1
|
Hegde S, Giotti B, Soong BY, Halasz L, Berichel JL, Magen A, Kloeckner B, Mattiuz R, Park MD, Marks A, Belabed M, Hamon P, Chin T, Troncoso L, Lee JJ, Ahimovic D, Bale M, Chung G, D'souza D, Angeliadis K, Dawson T, Kim-Schulze S, Flores RM, Kaufman AJ, Ginhoux F, Josefowicz SZ, Ma S, Tsankov AM, Marron TU, Brown BD, Merad M. Myeloid progenitor dysregulation fuels immunosuppressive macrophages in tumors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.24.600383. [PMID: 38979166 PMCID: PMC11230224 DOI: 10.1101/2024.06.24.600383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Monocyte-derived macrophages (mo-macs) drive immunosuppression in the tumor microenvironment (TME) and tumor-enhanced myelopoiesis in the bone marrow (BM) fuels these populations. Here, we performed paired transcriptome and chromatin analysis over the continuum of BM myeloid progenitors, circulating monocytes, and tumor-infiltrating mo-macs in mice and in patients with lung cancer to identify myeloid progenitor programs that fuel pro-tumorigenic mo-macs. Analyzing chromatin accessibility and histone mark changes, we show that lung tumors prime accessibility for Nfe2l2 (NRF2) in BM myeloid progenitors as a cytoprotective response to oxidative stress. NRF2 activity is sustained and increased during monocyte differentiation into mo-macs in the lung TME to regulate oxidative stress, in turn promoting metabolic adaptation, resistance to cell death, and contributing to immunosuppressive phenotype. NRF2 genetic deletion and pharmacological inhibition significantly reduced mo-macs' survival and immunosuppression in the TME, enabling NK and T cell therapeutic antitumor immunity and synergizing with checkpoint blockade strategies. Altogether, our study identifies a targetable epigenetic node of myeloid progenitor dysregulation that sustains immunoregulatory mo-macs in the TME.
Collapse
|
2
|
Ciudad MT, Quevedo R, Lamorte S, Jin R, Nzirorera N, Koritzinsky M, McGaha TL. Dabrafenib Alters MDSC Differentiation and Function by Activation of GCN2. CANCER RESEARCH COMMUNICATIONS 2024; 4:765-784. [PMID: 38421883 PMCID: PMC10936428 DOI: 10.1158/2767-9764.crc-23-0376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 12/12/2023] [Accepted: 02/27/2024] [Indexed: 03/02/2024]
Abstract
The effect of targeted therapeutics on anticancer immune responses is poorly understood. The BRAF inhibitor dabrafenib has been reported to activate the integrated stress response (ISR) kinase GCN2, and the therapeutic effect has been partially attributed to GCN2 activation. Because ISR signaling is a key component of myeloid-derived suppressor cell (MDSC) development and function, we measured the effect of dabrafenib on MDSC differentiation and suppressive activity. Our data showed that dabrafenib attenuated MDSC ability to suppress T-cell activity, which was associated with a GCN2-dependent block of the transition from monocytic progenitor to polymorphonuclear (PMN)-MDSCs and proliferative arrest resulting in PMN-MDSC loss. Transcriptional profiling revealed that dabrafenib-driven GCN2 activation altered metabolic features in MDSCs enhancing oxidative respiration, and attenuated transcriptional programs required for PMN development. Moreover, we observed a broad downregulation of transcriptional networks associated with PMN developmental pathways, and increased activity of transcriptional regulons driven by Atf5, Mafg, and Zbtb7a. This transcriptional program alteration underlies the basis for PMN-MDSC developmental arrest, skewing immature MDSC development toward monocytic lineage cells. In vivo, we observed a pronounced reduction in PMN-MDSCs in dabrafenib-treated tumor-bearing mice suggesting that dabrafenib impacts MDSC populations systemically and locally, in the tumor immune infiltrate. Thus, our data reveal transcriptional networks that govern MDSC developmental programs, and the impact of GCN2 stress signaling on the innate immune landscape in tumors, providing novel insight into potentially beneficial off-target effects of dabrafenib. SIGNIFICANCE An important, but poorly understood, aspect of targeted therapeutics for cancer is the effect on antitumor immune responses. This article shows that off-target effects of dabrafenib activating the kinase GCN2 impact MDSC development and function reducing PMN-MDSCs in vitro and in vivo. This has important implications for our understanding of how this BRAF inhibitor impacts tumor growth and provides novel therapeutic target and combination possibilities.
Collapse
Affiliation(s)
- M. Teresa Ciudad
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Department of Immunology, University of Toronto, Toronto, Canada
| | - Rene Quevedo
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Department of Immunology, University of Toronto, Toronto, Canada
| | - Sara Lamorte
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Department of Immunology, University of Toronto, Toronto, Canada
| | - Robbie Jin
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Department of Immunology, University of Toronto, Toronto, Canada
| | - Nadine Nzirorera
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Department of Immunology, University of Toronto, Toronto, Canada
| | - Marianne Koritzinsky
- Princess Margaret Cancer Center, University Health Network, Toronto, Canada
- Institute of Medical Science, University of Toronto, Toronto, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Tracy L. McGaha
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Department of Immunology, University of Toronto, Toronto, Canada
| |
Collapse
|
3
|
Pan J, Ye F, Li H, Yu C, Mao J, Xiao Y, Chen H, Wu J, Li J, Fei L, Wu Y, Meng X, Guo G, Wang Y. Dissecting the immune discrepancies in mouse liver allograft tolerance and heart/kidney allograft rejection. Cell Prolif 2024; 57:e13555. [PMID: 37748771 PMCID: PMC10905343 DOI: 10.1111/cpr.13555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 08/23/2023] [Accepted: 09/15/2023] [Indexed: 09/27/2023] Open
Abstract
The liver is the most tolerogenic of transplanted organs. However, the mechanisms underlying liver transplant tolerance are not well understood. The comparison between liver transplantation tolerance and heart/kidney transplantation rejection will deepen our understanding of tolerance and rejection in solid organs. Here, we built a mouse model of liver, heart and kidney allograft and performed single-cell RNA sequencing of 66,393 cells to describe the cell composition and immune cell interactions at the early stage of tolerance or rejection. We also performed bulk RNA-seq of mouse liver allografts from Day 7 to Day 60 post-transplantation to map the dynamic transcriptional variation in spontaneous tolerance. The transcriptome of lymphocytes and myeloid cells were characterized and compared in three types of organ allografts. Cell-cell interaction networks reveal the coordinated function of Kupffer cells, macrophages and their associated metabolic processes, including insulin receptor signalling and oxidative phosphorylation in tolerance induction. Cd11b+ dendritic cells (DCs) in liver allografts were found to inhibit cytotoxic T cells by secreting anti-inflammatory cytokines such as Il10. In summary, we profiled single-cell transcriptome analysis of mouse solid organ allografts. We characterized the immune microenvironment of mouse organ allografts in the acute rejection state (heart, kidney) and tolerance state (liver).
