression of Gfi-1 is associated with poor prognosis in patients with cervical cancer. In a tumor xenograft model, knockdown of Gfi-1 inhibited the tumor growth of Hela cells in vivo. Conclusion Our results reveal that Gfi-1 plays an important role in cervical cancer and Gfi-1/FBW7 axis serves as a potential therapeutic target for cervical cancer.
Collapse
Affiliation(s)
- Hongbing Cai
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, .,Hubei Clinical Cancer Study Center, .,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, People's Republic of China,
| | - Fan Zhang
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, .,Hubei Clinical Cancer Study Center, .,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, People's Republic of China,
| | - Zhen Li
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, .,Hubei Clinical Cancer Study Center, .,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, People's Republic of China,
| |
Collapse
|
36
|
Collins-McMillen D, Buehler J, Peppenelli M, Goodrum F. Molecular Determinants and the Regulation of Human Cytomegalovirus Latency and Reactivation. Viruses 2018; 10:E444. [PMID: 30127257 PMCID: PMC6116278 DOI: 10.3390/v10080444] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 08/16/2018] [Accepted: 08/17/2018] [Indexed: 02/06/2023] Open
Abstract
Human cytomegalovirus (HCMV) is a beta herpesvirus that establishes a life-long persistence in the host, like all herpesviruses, by way of a latent infection. During latency, viral genomes are maintained in a quieted state. Virus replication can be reactivated from latency in response to changes in cellular signaling caused by stress or differentiation. The past decade has brought great insights into the molecular basis of HCMV latency. Here, we review the complex persistence of HCMV with consideration of latent reservoirs, viral determinants and their host interactions, and host signaling and the control of cellular and viral gene expression that contributes to the establishment of and reactivation from latency.
Collapse
Affiliation(s)
| | - Jason Buehler
- BIO5 Institute, University of Arizona, Tucson, AZ 85721, USA.
| | | | - Felicia Goodrum
- BIO5 Institute, University of Arizona, Tucson, AZ 85721, USA.
- Department of Immunobiology, University of Arizona, Tucson, AZ 85721, USA.
| |
Collapse
|
37
|
Spatial regulation of expanded transcription in the Drosophila wing imaginal disc. PLoS One 2018; 13:e0201317. [PMID: 30063727 PMCID: PMC6067730 DOI: 10.1371/journal.pone.0201317] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 07/12/2018] [Indexed: 12/13/2022] Open
Abstract
Growth and patterning are coordinated during development to define organ size and shape. The growth, proliferation and differentiation of Drosophila wings are regulated by several conserved signaling pathways. Here, we show that the Salvador-Warts-Hippo (SWH) and Notch pathways converge on an enhancer in the expanded (ex) gene, which also responds to levels of the bHLH transcription factor Daughterless (Da). Separate cis-regulatory elements respond to Salvador-Warts-Hippo (SWH) and Notch pathways, to bHLH proteins, and to unidentified factors that repress ex transcription in the wing pouch and in the proneural region at the anterior wing margin. Senseless, a zinc-finger transcription factor acting in proneural regions, had a negative impact on ex transcription in the proneural region, but the transcriptional repressor Hairy had no effect. Our study suggests that a complex pattern of ex transcription results from integration of a uniform SWH signal with multiple other inputs, rather than from a pattern of SWH signaling.
Collapse
|
38
|
Reduced expression but not deficiency of GFI1 causes a fatal myeloproliferative disease in mice. Leukemia 2018; 33:110-121. [PMID: 29925903 PMCID: PMC6326955 DOI: 10.1038/s41375-018-0166-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 04/25/2018] [Accepted: 05/10/2018] [Indexed: 12/15/2022]
Abstract
Growth factor independent 1 (Gfi1) controls myeloid differentiation by regulating gene expression and limits the activation of p53 by facilitating its de-methylation at Lysine 372. In human myeloid leukemia, low GFI1 levels correlate with an inferior prognosis. Here, we show that knockdown (KD) of Gfi1 in mice causes a fatal myeloproliferative disease (MPN) that could progress to leukemia after additional mutations. Both KO and KD mice accumulate myeloid cells that show signs of metabolic stress and high levels of reactive oxygen species. However, only KO cells have elevated levels of Lysine 372 methylated p53. This suggests that in contrast to absence of GFI1, KD of GFI1 leads to the accumulation of myeloid cells because sufficient amount of GFI1 is present to impede p53-mediated cell death, leading to a fatal MPN. The combination of myeloid accumulation and the ability to counteract p53 activity under metabolic stress could explain the role of reduced GF1 expression in human myeloid leukemia.