Collapse
Affiliation(s)
- Jun Pan
- Department of Thyroid Surgery, the First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
| | - Fang Ye
- Liangzhu LaboratoryZhejiang UniversityHangzhouChina
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Hui Li
- Key Laboratory of Combined Multiorgan Transplantation, Ministry of Public Health, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Division of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
| | - Chengxuan Yu
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Jiajia Mao
- Kidney Disease Center, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
| | - Yanyu Xiao
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Haide Chen
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Junqing Wu
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Jiaqi Li
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Lijiang Fei
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Yijun Wu
- Department of Thyroid Surgery, the First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
| | - Xiaoming Meng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of PharmacyAnhui Medical University, The Key Laboratory of Anti‐inflammatory of Immune Medicines, Ministry of EducationHefeiChina
| | - Guoji Guo
- Liangzhu LaboratoryZhejiang UniversityHangzhouChina
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative MedicineDr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative MedicineHangzhouZhejiangChina
| | - Yingying Wang
- Kidney Disease Center, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
| |
Collapse
|
4
|
Zu S, Li YE, Wang K, Armand EJ, Mamde S, Amaral ML, Wang Y, Chu A, Xie Y, Miller M, Xu J, Wang Z, Zhang K, Jia B, Hou X, Lin L, Yang Q, Lee S, Li B, Kuan S, Liu H, Zhou J, Pinto-Duarte A, Lucero J, Osteen J, Nunn M, Smith KA, Tasic B, Yao Z, Zeng H, Wang Z, Shang J, Behrens MM, Ecker JR, Wang A, Preissl S, Ren B. Single-cell analysis of chromatin accessibility in the adult mouse brain. Nature 2023; 624:378-389. [PMID: 38092917 PMCID: PMC10719105 DOI: 10.1038/s41586-023-06824-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 11/01/2023] [Indexed: 12/17/2023]
Abstract
Recent advances in single-cell technologies have led to the discovery of thousands of brain cell types; however, our understanding of the gene regulatory programs in these cell types is far from complete1-4. Here we report a comprehensive atlas of candidate cis-regulatory DNA elements (cCREs) in the adult mouse brain, generated by analysing chromatin accessibility in 2.3 million individual brain cells from 117 anatomical dissections. The atlas includes approximately 1 million cCREs and their chromatin accessibility across 1,482 distinct brain cell populations, adding over 446,000 cCREs to the most recent such annotation in the mouse genome. The mouse brain cCREs are moderately conserved in the human brain. The mouse-specific cCREs-specifically, those identified from a subset of cortical excitatory neurons-are strongly enriched for transposable elements, suggesting a potential role for transposable elements in the emergence of new regulatory programs and neuronal diversity. Finally, we infer the gene regulatory networks in over 260 subclasses of mouse brain cells and develop deep-learning models to predict the activities of gene regulatory elements in different brain cell types from the DNA sequence alone. Our results provide a resource for the analysis of cell-type-specific gene regulation programs in both mouse and human brains.
Collapse
Affiliation(s)
- Songpeng Zu
- Department of Cellular and Molecular Medicine, University of California San Diego, School of Medicine, La Jolla, CA, USA
| | - Yang Eric Li
- Department of Cellular and Molecular Medicine, University of California San Diego, School of Medicine, La Jolla, CA, USA
- Department of Neurosurgery and Genetics, Washington University School of Medicine, St Louis, MO, USA
| | - Kangli Wang
- Department of Cellular and Molecular Medicine, University of California San Diego, School of Medicine, La Jolla, CA, USA
| | - Ethan J Armand
- Department of Cellular and Molecular Medicine, University of California San Diego, School of Medicine, La Jolla, CA, USA
| | - Sainath Mamde
- Department of Cellular and Molecular Medicine, University of California San Diego, School of Medicine, La Jolla, CA, USA
| | - Maria Luisa Amaral
- Department of Cellular and Molecular Medicine, University of California San Diego, School of Medicine, La Jolla, CA, USA
| | - Yuelai Wang
- Department of Cellular and Molecular Medicine, University of California San Diego, School of Medicine, La Jolla, CA, USA
| | - Andre Chu
- Department of Cellular and Molecular Medicine, University of California San Diego, School of Medicine, La Jolla, CA, USA
| | - Yang Xie
- Department of Cellular and Molecular Medicine, University of California San Diego, School of Medicine, La Jolla, CA, USA
| | - Michael Miller
- Center for Epigenomics, University of California San Diego, School of Medicine, La Jolla, CA, USA
| | - Jie Xu
- Department of Cellular and Molecular Medicine, University of California San Diego, School of Medicine, La Jolla, CA, USA
| | - Zhaoning Wang
- Department of Cellular and Molecular Medicine, University of California San Diego, School of Medicine, La Jolla, CA, USA
| | - Kai Zhang
- Department of Cellular and Molecular Medicine, University of California San Diego, School of Medicine, La Jolla, CA, USA
| | - Bojing Jia
- Department of Cellular and Molecular Medicine, University of California San Diego, School of Medicine, La Jolla, CA, USA
| | - Xiaomeng Hou
- Center for Epigenomics, University of California San Diego, School of Medicine, La Jolla, CA, USA
| | - Lin Lin
- Center for Epigenomics, University of California San Diego, School of Medicine, La Jolla, CA, USA
| | - Qian Yang
- Center for Epigenomics, University of California San Diego, School of Medicine, La Jolla, CA, USA
| | - Seoyeon Lee
- Department of Cellular and Molecular Medicine, University of California San Diego, School of Medicine, La Jolla, CA, USA
| | - Bin Li
- Department of Cellular and Molecular Medicine, University of California San Diego, School of Medicine, La Jolla, CA, USA
| | - Samantha Kuan
- Department of Cellular and Molecular Medicine, University of California San Diego, School of Medicine, La Jolla, CA, USA
| | - Hanqing Liu
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Jingtian Zhou
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | | | - Jacinta Lucero
- The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Julia Osteen
- The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Michael Nunn
- Howard Hughes Medical Institute, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | | | | | - Zizhen Yao
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Zihan Wang
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA, USA
| | - Jingbo Shang
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA, USA
| | | | - Joseph R Ecker
- Howard Hughes Medical Institute, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Allen Wang
- Center for Epigenomics, University of California San Diego, School of Medicine, La Jolla, CA, USA
| | - Sebastian Preissl
- Center for Epigenomics, University of California San Diego, School of Medicine, La Jolla, CA, USA
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Bing Ren
- Department of Cellular and Molecular Medicine, University of California San Diego, School of Medicine, La Jolla, CA, USA.