Collapse
|
39
|
Inactivation of Ezh2 Upregulates Gfi1 and Drives Aggressive Myc-Driven Group 3 Medulloblastoma. Cell Rep 2017; 18:2907-2917. [PMID: 28329683 DOI: 10.1016/j.celrep.2017.02.073] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 01/30/2017] [Accepted: 02/24/2017] [Indexed: 01/26/2023] Open
Abstract
The most aggressive of four medulloblastoma (MB) subgroups are cMyc-driven group 3 (G3) tumors, some of which overexpress EZH2, the histone H3K27 mono-, di-, and trimethylase of polycomb-repressive complex 2. Ezh2 has a context-dependent role in different cancers as an oncogene or tumor suppressor and retards tumor progression in a mouse model of G3 MB. Engineered deletions of Ezh2 in G3 MBs by gene editing nucleases accelerated tumorigenesis, whereas Ezh2 re-expression reversed attendant histone modifications and slowed tumor progression. Candidate oncogenic drivers suppressed by Ezh2 included Gfi1, a proto-oncogene frequently activated in human G3 MBs. Gfi1 disruption antagonized the tumor-promoting effects of Ezh2 loss; conversely, Gfi1 overexpression collaborated with Myc to bypass effects of Trp53 inactivation in driving MB progression in primary cerebellar neuronal progenitors. Although negative regulation of Gfi1 by Ezh2 may restrain MB development, Gfi1 activation can bypass these effects.
Collapse
|
40
|
Rabbolini DJ, Morel-Kopp MC, Chen Q, Gabrielli S, Dunlop LC, Chew LP, Blair N, Brighton TA, Singh N, Ng AP, Ward CM, Stevenson WS. Thrombocytopenia and CD34 expression is decoupled from α-granule deficiency with mutation of the first growth factor-independent 1B zinc finger. J Thromb Haemost 2017; 15:2245-2258. [PMID: 28880435 DOI: 10.1111/jth.13843] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Indexed: 01/23/2023]
Abstract
Essentials The phenotypes of different growth factor-independent 1B (GFI1B) variants are not established. GFI1B variants produce heterogeneous clinical phenotypes dependent on the site of mutation. Mutation of the first non-DNA-binding zinc-finger causes a mild platelet and clinical phenotype. GFI1B regulates the CD34 promoter; platelet CD34 expression is an indicator of GFI1B mutation. SUMMARY Background Mutation of the growth factor-independent 1B (GFI1B) fifth DNA-binding zinc-finger domain causes macrothrombocytopenia and α-granule deficiency leading to clinical bleeding. The phenotypes associated with GFI1B variants disrupting non-DNA-binding zinc-fingers remain uncharacterized. Objectives To determine the functional and phenotypic consequences of GFI1B variants disrupting non-DNA-binding zinc-finger domains. Methods The GFI1B C168F variant and a novel GFI1B c.2520 + 1_2520 + 8delGTGGGCAC splice variant were identified in four unrelated families. Phenotypic features, DNA-binding properties and transcriptional effects were determined and compared with those in individuals with a GFI1B H294 fs mutation of the fifth DNA-binding zinc-finger. Patient-specific induced pluripotent stem cell (iPSC)-derived megakaryocytes were generated to facilitate disease modeling. Results The DNA-binding GFI1B variant C168F, which is predicted to disrupt the first non-DNA-binding zinc-finger domain, is associated with macrothrombocytopenia without α-granule deficiency or bleeding symptoms. A GFI1B splice variant, c.2520 + 1_2520 + 8delGTGGGCAC, which generates a short GFI1B isoform that lacks non-DNA-binding zinc-fingers 1 and 2, is associated with increased platelet CD34 expression only, without quantitative or morphologic platelet abnormalities. GFI1B represses the CD34 promoter, and this repression is attenuated by different GFI1B zinc-finger mutations, suggesting that deregulation of CD34 expression occurs at a direct transcriptional level. Patient-specific iPSC-derived megakaryocytes phenocopy these observations. Conclusions Disruption of GFI1B non-DNA-binding zinc-finger 1 is associated with mild to moderate thrombocytopenia without α-granule deficiency or bleeding symptomatology, indicating that the site of GFI1B mutation has important phenotypic implications. Platelet CD34 expression appears to be a common feature of perturbed GFI1B function, and may have diagnostic utility.
Collapse
Affiliation(s)
- D J Rabbolini
- Northern Blood Research Centre, Kolling Institute of Medical Research, University of Sydney, Sydney, Australia
| | - M-C Morel-Kopp
- Northern Blood Research Centre, Kolling Institute of Medical Research, University of Sydney, Sydney, Australia
| | - Q Chen
- Northern Blood Research Centre, Kolling Institute of Medical Research, University of Sydney, Sydney, Australia
| | - S Gabrielli
- Northern Blood Research Centre, Kolling Institute of Medical Research, University of Sydney, Sydney, Australia
| | - L C Dunlop
- Department of Haematology, Liverpool Hospital, Sydney, Australia
| | - L P Chew
- Department of Haematology, Sarawak General Hospital, Sarawak, Malaysia
| | - N Blair
- Department of Neurogenetics, The Royal North Shore Hospital, Sydney, Australia
| | - T A Brighton
- Department of Haematology, Prince of Wales Hospital, Sydney, Australia
| | - N Singh
- Department of Haematology and Transfusion Medicine, Royal North Shore Hospital, Sydney, Australia
| | - A P Ng
- Department of Cancer and Haematology, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Australia
| | - C M Ward
- Northern Blood Research Centre, Kolling Institute of Medical Research, University of Sydney, Sydney, Australia
| | - W S Stevenson
- Northern Blood Research Centre, Kolling Institute of Medical Research, University of Sydney, Sydney, Australia
| |
Collapse
|
41
|
Rabbolini DJ, Morel-Kopp MC, Ward CM, Stevenson WS. GFI1B variants associated with thrombocytopenia. Platelets 2017; 28:525-527. [PMID: 28580815 DOI: 10.1080/09537104.2017.1317734] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- David J Rabbolini
- a Department of Haematology and Transfusion Medicine , Royal North Shore Hospital , Sydney , Australia.,b Northern Blood Research Centre , Kolling Institute of Medical Research, University of Sydney , Sydney , Australia
| | - Marie-Christine Morel-Kopp
- a Department of Haematology and Transfusion Medicine , Royal North Shore Hospital , Sydney , Australia.,b Northern Blood Research Centre , Kolling Institute of Medical Research, University of Sydney , Sydney , Australia
| | - Christopher M Ward
- a Department of Haematology and Transfusion Medicine , Royal North Shore Hospital , Sydney , Australia.,b Northern Blood Research Centre , Kolling Institute of Medical Research, University of Sydney , Sydney , Australia
| | - William S Stevenson
- a Department of Haematology and Transfusion Medicine , Royal North Shore Hospital , Sydney , Australia.,b Northern Blood Research Centre , Kolling Institute of Medical Research, University of Sydney , Sydney , Australia
| |
Collapse
|
42
|
Schulze H, Schlagenhauf A, Manukjan G, Beham-Schmid C, Andres O, Klopocki E, König EM, Haidl H, Panzer S, Althaus K, Muntean WE, Schwinger W, Urban C, Greinacher A, Bakchoul T, Seidel MG. Recessive grey platelet-like syndrome with unaffected erythropoiesis in the absence of the splice isoform GFI1B-p37. Haematologica 2017; 102:e375-e378. [PMID: 28550182 DOI: 10.3324/haematol.2017.167957] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Harald Schulze
- Chair of Experimental Biomedicine I, University Hospital Würzburg, Germany
| | - Axel Schlagenhauf
- Department of Pediatrics and Adolescent Medicine, Medical University Graz, Austria
| | - Georgi Manukjan
- Chair of Experimental Biomedicine I, University Hospital Würzburg, Germany
| | | | - Oliver Andres
- University Children's Hospital, University Hospital Würzburg, Germany
| | - Eva Klopocki
- Institute of Human Genetics, Biocenter, University of Würzburg, Germany
| | - Eva-Maria König
- Institute of Human Genetics, Biocenter, University of Würzburg, Germany
| | - Harald Haidl
- Department of Pediatrics and Adolescent Medicine, Medical University Graz, Austria
| | - Simon Panzer
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Vienna, Austria
| | - Karina Althaus
- Institute of Immunology and Transfusion Medicine, Universitätsmedizin Greifswald, Germany
| | - Wolfgang E Muntean
- Department of Pediatrics and Adolescent Medicine, Medical University Graz, Austria
| | - Wolfgang Schwinger
- Division of Pediatric Hematology-Oncology, Department of Pediatrics and Adolescent Medicine, Medical University Graz, Austria
| | - Christian Urban
- Division of Pediatric Hematology-Oncology, Department of Pediatrics and Adolescent Medicine, Medical University Graz, Austria
| | - Andreas Greinacher