- Center for Epigenomics, University of California San Diego, School of Medicine, La Jolla, CA, USA.
| |
Collapse
|
5
|
Ciudad MT, Quevedo R, Lamorte S, Jin R, Nzirorera N, Koritzinsky M, McGaha TL. Dabrafenib alters MDSC differentiation and function by activation of GCN2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.09.552588. [PMID: 37645997 PMCID: PMC10461929 DOI: 10.1101/2023.08.09.552588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
The effect of targeted therapeutics on anti-cancer immune responses is poorly understood. The BRAF inhibitor dabrafenib has been reported to activate the integrated stress response (ISR) kinase GCN2, and the therapeutic effect has been partially attributed to GCN2 activation. Since ISR signaling is a key component of myeloid-derived suppressor cell (MDSC) development and function, we measured the effect of dabrafenib on MDSC differentiation and suppressive activity. Our data showed that dabrafenib attenuated MDSC ability to suppress T cell activity, which was associated with a GCN2-dependent block of the transition from monocytic progenitor to polymorphonuclear (PMN)-MDSCs and proliferative arrest resulting in PMN-MDSC loss. Transcriptional profiling revealed that dabrafenib-driven GCN2 activation altered metabolic features in MDSCs enhancing oxidative respiration, and attenuated transcriptional programs required for PMN development. Moreover, we observed a broad downregulation of transcriptional networks associated with PMN developmental pathways, and increased activity of transcriptional regulons driven by Atf5 , Mafg , and Zbtb7a . This transcriptional program alteration underlies the basis for PMN-MDSC developmental arrest, skewing immature MDSC development towards monocytic lineage cells. In vivo , we observed a pronounced reduction in PMN-MDSCs in dabrafenib-treated tumor-bearing mice suggesting that dabrafenib impacts MDSC populations systemically and locally, in the tumor immune infiltrate. Thus, our data reveals transcriptional networks that govern MDSC developmental programs, and the impact of GCN2 stress signaling on the innate immune landscape in tumors, providing novel insight into potentially beneficial off target effects of dabrafenib.
Collapse
|
6
|
Jo D, Kim HK, Kim YK, Song J. Transcriptome Profile of Thyroid Glands in Bile Duct Ligation Mouse Model. Int J Mol Sci 2022; 23:ijms23158244. [PMID: 35897811 PMCID: PMC9332885 DOI: 10.3390/ijms23158244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/18/2022] [Accepted: 07/25/2022] [Indexed: 11/16/2022] Open
Abstract
Thyroid hormone (TH) contributes to multiple cellular mechanisms in the liver, muscle cells, adipose tissue, and brain, etc. In particular, the liver is an important organ in TH metabolism for the conversion of thyronine (T4) into triiodothyronine (T3) by the deiodinase enzyme. TH levels were significantly decreased and thyroid-stimulating hormone (TSH) levels were significantly increased in patients with liver failure compared with normal subjects. Among liver failure diseases, hepatic encephalopathy (HE) deserves more attention because liver damage and neuropathologies occur simultaneously. Although there is numerous evidence of TH dysregulation in the HE model, specific mechanisms and genetic features of the thyroid glands in the HE model are not fully understood. Here, we investigated the significantly different genes in the thyroid glands of a bile duct ligation (BDL) mouse model as the HE model, compared to the thyroid glands of the control mouse using RNA sequencing. We also confirmed the alteration in mRNA levels of thyroid gland function-related genes in the BDL mouse model. Furthermore, we evaluated the increased level of free T4 and TSH in the BDL mouse blood. Thus, we emphasize the potential roles of TH in liver metabolism and suggest that thyroid dysfunction-related genes in the HE model should be highlighted for finding the appropriate solution for an impaired thyroid system in HE.
Collapse
Affiliation(s)
- Danbi Jo
- Department of Anatomy, Chonnam National University Medical School, Seoyangro 264, Hwasun 58128, Korea;
- Biomedical Science Graduate Program (BMSGP), Chonnam National University, Seoyangro 264, Hwasun 58128, Korea
| | - Hee Kyung Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chonnam National University Medical School, Seoyangro 264, Hwasun 58128, Korea;
| | - Young-Kook Kim
- Department of Biochemistry, Chonnam National University Medical School, Seoyangro 264, Hwasun 58128, Korea;
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Seoyangro 264, Hwasun 58128, Korea;
- Biomedical Science Graduate Program (BMSGP), Chonnam National University, Seoyangro 264, Hwasun 58128, Korea
- Correspondence: ; Tel.: +82-61-379-2706
| |
Collapse
|
7
|
Hidalgo D, Bejder J, Pop R, Gellatly K, Hwang Y, Maxwell Scalf S, Eastman AE, Chen JJ, Zhu LJ, Heuberger JAAC, Guo S, Koury MJ, Nordsborg NB, Socolovsky M. EpoR stimulates rapid cycling and larger red cells during mouse and human erythropoiesis. Nat Commun 2021; 12:7334. [PMID: 34921133 PMCID: PMC8683474 DOI: 10.1038/s41467-021-27562-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 11/19/2021] [Indexed: 11/08/2022] Open
Abstract
The erythroid terminal differentiation program couples sequential cell divisions with progressive reductions in cell size. The erythropoietin receptor (EpoR) is essential for erythroblast survival, but its other functions are not well characterized. Here we use Epor-/- mouse erythroblasts endowed with survival signaling to identify novel non-redundant EpoR functions. We find that, paradoxically, EpoR signaling increases red cell size while also increasing the number and speed of erythroblast cell cycles. EpoR-regulation of cell size is independent of established red cell size regulation by iron. High erythropoietin (Epo) increases red cell size in wild-type mice and in human volunteers. The increase in mean corpuscular volume (MCV) outlasts the duration of Epo treatment and is not the result of increased reticulocyte number. Our work shows that EpoR signaling alters the relationship between cycling and cell size. Further, diagnostic interpretations of increased MCV should now include high Epo levels and hypoxic stress.
Collapse
Affiliation(s)
- Daniel Hidalgo
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jacob Bejder
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Ramona Pop
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Harvard Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Kyle Gellatly
- Program in Bioinformatics and Computational Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Yung Hwang
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - S Maxwell Scalf
- Department of Cell Biology and Yale Stem Cell Center, Yale University, New Haven, CT, USA
| | - Anna E Eastman
- Department of Cell Biology and Yale Stem Cell Center, Yale University, New Haven, CT, USA
| | - Jane-Jane Chen
- Institute for Medical Engineering & Science, MIT, Cambridge, MA, USA
| | - Lihua Julie Zhu
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Program in Bioinformatics and Computational Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Department of Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | | | - Shangqin Guo
- Department of Cell Biology and Yale Stem Cell Center, Yale University, New Haven, CT, USA
| | - Mark J Koury
- Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Merav Socolovsky
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
8
|
Identification of Critical Genes and lncRNAs in Osteolysis after Total Hip Arthroplasty and Osteoarthritis by RNA Sequencing. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6681925. [PMID: 33791375 PMCID: PMC7984875 DOI: 10.1155/2021/6681925] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 02/20/2021] [Accepted: 03/01/2021] [Indexed: 01/11/2023]
Abstract
Total hip arthroplasty (THA) is a cost-effective treatment for osteoarthritis (OA), and osteolysis is a common complication of THA. This study was aimed at exploring the relevant molecular biomarkers for osteolysis after THA. We performed RNA sequence to identify and characterize expressed mRNAs and lncRNAs in OA and osteolysis. Differentially expressed mRNAs (DEmRNAs) and lncRNAs (DElncRNAs) in OA and osteolysis were acquired, as well as shared DEmRNAs/DElncRNAs in OA and osteolysis and osteolysis-specific DEmRNAs/DElncRNAs. Then, shared and osteolysis-specific DElncRNA-DEmRNA coexpression networks were constructed to further investigate the function of DElncRNAs and DEmRNAs in OA and osteolysis. In total, 343 DEmRNAs and 25 DElncRNAs in OA, 908 DEmRNAs and 107 DElncRNAs in osteolysis, and 406 DEmRNAs and 46 DElncRNAs between OA and osteolysis were acquired. A total of 136 shared DEmRNAs and 9 shared DElncRNAs in OA and osteolysis and 736 osteolysis-specific DEmRNAs and 103 osteolysis-specific DElncRNAs were acquired. Then, 128 shared DElncRNA-DEmRNA coexpression pairs and 522 osteolysis-specific DElncRNA-DEmRNA coexpression pairs were identified. The present study highlighted the roles of four interaction pairs, including two shared lncRNA-mRNA interaction pairs in OA and osteolysis (AC111000.4 and AC016831.6), which may function in the immune process of OA and osteolysis by regulating CD8A and CD8B, respectively, and two osteolysis-specific interaction pairs (AC090607.4-FOXO3 and TAL1-ABALON), which may play an important role in osteoclastogenesis.