- Institute of Immunology and Transfusion Medicine, Universitätsmedizin Greifswald, Germany
| | - Tamam Bakchoul
- Institute of Immunology and Transfusion Medicine, Universitätsmedizin Greifswald, Germany
| | - Markus G Seidel
- Division of Pediatric Hematology-Oncology, Department of Pediatrics and Adolescent Medicine, Medical University Graz, Austria.,Research Unit Pediatric Hematology and Immunology, Medical University Graz, Austria
| |
Collapse
|
43
|
Uchiyama Y, Ogawa Y, Kunishima S, Shiina M, Nakashima M, Yanagisawa K, Yokohama A, Imagawa E, Miyatake S, Mizuguchi T, Takata A, Miyake N, Ogata K, Handa H, Matsumoto N. A novel
GFI
1B
mutation at the first zinc finger domain causes congenital macrothrombocytopenia. Br J Haematol 2017; 181:843-847. [DOI: 10.1111/bjh.14710] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Yuri Uchiyama
- Department of Human Genetics Yokohama City University Graduate School of Medicine Yokohama Japan
- Department of Medicine and Clinical Science Gunma University Graduate School of Medicine Gunma Japan
| | - Yoshiyuki Ogawa
- Department of Medicine and Clinical Science Gunma University Graduate School of Medicine Gunma Japan
| | - Shinji Kunishima
- Department of Advanced Diagnosis Clinical Research Centre National Hospital Organization Nagoya Medical Centre Nagoya Japan
| | - Masaaki Shiina
- Department of Biochemistry Yokohama City University Graduate School of Medicine YokohamaJapan
| | - Mitsuko Nakashima
- Department of Human Genetics Yokohama City University Graduate School of Medicine Yokohama Japan
| | - Kunio Yanagisawa
- Department of Medicine and Clinical Science Gunma University Graduate School of Medicine Gunma Japan
| | - Akihiko Yokohama
- Division of Blood Transfusion Service Gunma University Hospital Gunma Japan
| | - Eri Imagawa
- Department of Human Genetics Yokohama City University Graduate School of Medicine Yokohama Japan
| | - Satoko Miyatake
- Department of Human Genetics Yokohama City University Graduate School of Medicine Yokohama Japan
| | - Takeshi Mizuguchi
- Department of Human Genetics Yokohama City University Graduate School of Medicine Yokohama Japan
| | - Atsushi Takata
- Department of Human Genetics Yokohama City University Graduate School of Medicine Yokohama Japan
| | - Noriko Miyake
- Department of Human Genetics Yokohama City University Graduate School of Medicine Yokohama Japan
| | - Kazuhiro Ogata
- Department of Biochemistry Yokohama City University Graduate School of Medicine YokohamaJapan
| | - Hiroshi Handa
- Department of Medicine and Clinical Science Gunma University Graduate School of Medicine Gunma Japan
| | - Naomichi Matsumoto
- Department of Human Genetics Yokohama City University Graduate School of Medicine Yokohama Japan
| |
Collapse
|
44
|
Scheer S, Zaph C. The Lysine Methyltransferase G9a in Immune Cell Differentiation and Function. Front Immunol 2017; 8:429. [PMID: 28443098 PMCID: PMC5387087 DOI: 10.3389/fimmu.2017.00429] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 03/27/2017] [Indexed: 12/14/2022] Open
Abstract
G9a (KMT1C, EHMT2) is a lysine methyltransferase (KMT) whose primary function is to di-methylate lysine 9 of histone H3 (H3K9me2). G9a-dependent H3K9me2 is associated with gene silencing and acts primarily through the recruitment of H3K9me2-binding proteins that prevent transcriptional activation. Gene repression via G9a-dependent H3K9me2 is critically required in embryonic stem (ES) cells for the development of cellular lineages by repressing expression of pluripotency factors. In the immune system, lymphoid cells such as T cells and innate lymphoid cells (ILCs) can differentiate from a naïve state into one of several effector lineages that require both activating and repressive mechanisms to maintain the correct gene expression program. Furthermore, the long-term immunity to re-infection is mediated by memory T cells, which also require specific gene expression and repression to maintain a quiescent state. In this review, we examine the molecular machinery of G9a-dependent functions, address the role of G9a in lymphoid cell differentiation and function, and identify potential functions of T cells and ILCs that may be controlled by G9a. Together, this review will highlight the dynamic nature of G9a-dependent H3K9me2 in the immune system and shed light on the nature of repressive epigenetic modifications in cellular lineage choice.