Collapse
|
9
|
Genetic variability of the U5 and downstream sequence of major HIV-1 subtypes and circulating recombinant forms. Sci Rep 2020; 10:13214. [PMID: 32764600 PMCID: PMC7411029 DOI: 10.1038/s41598-020-70083-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 07/03/2020] [Indexed: 11/08/2022] Open
Abstract
The critical role of the regulatory elements at the 5′ end of the HIV-1 genome in controlling the life cycle of HIV-1 indicates that this region significantly influences virus fitness and its biological properties. In this study, we performed a detailed characterization of strain-specific variability of sequences from the U5 to upstream of the gag gene start codon of diverse HIV-1 strains by using next-generation sequencing (NGS) techniques. Overall, we found that this region of the HIV-1 genome displayed a low degree of intra-strain variability. On the other hand, inter-strain variability was found to be as high as that reported for gag and env genes (13–17%). We observed strain-specific single point and clustered mutations in the U5, PBS, and gag leader sequences (GLS), generating potential strain-specific transcription factor binding sites (TFBS). Using an infrared gel shift assay, we demonstrated the presence of potential TFBS such as E-box in CRF22_01A, and Stat 6 in subtypes A and G, as well as in their related CRFs. The strain-specific variation found in the sequence corresponding at the RNA level to functional domains of the 5ʹ UTR, could also potentially impact the secondary/tertiary structural rearrangement of this region. Thus, the variability observed in this 5′ end of the genomic region of divergent HIV-1 strains strongly suggests that functions of this region might be affected in a strain-specific manner. Our findings provide new insights into DNA–protein interactions that regulate HIV-1 replication and the influence of strain characterization on the biology of HIV-1 infection.
Collapse
|
10
|
Godavarthy PS, Kumar R, Herkt SC, Pereira RS, Hayduk N, Weissenberger ES, Aggoune D, Manavski Y, Lucas T, Pan KT, Voutsinas JM, Wu Q, Müller MC, Saussele S, Oellerich T, Oehler VG, Lausen J, Krause DS. The vascular bone marrow niche influences outcome in chronic myeloid leukemia via the E-selectin - SCL/TAL1 - CD44 axis. Haematologica 2019; 105:136-147. [PMID: 31018977 PMCID: PMC6939533 DOI: 10.3324/haematol.2018.212365] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 04/23/2019] [Indexed: 12/12/2022] Open
Abstract
The endosteal bone marrow niche and vascular endothelial cells provide sanctuaries for leukemic cells. In murine chronic myeloid leukemia (CML) CD44 on leukemia cells and E-selectin on bone marrow endothelium are essential mediators for the engraftment of leukemic stem cells. We hypothesized that non-adhesion of CML-initiating cells to E-selectin on the bone marrow endothelium may lead to superior eradication of leukemic stem cells in CML after treatment with imatinib than imatinib alone. Indeed, here we show that treatment with the E-selectin inhibitor GMI-1271 in combination with imatinib prolongs survival of mice with CML via decreased contact time of leukemia cells with bone marrow endothelium. Non-adhesion of BCR-ABL1+ cells leads to an increase of cell cycle progression and an increase of expression of the hematopoietic transcription factor and proto-oncogene Scl/Tal1 in leukemia-initiating cells. We implicate SCL/TAL1 as an indirect phosphorylation target of BCR-ABL1 and as a negative transcriptional regulator of CD44 expression. We show that increased SCL/TAL1 expression is associated with improved outcome in human CML. These data demonstrate the BCR-ABL1-specific, cell-intrinsic pathways leading to altered interactions with the vascular niche via the modulation of adhesion molecules - which could be exploited therapeutically in the future.
Collapse
Affiliation(s)
| | - Rahul Kumar
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Stefanie C Herkt
- Institute for Transfusion Medicine DRK- Blutspendedienst Baden-Württemberg - Hessen, Frankfurt am Main, Germany
| | - Raquel S Pereira
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Nina Hayduk
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Eva S Weissenberger
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Djamel Aggoune
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Yosif Manavski
- Institute of Cardiovascular Regeneration, Center for Molecular Medicine, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Tina Lucas
- Institute of Cardiovascular Regeneration, Center for Molecular Medicine, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Kuan-Ting Pan
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Jenna M Voutsinas
- Fred Hutchinson Cancer Research Center, Clinical Research Division, Biostatistics, Seattle, WA, USA
| | - Qian Wu
- Fred Hutchinson Cancer Research Center, Clinical Research Division, Biostatistics, Seattle, WA, USA
| | | | - Susanne Saussele
- Department of Hematology and Oncology, University Hospital Mannheim, Heidelberg University, Mannheim, Germany
| | - Thomas Oellerich
- Department of Internal Medicine, Hematology/Oncology, Goethe University, Frankfurt am Main, Germany.,German Cancer Research Center and German Cancer Consortium, Heidelberg, Germany
| | - Vivian G Oehler
- Fred Hutchinson Cancer Research Center, Clinical Research Division, Division of Hematology, University of Washington Medical Center, Seattle, WA, USA
| | - Joern Lausen
- Institute for Transfusion Medicine DRK- Blutspendedienst Baden-Württemberg - Hessen, Frankfurt am Main, Germany
| | - Daniela S Krause
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany .,German Cancer Research Center and German Cancer Consortium, Heidelberg, Germany.,Faculty of Medicine, Johann Wolfgang Goethe University, Frankfurt.,Frankfurt Cancer Institute, Frankfurt, Germany
| |
Collapse
|
11
|
Vagapova ER, Spirin PV, Lebedev TD, Prassolov VS. The Role of TAL1 in Hematopoiesis and Leukemogenesis. Acta Naturae 2018; 10:15-23. [PMID: 29713515 PMCID: PMC5916730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
TAL1 (SCL/TAL1, T-cell acute leukemia protein 1) is a transcription factor that is involved in the process of hematopoiesis and leukemogenesis. It participates in blood cell formation, forms mesoderm in early embryogenesis, and regulates hematopoiesis in adult organisms. TAL1 is essential in maintaining the multipotency of hematopoietic stem cells (HSC) and keeping them in quiescence (stage G0). TAL1 forms complexes with various transcription factors, regulating hematopoiesis (E2A/HEB, GATA1-3, LMO1-2, Ldb1, ETO2, RUNX1, ERG, FLI1). In these complexes, TAL1 regulates normal myeloid differentiation, controls the proliferation of erythroid progenitors, and determines the choice of the direction of HSC differentiation. The transcription factors TAL1, E2A, GATA1 (or GATA2), LMO2, and Ldb1 are the major components of the SCL complex. In addition to normal hematopoiesis, this complex may also be involved in the process of blood cell malignant transformation. Upregulation of C-KIT expression is one of the main roles played by the SCL complex. Today, TAL1 and its partners are considered promising therapeutic targets in the treatment of T-cell acute lymphoblastic leukemia.
Collapse
Affiliation(s)
- E. R. Vagapova
- The Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova Str. 32, Moscow,119991, Russia
| | - P. V. Spirin
- The Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova Str. 32, Moscow,119991, Russia
| | - T. D. Lebedev
- The Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova Str. 32, Moscow,119991, Russia
| | - V. S. Prassolov
- The Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova Str. 32, Moscow,119991, Russia
| |
Collapse
|
12
|
Ramírez C, Mendoza L. Phenotypic stability and plasticity in GMP-derived cells as determined by their underlying regulatory network. Bioinformatics 2017; 34:1174-1182. [DOI: 10.1093/bioinformatics/btx736] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 11/23/2017] [Indexed: 12/30/2022] Open
Affiliation(s)
- Carlos Ramírez
- Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Cd. Mx., México
| | - Luis Mendoza
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Cd. Mx., México
| |
Collapse
|
13
|
Silva IAL, Conceição N, Gagnon É, Caiado H, Brown JP, Gianfrancesco F, Michou L, Cancela ML. Effect of genetic variants of OPTN in the pathophysiology of Paget's disease of bone. Biochim Biophys Acta Mol Basis Dis 2017; 1864:143-151. [PMID: 28993189 DOI: 10.1016/j.bbadis.2017.10.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 10/02/2017] [Accepted: 10/04/2017] [Indexed: 01/14/2023]
Abstract
Paget's disease of bone (PDB) is the second most frequent metabolic bone disease after osteoporosis. Genetic factors play an important role in PDB, but to date PDB causing mutations were identified only in the Sequestosome 1 gene at the PDB3 locus. OPTN has been recently associated with PDB, however little is known about the effect of genetic variants in this gene in PDB pathophysiology. By sequencing OPTN in SQSTM1 non-carriers PDB patients we found 16 SNPs in regulatory, coding and non-coding regions. One of those was found to be associated with PDB in our cohort - rs2234968. Our results show that rs2238968 effect may be explained by a change in OPTN splicing that give rise to a predicted truncated protein. We also performed functional studies on the variants located in OPTN promoter - rs3829923 and the rare variant -9906 - to investigate putative regulators of OPTN. Our results show that OPTN expression seems to be regulated by SP1, RXR, E47, and the E2F family. In conclusion, our work suggests a potential pathophysiological role of SNPs in OPTN, giving a new perspective about the regulatory mechanisms of this gene. Ultimately we discovered a new variant associated with PDB in OPTN, reinforcing the relevance of this gene for the development of this bone disease.
Collapse
Affiliation(s)
- Iris A L Silva
- PhD program in Biomedical Sciences and Medicine, University of Algarve, Faro 8005-139, Portugal; Centre of Marine Sciences (CCMAR), University of Algarve, Faro 8005-139, Portugal
| | - Natércia Conceição
- Centre of Marine Sciences (CCMAR), University of Algarve, Faro 8005-139, Portugal; Department of Biomedical Sciences and Medicine, University of Algarve, Faro 8005-139, Portugal
| | - Édith Gagnon
- Research centre of the CHU de Québec-Université Laval, Québec City, QC, Canada
| | - Helena Caiado
- Centre of Marine Sciences (CCMAR), University of Algarve, Faro 8005-139, Portugal; PhD program in Regenerative Medicine, University of Algarve, Faro 8005-139, Portugal
| | - Jacques P Brown
- Research centre of the CHU de Québec-Université Laval, Québec City, QC, Canada; Division of Rheumatology, Department of Medicine, Université Laval and Department of Rheumatology, CHU de Québec-Université Laval, Québec City, QC, Canada
| | - Fernando Gianfrancesco
- Institute of Genetics and Biophysics "Adriano Buzzati-Traverso", National Research Council of Italy, 80131 Naples, Italy
| | - Laëtitia Michou
- Research centre of the CHU de Québec-Université Laval, Québec City, QC, Canada; Division of Rheumatology, Department of Medicine, Université Laval and Department of Rheumatology, CHU de Québec-Université Laval, Québec City, QC, Canada.
| | - M Leonor Cancela
- Centre of Marine Sciences (CCMAR), University of Algarve, Faro 8005-139, Portugal; Department of Biomedical Sciences and Medicine, University of Algarve, Faro 8005-139, Portugal.
| |
Collapse
|
14
|
SCL/TAL1: a multifaceted regulator from blood development to disease. Blood 2017; 129:2051-2060. [DOI: 10.1182/blood-2016-12-754051] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 01/17/2017] [Indexed: 12/12/2022] Open
Abstract
Abstract
SCL/TAL1 (stem cell leukemia/T-cell acute lymphoblastic leukemia [T-ALL] 1) is an essential transcription factor in normal and malignant hematopoiesis. It is required for specification of the blood program during development, adult hematopoietic stem cell survival and quiescence, and terminal maturation of select blood lineages. Following ectopic expression, SCL contributes to oncogenesis in T-ALL. Remarkably, SCL’s activities are all mediated through nucleation of a core quaternary protein complex (SCL:E-protein:LMO1/2 [LIM domain only 1 or 2]:LDB1 [LIM domain-binding protein 1]) and dynamic recruitment of conserved combinatorial associations of additional regulators in a lineage- and stage-specific context. The finely tuned control of SCL’s regulatory functions (lineage priming, activation, and repression of gene expression programs) provides insight into fundamental developmental and transcriptional mechanisms, and highlights mechanistic parallels between normal and oncogenic processes. Importantly, recent discoveries are paving the way to the development of innovative therapeutic opportunities in SCL+ T-ALL.