Collapse
Affiliation(s)
- Sebastian Scheer
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Colby Zaph
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| |
Collapse
|
45
|
Anguita E, Candel FJ, Chaparro A, Roldán-Etcheverry JJ. Transcription Factor GFI1B in Health and Disease. Front Oncol 2017; 7:54. [PMID: 28401061 PMCID: PMC5368270 DOI: 10.3389/fonc.2017.00054] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 03/13/2017] [Indexed: 12/13/2022] Open
Abstract
Many human diseases arise through dysregulation of genes that control key cell fate pathways. Transcription factors (TFs) are major cell fate regulators frequently involved in cancer, particularly in leukemia. The GFI1B gene, coding a TF, was identified by sequence homology with the oncogene growth factor independence 1 (GFI1). Both GFI1 and GFI1B have six C-terminal C2H2 zinc fingers and an N-terminal SNAG (SNAIL/GFI1) transcriptional repression domain. Gfi1 is essential for neutrophil differentiation in mice. In humans, GFI1 mutations are associated with severe congenital neutropenia. Gfi1 is also required for B and T lymphopoiesis. However, knockout mice have demonstrated that Gfi1b is required for development of both erythroid and megakaryocytic lineages. Consistent with this, human mutations of GFI1B produce bleeding disorders with low platelet count and abnormal function. Loss of Gfi1b in adult mice increases the absolute numbers of hematopoietic stem cells (HSCs) that are less quiescent than wild-type HSCs. In keeping with this key role in cell fate, GFI1B is emerging as a gene involved in cancer, which also includes solid tumors. In fact, abnormal activation of GFI1B and GFI1 has been related to human medulloblastoma and is also likely to be relevant in blood malignancies. Several pieces of evidence supporting this statement will be detailed in this mini review.
Collapse
Affiliation(s)
- Eduardo Anguita
- Hematology Department, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain; Department of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - Francisco J Candel
- Microbiology Department, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC) , Madrid , Spain
| | - Alberto Chaparro
- Hematology Department, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain; Department of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - Juan J Roldán-Etcheverry
- Hematology Department, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain; Department of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain
| |
Collapse
|
46
|
Adamik J, Jin S, Sun Q, Zhang P, Weiss KR, Anderson JL, Silbermann R, Roodman GD, Galson DL. EZH2 or HDAC1 Inhibition Reverses Multiple Myeloma-Induced Epigenetic Suppression of Osteoblast Differentiation. Mol Cancer Res 2017; 15:405-417. [PMID: 28119431 DOI: 10.1158/1541-7786.mcr-16-0242-t] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 12/16/2016] [Accepted: 12/21/2016] [Indexed: 01/12/2023]
Abstract
In multiple myeloma, osteolytic lesions rarely heal because of persistent suppressed osteoblast differentiation resulting in a high fracture risk. Herein, chromatin immunoprecipitation analyses reveal that multiple myeloma cells induce repressive epigenetic histone changes at the Runx2 locus that prevent osteoblast differentiation. The most pronounced multiple myeloma-induced changes were at the Runx2-P1 promoter, converting it from a poised bivalent state to a repressed state. Previously, it was observed that multiple myeloma induces the transcription repressor GFI1 in osteoblast precursors, which correlates with decreased Runx2 expression, thus prompting detailed characterization of the multiple myeloma and TNFα-dependent GFI1 response element within the Runx2-P1 promoter. Further analyses reveal that multiple myeloma-induced GFI1 binding to Runx2 in osteoblast precursors and recruitment of the histone modifiers HDAC1, LSD1, and EZH2 is required to establish and maintain Runx2 repression in osteogenic conditions. These GFI1-mediated repressive chromatin changes persist even after removal of multiple myeloma. Ectopic GFI1 is sufficient to bind to Runx2, recruit HDAC1 and EZH2, increase H3K27me3 on the gene, and prevent osteogenic induction of endogenous Runx2 expression. Gfi1 knockdown in MC4 cells blocked multiple myeloma-induced recruitment of HDAC1 and EZH2 to Runx2, acquisition of repressive chromatin architecture, and suppression of osteoblast differentiation. Importantly, inhibition of EZH2 or HDAC1 activity in pre-osteoblasts after multiple myeloma exposure in vitro or in osteoblast precursors from patients with multiple myeloma reversed the repressive chromatin architecture at Runx2 and rescued osteoblast differentiation.Implications: This study suggests that therapeutically targeting EZH2 or HDAC1 activity may reverse the profound multiple myeloma-induced osteoblast suppression and allow repair of the lytic lesions. Mol Cancer Res; 15(4); 405-17. ©2017 AACR.