Collapse
|
15
|
Abstract
SCL, a transcription factor of the basic helix-loop-helix family, is a master regulator of hematopoiesis. Scl specifies lateral plate mesoderm to a hematopoietic fate and establishes boundaries by inhibiting the cardiac lineage. A combinatorial interaction between Scl and Vegfa/Flk1 sets in motion the first wave of primitive hematopoiesis. Subsequently, definitive hematopoietic stem cells (HSCs) emerge from the embryo proper via an endothelial-to-hematopoietic transition controlled by Runx1, acting with Scl and Gata2. Past this stage, Scl in steady state HSCs is redundant with Lyl1, a highly homologous factor. However, Scl is haploinsufficient in stress response, when a rare subpopulation of HSCs with very long term repopulating capacity is called into action. SCL activates transcription by recruiting a core complex on DNA that necessarily includes E2A/HEB, GATA1-3, LIM-only proteins LMO1/2, LDB1, and an extended complex comprising ETO2, RUNX1, ERG, or FLI1. These interactions confer multifunctionality to a complex that can control cell proliferation in erythroid progenitors or commitment to terminal differentiation through variations in single component. Ectopic SCL and LMO1/2 expression in immature thymocytes activates of a stem cell gene network and reprogram cells with a finite lifespan into self-renewing preleukemic stem cells (pre-LSCs), an initiating event in T-cell acute lymphoblastic leukemias. Interestingly, fate conversion of fibroblasts to hematoendothelial cells requires not only Scl and Lmo2 but also Gata2, Runx1, and Erg, indicating a necessary collaboration between these transcription factors for hematopoietic reprogramming. Nonetheless, full reprogramming into self-renewing multipotent HSCs may require additional factors and most likely, a permissive microenvironment.
Collapse
Affiliation(s)
- T Hoang
- Laboratory of Hematopoiesis and Leukemia, Institute of Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, QC, Canada.
| | - J A Lambert
- Laboratory of Hematopoiesis and Leukemia, Institute of Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, QC, Canada
| | - R Martin
- Laboratory of Hematopoiesis and Leukemia, Institute of Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, QC, Canada
| |
Collapse
|
16
|
Wehrspaun CC, Haerty W, Ponting CP. Microglia recapitulate a hematopoietic master regulator network in the aging human frontal cortex. Neurobiol Aging 2015; 36:2443.e9-2443.e20. [PMID: 26002684 PMCID: PMC4503803 DOI: 10.1016/j.neurobiolaging.2015.04.008] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 02/18/2015] [Accepted: 04/17/2015] [Indexed: 12/11/2022]
Abstract
Microglia form the immune system of the brain. Previous studies in cell cultures and animal models suggest altered activation states and cellular senescence in the aged brain. Instead, we analyzed 3 transcriptome data sets from the postmortem frontal cortex of 381 control individuals to show that microglia gene markers assemble into a transcriptional module in a gene coexpression network. These markers predominantly represented M1 and M1/M2b activation phenotypes. Expression of genes in this module generally declines over the adult life span. This decrease was more pronounced in microglia surface receptors for microglia and/or neuron crosstalk than in markers for activation state phenotypes. In addition to these receptors for exogenous signals, microglia are controlled by brain-expressed regulatory factors. We identified a subnetwork of transcription factors, including RUNX1, IRF8, PU.1, and TAL1, which are master regulators (MRs) for the age-dependent microglia module. The causal contributions of these MRs on the microglia module were verified using publicly available ChIP-Seq data. Interactions of these key MRs were preserved in a protein-protein interaction network. Importantly, these MRs appear to be essential for regulating microglia homeostasis in the adult human frontal cortex in addition to their crucial roles in hematopoiesis and myeloid cell-fate decisions during embryogenesis.
Collapse
Affiliation(s)
- Claudia C Wehrspaun
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK; Section on Neuropathology, Clinical Brain Disorders Branch, Genes, Cognition and Psychosis Program, IRP, NIMH, NIH, Bethesda, MD, USA.
| | - Wilfried Haerty
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK; Department of Physiology, Anatomy and Genetics, MRC Functional Genomics Unit, University of Oxford, UK
| | - Chris P Ponting
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK; Department of Physiology, Anatomy and Genetics, MRC Functional Genomics Unit, University of Oxford, UK
| |
Collapse
|
17
|
Kolodziej S, Kuvardina ON, Oellerich T, Herglotz J, Backert I, Kohrs N, Buscató EL, Wittmann SK, Salinas-Riester G, Bonig H, Karas M, Serve H, Proschak E, Lausen J. PADI4 acts as a coactivator of Tal1 by counteracting repressive histone arginine methylation. Nat Commun 2014; 5:3995. [PMID: 24874575 PMCID: PMC4050257 DOI: 10.1038/ncomms4995] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 04/28/2014] [Indexed: 01/26/2023] Open
Abstract
The transcription factor Tal1 is a
critical activator or repressor of gene expression in hematopoiesis and leukaemia.
The mechanism by which Tal1
differentially influences transcription of distinct genes is not fully understood.
Here we show that Tal1 interacts
with the peptidylarginine deiminase
IV (PADI4). We
demonstrate that PADI4 can act as
an epigenetic coactivator through influencing H3R2me2a. At the Tal1/PADI4 target gene IL6ST the repressive H3R2me2a mark triggered by
PRMT6 is counteracted by
PADI4, which augments the
active H3K4me3 mark and thus increases IL6ST expression. In contrast, at the CTCF promoter PADI4 acts as a repressor. We propose that
the influence of PADI4 on
IL6ST transcription
plays a role in the control of IL6ST expression during lineage differentiation of
hematopoietic stem/progenitor cells. These results open the possibility to
pharmacologically influence Tal1
in leukaemia. Peptidylarginine deiminase 4 (PADI4) is a transcriptional
co-regulator that converts arginine residues at histone tails to citrulline. The authors
show that PADI4 interacts with the central haematopoietic transcription factor TAL1 to
regulate gene expression in an erythroleukemia cell line.
Collapse
Affiliation(s)
- Stephan Kolodziej
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Paul-Ehrlich-Strasse 42-44, D-60596 Frankfurt am Main, Germany
| | - Olga N Kuvardina
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Paul-Ehrlich-Strasse 42-44, D-60596 Frankfurt am Main, Germany
| | - Thomas Oellerich
- Department of Medicine, Hematology/Oncology, Johann-Wolfgang-Goethe University, Theodor-Stern-Kai 7, D-60590 Frankfurt am Main, Germany
| | - Julia Herglotz
- 1] Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Paul-Ehrlich-Strasse 42-44, D-60596 Frankfurt am Main, Germany [2]
| | - Ingo Backert
- 1] Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Paul-Ehrlich-Strasse 42-44, D-60596 Frankfurt am Main, Germany [2]
| | - Nicole Kohrs
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Paul-Ehrlich-Strasse 42-44, D-60596 Frankfurt am Main, Germany
| | - Estel la Buscató
- Institute of Pharmaceutical Chemistry, Johann-Wolfgang-Goethe University, Max-von-Laue-Strasse 9, D-60438 Frankfurt am Main, Germany
| | - Sandra K Wittmann
- Institute of Pharmaceutical Chemistry, Johann-Wolfgang-Goethe University, Max-von-Laue-Strasse 9, D-60438 Frankfurt am Main, Germany
| | - Gabriela Salinas-Riester
- Medical-University Goettingen, Transcriptome Analysis Laboratory, Justus-von-Liebig-Weg 11, D-37077 Goettingen, Germany
| | - Halvard Bonig
- German Red Cross Blood Service and Institute for Transfusion Medicine and Immunohematology, Johann-Wolfgang-Goethe University, Sandhofstrasse 1, D-60528 Frankfurt am Main, Germany
| | - Michael Karas
- Institute of Pharmaceutical Chemistry, Johann-Wolfgang-Goethe University, Max-von-Laue-Strasse 9, D-60438 Frankfurt am Main, Germany
| | - Hubert Serve
- 1] Department of Medicine, Hematology/Oncology, Johann-Wolfgang-Goethe University, Theodor-Stern-Kai 7, D-60590 Frankfurt am Main, Germany [2] German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Ewgenij Proschak
- 1] Institute of Pharmaceutical Chemistry, Johann-Wolfgang-Goethe University, Max-von-Laue-Strasse 9, D-60438 Frankfurt am Main, Germany [2] German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Jörn Lausen
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Paul-Ehrlich-Strasse 42-44, D-60596 Frankfurt am Main, Germany
| |
Collapse
|
18
|
Abstract
SCL/TAL1, a tissue-specific transcription factor of the basic helix-loop-helix family, and c-Kit, a tyrosine kinase receptor, control hematopoietic stem cell survival and quiescence. Here we report that SCL levels are limiting for the clonal expansion of Kit⁺ multipotent and erythroid progenitors. In addition, increased SCL expression specifically enhances the sensitivity of these progenitors to steel factor (KIT ligand) without affecting interleukin-3 response, whereas a DNA-binding mutant antagonizes KIT function and induces apoptosis in progenitors. Furthermore, a twofold increase in SCL levels in mice bearing a hypomorphic Kit allele (W41/41) corrects their hematocrits and deficiencies in erythroid progenitor numbers. At the molecular level, we found that SCL and c-Kit signaling control a common gene expression signature, of which 19 genes are associated with apoptosis. Half of those were decreased in purified megakaryocyte/erythroid progenitors (MEPs) from W41/41 mice and rescued by the SCL transgene. We conclude that Scl operates downstream of Kit to support the survival of MEPs. Finally, higher SCL expression upregulates Kit in normal bone marrow cells and increases chimerism after bone marrow transplantation, indicating that Scl is also upstream of Kit. We conclude that Scl and Kit establish a positive feedback loop in multipotent and MEPs.
Collapse
|
19
|
Li Y, Deng C, Hu X, Patel B, Fu X, Qiu Y, Brand M, Zhao K, Huang S. Dynamic interaction between TAL1 oncoprotein and LSD1 regulates TAL1 function in hematopoiesis and leukemogenesis. Oncogene 2012; 31:5007-18. [PMID: 22310283 PMCID: PMC3510314 DOI: 10.1038/onc.2012.8] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2011] [Revised: 12/15/2011] [Accepted: 12/30/2011] [Indexed: 12/29/2022]
Abstract
TAL1/SCL is a hematopoietic-specific oncogene and its activity is regulated by associated transcriptional co-activators and corepressors. Dysregulation of TAL1 activity has been associated with T-cell leukemogenesis. However, it remains unclear how the interactions between TAL1 and corepressors versus co-activators are properly regulated. Here, we reported that protein kinase A (PKA)-mediated phosphorylation regulates TAL1 interaction with the lysine-specific demethylase (LSD1) that removes methyl group from methylated Lys 4 on histone H3 tails. Phosphorylation of serine 172 in TAL1 specifically destabilizes the TAL1-LSD1 interaction leading to promoter H3K4 hypermethylation and activation of target genes that have been suppressed in normal and malignant hematopoiesis. Knockdown of TAL1 or LSD1 led to a derepression of the TAL1 target genes in T-cell acute lymphoblast leukemia (T-ALL) Jurkat cells, which is accompanied by elevating promoter H3K4 methylation. Similarly, treatment of PKA activator forskolin resulted in derepression of target genes by reducing its interaction with LSD1 while PKA inhibitor H89 represses them by suppressing H3K4 methylation levels. Consistent with the dual roles of TAL1 in transcription, TAL1-associated LSD1 is decreased while recruitment of hSET1 is increased at the TAL1 targets during erythroid differentiation. This process is accompanied by a dramatic increase in H3K4 methylation. Thus, our data revealed a novel interplay between PKA phosphorylation and TAL1-mediated epigenetic regulation that regulates hematopoietic transcription and differentiation programs during hematopoiesis and leukemogenesis.
Collapse
Affiliation(s)
- Ying Li
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, FL 32610
- College of Life Science, Jilin University, Changchun 130023, China
| | - Changwang Deng
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, FL 32610
| | - Xin Hu
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, FL 32610
- College of Life Science, Jilin University, Changchun 130023, China
| | - Bhavita Patel
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, FL 32610
| | - Xueqi Fu
- College of Life Science, Jilin University, Changchun 130023, China
| | - Yi Qiu
- Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL 32610
- Shands Cancer Center, University of Florida College of Medicine, Gainesville, FL 32610
| | - Marjorie Brand
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Keji Zhao
- Laboratory of Molecular Immunology, NHLBI, NIH, Bethesda, MD
| | - Suming Huang
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, FL 32610
- Shands Cancer Center, University of Florida College of Medicine, Gainesville, FL 32610
| |
Collapse
|
20
|
Jopling C, Suñé G, Faucherre A, Fabregat C, Izpisua Belmonte JC. Hypoxia induces myocardial regeneration in zebrafish. Circulation 2012; 126:3017-27. [PMID: 23151342 DOI: 10.1161/circulationaha.112.107888] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Hypoxia plays an important role in many biological/pathological processes. In particular, hypoxia is associated with cardiac ischemia. which, although initially inducing a protective response, will ultimately lead to the death of cardiomyocytes and loss of tissue, severely affecting cardiac functionality. Although myocardial damage/loss remains an insurmountable problem for adult mammals, the same is not true for adult zebrafish, which are able to completely regenerate their heart after extensive injury. Myocardial regeneration in zebrafish involves the dedifferentiation and proliferation of cardiomyocytes to replace the damaged/missing tissue; at present, however, little is known about what factors regulate this process. METHODS AND RESULTS We surmised that ventricular amputation would lead to hypoxia induction in the myocardium of zebrafish and that this may play a role in regulating the regeneration of the missing cardiac tissue. Using a combination of O(2) perturbation, conditional transgenics, in vitro cell culture, and microarray analysis, we found that hypoxia induces cardiomyocytes to dedifferentiate and proliferate during heart regeneration in zebrafish and have identified a number of genes that could play a role in this process. CONCLUSION These results indicate that hypoxia plays a positive role during heart regeneration, which should be taken into account in future strategies aimed at inducing heart regeneration in humans.