Collapse
Affiliation(s)
- Juraj Adamik
- Department of Medicine, Division of Hematology-Oncology, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Shunqian Jin
- Department of Medicine, Division of Hematology-Oncology, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Quanhong Sun
- Department of Medicine, Division of Hematology-Oncology, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Peng Zhang
- Department of Medicine, Division of Hematology-Oncology, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Kurt R Weiss
- Department of Orthopaedic Surgery, University of Pittsburgh Medical Center, Cancer Stem Cell Laboratory, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Judith L Anderson
- Department of Medicine, Division of Hematology-Oncology, Indiana University, Indianapolis, Indiana
| | - Rebecca Silbermann
- Department of Medicine, Division of Hematology-Oncology, Indiana University, Indianapolis, Indiana
| | - G David Roodman
- Department of Medicine, Division of Hematology-Oncology, Indiana University, Indianapolis, Indiana. .,Richard L. Roudebush VA Medical Center, Indianapolis, Indiana
| | - Deborah L Galson
- Department of Medicine, Division of Hematology-Oncology, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania. .,McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
47
|
Liu XF, Hummel M, Abecassis M. Epigenetic regulation of cellular and cytomegalovirus genes during myeloid cell development. ACTA ACUST UNITED AC 2017; 3. [PMID: 28707002 DOI: 10.18103/imr.v3i3.385] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Myeloid cells are important cell types that carry human cytomegalovirus. Latent viral DNA is present in CD34+ progenitor cells and their derived monocytes. However, differentiation of latently infected monocytes to mature macrophages or dendritic cells causes reactivation of latent viruses. During hematopoietic development, pluripotent genes are repressed, and lineage specific genes are activated in a step-wise manner. This process is governed by cell-type specific chromatin states. Enhancers in the hematopoietic system are highly dynamic and established by pioneer (first tier) transcription factors (TFs), which set the stage for second and third tier TF binding. In this review, we examine the epigenetic mechanisms that regulate myeloid cell development, cell identity, and activation with a special focus on factors that regulate viral gene expression and the status of viral infection in myeloid cells.
Collapse
Affiliation(s)
- Xue-Feng Liu
- Comprehensive Transplant Center, Division of Organ Transplantation, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611
| | - Mary Hummel
- Comprehensive Transplant Center, Division of Organ Transplantation, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611
| | - Michael Abecassis
- Comprehensive Transplant Center, Division of Organ Transplantation, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611
| |
Collapse
|
48
|
Desnues B, Macedo AB, Ordoñez-Rueda D, Roussel-Queval A, Malissen B, Bruhns P, Malissen M, Alexopoulou L. The transcriptional repressor Gfi1 prevents lupus autoimmunity by restraining TLR7 signaling. Eur J Immunol 2016; 46:2801-2811. [PMID: 27600904 DOI: 10.1002/eji.201646573] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 07/28/2016] [Accepted: 09/03/2016] [Indexed: 01/16/2023]
Abstract
The transcriptional repressor growth factor independence 1 (Gfi1) is important in myeloid and lymphoid differentiation. In the current study we evaluated the involvement of Gfi1 in systemic lupus erythematosus (SLE). We found that Genista mice, which carry a hypomorphic mutation in the gfi1 gene or Gfi1-deficient (Gfi1-/- ) mice develop signs of spontaneous lupus autoimmunity, including increased serum levels of IgM and IgG2a, autoantibodies against RNA and DNA, glomerular immunodeposits and increased frequencies of plasmablasts, germinal center (GC) B cells and age-associated B cells (ABCs). On the contrary, Genista mice deprived of TLR7 did not show any of these phenotypes, suggesting that the observed lupus autoimmunity in Genista mice is TLR7-dependent. Moreover, Genista mice showed an increased activation of dendritic cells (DCs), B and T cells that was dependent on TLR7 for DCs and B cells, but not for T cells. Upon TLR7 or TLR4 stimulation Genista DCs produced increased amounts of TNF, IL-6 and IFN-β and showed increased NF-κB phosphorylation and IRF7 nuclear translocation, suggesting that Gfi1 controls the NF-κB and type I IFN signaling pathway downstream of TLRs. Our data reveal that Gfi1 plays a critical role in the prevention of spontaneous lupus autoimmunity by negatively regulating TLR7 signaling.