Collapse
Affiliation(s)
- Chris Jopling
- The Salk Institute for Biological Studies, 10010 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | | | | | | | | |
Collapse
|
21
|
Wang L, Jia Y, Rogers H, Wu YP, Huang S, Noguchi CT. GATA-binding protein 4 (GATA-4) and T-cell acute leukemia 1 (TAL1) regulate myogenic differentiation and erythropoietin response via cross-talk with Sirtuin1 (Sirt1). J Biol Chem 2012; 287:30157-69. [PMID: 22773876 DOI: 10.1074/jbc.m112.376640] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Erythropoietin (EPO), the cytokine required for erythrocyte production, contributes to muscle progenitor cell proliferation and delay myogenic differentiation. However, the underlying mechanism is not yet fully understood. Here, we report that EPO changes the skeletal myogenic regulatory factor expression program and delays differentiation via induction of GATA-4 and the basic helix-loop-helix TAL1 and that knockdown of both factors promotes differentiation. EPO increases the Sirt1 level, a NAD(+)-dependent deacetylase, and also induces the NAD(+)/NADH ratio that further increases Sirt1 activity. Sirt1 knockdown reduced GATA-4 and TAL1 expression, impaired EPO effect on delayed myogenic differentiation, and the Sirt1 knockdown effect was abrogated when combined with overexpression of GATA-4 or TAL1. GATA-4 interacts with Sirt1 and targets Sirt1 to the myogenin promoter and represses myogenin expression, whereas TAL1 inhibits myogenin expression by decreasing MyoD binding to and activation of the myogenin promoter. Sirt1 was found to bind to the GATA-4 promoter to directly regulate GATA-4 expression and GATA-4 binds to the TAL1 promoter to regulate TAL1 expression positively. These data suggest that GATA-4, TAL1, and Sirt1 cross-talk each other to regulate myogenic differentiation and mediate EPO activity during myogenic differentiation with Sirt1 playing a role upstream of GATA-4 and TAL1. Taken together, our findings reveal a novel role for GATA-4 and TAL1 to affect skeletal myogenic differentiation and EPO response via cross-talk with Sirt1.
Collapse
Affiliation(s)
- Li Wang
- Molecular Medicine Branch, NIDDK, National Institutes of Health, Bethesda, MD 20892-1822, USA
| | | | | | | | | | | |
Collapse
|
22
|
Courtial N, Smink JJ, Kuvardina ON, Leutz A, Göthert JR, Lausen J. Tal1 regulates osteoclast differentiation through suppression of the master regulator of cell fusion DC-STAMP. FASEB J 2011; 26:523-32. [PMID: 21990371 DOI: 10.1096/fj.11-190850] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The balance between bone-forming osteoblasts and bone-resorbing osteoclasts is crucial to bone homeostasis, an equilibrium that is disturbed in many bone diseases. The transcription factor Tal1 is involved in the establishment of hematopoietic stem cells in the embryo and is a master regulator of hematopoietic gene expression in the adult. Here, we show that Tal1 is expressed in osteoclasts and that loss of Tal1 in osteoclast progenitors leads to altered expression of >1200 genes. We found that DC-STAMP, a key regulator of osteoclast cell fusion, is a direct target gene of Tal1 and show that Tal1 represses DC-STAMP expression by counteracting the activating function of the transcription factors PU.1 and MITF. The identification of Tal1 as a factor involved in cell fusion contributes to the understanding of osteoclast-associated diseases, including osteoporosis.
Collapse
Affiliation(s)
- Nadine Courtial
- Georg-Speyer-Haus, Institute for Biomedical Research, Frankfurt, Germany
| | | | | | | | | | | |
Collapse
|
23
|
Adult hematopoiesis is regulated by TIF1γ, a repressor of TAL1 and PU.1 transcriptional activity. Cell Stem Cell 2011; 8:412-25. [PMID: 21474105 DOI: 10.1016/j.stem.2011.02.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2010] [Revised: 12/11/2010] [Accepted: 02/08/2011] [Indexed: 12/30/2022]
Abstract
Crosstalk between transcription factors and cytokines precisely regulates tissue homeostasis. Transcriptional intermediary factor 1γ (TIF1γ) regulates vertebrate hematopoietic development, can control transcription elongation, and is a component of the TGF-β signaling pathway. Here we show that deletion of TIF1γ in adult hematopoiesis is compatible with life and long-term maintenance of essential blood cell lineages. However, loss of TIF1γ results in deficient long-term hematopoietic stem cell (LT-HSC) transplantation activity, deficient short-term HSC (ST-HSC) bone marrow retention, and priming ST-HSCs to myelomonocytic lineage. These defects are hematopoietic cell-autonomous, and priming of TIF1γ-deficient ST-HSCs can be partially rescued by wild-type hematopoietic cells. TIF1γ can form complexes with TAL1 or PU.1-two essential DNA-binding proteins in hematopoiesis-occupy specific subsets of their DNA binding sites in vivo, and repress their transcriptional activity. These results suggest a regulation of adult hematopoiesis through TIF1γ-mediated transcriptional repression of TAL1 and PU.1 target genes.
Collapse
|
24
|
SCL-mediated regulation of the cell-cycle regulator p21 is critical for murine megakaryopoiesis. Blood 2011; 118:723-35. [DOI: 10.1182/blood-2011-01-328765] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Abstract
Megakaryopoiesis is a complex process that involves major cellular and nuclear changes and relies on controlled coordination of cellular proliferation and differentiation. These mechanisms are orchestrated in part by transcriptional regulators. The key hematopoietic transcription factor stem cell leukemia (SCL)/TAL1 is required in early hematopoietic progenitors for specification of the megakaryocytic lineage. These early functions have, so far, prevented full investigation of its role in megakaryocyte development in loss-of-function studies. Here, we report that SCL critically controls terminal megakaryocyte maturation. In vivo deletion of Scl specifically in the megakaryocytic lineage affects all key attributes of megakaryocyte progenitors (MkPs), namely, proliferation, ploidization, cytoplasmic maturation, and platelet release. Genome-wide expression analysis reveals increased expression of the cell-cycle regulator p21 in Scl-deleted MkPs. Importantly, p21 knockdown-mediated rescue of Scl-mutant MkPs shows full restoration of cell-cycle progression and partial rescue of the nuclear and cytoplasmic maturation defects. Therefore, SCL-mediated transcriptional control of p21 is essential for terminal maturation of MkPs. Our study provides a mechanistic link between a major hematopoietic transcriptional regulator, cell-cycle progression, and megakaryocytic differentiation.
Collapse
|
25
|
Fang X, Yoon JG, Li L, Tsai YS, Zheng S, Hood L, Goodlett DR, Foltz G, Lin B. Landscape of the SOX2 protein-protein interactome. Proteomics 2011; 11:921-34. [PMID: 21280222 DOI: 10.1002/pmic.201000419] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2010] [Revised: 11/19/2010] [Accepted: 12/05/2010] [Indexed: 01/21/2023]
Abstract
SOX2 is a key gene implicated in maintaining the stemness of embryonic and adult stem cells that appears to re-activate in several human cancers including glioblastoma multiforme. Using immunoprecipitation (IP)/MS/MS, we identified 144 proteins that are putative SOX2 interacting proteins. Of note, SOX2 was found to interact with several heterogeneous nuclear ribonucleoprotein family proteins, including HNRNPA2B1, HNRNPA3, HNRNPC, HNRNPK, HNRNPL, HNRNPM, HNRNPR, HNRNPU, as well as other ribonucleoproteins, DNA repair proteins and helicases. Gene ontology (GO) analysis revealed that the SOX2 interactome was enriched for GO terms GO:0030529 ribonucleoprotein complex and GO:0004386 helicase activity. These findings indicate that SOX2 associates with the heterogeneous nuclear ribonucleoprotein complex, suggesting a possible role for SOX2 in post-transcriptional regulation in addition to its function as a transcription factor.
Collapse
Affiliation(s)
- Xuefeng Fang
- Swedish Neuroscience Institute, Swedish Medical Center, Seattle, WA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|