Collapse
Affiliation(s)
- Benoit Desnues
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| | | | | | | | | | - Pierre Bruhns
- Unité des Anticorps en Thérapie et Pathologie, Département d'Immunologie, Institut Pasteur, Paris, France.,INSERM, U1222, Paris, France
| | - Marie Malissen
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| | | |
Collapse
|
49
|
Sudden infant death syndrome: exposure to cigarette smoke leads to hypomethylation upstream of the growth factor independent 1 (GFI1) gene promoter. Forensic Sci Med Pathol 2016; 12:399-406. [PMID: 27677632 DOI: 10.1007/s12024-016-9812-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/06/2016] [Indexed: 01/01/2023]
Abstract
PURPOSE Smoking during pregnancy has long been known as an important risk factor for sudden infant death syndrome (SIDS). However, the precise relationship between the smoking behavior of the mother and SIDS still remains unclear. In this study, the influence of prenatal smoking exposure on the childrens' DNA methylation state of a CpG island located upstream of the promoter of the growth factor independent 1 (GFI1) gene was analyzed. METHODS Blood samples of well-defined SIDS cases with non-smoking mothers (n = 11), SIDS cases with smoking mothers during pregnancy (n = 11), and non-SIDS cases (n = 6) were obtained from a previous study and methylation states were determined by bisulfite sequencing. RESULTS Significant hypomethylation was observed in this CpG island in SIDS cases with cigarette smoke exposure compared to non-exposed cases. The strongest effect in this CpG island was observed for 49 CpG sites located within a transcription factor binding site. Coding for a transcriptional repressor, GFI1 plays an important role in various developmental processes. Alterations in the GFI1 expression might be linked to various conditions that are known to be associated with SIDS, such as dysregulated hematopoiesis and excessive inflammatory response. CONCLUSION Data obtained in this study show that analysis of methylation states in cases of sudden infant death syndrome might provide a further important piece of knowledge toward understanding SIDS, and should be investigated in further studies.
Collapse
|
50
|
Daly ME. Transcription factor defects causing platelet disorders. Blood Rev 2016; 31:1-10. [PMID: 27450272 DOI: 10.1016/j.blre.2016.07.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 06/10/2016] [Accepted: 07/12/2016] [Indexed: 01/19/2023]
Abstract
Recent years have seen increasing recognition of a subgroup of inherited platelet function disorders which are due to defects in transcription factors that are required to regulate megakaryopoiesis and platelet production. Thus, germline mutations in the genes encoding the haematopoietic transcription factors RUNX1, GATA-1, FLI1, GFI1b and ETV6 have been associated with both quantitative and qualitative platelet abnormalities, and variable bleeding symptoms in the affected patients. Some of the transcription factor defects are also associated with an increased predisposition to haematologic malignancies (RUNX1, ETV6), abnormal erythropoiesis (GATA-1, GFI1b, ETV6) and immune dysfunction (FLI1). The persistence of MYH10 expression in platelets is a surrogate marker for FLI1 and RUNX1 defects. Characterisation of the transcription factor defects that give rise to platelet function disorders, and of the genes that are differentially regulated as a result, are yielding insights into the roles of these genes in platelet formation and function.
Collapse
Affiliation(s)
- Martina E Daly
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield Medical School, Beech Hill Road, Sheffield, S10 2RX, UK.
| |
Collapse
|